87 results on '"Bornancin F"'
Search Results
2. The Rhodopsin-Transducin-cGMP-Phosphodiesterase Cascade of Visual Transduction
- Author
-
Bornancin, F., Pfister, C., Chabre, M., Borsellino, Antonio, editor, Cervetto, Luigi, editor, and Torre, Vincent, editor
- Published
- 1990
- Full Text
- View/download PDF
3. The Transitory Complexes Between Photoexcited Rhodopsin and Transducin. Biochemical and Spectroscopic Studies
- Author
-
Bornancin, F., primary and Pfister, C., additional
- Published
- 1992
- Full Text
- View/download PDF
4. Phosphorylation et régulation de la protéine-kinase C.
- Author
-
Bornancin, F, primary
- Published
- 1998
- Full Text
- View/download PDF
5. Threonine-497 is a critical site for permissive activation of protein kinase Cα
- Author
-
Cazaubon, S, primary, Bornancin, F, additional, and Parker, P J, additional
- Published
- 1994
- Full Text
- View/download PDF
6. ADP-ribosylation of Gs by cholera toxin is potentiated by agonist activation of beta-adrenergic receptors in the absence of GTP
- Author
-
Bornancin, F., primary, Audigier, Y., additional, and Chabre, M., additional
- Published
- 1993
- Full Text
- View/download PDF
7. 12-O-Tetradecanoylphorbol-13-acetate-induced dephosphorylation of protein kinase Calpha correlates with the presence of a membrane-associated protein phosphatase 2A heterotrimer.
- Author
-
Hansra, G, Bornancin, F, Whelan, R, Hemmings, B A, and Parker, P J
- Abstract
Protein kinase C signaling is desensitized through a combination of dephosphorylation and proteolysis in intact cells. The process of dephosphorylation is analyzed here, as well as its relationship to degradation. It is established for protein kinase Calpha that dephosphorylation occurs in a membrane compartment following activation and temporally preceding significant degradation. The phosphatase responsible for the dephosphorylation appears to be a heterotrimeric type 2A phosphatase, which is shown to be in part constitutively membrane associated. Consistent with a role for this activity, okadaic acid is shown to inhibit the phorbol ester-induced dephosphorylation of protein kinase C that occurs in intact cells. Furthermore, phorbol ester-induced down-regulation of protein kinase Calpha is shown not to be dependent on the rate of dephosphorylation, indicating that these desensitizing pathways may operate in parallel.
- Published
- 1996
8. Phosphorylation of protein kinase C-alpha on serine 657 controls the accumulation of active enzyme and contributes to its phosphatase-resistant state.
- Author
-
Bornancin, F and Parker, P J
- Abstract
Serine 657 in protein kinase C-alpha (PKCalpha) is a site of phosphorylation on expression of the recombinant protein in mammalian cells. To define the function of this phosphorylation, PKCalpha species with mutations of this site were investigated. The alanine mutant, S657A PKCalpha, displayed slow phosphate accumulation in pulse-chase experiments, indicating a rate-limiting role in the initial phase of phosphorylation. Consistent with this, the aspartic acid mutant, S657D PKCalpha, showed an increased rate of phosphate accumulation. Both the S657D and S657A PKCalpha mutants were slow to accumulate as fully phosphorylated forms during a second phase of phosphorylation. This latter property is shown to correlate with an increased phosphatase sensitivity and decreased protein kinase activity for these two PKCalpha mutants. It is further shown that once fully phosphorylated, the S657D PKCalpha mutant displays WT PKCalpha properties with respect to thermal stability and phosphatase sensitivity in vitro and in vivo; in contrast, the S657A PKCalpha mutant remains sensitive. The properties of the Ser-657 site PKCalpha mutants define functional roles for this phosphorylation in both the accumulation of phosphate on PKCalpha as well as in its agonist-induced dephosphorylation. These results are discussed in the context of a working model of PKCalpha behavior, providing insight into the workings of other kinases with equivalent sites of phosphorylation.
- Published
- 1997
9. Visual Signal Transduction; The Cycle of Transducin Shuttling between Rhodopsin and cGMP Phosphodiesterase
- Author
-
Chabre, M., primary, Bigay, J., additional, Bruckert, F., additional, Bornancin, F., additional, Deterre, P., additional, Pfister, C., additional, and Vuong, T.M., additional
- Published
- 1988
- Full Text
- View/download PDF
10. Ovalbumin-induced plasma interleukin-4 levels are reduced in ceramide kinase-deficient DO11.10 RAG1-/- mice
- Author
-
Baumruker Thomas, Urtz Nicole, Niwa Satoru, Billich Andreas, and Bornancin Frédéric
- Subjects
Nutritional diseases. Deficiency diseases ,RC620-627 - Abstract
Abstract Ceramide kinase (CERK) produces the bioactive lipid ceramide-1-phosphate (C1P) and is a key regulator of ceramide and dihydroceramide levels. It is likely that CERK and C1P play a role in inflammatory processes but the cells involved and the mechanisms used remain to be clarified. In particular, the impact of CERK on T-cell biology has not been studied so far. Here, we used Cerk-/- mice backcrossed with DO11.10/RAG1-/- mice to probe the effect of CERK ablation on T-cell activation. Levels of interleukin (IL)-2, IL-4, IL-5, IL-13, of tumor necrosis factor (TNF)-α, and of interferon (INF)-γ were recorded following ovalbumin challenge in vivo and using ovalbumin-treated splenocytes ex- vivo. Absence of CERK led to a significant decrease in the production of IL-4, thus suggesting that CERK may polarize T cells towards the TH2 cell subtype. However, the importance of CERK to TH2 cell biology will have to be investigated further because in a model of asthma, which is TH2-cell driven, Cerk-/- mice responded like wild-type animals.
- Published
- 2010
- Full Text
- View/download PDF
11. Ligandability Assessment of IL-1β by Integrated Hit Identification Approaches.
- Author
-
Vulpetti A, Rondeau JM, Bellance MH, Blank J, Boesch R, Boettcher A, Bornancin F, Buhr S, Connor LE, Dumelin CE, Esser O, Hediger M, Hintermann S, Hommel U, Koch E, Lapointe G, Leder L, Lehmann S, Lehr P, Meier P, Muller L, Ostermeier D, Ramage P, Schiebel-Haddad S, Smith AB, Stojanovic A, Velcicky J, Yamamoto R, and Hurth K
- Subjects
- Humans, Drug Discovery, Ligands, Receptors, Interleukin-1 Type I metabolism, Receptors, Interleukin-1 Type I antagonists & inhibitors, Small Molecule Libraries chemistry, Small Molecule Libraries pharmacology, Structure-Activity Relationship, DNA chemistry, Gene Library, Interleukin-1beta metabolism
- Abstract
Human interleukin-1β (IL-1β) is a pro-inflammatory cytokine that plays a critical role in the regulation of the immune response and the development of various inflammatory diseases. In this publication, we disclose our efforts toward the discovery of IL-1β binders that interfere with IL-1β signaling. To this end, several technologies were used in parallel, including fragment-based screening (FBS), DNA-encoded library (DEL) technology, peptide discovery platform (PDP), and virtual screening. The utilization of distinct technologies resulted in the identification of new chemical entities exploiting three different sites on IL-1β, all of them also inhibiting the interaction with the IL-1R1 receptor. Moreover, we identified lysine 103 of IL-1β as a target residue suitable for the development of covalent, low-molecular-weight IL-1β antagonists.
- Published
- 2024
- Full Text
- View/download PDF
12. HDAC6/aggresome processing pathway importance for inflammasome formation is context-dependent.
- Author
-
Wang L, Shi S, Unterreiner A, Kapetanovic R, Ghosh S, Sanchez J, Aslani S, Xiong Y, Hsu CL, Donovan KA, Farady CJ, Fischer ES, Bornancin F, and Matthias P
- Subjects
- Animals, Humans, Mice, Cell Line, Histone Deacetylase 6 genetics, Histone Deacetylase 6 metabolism, Interleukin-1beta genetics, Interleukin-1beta metabolism, Macrophages metabolism, NLR Family, Pyrin Domain-Containing 3 Protein genetics, NLR Family, Pyrin Domain-Containing 3 Protein metabolism, Protein Transport physiology, Inflammasomes metabolism, Leukocytes, Mononuclear metabolism
- Abstract
The inflammasome is a large multiprotein complex that assembles in the cell cytoplasm in response to stress or pathogenic infection. Its primary function is to defend the cell and promote the secretion of pro-inflammatory cytokines, including IL-1β and IL-18. Previous research has shown that in immortalized bone marrow-derived macrophages (iBMDMs) inflammasome assembly is dependent on the deacetylase HDAC6 and the aggresome processing pathway (APP), a cellular pathway involved in the disposal of misfolded proteins. Here we used primary BMDMs from mice in which HDAC6 is ablated or impaired and found that inflammasome activation was largely normal. We also used human peripheral blood mononuclear cells and monocyte cell lines expressing a synthetic protein blocking the HDAC6-ubiquitin interaction and impairing the APP and found that inflammasome activation was moderately affected. Finally, we used a novel HDAC6 degrader and showed that inflammasome activation was partially impaired in human macrophage cell lines with depleted HDAC6. Our results therefore show that HDAC6 importance in inflammasome activation is context-dependent., Competing Interests: Conflict of interest The authors declare the following financial interests/personal relationships which may be considered as potential competing interests: Part of the results presented herein have been used in the patent application EP-A-20213494.6. A. U., S. A., C. J. F. and F. B. are employees of Novartis. E. S. F is a founder, scientific advisory board (SAB) member, and equity holder of Civetta Therapeutics, Lighthorse Therapeutics, Proximity Therapeutics, and Neomorph, Inc. (also board of directors). He is an equity holder and SAB member for Avilar Therapeutics and Photys Therapeutics and a consultant to Novartis, Sanofi, EcoR1 Capital, Ajax Therapeutics, Odyssey Therapeutics and Deerfield. The Fischer lab receives or has received research funding from Deerfield, Novartis, Ajax, Interline and Astellas. K. A. D is a consultant to Kronos Bio and Neomorph Inc., (Copyright © 2024 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
13. A phospho-harmonic orchestra plays the NLRP3 score.
- Author
-
Bornancin F and Dekker C
- Subjects
- Phosphorylation, Protein Processing, Post-Translational, Proteins metabolism, Inflammasomes metabolism, NLR Family, Pyrin Domain-Containing 3 Protein metabolism
- Abstract
NLRP3 is a prototypical sensor protein connecting cellular stress to pro-inflammatory signaling. A complex array of regulatory steps is required to switch NLRP3 from an inactive state into a primed entity that is poised to assemble an inflammasome. Accumulating evidence suggests that post-translational mechanisms are critical. In particular, phosphorylation/dephosphorylation and ubiquitylation/deubiquitylation reactions have been reported to regulate NLRP3. Taken individually, several post-translational modifications appear to be essential. However, it remains difficult to understand how they may be coordinated, whether there is a unique sequence of regulatory steps accounting for the functional maturation of NLRP3, or whether the sequence is subject to variations depending on cell type, the stimulus, and other parameters such as the cellular context. This review will focus on the regulation of the NLRP3 inflammasome by phosphorylation and dephosphorylation, and on kinases and phosphatases that have been reported to modulate NLRP3 activity. The aim is to try to integrate the current understanding and highlight potential gaps for further studies., Competing Interests: Both authors are employees and/or shareholders of Novartis Pharma AG., (Copyright © 2023 Bornancin and Dekker.)
- Published
- 2023
- Full Text
- View/download PDF
14. Discovery of a selective and biologically active low-molecular weight antagonist of human interleukin-1β.
- Author
-
Hommel U, Hurth K, Rondeau JM, Vulpetti A, Ostermeier D, Boettcher A, Brady JP, Hediger M, Lehmann S, Koch E, Blechschmidt A, Yamamoto R, Tundo Dottorello V, Haenni-Holzinger S, Kaiser C, Lehr P, Lingel A, Mureddu L, Schleberger C, Blank J, Ramage P, Freuler F, Eder J, and Bornancin F
- Subjects
- Humans, Interleukin-1beta, Molecular Weight, Binding Sites, Biophysics, Cytokines, Thinness
- Abstract
Human interleukin-1β (hIL-1β) is a pro-inflammatory cytokine involved in many diseases. While hIL-1β directed antibodies have shown clinical benefit, an orally available low-molecular weight antagonist is still elusive, limiting the applications of hIL-1β-directed therapies. Here we describe the discovery of a low-molecular weight hIL-1β antagonist that blocks the interaction with the IL-1R1 receptor. Starting from a low affinity fragment-based screening hit 1, structure-based optimization resulted in a compound (S)-2 that binds and antagonizes hIL-1β with single-digit micromolar activity in biophysical, biochemical, and cellular assays. X-ray analysis reveals an allosteric mode of action that involves a hitherto unknown binding site in hIL-1β encompassing two loops involved in hIL-1R1/hIL-1β interactions. We show that residues of this binding site are part of a conformationally excited state of the mature cytokine. The compound antagonizes hIL-1β function in cells, including primary human fibroblasts, demonstrating the relevance of this discovery for future development of hIL-1β directed therapeutics., (© 2023. Springer Nature Limited.)
- Published
- 2023
- Full Text
- View/download PDF
15. Remibrutinib (LOU064) inhibits neuroinflammation driven by B cells and myeloid cells in preclinical models of multiple sclerosis.
- Author
-
Nuesslein-Hildesheim B, Ferrero E, Schmid C, Huck C, Smith P, Tisserand S, Rubert J, Bornancin F, Eichlisberger D, and Cenni B
- Subjects
- Humans, Animals, Mice, Neuroinflammatory Diseases, Myeloid Cells, Agammaglobulinaemia Tyrosine Kinase, Antigen-Antibody Complex, Anti-Inflammatory Agents, Multiple Sclerosis drug therapy, Encephalomyelitis, Autoimmune, Experimental drug therapy
- Abstract
Background: Bruton's tyrosine kinase (BTK) is a key signaling node in B cell receptor (BCR) and Fc receptor (FcR) signaling. BTK inhibitors (BTKi) are an emerging oral treatment option for patients suffering from multiple sclerosis (MS). Remibrutinib (LOU064) is a potent, highly selective covalent BTKi with a promising preclinical and clinical profile for MS and other autoimmune or autoallergic indications., Methods: The efficacy and mechanism of action of remibrutinib was assessed in two different experimental autoimmune encephalomyelitis (EAE) mouse models for MS. The impact of remibrutinib on B cell-driven EAE pathology was determined after immunization with human myelin oligodendrocyte glycoprotein (HuMOG). The efficacy on myeloid cell and microglia driven neuroinflammation was determined in the RatMOG EAE. In addition, we assessed the relationship of efficacy to BTK occupancy in tissue, ex vivo T cell response, as well as single cell RNA-sequencing (scRNA-seq) in brain and spinal cord tissue., Results: Remibrutinib inhibited B cell-dependent HuMOG EAE in dose-dependent manner and strongly reduced neurological symptoms. At the efficacious oral dose of 30 mg/kg, remibrutinib showed strong BTK occupancy in the peripheral immune organs and in the brain of EAE mice. Ex vivo MOG-specific T cell recall response was reduced, but not polyclonal T cell response, indicating absence of non-specific T cell inhibition. Remibrutinib also inhibited RatMOG EAE, suggesting that myeloid cell and microglia inhibition contribute to its efficacy in EAE. Remibrutinib did not reduce B cells, total Ig levels nor MOG-specific antibody response. In brain and spinal cord tissue a clear anti-inflammatory effect in microglia was detected by scRNA-seq. Finally, remibrutinib showed potent inhibition of in vitro immune complex-driven inflammatory response in human microglia., Conclusion: Remibrutinib inhibited EAE models by a two-pronged mechanism based on inhibition of pathogenic B cell autoreactivity, as well as direct anti-inflammatory effects in microglia. Remibrutinib showed efficacy in both models in absence of direct B cell depletion, broad T cell inhibition or reduction of total Ig levels. These findings support the view that remibrutinib may represent a novel treatment option for patients with MS., (© 2023. BioMed Central Ltd., part of Springer Nature.)
- Published
- 2023
- Full Text
- View/download PDF
16. HDAC6/aggresome processing pathway importance for inflammasome formation is context dependent.
- Author
-
Wang L, Unterreiner A, Kapetanovic R, Aslani S, Xiong Y, Donovan KA, Farady CJ, Fischer ES, Bornancin F, and Matthias P
- Abstract
The inflammasome is a large multiprotein complex that assembles in the cell cytoplasm in response to stress or pathogenic infection. Its primary function is to defend the cell and promote the secretion of pro-inflammatory cytokines, including IL-1β and IL-18. It was shown that in immortalized bone marrow derived macrophages (iBMDMs) inflammasome assembly is dependent on the deacetylase HDAC6 and the aggresome processing pathway (APP), a cellular pathway involved in the disposal of misfolded proteins. Here we used primary BMDMs from mice in which HDAC6 is ablated or impaired and found that inflammasome activation was largely normal. We also used human peripheral blood mononuclear cells and monocytes cell lines expressing a synthetic protein blocking HDAC6-ubiquitin interaction and impairing the APP and found that inflammasome activation was moderately affected. Finally, we used a novel HDAC6 degrader and showed that inflammasome activation was partially impaired in human macrophage cell lines with depleted HDAC6. Our results therefore show that HDAC6 importance in inflammasome activation is context dependent., Competing Interests: Declaration of interests Part of the results presented herein have been used in the patent application EP-A-20213494.6. A.U., S.A., C.J.F. and F.B. are employees of Novartis. E.S.F is a founder, scientific advisory board (SAB) member, and equity holder of Civetta Therapeutics, Lighthorse Therapeutics, Proximity Therapeutics, and Neomorph, Inc. (also board of directors). He is an equity holder and SAB member for Avilar Therapeutics and Photys Therapeutics and a consultant to Novartis, Sanofi, EcoR1 Capital, Ajax Therapeutics, Odyssey Therapeutics and Deerfield. The Fischer lab receives or has received research funding from Deerfield, Novartis, Ajax, Interline and Astellas. K.A.D is a consultant to Kronos Bio and Neomorph Inc.
- Published
- 2023
- Full Text
- View/download PDF
17. Integrating knowledge of protein sequence with protein function for the prediction and validation of new MALT1 substrates.
- Author
-
Bell PA, Scheuermann S, Renner F, Pan CL, Lu HY, Turvey SE, Bornancin F, Régnier CH, and Overall CM
- Abstract
We developed a bioinformatics-led substrate discovery workflow to expand the known substrate repertoire of MALT1. Our approach, termed GO-2-Substrates, integrates protein function information, including GO terms from known substrates, with protein sequences to rank substrate candidates by similarity. We applied GO-2-Substrates to MALT1, a paracaspase and master regulator of NF-κB signalling in adaptive immune responses. With only 12 known substrates, the evolutionarily conserved paracaspase functions and phenotypes of Malt1
-/- mice strongly implicate the existence of undiscovered substrates. We tested the ranked predictions from GO-2-Substrates of new MALT1 human substrates by co-expression of candidates transfected with the oncogenic constitutively active cIAP2-MALT1 fusion protein or CARD11/BCL10/MALT1 active signalosome. We identified seven new MALT1 substrates by the co-transfection screen: TANK, TAB3, CASP10, ZC3H12D, ZC3H12B, CILK1 and ILDR2. Using catalytically inactive cIAP2-MALT1 (Cys464Ala), a MALT1 inhibitor, MLT-748, and noncleavable P1-Arg to Ala mutant versions of each substrate in dual transfections, we validated the seven new substrates in vitro. We confirmed the cleavage of endogenous TANK and the RNase ZC3H12D in B cells by Western blotting and mining TAILS N -terminomics datasets, where we also uncovered evidence for these and 12 other candidate substrates by endogenous MALT1. Thus, protein function information improves substrate predictions. The new substrates and other high-ranked MALT1 candidate substrates should open new biological frontiers for further validation and exploration of the function of MALT1 within and beyond NF-κB regulation., Competing Interests: 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., (© 2022 The Author(s).)- Published
- 2022
- Full Text
- View/download PDF
18. MALT1 Is a Targetable Driver of Epithelial-to-Mesenchymal Transition in Claudin-Low, Triple-Negative Breast Cancer.
- Author
-
Lee JL, Ekambaram P, Carleton NM, Hu D, Klei LR, Cai Z, Myers MI, Hubel NE, Covic L, Agnihotri S, Krappmann D, Bornancin F, Lee AV, Oesterreich S, McAllister-Lucas LM, and Lucas PC
- Subjects
- Animals, Cell Line, Tumor, Cell Movement, Claudins pharmacology, Claudins therapeutic use, Epithelial-Mesenchymal Transition, Humans, Mice, Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein genetics, Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein metabolism, Receptor, PAR-1 therapeutic use, Triple Negative Breast Neoplasms metabolism
- Abstract
MALT1 is the effector protein of the CARMA/Bcl10/MALT1 (CBM) signalosome, a multiprotein complex that drives pro-inflammatory signaling pathways downstream of a diverse set of receptors. Although CBM activity is best known for its role in immune cells, emerging evidence suggests that it plays a key role in the pathogenesis of solid tumors, where it can be activated by selected G protein-coupled receptors (GPCR). Here, we demonstrated that overexpression of GPCRs implicated in breast cancer pathogenesis, specifically the receptors for Angiotensin II and thrombin (AT1R and PAR1), drove a strong epithelial-to-mesenchymal transition (EMT) program in breast cancer cells that is characteristic of claudin-low, triple-negative breast cancer (TNBC). In concert, MALT1 was activated in these cells and contributed to the dramatic EMT phenotypic changes through regulation of master EMT transcription factors including Snail and ZEB1. Importantly, blocking MALT1 signaling, through either siRNA-mediated depletion of MALT1 protein or pharmacologic inhibition of its activity, was effective at partially reversing the molecular and phenotypic indicators of EMT. Treatment of mice with mepazine, a pharmacologic MALT1 inhibitor, reduced growth of PAR1+, MDA-MB-231 xenografts and had an even more dramatic effect in reducing the burden of metastatic disease. These findings highlight MALT1 as an attractive therapeutic target for claudin-low TNBCs harboring overexpression of one or more selected GPCRs., Implications: This study nominates a GPCR/MALT1 signaling axis as a pathway that can be pharmaceutically targeted to abrogate EMT and metastatic progression in TNBC, an aggressive form of breast cancer that currently lacks targeted therapies., (©2021 American Association for Cancer Research.)
- Published
- 2022
- Full Text
- View/download PDF
19. Evaluation of protein kinase D auto-phosphorylation as biomarker for NLRP3 inflammasome activation.
- Author
-
Heiser D, Rubert J, Unterreiner A, Maurer C, Kamke M, Bodendorf U, Farady CJ, Roediger B, and Bornancin F
- Subjects
- Biomarkers metabolism, Caspase 1 metabolism, Cell Line, Tumor, Humans, Inflammation metabolism, Intracellular Signaling Peptides and Proteins metabolism, Phosphate-Binding Proteins metabolism, Phosphorylation, Pyrin metabolism, U937 Cells, Inflammasomes metabolism, NLR Family, Pyrin Domain-Containing 3 Protein metabolism, Protein Kinase C metabolism
- Abstract
Background: The NLRP3 inflammasome is a critical component of sterile inflammation, which is involved in many diseases. However, there is currently no known proximal biomarker for measuring NLRP3 activation in pathological conditions. Protein kinase D (PKD) has emerged as an important NLRP3 kinase that catalyzes the release of a phosphorylated NLRP3 species that is competent for inflammasome complex assembly., Methods: To explore the potential for PKD activation to serve as a selective biomarker of the NLRP3 pathway, we tested various stimulatory conditions in THP-1 and U937 cell lines, probing the inflammasome space beyond NLRP3. We analyzed the correlation between PKD activation (monitored by its auto-phosphorylation) and functional inflammasome readouts., Results: PKD activation/auto-phosphorylation always preceded cleavage of caspase-1 and gasdermin D, and treatment with the PKD inhibitor CRT0066101 could block NLRP3 inflammasome assembly and interleukin-1β production. Conversely, blocking NLRP3 either genetically or using the MCC950 inhibitor prevented PKD auto-phosphorylation, indicating a bidirectional functional crosstalk between NLRP3 and PKD. Further assessments of the pyrin and NLRC4 pathways, however, revealed that PKD auto-phosphorylation can be triggered by a broad range of stimuli unrelated to NLRP3 inflammasome assembly., Conclusion: Although PKD and NLRP3 become functionally interconnected during NLRP3 activation, the promiscuous reactivity of PKD challenges its potential use for tracing the NLRP3 inflammasome pathway., Competing Interests: All authors are employees at Novartis Institutes for Biomedical Research (NIBR) at time of studies. This does not alter adherence to PLOS ONE policies on sharing data and materials. None of the authors have competing interest relating to employment,consultancy, patents, products in development, marketed product or else.
- Published
- 2021
- Full Text
- View/download PDF
20. CARD10 cleavage by MALT1 restricts lung carcinoma growth in vivo.
- Author
-
Israël L, Glück A, Berger M, Coral M, Ceci M, Unterreiner A, Rubert J, Bardet M, Ginster S, Golding-Ochsenbein AM, Martin K, Hoyler T, Calzascia T, Wieczorek G, Hillenbrand R, Ferretti S, Ferrero E, and Bornancin F
- Abstract
CARD-CC complexes involving BCL10 and MALT1 are major cellular signaling hubs. They govern NF-κB activation through their scaffolding properties as well as MALT1 paracaspase function, which cleaves substrates involved in NF-κB regulation. In human lymphocytes, gain-of-function defects in this pathway lead to lymphoproliferative disorders. CARD10, the prototypical CARD-CC protein in non-hematopoietic cells, is overexpressed in several cancers and has been associated with poor prognosis. However, regulation of CARD10 remains poorly understood. Here, we identified CARD10 as the first MALT1 substrate in non-hematopoietic cells and showed that CARD10 cleavage by MALT1 at R587 dampens its capacity to activate NF-κB. Preventing CARD10 cleavage in the lung tumor A549 cell line increased basal levels of IL-6 and extracellular matrix components in vitro, and led to increased tumor growth in a mouse xenograft model, suggesting that CARD10 cleavage by MALT1 might be a built-in mechanism controlling tumorigenicity.
- Published
- 2021
- Full Text
- View/download PDF
21. Pharmacological inhibition of IKKβ dampens NLRP3 inflammasome activation after priming in the human myeloid cell line THP-1.
- Author
-
Unterreiner A, Rubert J, Kauffmann M, Fruhauf A, Heiser D, Erbel P, Schlapbach A, Eder J, Bodendorf U, Boettcher A, Farady CJ, and Bornancin F
- Subjects
- Amides pharmacology, Caspase 1 metabolism, Humans, Immunity, Innate drug effects, Inflammasomes drug effects, Inflammasomes immunology, Interleukin-1beta biosynthesis, NF-kappa B metabolism, Nigericin pharmacology, Protein Kinase Inhibitors pharmacology, THP-1 Cells, Thiophenes pharmacology, I-kappa B Kinase antagonists & inhibitors, Inflammasomes metabolism, NLR Family, Pyrin Domain-Containing 3 Protein metabolism
- Abstract
The NLRP3 inflammasome is a critical component of the innate immune response to sterile inflammation. Its regulation involves a priming step, required for up-regulation of inflammasome protagonists and an activation step leading to NLRP3 inflammasome complex assembly, which triggers caspase-1 activity. The IκKβ kinase regulates canonical NF-κB, a key pathway involved in transcriptional priming. We found that IκKβ also regulates the activation and function of the NLRP3 inflammasome beyond the priming step. Two unrelated IκKβ inhibitors, AFN700 and TPCA-1, when applied after priming, fully blocked IL-1β secretion triggered by nigericin in THP-1 cells. Both inhibitors prevented neither inflammasome assembly, as monitored by measuring the formation of ASC specks, nor the generation of caspase-1 p20, a hallmark of caspase-1 activity, but they impaired the initial cleavage and activation of procaspase-1. These data thus indicate that IκKβ activity is required for efficient activation of NLRP3, suggesting that IκKβ may fulfill a dual role in coupling priming and activation of the NLRP3 inflammasome., 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 © 2021 Elsevier Inc. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
22. Optimization of the In Vivo Potency of Pyrazolopyrimidine MALT1 Protease Inhibitors by Reducing Metabolism and Increasing Potency in Whole Blood.
- Author
-
Quancard J, Simic O, Pissot Soldermann C, Aichholz R, Blatter M, Renatus M, Erbel P, Melkko S, Endres R, Sorge M, Kieffer L, Wagner T, Beltz K, Mcsheehy P, Wartmann M, Régnier CH, Calzascia T, Radimerski T, Bigaud M, Weiss A, Bornancin F, and Schlapbach A
- Subjects
- Animals, Antineoplastic Agents chemical synthesis, Antineoplastic Agents metabolism, Antineoplastic Agents pharmacokinetics, Antineoplastic Agents therapeutic use, Caspase Inhibitors chemical synthesis, Caspase Inhibitors metabolism, Caspase Inhibitors pharmacokinetics, Cell Line, Tumor, Female, Half-Life, Humans, Mice, Inbred BALB C, Mice, SCID, Microsomes, Liver metabolism, Neoplasms drug therapy, Pyrazoles chemical synthesis, Pyrazoles metabolism, Pyrazoles pharmacokinetics, Pyrimidines chemical synthesis, Pyrimidines metabolism, Pyrimidines pharmacokinetics, Rats, Sprague-Dawley, Sheep, Urea chemical synthesis, Urea metabolism, Urea pharmacokinetics, Xenograft Model Antitumor Assays, Blood metabolism, Caspase Inhibitors therapeutic use, Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein antagonists & inhibitors, Pyrazoles therapeutic use, Pyrimidines therapeutic use, Urea therapeutic use
- Abstract
The paracaspase MALT1 has gained increasing interest as a target for the treatment of subsets of lymphomas as well as autoimmune diseases, and there is a need for suitable compounds to explore the therapeutic potential of this target. Here, we report the optimization of the in vivo potency of pyrazolopyrimidines, a class of highly selective allosteric MALT1 inhibitors. High doses of the initial lead compound led to tumor stasis in an activated B-cell-like (ABC) diffuse large B-cell lymphoma (DLBCL) xenograft model, but this compound suffered from a short in vivo half-life and suboptimal potency in whole blood. Guided by metabolism studies, we identified compounds with reduced metabolic clearance and increased in vivo half-life. In the second optimization step, masking one of the hydrogen-bond donors of the central urea moiety through an intramolecular interaction led to improved potency in whole blood. This was associated with improved in vivo potency in a mechanistic model of B cell activation. The optimized compound led to tumor regression in a CARD11 mutant ABC-DLBCL lymphoma xenograft model.
- Published
- 2020
- Full Text
- View/download PDF
23. Discovery of Potent, Highly Selective, and In Vivo Efficacious, Allosteric MALT1 Inhibitors by Iterative Scaffold Morphing.
- Author
-
Pissot Soldermann C, Simic O, Renatus M, Erbel P, Melkko S, Wartmann M, Bigaud M, Weiss A, McSheehy P, Endres R, Santos P, Blank J, Schuffenhauer A, Bold G, Buschmann N, Zoller T, Altmann E, Manley PW, Dix I, Buchdunger E, Scesa J, Quancard J, Schlapbach A, Bornancin F, Radimerski T, and Régnier CH
- Subjects
- Animals, Antineoplastic Agents chemical synthesis, Antineoplastic Agents pharmacology, Antineoplastic Agents therapeutic use, Caspase Inhibitors chemical synthesis, Caspase Inhibitors pharmacology, Drug Discovery, Female, Humans, Immunity, Humoral drug effects, Male, Mice, Inbred BALB C, Molecular Structure, Pyrazoles chemical synthesis, Pyrazoles pharmacology, Pyrazoles therapeutic use, Pyrimidines chemical synthesis, Pyrimidines pharmacology, Pyrimidines therapeutic use, Rats, Sprague-Dawley, Structure-Activity Relationship, T-Lymphocytes drug effects, Urea pharmacology, Xenograft Model Antitumor Assays, Caspase Inhibitors therapeutic use, Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein antagonists & inhibitors, Neoplasms drug therapy, Urea analogs & derivatives, Urea therapeutic use
- Abstract
MALT1 plays a central role in immune cell activation by transducing NF-κB signaling, and its proteolytic activity represents a key node for therapeutic intervention. Two cycles of scaffold morphing of a high-throughput biochemical screening hit resulted in the discovery of MLT-231, which enabled the successful pharmacological validation of MALT1 allosteric inhibition in preclinical models of humoral immune responses and B-cell lymphomas. Herein, we report the structural activity relationships (SARs) and analysis of the physicochemical properties of a pyrazolopyrimidine-derived compound series. In human T-cells and B-cell lymphoma lines, MLT-231 potently and selectively inhibits the proteolytic activity of MALT1 in NF-κB-dependent assays. Both in vitro and in vivo profiling of MLT-231 support further optimization of this in vivo tool compound toward preclinical characterization.
- Published
- 2020
- Full Text
- View/download PDF
24. Requirement of Mucosa-Associated Lymphoid Tissue Lymphoma Translocation Protein 1 Protease Activity for Fcγ Receptor-Induced Arthritis, but Not Fcγ Receptor-Mediated Platelet Elimination, in Mice.
- Author
-
Martin K, Touil R, Cvijetic G, Israel L, Kolb Y, Sarret S, Valeaux S, Degl'Innocenti E, Le Meur T, Caesar N, Bardet M, Beerli C, Zerwes HG, Kovarik J, Beltz K, Schlapbach A, Quancard J, Régnier CH, Bigaud M, Junt T, Wieczorek G, Isnardi I, Littlewood-Evans A, Bornancin F, and Calzascia T
- Subjects
- Animals, Antigen-Antibody Complex metabolism, Blood Platelets metabolism, Cytokines immunology, Disease Models, Animal, Humans, Mice, Myeloid Cells metabolism, Arthritis, Experimental immunology, Arthritis, Rheumatoid immunology, Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein immunology, Receptors, IgG immunology
- Abstract
Objective: Fcγ receptors (FcγR) play important roles in both protective and pathogenic immune responses. The assembly of the CBM signalosome encompassing caspase recruitment domain-containing protein 9, B cell CLL/lymphoma 10, and mucosa-associated lymphoid tissue lymphoma translocation protein 1 (MALT-1) is required for optimal FcγR-induced canonical NF-κB activation and proinflammatory cytokine release. This study was undertaken to clarify the relevance of MALT-1 protease activity in FcγR-driven events and evaluate the therapeutic potential of selective MALT-1 protease inhibitors in FcγR-mediated diseases., Methods: Using genetic and pharmacologic disruption of MALT-1 scaffolding and enzymatic activity, we assessed the relevance of MALT-1 function in murine and human primary myeloid cells upon stimulation with immune complexes (ICs) and in murine models of autoantibody-driven arthritis and immune thrombocytopenic purpura (ITP)., Results: MALT-1 protease function is essential for optimal FcγR-induced production of proinflammatory cytokines by various murine and human myeloid cells stimulated with ICs. In contrast, MALT-1 protease inhibition did not affect the Syk-dependent, FcγR-mediated production of reactive oxygen species or leukotriene B
4 . Notably, pharmacologic MALT-1 protease inhibition in vivo reduced joint inflammation in the murine K/BxN serum-induced arthritis model (mean area under the curve for paw swelling of 45.42% versus 100% in control mice; P = 0.0007) but did not affect platelet depletion in a passive model of ITP., Conclusion: Our findings indicate a specific contribution of MALT-1 protease activity to FcγR-mediated events and suggest that MALT-1 protease inhibitors have therapeutic potential in a subset of FcγR-driven inflammatory disorders., (© 2020, American College of Rheumatology.)- Published
- 2020
- Full Text
- View/download PDF
25. Pharmacological Inhibition of MALT1 Protease Leads to a Progressive IPEX-Like Pathology.
- Author
-
Martin K, Junker U, Tritto E, Sutter E, Rubic-Schneider T, Morgan H, Niwa S, Li J, Schlapbach A, Walker D, Bigaud M, Beerli C, Littlewood-Evans A, Rudolph B, Laisney M, Ledieu D, Beltz K, Quancard J, Bornancin F, Zamurovic Ribrioux N, and Calzascia T
- Subjects
- Animals, Diabetes Mellitus, Type 1 etiology, Dogs, Female, Humans, Immune System Diseases etiology, Inflammation chemically induced, Male, Mice, Mice, Inbred C57BL, Rats, Rats, Inbred Lew, Rats, Wistar, T-Lymphocytes, Regulatory immunology, Diabetes Mellitus, Type 1 congenital, Diarrhea etiology, Genetic Diseases, X-Linked etiology, Immune System Diseases congenital, Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein antagonists & inhibitors, T-Lymphocytes, Regulatory drug effects
- Abstract
Genetic disruption or short-term pharmacological inhibition of MALT1 protease is effective in several preclinical models of autoimmunity and B cell malignancies. Despite these protective effects, the severe reduction in regulatory T cells (Tregs) and the associated IPEX-like pathology occurring upon congenital disruption of the MALT1 protease in mice has raised concerns about the long-term safety of MALT1 inhibition. Here we describe the results of a series of toxicology studies in rat and dog species using MLT-943, a novel potent and selective MALT1 protease inhibitor. While MLT-943 effectively prevented T cell-dependent B cell immune responses and reduced joint inflammation in the collagen-induced arthritis rat pharmacology model, in both preclinical species, pharmacological inhibition of MALT1 was associated with a rapid and dose-dependent reduction in Tregs and resulted in the progressive appearance of immune abnormalities and clinical signs of an IPEX-like pathology. At the 13-week time point, rats displayed severe intestinal inflammation associated with mast cell activation, high serum IgE levels, systemic T cell activation and mononuclear cell infiltration in multiple tissues. Importantly, using thymectomized rats we demonstrated that MALT1 protease inhibition affects peripheral Treg frequency independently of effects on thymic Treg output and development. Our data confirm the therapeutic potential of MALT1 protease inhibitors but highlight the safety risks and challenges to consider before potential application of such inhibitors into the clinic., (Copyright © 2020 Martin, Junker, Tritto, Sutter, Rubic-Schneider, Morgan, Niwa, Li, Schlapbach, Walker, Bigaud, Beerli, Littlewood-Evans, Rudolph, Laisney, Ledieu, Beltz, Quancard, Bornancin, Zamurovic Ribrioux and Calzascia.)
- Published
- 2020
- Full Text
- View/download PDF
26. Classification and Nomenclature of Metacaspases and Paracaspases: No More Confusion with Caspases.
- Author
-
Minina EA, Staal J, Alvarez VE, Berges JA, Berman-Frank I, Beyaert R, Bidle KD, Bornancin F, Casanova M, Cazzulo JJ, Choi CJ, Coll NS, Dixit VM, Dolinar M, Fasel N, Funk C, Gallois P, Gevaert K, Gutierrez-Beltran E, Hailfinger S, Klemenčič M, Koonin EV, Krappmann D, Linusson A, Machado MFM, Madeo F, Megeney LA, Moschou PN, Mottram JC, Nyström T, Osiewacz HD, Overall CM, Pandey KC, Ruland J, Salvesen GS, Shi Y, Smertenko A, Stael S, Ståhlberg J, Suárez MF, Thome M, Tuominen H, Van Breusegem F, van der Hoorn RAL, Vardi A, Zhivotovsky B, Lam E, and Bozhkov PV
- Subjects
- Animals, Caspases chemistry, Caspases metabolism, Consensus, Humans, Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein chemistry, Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein metabolism, Plant Proteins chemistry, Plant Proteins metabolism, Protein Conformation, Structure-Activity Relationship, Caspases classification, Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein classification, Plant Proteins classification, Terminology as Topic
- Published
- 2020
- Full Text
- View/download PDF
27. Malt1 Protease Deficiency in Mice Disrupts Immune Homeostasis at Environmental Barriers and Drives Systemic T Cell-Mediated Autoimmunity.
- Author
-
Martin K, Touil R, Kolb Y, Cvijetic G, Murakami K, Israel L, Duraes F, Buffet D, Glück A, Niwa S, Bigaud M, Junt T, Zamurovic N, Smith P, McCoy KD, Ohashi PS, Bornancin F, and Calzascia T
- Subjects
- Animals, Mice, Mice, Inbred C57BL, Mice, Knockout, Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein deficiency, Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein genetics, Autoimmunity immunology, Homeostasis immunology, Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein immunology, T-Lymphocytes, Regulatory immunology
- Abstract
The paracaspase Malt1 is a key regulator of canonical NF-κB activation downstream of multiple receptors in both immune and nonimmune cells. Genetic disruption of Malt1 protease function in mice and MALT1 mutations in humans results in reduced regulatory T cells and a progressive multiorgan inflammatory pathology. In this study, we evaluated the altered immune homeostasis and autoimmune disease in Malt1 protease-deficient (Malt1PD) mice and the Ags driving disease manifestations. Our data indicate that B cell activation and IgG1/IgE production is triggered by microbial and dietary Ags preferentially in lymphoid organs draining mucosal barriers, likely as a result of dysregulated mucosal immune homeostasis. Conversely, the disease was driven by a polyclonal T cell population directed against self-antigens. Characterization of the Malt1PD T cell compartment revealed expansion of T effector memory cells and concomitant loss of a CD4
+ T cell population that phenotypically resembles anergic T cells. Therefore, we propose that the compromised regulatory T cell compartment in Malt1PD animals prevents the efficient maintenance of anergy and supports the progressive expansion of pathogenic, IFN-γ-producing T cells. Overall, our data revealed a crucial role of the Malt1 protease for the maintenance of intestinal and systemic immune homeostasis, which might provide insights into the mechanisms underlying IPEX-related diseases associated with mutations in MALT1 ., (Copyright © 2019 by The American Association of Immunologists, Inc.)- Published
- 2019
- Full Text
- View/download PDF
28. Inhibition of MALT1 paracaspase activity improves lesion recovery following spinal cord injury.
- Author
-
Zhang H, Sun G, Li X, Fu Z, Guo C, Cao G, Wang B, Wang Q, Yang S, Li D, Xia X, Li P, Zhu J, Zhou W, Zheng L, Li J, Zhang L, Hao J, Zhou L, Bornancin F, Li Z, Yin Z, and Gao Y
- Abstract
Spinal cord injury (SCI) is a devastating traumatic injury that causes persistent, severe motor and sensory dysfunction. Immune responses are involved in functional recovery after SCI. Mucosa-associated lymphoid tissue lymphoma translocation 1 (MALT1) has been shown to regulate the survival and differentiation of immune cells and to play a critical role in many diseases, but its function in lesion recovery after SCI remains unclear. In this paper, we generated KI (knock in) mice with a point mutation (C472G) in the active center of MALT1 and found that the KI mice exhibited improved functional recovery after SCI. Fewer macrophages were recruited to the injury site in KI mice and these macrophages differentiated into anti-inflammatory macrophages. Moreover, macrophages from KI mice exhibited reduced phosphorylation of p65, which in turn resulted in decreased SOCS3 expression and increased pSTAT6 levels. Similar results were obtained upon inhibition of MALT1 paracaspase with the small molecule inhibitor "MI-2" or the more specific inhibitor "MLT-827". In patients with SCI, peripheral blood mononuclear cells (PBMC) displayed increased MALT1 paracaspase. Human macrophages showed reduced pro-inflammatory and increased anti-inflammatory characteristics following the inhibition of MALT1 paracaspase. These findings suggest that inhibition of MALT1 paracaspase activity in the clinic may improve lesion recovery in subjects with SCI., (Copyright © 2019 Science China Press. Published by Elsevier B.V. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
29. Editorial: CARMA Proteins: Playing a Hand of Four CARDs.
- Author
-
Bornancin F and Snow AL
- Subjects
- Animals, CARD Signaling Adaptor Proteins chemistry, Humans, CARD Signaling Adaptor Proteins genetics, CARD Signaling Adaptor Proteins metabolism
- Published
- 2019
- Full Text
- View/download PDF
30. An allosteric MALT1 inhibitor is a molecular corrector rescuing function in an immunodeficient patient.
- Author
-
Quancard J, Klein T, Fung SY, Renatus M, Hughes N, Israël L, Priatel JJ, Kang S, Blank MA, Viner RI, Blank J, Schlapbach A, Erbel P, Kizhakkedathu J, Villard F, Hersperger R, Turvey SE, Eder J, Bornancin F, and Overall CM
- Subjects
- Gene Expression Regulation, Humans, Immunologic Deficiency Syndromes genetics, Immunologic Deficiency Syndromes therapy, Lymphocytes metabolism, MAP Kinase Signaling System genetics, MAP Kinase Signaling System physiology, Male, Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein genetics, Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein ultrastructure, NF-kappa B metabolism, Neoplasm Proteins, Signal Transduction, Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein antagonists & inhibitors, Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein metabolism
- Abstract
MALT1 paracaspase is central for lymphocyte antigen-dependent responses including NF-κB activation. We discovered nanomolar, selective allosteric inhibitors of MALT1 that bind by displacing the side chain of Trp580, locking the protease in an inactive conformation. Interestingly, we had previously identified a patient homozygous for a MALT1 Trp580-to-serine mutation who suffered from combined immunodeficiency. We show that the loss of tryptophan weakened interactions between the paracaspase and C-terminal immunoglobulin MALT1 domains resulting in protein instability, reduced protein levels and functions. Upon binding of allosteric inhibitors of increasing potency, we found proportionate increased stabilization of MALT1-W580S to reach that of wild-type MALT1. With restored levels of stable MALT1 protein, the most potent of the allosteric inhibitors rescued NF-κB and JNK signaling in patient lymphocytes. Following compound washout, MALT1 substrate cleavage was partly recovered. Thus, a molecular corrector rescues an enzyme deficiency by substituting for the mutated residue, inspiring new potential precision therapies to increase mutant enzyme activity in other deficiencies.
- Published
- 2019
- Full Text
- View/download PDF
31. Myeloid Innate Signaling Pathway Regulation by MALT1 Paracaspase Activity.
- Author
-
Unterreiner A, Touil R, Anastasi D, Dubois N, Niwa S, Calzascia T, and Bornancin F
- Subjects
- Animals, Humans, Mice, Signal Transduction, Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein metabolism
- Abstract
Besides its function in lymphoid cells, which has been addressed by numerous studies, the paracaspase MALT1 also plays an important role in innate cells downstream of pattern recognition receptors. Best studied are the Dectin-1 and Dectin-2 members of the C-type lectin-like receptor family that induce a SYK- and CARD9-dependent signaling cascade leading to NF-κB activation, in a MALT1-dependent manner. By contrast, Toll-like receptors (TLR), such as TLR-4, propagate NF-κB activation but signal via an MYD88/IRAK-dependent cascade. Nonetheless, whether MALT1 might contribute to TLR-4 signaling has remained unclear. Recent evidence with MLT-827, a potent and selective inhibitor of MALT1 paracaspase activity, indicates that TNF- production downstream of TLR-4 in human myeloid cells is independent of MALT1, as opposed to TNF- production downstream of Dectin-1, which is MALT1 dependent. Here, we addressed the selective involvement of MALT1 in pattern recognition sensing further, using a variety of human and mouse cellular preparations, and stimulation of Dectin-1, MINCLE or TLR-4 pathways. We also provided additional insights by exploring cytokines beyond TNF-, and by comparing MLT-827 to a SYK inhibitor (Cpd11) and to an IKK inhibitor (AFN700). Collectively, the data provided further evidence for the MALT1-dependency of C-type lectin-like receptor -signaling by contrast to TLR-signaling.
- Published
- 2019
- Full Text
- View/download PDF
32. MALT1 activation by TRAF6 needs neither BCL10 nor CARD11.
- Author
-
Bardet M, Seeholzer T, Unterreiner A, Woods S, Krappmann D, and Bornancin F
- Subjects
- B-Cell CLL-Lymphoma 10 Protein deficiency, CARD Signaling Adaptor Proteins deficiency, CRISPR-Cas Systems, Enzyme Activation drug effects, Gene Editing methods, Gene Expression Profiling, Gene Expression Regulation, Guanylate Cyclase deficiency, HEK293 Cells, Humans, Intracellular Signaling Peptides and Proteins, Jurkat Cells, Lymphocyte Activation drug effects, Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein metabolism, Signal Transduction, TNF Receptor-Associated Factor 6 metabolism, Tetradecanoylphorbol Acetate pharmacology, B-Cell CLL-Lymphoma 10 Protein genetics, CARD Signaling Adaptor Proteins genetics, Guanylate Cyclase genetics, Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein genetics, TNF Receptor-Associated Factor 6 genetics
- Abstract
The MALT1 (Mucosa associated lymphoid tissue lymphoma translocation protein 1) paracaspase couples antigen receptors on lymphocytes to downstream signaling events. Activation of MALT1 is known to involve stimulus-dependent CBM complex formation, that is, the recruitment of BCL10-bound MALT1 to a CARD-Coiled Coil protein. Beyond this canonical, CBM-dependent mechanism of MALT1 activation, recent studies suggest that MALT1 protease activity may be triggered by alternative mechanisms. For instance, the E3-ligase TRAF6 can activate MALT1 proteolytic function and induce MALT1 auto-cleavage. However, the interplay between CBM and TRAF6 with regard to MALT1 activation has remained incompletely elucidated. Here, by generating CRISPR/Cas9-derived knock-out Jurkat T-cells, we show that TRAF6 was dispensable for CARD11/BCL10-dependent MALT1 activation upon T-cell stimulation. However, ectopically-expressed TRAF6 could induce MALT1 activity in Jurkat T-cells devoid of either CARD11 or BCL10. These data provide unequivocal evidence that TRAF6-mediated MALT1 activation does not require the upstream scaffold CARD11 or the interaction between MALT1 and BCL10. Thus, TRAF6 may be part of a previously unidentified non-canonical pathway that triggers MALT1 protease activity independently of canonical CBM signalosomes., (Copyright © 2018 Elsevier Inc. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
33. Pharmacological inhibition of MALT1 protease activity suppresses endothelial activation via enhancing MCPIP1 expression.
- Author
-
Li Y, Huang S, Huang X, Li X, Falcon A, Soutar A, Bornancin F, Jiang Z, Xin HB, and Fu M
- Subjects
- Animals, Caspases metabolism, Cell Line, Human Umbilical Vein Endothelial Cells, Humans, Lymphocyte Activation drug effects, Macrophages drug effects, Macrophages metabolism, Mice, Mice, Inbred C57BL, NF-kappa B metabolism, Proteolysis drug effects, Signal Transduction drug effects, Endothelial Cells drug effects, Endothelial Cells metabolism, Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein antagonists & inhibitors, Ribonucleases metabolism, Transcription Factors metabolism
- Abstract
Mucosa associated lymphoid tissue lymphoma translocation protein 1 (MALT1) is not only an intracellular signaling scaffold protein but also a paracaspase that plays a key role in the signal transduction and cellular activation of lymphocytes and macrophages. However, its role in endothelial cells remains unknown. Here we report that pharmacological inhibition of MALT1 protease activity strongly suppresses endothelial activation via enhancing MCPIP1 expression. Treatment with MALT1 protease inhibitors selectively inhibited TNFα-induced VCAM-1 expression in HUVECs and LPS-induced VCAM-1 expression in mice. In addition, Inhibition of MALT1 protease activity also significantly inhibited TNFα-induced adhesion of THP-1 monocytic cells to HUVECs. To explore the mechanisms, MALT1 inhibitors does not affect the activation of NF-κB signaling pathway in HUVEC. However, they can stabilize MCPIP1 protein and significantly enhance MCPIP1 protein level in endothelial cells. These results suggest that MALT1 paracaspase also targets MCPIP1 and degrade MCPIP1 protein in endothelial cells similar as it does in immune cells. Taken together, the study suggest inhibition of MALT1 protease activity may represent a new strategy for prevention/therapy of vascular inflammatory diseases such as atherosclerosis., (Copyright © 2018 Elsevier Inc. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
34. A CARD10-Dependent Tonic Signalosome Activates MALT1 Paracaspase and Regulates IL-17/TNF-α-Driven Keratinocyte Inflammation.
- Author
-
Israël L, Bardet M, Huppertz A, Mercado N, Ginster S, Unterreiner A, Schlierf A, Goetschy JF, Zerwes HG, Roth L, Kolbinger F, and Bornancin F
- Subjects
- CARD Signaling Adaptor Proteins biosynthesis, Humans, Inflammation metabolism, Inflammation pathology, Interleukin-17 biosynthesis, Keratinocytes pathology, Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein biosynthesis, RNA genetics, Tumor Necrosis Factor-alpha biosynthesis, CARD Signaling Adaptor Proteins genetics, Gene Expression Regulation, Inflammation genetics, Interleukin-17 genetics, Keratinocytes metabolism, Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein genetics, Tumor Necrosis Factor-alpha genetics
- Published
- 2018
- Full Text
- View/download PDF
35. N-aryl-piperidine-4-carboxamides as a novel class of potent inhibitors of MALT1 proteolytic activity.
- Author
-
Schlapbach A, Revesz L, Pissot Soldermann C, Zoller T, Régnier CH, Bornancin F, Radimerski T, Blank J, Schuffenhauer A, Renatus M, Erbel P, Melkko S, Heng R, Simic O, Endres R, Wartmann M, and Quancard J
- Subjects
- Animals, Antineoplastic Agents chemical synthesis, Antineoplastic Agents chemistry, Cell Line, Tumor, Cell Proliferation drug effects, Dose-Response Relationship, Drug, Hepatocytes drug effects, Humans, Jurkat Cells, Microsomes drug effects, Molecular Structure, Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein metabolism, Piperidines chemical synthesis, Piperidines chemistry, Proteolysis drug effects, Rats, Structure-Activity Relationship, Antineoplastic Agents pharmacology, Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein antagonists & inhibitors, Piperidines pharmacology
- Abstract
Starting from a weak screening hit, potent and selective inhibitors of the MALT1 protease function were elaborated. Advanced compounds displayed high potency in biochemical and cellular assays. Compounds showed activity in a mechanistic Jurkat T cell activation assay as well as in the B-cell lymphoma line OCI-Ly3, which suggests potential use of MALT1 inhibitors in the treatment of autoimmune diseases as well as B-cell lymphomas with a dysregulated NF-κB pathway. Initially, rat pharmacokinetic properties of this compound series were dominated by very high clearance which could be linked to amide cleavage. Using a rat hepatocyte assay a good in vitro-in vivo correlation could be established which led to the identification of compounds with improved PK properties., (Copyright © 2018 Elsevier Ltd. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
36. Ways and waves of MALT1 paracaspase activation.
- Author
-
Israël L and Bornancin F
- Subjects
- Caspases, Liver, Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein, Natural Killer T-Cells, Pyroptosis
- Published
- 2018
- Full Text
- View/download PDF
37. The T-cell fingerprint of MALT1 paracaspase revealed by selective inhibition.
- Author
-
Bardet M, Unterreiner A, Malinverni C, Lafossas F, Vedrine C, Boesch D, Kolb Y, Kaiser D, Glück A, Schneider MA, Katopodis A, Renatus M, Simic O, Schlapbach A, Quancard J, Régnier CH, Bold G, Pissot-Soldermann C, Carballido JM, Kovarik J, Calzascia T, and Bornancin F
- Subjects
- Cell Proliferation, Cell Survival, Cells, Cultured, Gene Expression Regulation, Humans, Immunomodulation, Interleukin-2 metabolism, Interleukin-2 Receptor alpha Subunit metabolism, Lymphocyte Activation, Proteolysis, Receptors, Antigen, T-Cell metabolism, Signal Transduction, Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein metabolism, NF-kappa B metabolism, T-Lymphocytes immunology
- Abstract
Mucosa-associated lymphoid tissue lymphoma translocation protein 1 (MALT1) is essential for immune responses triggered by antigen receptors but the contribution of its paracaspase activity is not fully understood. Here, we studied how MALT1 proteolytic function regulates T-cell activation and fate after engagement of the T-cell receptor pathway. We show that MLT-827, a potent and selective MALT1 paracaspase inhibitor, does not prevent the initial phase of T-cell activation, in contrast to the pan-protein kinase C inhibitor AEB071. However, MLT-827 strongly impacted cell expansion after activation. We demonstrate this is the consequence of profound inhibition of IL-2 production as well as reduced expression of the IL-2 receptor alpha subunit (CD25), resulting from defective canonical NF-κB activation and accelerated mRNA turnover mechanisms. Accordingly, MLT-827 revealed a unique transcriptional fingerprint of MALT1 protease activity, providing evidence for broad control of T-cell signaling pathways. Altogether, this first report with a potent and selective inhibitor elucidates how MALT1 paracaspase activity integrates several T-cell activation pathways and indirectly controls gamma-chain receptor dependent survival, to impact on T-cell expansion., (© 2017 Australasian Society for Immunology Inc.)
- Published
- 2018
- Full Text
- View/download PDF
38. Selective MALT1 paracaspase inhibition does not block TNF-α production downstream of TLR4 in myeloid cells.
- Author
-
Unterreiner A, Stoehr N, Huppertz C, Calzascia T, Farady CJ, and Bornancin F
- Subjects
- Caspases, Myeloid Cells, NF-kappa B, Signal Transduction, Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein, Tumor Necrosis Factor-alpha
- Published
- 2017
- Full Text
- View/download PDF
39. Two Antagonistic MALT1 Auto-Cleavage Mechanisms Reveal a Role for TRAF6 to Unleash MALT1 Activation.
- Author
-
Ginster S, Bardet M, Unterreiner A, Malinverni C, Renner F, Lam S, Freuler F, Gerrits B, Voshol J, Calzascia T, Régnier CH, Renatus M, Nikolay R, Israël L, and Bornancin F
- Subjects
- Adaptor Proteins, Signal Transducing genetics, Adaptor Proteins, Signal Transducing metabolism, B-Cell CLL-Lymphoma 10 Protein, Blotting, Western, CARD Signaling Adaptor Proteins genetics, CARD Signaling Adaptor Proteins metabolism, Caspases genetics, Cell Line, Cells, Cultured, Electrophoresis, Polyacrylamide Gel, Guanylate Cyclase genetics, Guanylate Cyclase metabolism, HEK293 Cells, Humans, Immunoblotting, Jurkat Cells, Lymphocytes metabolism, Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein, Mutagenesis, Neoplasm Proteins genetics, Protein Isoforms genetics, Signal Transduction genetics, Signal Transduction physiology, TNF Receptor-Associated Factor 6 genetics, Ubiquitination genetics, Ubiquitination physiology, Caspases metabolism, Neoplasm Proteins metabolism, Protein Isoforms metabolism, T-Lymphocytes metabolism, TNF Receptor-Associated Factor 6 metabolism
- Abstract
The paracaspase MALT1 has arginine-directed proteolytic activity triggered by engagement of immune receptors. Recruitment of MALT1 into activation complexes is required for MALT1 proteolytic function. Here, co-expression of MALT1 in HEK293 cells, either with activated CARD11 and BCL10 or with TRAF6, was used to explore the mechanism of MALT1 activation at the molecular level. This work identified a prominent self-cleavage site of MALT1 isoform A (MALT1A) at R781 (R770 in MALT1B) and revealed that TRAF6 can activate MALT1 independently of the CBM. Intramolecular cleavage at R781/R770 removes a C-terminal TRAF6-binding site in both MALT1 isoforms, leaving MALT1B devoid of the two key interaction sites with TRAF6. A previously identified auto-proteolysis site of MALT1 at R149 leads to deletion of the death-domain, thereby abolishing interaction with BCL10. By using MALT1 isoforms and cleaved fragments thereof, as well as TRAF6 WT and mutant forms, this work shows that TRAF6 induces N-terminal auto-proteolytic cleavage of MALT1 at R149 and accelerates MALT1 protein turnover. The MALT1 fragment generated by N-terminal self-cleavage at R149 was labile and displayed enhanced signaling properties that required an intact K644 residue, previously shown to be a site for mono-ubiquitination of MALT1. Conversely, C-terminal self-cleavage at R781/R770 hampered the ability for self-cleavage at R149 and stabilized MALT1 by hindering interaction with TRAF6. C-terminal self-cleavage had limited impact on MALT1A but severely reduced MALT1B proteolytic and signaling functions. It also abrogated NF-κB activation by N-terminally cleaved MALT1A. Altogether, this study provides further insights into mechanisms that regulate the scaffolding and activation cycle of MALT1. It also emphasizes the reduced functional capacity of MALT1B as compared to MALT1A., Competing Interests: All co-authors work or used to work for Novartis. However, this does not alter our adherence to PLoS ONE policies on sharing data and materials.
- Published
- 2017
- Full Text
- View/download PDF
40. Deficiency of MALT1 paracaspase activity results in unbalanced regulatory and effector T and B cell responses leading to multiorgan inflammation.
- Author
-
Bornancin F, Renner F, Touil R, Sic H, Kolb Y, Touil-Allaoui I, Rush JS, Smith PA, Bigaud M, Junker-Walker U, Burkhart C, Dawson J, Niwa S, Katopodis A, Nuesslein-Hildesheim B, Weckbecker G, Zenke G, Kinzel B, Traggiai E, Brenner D, Brüstle A, St Paul M, Zamurovic N, McCoy KD, Rolink A, Régnier CH, Mak TW, Ohashi PS, Patel DD, and Calzascia T
- Subjects
- Animals, B-Lymphocytes, Regulatory pathology, Caspases genetics, Cell Differentiation genetics, Encephalomyelitis, Autoimmune, Experimental genetics, Encephalomyelitis, Autoimmune, Experimental pathology, Humans, Immunoglobulin E genetics, Immunoglobulin E immunology, Immunoglobulin G genetics, Immunoglobulin G immunology, Inflammation genetics, Inflammation immunology, Inflammation pathology, Interleukin-10 genetics, Interleukin-10 immunology, Interleukin-2 genetics, Interleukin-2 immunology, Mice, Mice, Knockout, Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein, Neoplasm Proteins genetics, T-Lymphocytes, Regulatory pathology, Th1 Cells immunology, Th1 Cells pathology, Th17 Cells immunology, Th17 Cells pathology, B-Lymphocytes, Regulatory immunology, Caspases immunology, Cell Differentiation immunology, Cell Proliferation, Encephalomyelitis, Autoimmune, Experimental immunology, Neoplasm Proteins immunology, T-Lymphocytes, Regulatory immunology
- Abstract
The paracaspase MALT1 plays an important role in immune receptor-driven signaling pathways leading to NF-κB activation. MALT1 promotes signaling by acting as a scaffold, recruiting downstream signaling proteins, as well as by proteolytic cleavage of multiple substrates. However, the relative contributions of these two different activities to T and B cell function are not well understood. To investigate how MALT1 proteolytic activity contributes to overall immune cell regulation, we generated MALT1 protease-deficient mice (Malt1(PD/PD)) and compared their phenotype with that of MALT1 knockout animals (Malt1(-/-)). Malt1(PD/PD) mice displayed defects in multiple cell types including marginal zone B cells, B1 B cells, IL-10-producing B cells, regulatory T cells, and mature T and B cells. In general, immune defects were more pronounced in Malt1(-/-) animals. Both mouse lines showed abrogated B cell responses upon immunization with T-dependent and T-independent Ags. In vitro, inactivation of MALT1 protease activity caused reduced stimulation-induced T cell proliferation, impaired IL-2 and TNF-α production, as well as defective Th17 differentiation. Consequently, Malt1(PD/PD) mice were protected in a Th17-dependent experimental autoimmune encephalomyelitis model. Surprisingly, Malt1(PD/PD) animals developed a multiorgan inflammatory pathology, characterized by Th1 and Th2/0 responses and enhanced IgG1 and IgE levels, which was delayed by wild-type regulatory T cell reconstitution. We therefore propose that the pathology characterizing Malt1(PD/PD) animals arises from an immune imbalance featuring pathogenic Th1- and Th2/0-skewed effector responses and reduced immunosuppressive compartments. These data uncover a previously unappreciated key function of MALT1 protease activity in immune homeostasis and underline its relevance in human health and disease., (Copyright © 2015 by The American Association of Immunologists, Inc.)
- Published
- 2015
- Full Text
- View/download PDF
41. The ceramide kinase inhibitor NVP-231 inhibits breast and lung cancer cell proliferation by inducing M phase arrest and subsequent cell death.
- Author
-
Pastukhov O, Schwalm S, Zangemeister-Wittke U, Fabbro D, Bornancin F, Japtok L, Kleuser B, Pfeilschifter J, and Huwiler A
- Subjects
- Adenocarcinoma, Blotting, Western, Cell Survival, DNA biosynthesis, DNA drug effects, Flow Cytometry, Humans, MCF-7 Cells, Staurosporine pharmacology, Apoptosis drug effects, Benzothiazoles pharmacology, Breast Neoplasms, Bridged-Ring Compounds pharmacology, Cell Proliferation drug effects, Enzyme Inhibitors pharmacology, Lung Neoplasms, M Phase Cell Cycle Checkpoints drug effects, Phosphotransferases (Alcohol Group Acceptor) antagonists & inhibitors, Tumor Stem Cell Assay
- Abstract
Background and Purpose: Ceramide kinase (CerK) catalyzes the generation of ceramide-1-phosphate which may regulate various cellular functions, including inflammatory reactions and cell growth. Here, we studied the effect of a recently developed CerK inhibitor, NVP-231, on cancer cell proliferation and viability and investigated the role of cell cycle regulators implicated in these responses., Experimental Approach: The breast and lung cancer cell lines MCF-7 and NCI-H358 were treated with increasing concentrations of NVP-231 and DNA synthesis, colony formation and cell death were determined. Flow cytometry was performed to analyse cell cycle distribution of cells and Western blot analysis was used to detect changes in cell cycle regulator expression and activation., Key Results: In both cell lines, NVP-231 concentration-dependently reduced cell viability, DNA synthesis and colony formation. Moreover it induced apoptosis, as measured by increased DNA fragmentation and caspase-3 and caspase-9 cleavage. Cell cycle analysis revealed that NVP-231 decreased the number of cells in S phase and induced M phase arrest with an increased mitotic index, as determined by increased histone H3 phosphorylation. The effect on the cell cycle was even more pronounced when NVP-231 treatment was combined with staurosporine. Finally, overexpression of CerK protected, whereas down-regulation of CerK with siRNA sensitized, cells for staurosporine-induced apoptosis., Conclusions and Implications: Our data demonstrate for the first time a crucial role for CerK in the M phase control in cancer cells and suggest its targeted inhibition, using drugs such as NVP-231, in combination with conventional pro-apoptotic chemotherapy., (© 2014 The British Pharmacological Society.)
- Published
- 2014
- Full Text
- View/download PDF
42. Structural determinants of MALT1 protease activity.
- Author
-
Wiesmann C, Leder L, Blank J, Bernardi A, Melkko S, Decock A, D'Arcy A, Villard F, Erbel P, Hughes N, Freuler F, Nikolay R, Alves J, Bornancin F, and Renatus M
- Subjects
- Adaptor Proteins, Signal Transducing genetics, Adaptor Proteins, Signal Transducing metabolism, Amino Acid Sequence, Animals, B-Cell CLL-Lymphoma 10 Protein, Catalytic Domain, Cells, Cultured, Dimerization, Enzyme Activation, HEK293 Cells, Humans, Ligands, Mice, Models, Molecular, Molecular Sequence Data, Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein, Mutation, NF-kappa B genetics, NF-kappa B metabolism, Protein Binding, Protein Interaction Domains and Motifs genetics, Protein Structure, Tertiary, Receptors, Antigen chemistry, Receptors, Antigen genetics, Receptors, Antigen metabolism, Signal Transduction, Structure-Activity Relationship, Caspases chemistry, Caspases metabolism, Neoplasm Proteins chemistry, Neoplasm Proteins metabolism
- Abstract
The formation of the CBM (CARD11-BCL10-MALT1) complex is pivotal for antigen-receptor-mediated activation of the transcription factor NF-κB. Signaling is dependent on MALT1 (mucosa-associated lymphoid tissue lymphoma translocation protein 1), which not only acts as a scaffolding protein but also possesses proteolytic activity mediated by its caspase-like domain. It remained unclear how the CBM activates MALT1. Here, we provide biochemical and structural evidence that MALT1 activation is dependent on its dimerization and show that mutations at the dimer interface abrogate activity in cells. The unliganded protease presents itself in a dimeric yet inactive state and undergoes substantial conformational changes upon substrate binding. These structural changes also affect the conformation of the C-terminal Ig-like domain, a domain that is required for MALT1 activity. Binding to the active site is coupled to a relative movement of caspase and Ig-like domains. MALT1 binding partners thus may have the potential of tuning MALT1 protease activity without binding directly to the caspase domain., (Copyright © 2012 Elsevier Ltd. All rights reserved.)
- Published
- 2012
- Full Text
- View/download PDF
43. Ceramide kinase: the first decade.
- Author
-
Bornancin F
- Subjects
- Amino Acid Motifs, Animals, Cell Proliferation, Cells, Cultured, Gene Expression Regulation, Humans, Immune System metabolism, Inflammation Mediators metabolism, Neoplasms genetics, Neoplasms pathology, Nervous System metabolism, Phosphotransferases (Alcohol Group Acceptor) antagonists & inhibitors, Phosphotransferases (Alcohol Group Acceptor) genetics, Protein Structure, Tertiary, Protein Transport, Signal Transduction, Sphingolipids metabolism, Up-Regulation, Ceramides metabolism, Phosphotransferases (Alcohol Group Acceptor) metabolism
- Abstract
It has been some 20 years since the initial discovery of ceramide 1-phosphate (C1P) and nearly a decade since ceramide kinase (CERK) was cloned. Many studies have shown that C1P is important for membrane biology and for the regulation of membrane-bound proteins, and the CERK enzyme has appeared to be tightly regulated in order to control both ceramide levels and production of C1P. Furthermore, C1P made by CERK has emerged as a genuine signalling entity. However, it represents only part of the C1P pool that is available in the cell, therefore suggesting that alternative unknown C1P-producing mechanisms may also play a role. Recent technological developments for measuring complex sphingolipids in biological samples, together with the availability of Cerk-deficient animals as well as potent CERK inhibitors, have now provided new grounds for investigating C1P biology further. Here, we will review the current understanding of CERK and C1P in terms of biochemistry and functional implications, with particular attention to C1P produced by CERK., (Copyright © 2010 Elsevier Inc. All rights reserved.)
- Published
- 2011
- Full Text
- View/download PDF
44. Transcriptional repression of ceramide kinase in LPS-challenged macrophages.
- Author
-
Rovina P, Jaritz M, and Bornancin F
- Subjects
- Animals, Cell Line, Down-Regulation, Macrophage Activation, Macrophages enzymology, Mice, Mice, Inbred BALB C, Gene Expression Regulation, Lipopolysaccharides immunology, Macrophages immunology, Phosphotransferases (Alcohol Group Acceptor) genetics, Transcription, Genetic
- Abstract
Ceramide kinase (CERK) produces the bioactive lipid ceramide 1-phosphate (C1P). Both CERK and C1P have been identified as mediators of cell growth and survival. Recent evidence showed that CERK is down-regulated during M1-type macrophage activation, which is known to promote cell growth arrest. However, the mechanism has not been investigated yet and, in particular, whether growth arrest might be the signal for down-regulation of CERK is currently unknown. Here, we found that LPS-mediated TLR-4 engagement reduces Cerk mRNA levels in mouse primary macrophages. Reporter gene assays in RAW264.7 macrophages showed that LPS inhibits the transcriptional activity of the Cerk proximal promoter. The G1-cell cycle blocker mimosine did not inhibit Cerk transcription, suggesting that transcriptional repression of Cerk by LPS is not a primary consequence of LPS-induced cell cycle blockade., (Copyright © 2010 Elsevier Inc. All rights reserved.)
- Published
- 2010
- Full Text
- View/download PDF
45. Cleavage by MALT1 induces cytosolic release of A20.
- Author
-
Malinverni C, Unterreiner A, Staal J, Demeyer A, Galaup M, Luyten M, Beyaert R, and Bornancin F
- Subjects
- Cell Line, Tumor, DNA-Binding Proteins, Humans, Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein, Peptide Fragments analysis, Peptide Fragments metabolism, Protein Transport, Tumor Necrosis Factor alpha-Induced Protein 3, Caspases metabolism, Cytosol enzymology, Intracellular Signaling Peptides and Proteins metabolism, Neoplasm Proteins metabolism, Nuclear Proteins metabolism
- Abstract
The MALT1 paracaspase has arginine-directed proteolytic activity. A20 is a dual ubiquitin-editing enzyme involved in termination of NF-κB signaling. Upon T- or B-cell receptor engagement human (h) A20 is cleaved by MALT1 after arginine 439, yielding an N-terminal fragment (hA20p50) and a C-terminal one (hA20p37). The hA20p50 fragment has never been detected directly, thus limiting insight into the functional consequences of MALT1-mediated cleavage of A20. Here, various antibodies were tested, including newly generated hA20p50 and hA20p37 specific antibodies, leading to detection of the hA20p50 fragment produced after MALT1-mediated cleavage of ectopically expressed as well as endogenous A20 proteins. The properties of both A20 fragments, generated upon co-expression with a constitutively active MALT1 protein, were further studied by sub-cellular fractionation and fluorescence microscopy. In contrast to full-length A20 which is particulate and insoluble, we found hA20p50 to be soluble and readily released into the cytosol whereas hA20p37 was partially soluble, thus suggesting loss of compartmentalization as a possible mechanism for MALT1-mediated dampening of A20 function., (Copyright © 2010 Elsevier Inc. All rights reserved.)
- Published
- 2010
- Full Text
- View/download PDF
46. Modulation of ceramide metabolism in mouse primary macrophages.
- Author
-
Rovina P, Graf C, and Bornancin F
- Subjects
- Animals, Dexamethasone pharmacology, Lipopolysaccharides immunology, Macrophages drug effects, Macrophages enzymology, Mice, Mice, Inbred BALB C, Ceramides metabolism, Macrophage Activation, Macrophages immunology, Phosphotransferases (Alcohol Group Acceptor) metabolism
- Abstract
Ceramide kinase (CERK) produces the bioactive lipid ceramide 1-phosphate (C1P) and is, together with glucosylceramide synthase (GCS) and sphingomyelin synthases (SMS-1 and -2), a key regulator of ceramide metabolism. Here, we used a previously validated assay for measuring CERK, GCS, and SMS activities simultaneously, to study the regulation of ceramide metabolism in mouse macrophages. Elicitation of peritoneal macrophages as well as differentiation of bone marrow-derived monocytes into macrophages led to "ceramide anabolic switching" by re-directing ceramide anabolism towards C1P synthesis by CERK. In contrast, macrophage activation by lipopolysaccharide (LPS) evoked a "ceramide anabolic switch" going in the opposite direction, i.e. featuring up-regulation of GCS and SMS and down-regulation of CERK. The LPS effects were partially blocked by dexamethasone, a known macrophage de-activator. Altogether, the data reveal a contrasting regulation of ceramide metabolism enzymes during macrophage biological responses., (Copyright 2010 Elsevier Inc. All rights reserved.)
- Published
- 2010
- Full Text
- View/download PDF
47. Ovalbumin-induced plasma interleukin-4 levels are reduced in ceramide kinase-deficient DO11.10 RAG1-/- mice.
- Author
-
Niwa S, Urtz N, Baumruker T, Billich A, and Bornancin F
- Subjects
- Animals, Asthma genetics, Asthma metabolism, Bronchoalveolar Lavage Fluid, Gene Expression Regulation, Enzymologic, Homeodomain Proteins genetics, Interferon-gamma metabolism, Mice, Mice, Transgenic, Spleen cytology, T-Lymphocytes cytology, Th2 Cells metabolism, Tumor Necrosis Factor-alpha metabolism, Interleukin-4 blood, Ovalbumin metabolism, Phosphotransferases (Alcohol Group Acceptor) genetics
- Abstract
Ceramide kinase (CERK) produces the bioactive lipid ceramide-1-phosphate (C1P) and is a key regulator of ceramide and dihydroceramide levels. It is likely that CERK and C1P play a role in inflammatory processes but the cells involved and the mechanisms used remain to be clarified. In particular, the impact of CERK on T-cell biology has not been studied so far. Here, we used Cerk-/- mice backcrossed with DO11.10/RAG1-/- mice to probe the effect of CERK ablation on T-cell activation. Levels of interleukin (IL)-2, IL-4, IL-5, IL-13, of tumor necrosis factor (TNF)-alpha, and of interferon (INF)-gamma were recorded following ovalbumin challenge in vivo and using ovalbumin-treated splenocytes ex- vivo. Absence of CERK led to a significant decrease in the production of IL-4, thus suggesting that CERK may polarize T cells towards the TH2 cell subtype. However, the importance of CERK to TH2 cell biology will have to be investigated further because in a model of asthma, which is TH2-cell driven, Cerk-/- mice responded like wild-type animals.
- Published
- 2010
- Full Text
- View/download PDF
48. Ceramide kinase profiling by mass spectrometry reveals a conserved phosphorylation pattern downstream of the catalytic site.
- Author
-
Chen WQ, Graf C, Zimmel D, Rovina P, Krapfenbauer K, Jaritz M, Parker PJ, Lubec G, and Bornancin F
- Subjects
- Amino Acid Sequence, Animals, COS Cells, Catalytic Domain, Chlorocebus aethiops, Humans, Molecular Sequence Data, Phosphoproteins genetics, Phosphoproteins metabolism, Phosphorylation, Phosphotransferases (Alcohol Group Acceptor) chemistry, Phosphotransferases (Alcohol Group Acceptor) genetics, Point Mutation, Recombinant Proteins chemistry, Recombinant Proteins genetics, Recombinant Proteins metabolism, Sequence Alignment, Serine metabolism, Mass Spectrometry methods, Phosphotransferases (Alcohol Group Acceptor) metabolism, Protein Processing, Post-Translational
- Abstract
Ceramide kinase (CERK) is essential for production of ceramide-1-phosphate (C1P), a bioactive lipid whose formation critically modulates ceramide levels. To explore how CERK is regulated, we used insect cell-expressed, recombinant hCERK and searched for post-translational modifications, using mass-spectrometry techniques. This led to identification of two phosphorylated serine residues, at positions 340 and 408. Point mutations preventing phosphorylation at either of these sites did not lead to detectable changes in subcellular localization or activity. However, preventing phosphorylation at S340 resulted in CERK instability as revealed by the behavior of the S340A mutant protein under various assay conditions in vitro. Phosphorylation of a cognate serine residue in sphingosine kinases was previously shown to be important. Therefore, phosphorylation within a conserved "regulation loop" downstream of the catalytic domain emerges as a new paradigm for regulation of kinases of the diacylglycerol kinase family. This "regulation loop" is reminiscent of the "activation loop" that controls AGC protein kinases, being a similar distance from the critical ATP binding site determinants in the primary sequence.
- Published
- 2010
- Full Text
- View/download PDF
49. 4,5,6-Trisubstituted piperidinones as conformationally restricted ceramide analogues: synthesis and evaluation as inhibitors of sphingosine and ceramide kinases and as NKT cell-stimulatory antigens.
- Author
-
Mathew T, Cavallari M, Billich A, Bornancin F, Nussbaumer P, De Libero G, and Vasella A
- Subjects
- Antigens, CD1d immunology, Antigens, CD1d metabolism, Ceramides metabolism, Ceramides pharmacology, Cytokines metabolism, Enzyme Inhibitors chemistry, Enzyme Inhibitors pharmacology, Humans, Lipid Metabolism immunology, Lipid Metabolism physiology, Models, Chemical, Molecular Conformation, Natural Killer T-Cells cytology, Natural Killer T-Cells immunology, Phosphotransferases (Alcohol Group Acceptor) metabolism, Piperidines chemical synthesis, Receptors, Antigen, T-Cell immunology, Receptors, Antigen, T-Cell metabolism, Ceramides chemistry, Enzyme Inhibitors chemical synthesis, Natural Killer T-Cells drug effects, Phosphotransferases (Alcohol Group Acceptor) antagonists & inhibitors, Piperidines chemistry, Piperidines pharmacology
- Abstract
The conformationally based piperidinone sphingosine analogues 7, 8, 15, and 16 were synthesized from allylic alcohol 34 via lactams 31 and 32. The L-arabino diol 7 and the L-ribo diol 8 were transformed into the amino alcohols 17-24. The L-gluco ceramide analogues 43, 46a, and 47, and the L-altro ceramide analogues 51a and 52 were synthesized from either 31 or 32. The L-ribo diols 8 and 16, and the amino alcohols 19 and 20 inhibit sphingosine kinase 1 (SPHK1), while the L-arabino analogues 7, 15, 17, and 18 are inactive. The L-arabino and the L-ribo dimethylamines 21-24, the L-gluco ceramide analogues 43, 46a, and 47, and the L-altro ceramide analogues 51a and 52 did not block SPHK1. Neither the L-arabino diol 7 nor the L-ribo diol 8 inhibited SPHK2 or ceramide kinase. The L-arabino diols 7 and 15 stimulate invariant natural killer T (iNKT) cells when presented by living antigen-presenting cells (APC) and also by plate-bound human CD1d, whereas the L-ribo diols 8 and 16, the L-arabino amino alcohols 17-18, and the dimethylamines 21-22 did not activate iNKT cells. The L-gluco ceramide analogues 43, 46a, and 47 had strongly stimulatory effects on iNKT cells when presented by living APC and also by plate-bound human CD1d, whereas the L-altro ceramide analogue 52 activated only weakly. All activatory compounds induced preferentially the release of pro-inflammatory cytokines, indicating the formation of a stable CD1d--lipid--T-cell receptor complex.
- Published
- 2009
- Full Text
- View/download PDF
50. Subcellular localization of ceramide kinase and ceramide kinase-like protein requires interplay of their Pleckstrin Homology domain-containing N-terminal regions together with C-terminal domains.
- Author
-
Rovina P, Schanzer A, Graf C, Mechtcheriakova D, Jaritz M, and Bornancin F
- Subjects
- Amino Acid Motifs, Amino Acid Sequence, Cell Nucleolus enzymology, Conserved Sequence, Humans, Molecular Sequence Data, Protein Structure, Tertiary, Protein Transport, Retinal Degeneration enzymology, Retinal Degeneration pathology, Sequence Alignment, Sequence Deletion, Structure-Activity Relationship, Subcellular Fractions enzymology, Phosphotransferases (Alcohol Group Acceptor) chemistry, Phosphotransferases (Alcohol Group Acceptor) metabolism
- Abstract
Ceramide kinase (CERK) and the ceramide kinase-like protein (CERKL), two related members of the diacylglycerol kinase family, are ill-defined at the molecular level. In particular, what determines their distinctive subcellular localization is not well understood. Here we show that the Pleckstrin Homology (PH) domain of CERK, which is required for Golgi complex localization, can substitute for the N-terminal region of CERKL and allow for wild-type CERKL localization, which is typified by nucleolar accumulation. This demonstrates that determinants for localization of these two enzymes do not lie solely in their PH domain-containing N-terminal regions. Moreover, we present evidence for a previously unrecognized participation of CERK distal sequences in structural stability, localization and activity of the full-length protein. Progressive deletion of CERK and CERKL from the C-terminus revealed similar sequential organization in both proteins, with nuclear import signals in their N-terminal part, and nuclear export signals in their C-terminal part. Furthermore, mutagenesis of individual cysteine residues of a CERK-specific CXXXCXXC motif severely compromised both exportation of CERK from the nucleus and its association with the Golgi complex. Altogether, this work identifies conserved domains in CERK and CERKL as well as new determinants for their subcellular localization. It further suggests a nucleocytoplasmic shuttling mechanism for both proteins that may be defective in CERKL mutant proteins responsible for retinal degenerative diseases.
- Published
- 2009
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.