1
|
Brvar M, O'Neill TJ, Plettenburg O, Krappmann D. An updated patent review of MALT1 inhibitors (2021-present). Expert Opin Ther Pat 2025:1-18. [PMID: 40209204 DOI: 10.1080/13543776.2025.2484371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Revised: 03/01/2025] [Accepted: 03/21/2025] [Indexed: 04/12/2025]
Abstract
INTRODUCTION MALT1 paracaspase acts as a molecular scaffold and a proteolytic enzyme in immune cells. MALT1 has emerged as a promising drug target for cancer therapy, and especially for targeting MALT1 in aggressive lymphomas. Drug discovery programs have yielded potent and selective MALT1 protease inhibitors. First-in-class MALT1 inhibitors have been moved to early clinical trials to evaluate safety and efficacy. AREAS COVERED This review will provide an update regarding the mode of action, the chemical space and therapeutic use of MALT1 inhibitors based on recent patents and the scientific literature (05/2021-12/2024). EXPERT OPINION Allosteric inhibition is the preferred mode of action to inhibit the MALT1 protease. Chemical advances largely focus on improving binding and inhibition in the allosteric site of MALT1. New composition of matter has been generated, but a clinical proof for the safety and efficacy of allosteric MALT1 inhibitors is still pending. We still lack potent and selective competitive or covalent MALT1 inhibitors, indicating the challenges with targeting the active site. Further, MALT1 protein degraders and MALT1 scaffolding inhibitors have been developed, which may have distinct inhibitory profiles compared to allosteric MALT1 protease inhibitors, but more potent and selective compounds are needed to judge the feasibility and usefulness of these approaches.
Collapse
Affiliation(s)
- Matjaz Brvar
- Institute for Medicinal Chemistry, Molecular Targets and Therapeutics Center, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg, Germany
- Centre of Biomolecular Drug Research (BMWZ), Institute of Organic Chemistry, Leibniz-University Hannover, Hannover, Germany
| | - Thomas J O'Neill
- Research Unit Signaling and Translation, Group Signaling and Immunity, Molecular Targets and Therapeutics Center, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| | - Oliver Plettenburg
- Institute for Medicinal Chemistry, Molecular Targets and Therapeutics Center, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg, Germany
- Centre of Biomolecular Drug Research (BMWZ), Institute of Organic Chemistry, Leibniz-University Hannover, Hannover, Germany
- Laboratory of Nano- and Quantum Engineering, Leibniz University Hannover, Hannover, Germany
- Medicinal Chemistry and Drug Design, Institute of Lung Health (ILH), Gießen, Germany
| | - Daniel Krappmann
- Research Unit Signaling and Translation, Group Signaling and Immunity, Molecular Targets and Therapeutics Center, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
- Faculty of Biology, Ludwig-Maximilians-Universität München, Planegg-Martinsried, Germany
| |
Collapse
|
2
|
Dery KJ, Wong Z, Wei M, Kupiec-Weglinski JW. Mechanistic Insights into Alternative Gene Splicing in Oxidative Stress and Tissue Injury. Antioxid Redox Signal 2024; 41:890-909. [PMID: 37776178 PMCID: PMC11631805 DOI: 10.1089/ars.2023.0437] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Accepted: 09/17/2023] [Indexed: 10/01/2023]
Abstract
Significance: Oxidative stress (OS) and inflammation are inducers of tissue injury. Alternative splicing (AS) is an essential regulatory step for diversifying the eukaryotic proteome. Human diseases link AS to OS; however, the underlying mechanisms must be better understood. Recent Advances: Genome‑wide profiling studies identify new differentially expressed genes induced by OS-dependent ischemia/reperfusion injury. Overexpression of RNA-binding protein RBFOX1 protects against inflammation. Hypoxia-inducible factor-1α directs polypyrimidine tract binding protein 1 to regulate mouse carcinoembryonic antigen-related cell adhesion molecule 1 (Ceacam1) AS under OS conditions. Heterogeneous nuclear ribonucleoprotein L variant 1 contains an RGG/RG motif that coordinates with transcription factors to influence human CEACAM1 AS. Hypoxia intervention involving short interfering RNAs directed to long-noncoding RNA 260 polarizes M2 macrophages toward an anti-inflammatory phenotype and alleviates OS by inhibiting IL-28RA gene AS. Critical Issues: Protective mechanisms that eliminate reactive oxygen species (ROS) are important for resolving imbalances that lead to chronic inflammation. Defects in AS can cause ROS generation, cell death regulation, and the activation of innate and adaptive immune factors. We propose that AS pathways link redox regulation to the activation or suppression of the inflammatory response during cellular stress. Future Directions: Emergent studies using molecule-mediated RNA splicing are being conducted to exploit the immunogenicity of AS protein products. Deciphering the mechanisms that connect misspliced OS and pathologies should remain a priority. Controlled release of RNA directly into cells with clinical applications is needed as the demand for innovative nucleic acid delivery systems continues to be demonstrated.
Collapse
Affiliation(s)
- Kenneth J. Dery
- The Dumont-UCLA Transplantation Center, Division of Liver and Pancreas Transplantation, Department of Surgery, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Zeriel Wong
- The Dumont-UCLA Transplantation Center, Division of Liver and Pancreas Transplantation, Department of Surgery, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Megan Wei
- The Dumont-UCLA Transplantation Center, Division of Liver and Pancreas Transplantation, Department of Surgery, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Jerzy W. Kupiec-Weglinski
- The Dumont-UCLA Transplantation Center, Division of Liver and Pancreas Transplantation, Department of Surgery, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| |
Collapse
|
3
|
Moud BN, Ober F, O’Neill TJ, Krappmann D. MALT1 substrate cleavage: what is it good for? Front Immunol 2024; 15:1412347. [PMID: 38863711 PMCID: PMC11165066 DOI: 10.3389/fimmu.2024.1412347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Accepted: 05/07/2024] [Indexed: 06/13/2024] Open
Abstract
CARD-BCL10-MALT1 (CBM) signalosomes connect distal signaling of innate and adaptive immune receptors to proximal signaling pathways and immune activation. Four CARD scaffold proteins (CARD9, 10, 11, 14) can form seeds that nucleate the assembly of BCL10-MALT1 filaments in a cell- and stimulus-specific manner. MALT1 (also known as PCASP1) serves a dual function within the assembled CBM complexes. By recruiting TRAF6, MALT1 acts as a molecular scaffold that initiates IκB kinase (IKK)/NF-κB and c-Jun N-terminal kinase (JNK)/AP-1 signaling. In parallel, proximity-induced dimerization of the paracaspase domain activates the MALT1 protease which exerts its function by cleaving a set of specific substrates. While complete MALT1 ablation leads to immune deficiency, selective destruction of either scaffolding or protease function provokes autoimmune inflammation. Thus, balanced MALT1-TRAF6 recruitment and MALT1 substrate cleavage are critical to maintain immune homeostasis and to promote optimal immune activation. Further, MALT1 protease activity drives the survival of aggressive lymphomas and other non-hematologic solid cancers. However, little is known about the relevance of the cleavage of individual substrates for the pathophysiological functions of MALT1. Unbiased serendipity, screening and computational predictions have identified and validated ~20 substrates, indicating that MALT1 targets a quite distinct set of proteins. Known substrates are involved in CBM auto-regulation (MALT1, BCL10 and CARD10), regulation of signaling and adhesion (A20, CYLD, HOIL-1 and Tensin-3), or transcription (RelB) and mRNA stability/translation (Regnase-1, Roquin-1/2 and N4BP1), indicating that MALT1 often targets multiple proteins involved in similar cellular processes. Here, we will summarize what is known about the fate and functions of individual MALT1 substrates and how their cleavage contributes to the biological functions of the MALT1 protease. We will outline what is needed to better connect critical pathophysiological roles of the MALT1 protease with the cleavage of distinct substrates.
Collapse
Affiliation(s)
| | | | | | - Daniel Krappmann
- Research Unit Signaling and Translation, Group Signaling and Immunity, Molecular Targets and Therapeutics Center, Helmholtz Zentrum München – German Research Center for Environmental Health, Neuherberg, Germany
| |
Collapse
|
4
|
Giehler F, Ostertag MS, Sommermann T, Weidl D, Sterz KR, Kutz H, Moosmann A, Feller SM, Geerlof A, Biesinger B, Popowicz GM, Kirchmair J, Kieser A. Epstein-Barr virus-driven B cell lymphoma mediated by a direct LMP1-TRAF6 complex. Nat Commun 2024; 15:414. [PMID: 38195569 PMCID: PMC10776578 DOI: 10.1038/s41467-023-44455-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 12/14/2023] [Indexed: 01/11/2024] Open
Abstract
Epstein-Barr virus (EBV) latent membrane protein 1 (LMP1) drives viral B cell transformation and oncogenesis. LMP1's transforming activity depends on its C-terminal activation region 2 (CTAR2), which induces NF-κB and JNK by engaging TNF receptor-associated factor 6 (TRAF6). The mechanism of TRAF6 recruitment to LMP1 and its role in LMP1 signalling remains elusive. Here we demonstrate that TRAF6 interacts directly with a viral TRAF6 binding motif within CTAR2. Functional and NMR studies supported by molecular modeling provide insight into the architecture of the LMP1-TRAF6 complex, which differs from that of CD40-TRAF6. The direct recruitment of TRAF6 to LMP1 is essential for NF-κB activation by CTAR2 and the survival of LMP1-driven lymphoma. Disruption of the LMP1-TRAF6 complex by inhibitory peptides interferes with the survival of EBV-transformed B cells. In this work, we identify LMP1-TRAF6 as a critical virus-host interface and validate this interaction as a potential therapeutic target in EBV-associated cancer.
Collapse
Affiliation(s)
- Fabian Giehler
- Research Unit Signaling and Translation, Helmholtz Center Munich - German Research Center for Environmental Health, 85764, Neuherberg, Germany
- Research Unit Gene Vectors, Helmholtz Center Munich - German Research Center for Environmental Health, 81377, Munich, Germany
- German Center for Infection Research (DZIF), Partner Site Munich, Munich, Germany
| | - Michael S Ostertag
- Institute of Structural Biology, Helmholtz Center Munich - German Research Center for Environmental Health, 85764, Neuherberg, Germany
| | - Thomas Sommermann
- Immune Regulation and Cancer, Max Delbrück Center for Molecular Medicine, 13125, Berlin, Germany
| | - Daniel Weidl
- Institute of Clinical and Molecular Virology, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nuremberg, 91054, Erlangen, Germany
| | - Kai R Sterz
- Research Unit Gene Vectors, Helmholtz Center Munich - German Research Center for Environmental Health, 81377, Munich, Germany
| | - Helmut Kutz
- Research Unit Gene Vectors, Helmholtz Center Munich - German Research Center for Environmental Health, 81377, Munich, Germany
| | - Andreas Moosmann
- Research Unit Gene Vectors, Helmholtz Center Munich - German Research Center for Environmental Health, 81377, Munich, Germany
- German Center for Infection Research (DZIF), Partner Site Munich, Munich, Germany
- Department of Medicine III, University Hospital, Ludwig-Maximilians-University Munich, 81377, Munich, Germany
| | - Stephan M Feller
- Institute of Molecular Medicine, Martin-Luther-University Halle-Wittenberg, 06120, Halle, Germany
| | - Arie Geerlof
- Institute of Structural Biology, Helmholtz Center Munich - German Research Center for Environmental Health, 85764, Neuherberg, Germany
| | - Brigitte Biesinger
- Institute of Clinical and Molecular Virology, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nuremberg, 91054, Erlangen, Germany
| | - Grzegorz M Popowicz
- Institute of Structural Biology, Helmholtz Center Munich - German Research Center for Environmental Health, 85764, Neuherberg, Germany
| | - Johannes Kirchmair
- Universität Hamburg, Department of Informatics, Center for Bioinformatics (ZBH), 20146, Hamburg, Germany
- Department of Pharmaceutical Sciences, Division of Pharmaceutical Chemistry, University of Vienna, 1090, Vienna, Austria
| | - Arnd Kieser
- Research Unit Signaling and Translation, Helmholtz Center Munich - German Research Center for Environmental Health, 85764, Neuherberg, Germany.
- Research Unit Gene Vectors, Helmholtz Center Munich - German Research Center for Environmental Health, 81377, Munich, Germany.
- German Center for Infection Research (DZIF), Partner Site Munich, Munich, Germany.
| |
Collapse
|
5
|
Juilland M, Alouche N, Ubezzi I, Gonzalez M, Rashid HO, Scarpellino L, Erdmann T, Grau M, Lenz G, Luther SA, Thome M. Identification of Tensin-3 as a MALT1 substrate that controls B cell adhesion and lymphoma dissemination. Proc Natl Acad Sci U S A 2023; 120:e2301155120. [PMID: 38109544 PMCID: PMC10756297 DOI: 10.1073/pnas.2301155120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 10/24/2023] [Indexed: 12/20/2023] Open
Abstract
The protease MALT1 promotes lymphocyte activation and lymphomagenesis by cleaving a limited set of cellular substrates, most of which control gene expression. Here, we identified the integrin-binding scaffold protein Tensin-3 as a MALT1 substrate in activated human B cells. Activated B cells lacking Tensin-3 showed decreased integrin-dependent adhesion but exhibited comparable NF-κB1 and Jun N-terminal kinase transcriptional responses. Cells expressing a noncleavable form of Tensin-3, on the other hand, showed increased adhesion. To test the role of Tensin-3 cleavage in vivo, mice expressing a noncleavable version of Tensin-3 were generated, which showed a partial reduction in the T cell-dependent B cell response. Interestingly, human diffuse large B cell lymphomas and mantle cell lymphomas with constitutive MALT1 activity showed strong constitutive Tensin-3 cleavage and a decrease in uncleaved Tensin-3 levels. Moreover, silencing of Tensin-3 expression in MALT1-driven lymphoma promoted dissemination of xenografted lymphoma cells to the bone marrow and spleen. Thus, MALT1-dependent Tensin-3 cleavage reveals a unique aspect of the function of MALT1, which negatively regulates integrin-dependent B cell adhesion and facilitates metastatic spread of B cell lymphomas.
Collapse
Affiliation(s)
- Mélanie Juilland
- Department of Immunobiology, University of Lausanne, EpalingesCH-1066, Switzerland
| | - Nagham Alouche
- Department of Immunobiology, University of Lausanne, EpalingesCH-1066, Switzerland
| | - Ivana Ubezzi
- Department of Immunobiology, University of Lausanne, EpalingesCH-1066, Switzerland
| | - Montserrat Gonzalez
- Department of Immunobiology, University of Lausanne, EpalingesCH-1066, Switzerland
| | - Harun-Or Rashid
- Department of Immunobiology, University of Lausanne, EpalingesCH-1066, Switzerland
| | - Leonardo Scarpellino
- Department of Immunobiology, University of Lausanne, EpalingesCH-1066, Switzerland
| | - Tabea Erdmann
- Department of Medicine A for Hematology, Oncology and Pneumology, University Hospital Münster, MünsterD-48149, Germany
| | - Michael Grau
- Department of Medicine A for Hematology, Oncology and Pneumology, University Hospital Münster, MünsterD-48149, Germany
| | - Georg Lenz
- Department of Medicine A for Hematology, Oncology and Pneumology, University Hospital Münster, MünsterD-48149, Germany
| | - Sanjiv A. Luther
- Department of Immunobiology, University of Lausanne, EpalingesCH-1066, Switzerland
| | - Margot Thome
- Department of Immunobiology, University of Lausanne, EpalingesCH-1066, Switzerland
| |
Collapse
|
6
|
Wimberger N, Ober F, Avar G, Grau M, Xu W, Lenz G, Menden MP, Krappmann D. Oncogene-induced MALT1 protease activity drives posttranscriptional gene expression in malignant lymphomas. Blood 2023; 142:1985-2001. [PMID: 37623434 PMCID: PMC10733837 DOI: 10.1182/blood.2023021299] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 07/13/2023] [Accepted: 07/26/2023] [Indexed: 08/26/2023] Open
Abstract
Constitutive mucosa-associated lymphoid tissue lymphoma translocation protein 1 (MALT1) activity drives survival of malignant lymphomas addicted to chronic B-cell receptor signaling, oncogenic CARD11, or the API2-MALT1 (also BIRC3::MALT1) fusion oncoprotein. Although MALT1 scaffolding induces NF-κB-dependent survival signaling, MALT1 protease function is thought to augment NF-κB activation by cleaving signaling mediators and transcriptional regulators in B-cell lymphomas. However, the pathological role of MALT1 protease function in lymphomagenesis is not well understood. Here, we show that TRAF6 controls MALT1-dependent activation of NF-κB transcriptional responses but is dispensable for MALT1 protease activation driven by oncogenic CARD11. To uncouple enzymatic and nonenzymatic functions of MALT1, we analyzed TRAF6-dependent and -independent as well as MALT1 protease-dependent gene expression profiles downstream of oncogenic CARD11 and API2-MALT1. The data suggest that by cleaving and inactivating the RNA binding proteins Regnase-1 and Roquin-1/2, MALT1 protease induces posttranscriptional upregulation of many genes including NFKBIZ/IκBζ, NFKBID/IκBNS, and ZC3H12A/Regnase-1 in activated B-cell-like diffuse large B-cell lymphoma (ABC DLBCL). We demonstrate that oncogene-driven MALT1 activity in ABC DLBCL cells regulates NFKBIZ and NFKBID induction on an mRNA level via releasing a brake imposed by Regnase-1 and Roquin-1/2. Furthermore, MALT1 protease drives posttranscriptional gene induction in the context of the API2-MALT1 fusion created by the recurrent t(11;18)(q21;q21) translocation in MALT lymphoma. Thus, MALT1 paracaspase acts as a bifurcation point for enhancing transcriptional and posttranscriptional gene expression in malignant lymphomas. Moreover, the identification of MALT1 protease-selective target genes provides specific biomarkers for the clinical evaluation of MALT1 inhibitors.
Collapse
Affiliation(s)
- Nicole Wimberger
- Research Unit Signaling and Translation, Group Signaling and Immunity, Molecular Targets Therapeutic Center, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Franziska Ober
- Research Unit Signaling and Translation, Group Signaling and Immunity, Molecular Targets Therapeutic Center, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Göksu Avar
- Department of Computational Health, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Department of Biology, Ludwig Maximilian University Munich, Martinsried, Germany
| | - Michael Grau
- Department of Medicine A, Hematology, Oncology, and Pneumology, University Hospital Muenster, Muenster, Germany
| | - Wendan Xu
- Department of Medicine A, Hematology, Oncology, and Pneumology, University Hospital Muenster, Muenster, Germany
| | - Georg Lenz
- Department of Medicine A, Hematology, Oncology, and Pneumology, University Hospital Muenster, Muenster, Germany
| | - Michael P. Menden
- Department of Computational Health, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Department of Biology, Ludwig Maximilian University Munich, Martinsried, Germany
- Department of Biochemistry and Pharmacology, The University of Melbourne, Melbourne, Australia
| | - Daniel Krappmann
- Research Unit Signaling and Translation, Group Signaling and Immunity, Molecular Targets Therapeutic Center, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Department of Biology, Ludwig Maximilian University Munich, Martinsried, Germany
| |
Collapse
|
7
|
O'Neill TJ, Tofaute MJ, Krappmann D. Function and targeting of MALT1 paracaspase in cancer. Cancer Treat Rev 2023; 117:102568. [PMID: 37126937 DOI: 10.1016/j.ctrv.2023.102568] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 04/18/2023] [Accepted: 04/21/2023] [Indexed: 05/03/2023]
Abstract
The paracaspase MALT1 has emerged as a key regulator of immune signaling, which also promotes tumor development by both cancer cell-intrinsic and -extrinsic mechanisms. As an integral subunit of the CARD11-BCL10-MALT1 (CBM) signaling complex, MALT1 has an intriguing dual function in lymphocytes. MALT1 acts as a scaffolding protein to drive activation of NF-κB transcription factors and as a protease to modulate signaling and immune activation by cleavage of distinct substrates. Aberrant MALT1 activity is critical for NF-κB-dependent survival and proliferation of malignant cancer cells, which is fostered by paracaspase-catalyzed inactivation of negative regulators of the canonical NF-κB pathway like A20, CYLD and RelB. Specifically, B cell receptor-addicted lymphomas rely strongly on this cancer cell-intrinsic MALT1 protease function, but also survival, proliferation and metastasis of certain solid cancers is sensitive to MALT1 inhibition. Beyond this, MALT1 protease exercises a cancer cell-extrinsic role by maintaining the immune-suppressive function of regulatory T (Treg) cells in the tumor microenvironment (TME). MALT1 inhibition is able to convert immune-suppressive to pro-inflammatory Treg cells in the TME of solid cancers, thereby eliciting a robust anti-tumor immunity that can augment the effects of checkpoint inhibitors. Therefore, the cancer cell-intrinsic and -extrinsic tumor promoting MALT1 protease functions offer unique therapeutic opportunities, which has motivated the development of potent and selective MALT1 inhibitors currently under pre-clinical and clinical evaluation.
Collapse
Affiliation(s)
- Thomas J O'Neill
- Research Unit Signaling and Translation, Group Signaling and Immunity, Molecular Targets and Therapeutics Center, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| | - Marie J Tofaute
- Research Unit Signaling and Translation, Group Signaling and Immunity, Molecular Targets and Therapeutics Center, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| | - Daniel Krappmann
- Research Unit Signaling and Translation, Group Signaling and Immunity, Molecular Targets and Therapeutics Center, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany.
| |
Collapse
|
8
|
O’Neill TJ, Gewies A, Seeholzer T, Krappmann D. TRAF6 controls T cell homeostasis by maintaining the equilibrium of MALT1 scaffolding and protease functions. Front Immunol 2023; 14:1111398. [PMID: 36761777 PMCID: PMC9902345 DOI: 10.3389/fimmu.2023.1111398] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 01/10/2023] [Indexed: 01/26/2023] Open
Abstract
MALT1 is a core component of the CARD11-BCL10-MALT1 (CBM) signalosome, in which it acts as a scaffold and a protease to bridge T cell receptor (TCR) ligation to immune activation. As a scaffold, MALT1 binds to TRAF6, and T cell-specific TRAF6 ablation or destruction of MALT1-TRAF6 interaction provokes activation of conventional T (Tconv) effector cells. In contrast, MALT1 protease activity controls the development and suppressive function of regulatory T (Treg) cells in a T cell-intrinsic manner. Thus, complete loss of TRAF6 or selective inactivation of MALT1 catalytic function in mice skews the immune system towards autoimmune inflammation, but distinct mechanisms are responsible for these immune disorders. Here we demonstrate that TRAF6 deletion or MALT1 paracaspase inactivation are highly interdependent in causing the distinct immune pathologies. We crossed mice with T cell-specific TRAF6 ablation (Traf6-ΔT) and mice with a mutation rendering the MALT1 paracaspase dead in T cells (Malt1 PD-T) to yield Traf6-ΔT;Malt1 PD-T double mutant mice. These mice reveal that the autoimmune inflammation caused by TRAF6-ablation relies strictly on the function of the MALT1 protease to drive the activation of Tconv cells. Vice versa, despite the complete loss of Treg cells in Traf6-ΔT;Malt1 PD-T double mutant mice, inactivation of the MALT1 protease is unable to cause autoinflammation, because the Tconv effector cells are not activated in the absence of TRAF6. Consequentially, combined MALT1 paracaspase inactivation and TRAF6 deficiency in T cells mirrors the immunodeficiency seen upon T cell-specific MALT1 ablation.
Collapse
|
9
|
Jones AN, Graß C, Meininger I, Geerlof A, Klostermann M, Zarnack K, Krappmann D, Sattler M. Modulation of pre-mRNA structure by hnRNP proteins regulates alternative splicing of MALT1. SCIENCE ADVANCES 2022; 8:eabp9153. [PMID: 35921415 PMCID: PMC9348792 DOI: 10.1126/sciadv.abp9153] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/02/2023]
Abstract
Alternative splicing plays key roles for cell type-specific regulation of protein function. It is controlled by cis-regulatory RNA elements that are recognized by RNA binding proteins (RBPs). The MALT1 paracaspase is a key factor of signaling pathways that mediate innate and adaptive immune responses. Alternative splicing of MALT1 is critical for controlling optimal T cell activation. We demonstrate that MALT1 splicing depends on RNA structural elements that sequester the splice sites of the alternatively spliced exon7. The RBPs hnRNP U and hnRNP L bind competitively to stem-loop RNA structures that involve the 5' and 3' splice sites flanking exon7. While hnRNP U stabilizes RNA stem-loop conformations that maintain exon7 skipping, hnRNP L disrupts these RNA elements to facilitate recruitment of the essential splicing factor U2AF2, thereby promoting exon7 inclusion. Our data represent a paradigm for the control of splice site selection by differential RBP binding and modulation of pre-mRNA structure.
Collapse
Affiliation(s)
- Alisha N. Jones
- Institute of Structural Biology, Molecular Targets and Therapeutics Center, Helmholtz Zentrum München, Neuherberg, 85764 München, Germany
- Bavarian NMR Center, Department of Chemistry, Technical University of Munich, Garching, 85748 München, Germany
| | - Carina Graß
- Research Unit Cellular Signal Integration, Institute of Molecular Toxicology and Pharmacology, Molecular Targets and Therapeutics Center, Helmholtz Zentrum München, Neuherberg, 85764 München, Germany
| | - Isabel Meininger
- Research Unit Cellular Signal Integration, Institute of Molecular Toxicology and Pharmacology, Molecular Targets and Therapeutics Center, Helmholtz Zentrum München, Neuherberg, 85764 München, Germany
| | - Arie Geerlof
- Institute of Structural Biology, Molecular Targets and Therapeutics Center, Helmholtz Zentrum München, Neuherberg, 85764 München, Germany
| | - Melina Klostermann
- Buchmann Institute for Molecular Life Sciences (BMLS) & Faculty of Biological Sciences, Goethe University Frankfurt, Max-von-Laue-Str. 15, 60438 Frankfurt am Main, Germany
| | - Kathi Zarnack
- Buchmann Institute for Molecular Life Sciences (BMLS) & Faculty of Biological Sciences, Goethe University Frankfurt, Max-von-Laue-Str. 15, 60438 Frankfurt am Main, Germany
| | - Daniel Krappmann
- Research Unit Cellular Signal Integration, Institute of Molecular Toxicology and Pharmacology, Molecular Targets and Therapeutics Center, Helmholtz Zentrum München, Neuherberg, 85764 München, Germany
- Corresponding author. (D.K.); (M.S.)
| | - Michael Sattler
- Institute of Structural Biology, Molecular Targets and Therapeutics Center, Helmholtz Zentrum München, Neuherberg, 85764 München, Germany
- Bavarian NMR Center, Department of Chemistry, Technical University of Munich, Garching, 85748 München, Germany
- Corresponding author. (D.K.); (M.S.)
| |
Collapse
|
10
|
Bell PA, Scheuermann S, Renner F, Pan CL, Lu HY, Turvey SE, Bornancin F, Régnier CH, Overall CM. Integrating knowledge of protein sequence with protein function for the prediction and validation of new MALT1 substrates. Comput Struct Biotechnol J 2022; 20:4717-4732. [PMID: 36147669 PMCID: PMC9463181 DOI: 10.1016/j.csbj.2022.08.021] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 08/07/2022] [Accepted: 08/08/2022] [Indexed: 11/30/2022] Open
Affiliation(s)
- Peter A. Bell
- Centre for Blood Research, Life Sciences Centre, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
- Department of Oral Biological and Medical Sciences, Faculty of Dentistry, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Sophia Scheuermann
- Centre for Blood Research, Life Sciences Centre, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
- Department of Oral Biological and Medical Sciences, Faculty of Dentistry, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
- Department of Immunology, Eberhard Karl University Tübingen, 72076 Tübingen, Germany
- Department of Hematology and Oncology, University Hospital Tübingen, Children's Hospital, 72076 Tübingen, Germany
| | - Florian Renner
- Novartis Institutes for BioMedical Research, Novartis Campus, CH-4056 Basel, Switzerland
- Molecular Targeted Therapy - Discovery Oncology, Roche Pharma Research & Early Development, F. Hoffmann-La Roche Ltd, 4070 Basel, Switzerland
| | - Christina L. Pan
- Centre for Blood Research, Life Sciences Centre, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
- Department of Oral Biological and Medical Sciences, Faculty of Dentistry, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Henry Y. Lu
- Department of Pediatrics, British Columbia Children's Hospital, The University of British Columbia, Vancouver, BC V5Z 4H4, Canada
- Department of Experimental Medicine, Faculty of Medicine, The University of British Columbia, Vancouver, BC V5Z 1M9, Canada
| | - Stuart E. Turvey
- Department of Pediatrics, British Columbia Children's Hospital, The University of British Columbia, Vancouver, BC V5Z 4H4, Canada
- Department of Experimental Medicine, Faculty of Medicine, The University of British Columbia, Vancouver, BC V5Z 1M9, Canada
| | - Frédéric Bornancin
- Novartis Institutes for BioMedical Research, Novartis Campus, CH-4056 Basel, Switzerland
| | - Catherine H. Régnier
- Novartis Institutes for BioMedical Research, Novartis Campus, CH-4056 Basel, Switzerland
| | - Christopher M. Overall
- Centre for Blood Research, Life Sciences Centre, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
- Department of Oral Biological and Medical Sciences, Faculty of Dentistry, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
- Corresponding author at: Centre for Blood Research, Life Sciences Centre, University of British Columbia, Vancouver, BC V6T 1Z3, Canada.
| |
Collapse
|
11
|
Xu Z, Qiao S, Qian W, Zhu Y, Yan W, Shen S, Wang T. Card9 protects fungal peritonitis through regulating Malt1-mediated activation of autophagy in macrophage. Int Immunopharmacol 2022; 110:108941. [PMID: 35850054 DOI: 10.1016/j.intimp.2022.108941] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 06/02/2022] [Accepted: 06/07/2022] [Indexed: 11/05/2022]
Abstract
Fungal peritonitis is an inflammatory condition of the peritoneum which occurs secondary to peritoneal dialysis. Most cases of peritonitis are caused by microbial invasion into the peritoneal cavity, resulting in high morbidity and mortality. Unlike bacterial peritonitis, little is known on fungal peritonitis. Card9, an adapter protein, plays a critical role in anti-fungal immunity. In this study, by using zymosan-induced peritonitis and C. albicans-induced peritonitis mouse model, we demonstrated that fungal peritonitis was exacerbated in Card9-/- mice, compared with WT mice. Next, we found the autophagy activation of peritonealmacrophages was impaired in Card9-/- peritonitis mice. The autophagy agonist, MG132, ameliorated peritonitis in Card9-/- mice. The result of microarray analysis indicates Malt1 was significantly decreased in Card9-/- peritonitis mice. Furthermore, we demonstrated that Malt1 interacts with P62 and mediates the function of P62 to clear ubiquitinated proteins. After overexpression of Malt1, impaired autophagy activation caused by Card9 deficient was significantly rescued. Together, our results indicate that Card9 protects fungal peritonitis by regulating Malt1-mediated autophagy in macrophages. Our research provides a new idea for the pathogenesis of fungal peritonitis, which is of great significance for the clinical treatment of fungal peritonitis.
Collapse
Affiliation(s)
- Zhen Xu
- Department of Oncology, Yancheng First Hospital, Affiliated Hospital of Nanjing University Medical School, The First People's Hospital of Yancheng, Yancheng, Jiangsu 224001, China; The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing 210093, China; Jiangsu Key Laboratory of Molecular Medicine, Division of Immunology, Medical School, Nanjing University, Nanjing 210093, China
| | - Shuping Qiao
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing 210093, China; Jiangsu Key Laboratory of Molecular Medicine, Division of Immunology, Medical School, Nanjing University, Nanjing 210093, China
| | - Wei Qian
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing 210093, China; Jiangsu Key Laboratory of Molecular Medicine, Division of Immunology, Medical School, Nanjing University, Nanjing 210093, China
| | - Yanan Zhu
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing 210093, China; Jiangsu Key Laboratory of Molecular Medicine, Division of Immunology, Medical School, Nanjing University, Nanjing 210093, China
| | - Wenyue Yan
- Department of Oncology, Yancheng First Hospital, Affiliated Hospital of Nanjing University Medical School, The First People's Hospital of Yancheng, Yancheng, Jiangsu 224001, China.
| | - Sunan Shen
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing 210093, China; Jiangsu Key Laboratory of Molecular Medicine, Division of Immunology, Medical School, Nanjing University, Nanjing 210093, China.
| | - Tingting Wang
- Department of Oncology, Yancheng First Hospital, Affiliated Hospital of Nanjing University Medical School, The First People's Hospital of Yancheng, Yancheng, Jiangsu 224001, China; The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing 210093, China; Jiangsu Key Laboratory of Molecular Medicine, Division of Immunology, Medical School, Nanjing University, Nanjing 210093, China.
| |
Collapse
|
12
|
O'Neill TJ, Seeholzer T, Gewies A, Gehring T, Giesert F, Hamp I, Graß C, Schmidt H, Kriegsmann K, Tofaute MJ, Demski K, Poth T, Rosenbaum M, Schnalzger T, Ruland J, Göttlicher M, Kriegsmann M, Naumann R, Heissmeyer V, Plettenburg O, Wurst W, Krappmann D. TRAF6 prevents fatal inflammation by homeostatic suppression of MALT1 protease. Sci Immunol 2021; 6:eabh2095. [PMID: 34767456 DOI: 10.1126/sciimmunol.abh2095] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Thomas J O'Neill
- Research Unit Cellular Signal Integration, Institute of Molecular Toxicology and Pharmacology, Helmholtz Zentrum München-German Research Center for Environmental Health, 85764 Neuherberg, Germany
| | - Thomas Seeholzer
- Research Unit Cellular Signal Integration, Institute of Molecular Toxicology and Pharmacology, Helmholtz Zentrum München-German Research Center for Environmental Health, 85764 Neuherberg, Germany
| | - Andreas Gewies
- Research Unit Cellular Signal Integration, Institute of Molecular Toxicology and Pharmacology, Helmholtz Zentrum München-German Research Center for Environmental Health, 85764 Neuherberg, Germany
| | - Torben Gehring
- Research Unit Cellular Signal Integration, Institute of Molecular Toxicology and Pharmacology, Helmholtz Zentrum München-German Research Center for Environmental Health, 85764 Neuherberg, Germany
| | - Florian Giesert
- Institute for Developmental Genetics, Helmholtz Zentrum München-German Research Center for Environmental Health, 85764 Neuherberg, Germany
| | - Isabel Hamp
- Institute for Medicinal Chemistry, Helmholtz Zentrum München-German Research Center for Environmental Health, 30167 Hannover, Germany.,Centre of Biomolecular Drug Research (BMWZ), Institute of Organic Chemistry, Leibniz Universität Hannover, 30167 Hannover, Germany
| | - Carina Graß
- Research Unit Cellular Signal Integration, Institute of Molecular Toxicology and Pharmacology, Helmholtz Zentrum München-German Research Center for Environmental Health, 85764 Neuherberg, Germany
| | - Henrik Schmidt
- Institute for Immunology, Biomedical Center Munich, LMU Munich, 82152 Martinsried, Germany
| | - Katharina Kriegsmann
- Department of Hematology, Oncology and Rheumatology, University Hospital Heidelberg, 69120 Heidelberg, Germany
| | - Marie J Tofaute
- Research Unit Cellular Signal Integration, Institute of Molecular Toxicology and Pharmacology, Helmholtz Zentrum München-German Research Center for Environmental Health, 85764 Neuherberg, Germany
| | - Katrin Demski
- Research Unit Cellular Signal Integration, Institute of Molecular Toxicology and Pharmacology, Helmholtz Zentrum München-German Research Center for Environmental Health, 85764 Neuherberg, Germany
| | - Tanja Poth
- Center for Model System and Comparative Pathology (CMCP), Institute of Pathology, University Hospital Heidelberg, 69120 Heidelberg, Germany
| | - Marc Rosenbaum
- Institute of Clinical Chemistry and Pathobiochemistry, TUM School of Medicine, Technical University of Munich, 81675 Munich, Germany.,TranslaTUM, Center for Translational Cancer Research, Technical University of Munich, 81675 Munich, Germany
| | - Theresa Schnalzger
- Institute of Clinical Chemistry and Pathobiochemistry, TUM School of Medicine, Technical University of Munich, 81675 Munich, Germany.,TranslaTUM, Center for Translational Cancer Research, Technical University of Munich, 81675 Munich, Germany
| | - Jürgen Ruland
- Institute of Clinical Chemistry and Pathobiochemistry, TUM School of Medicine, Technical University of Munich, 81675 Munich, Germany.,TranslaTUM, Center for Translational Cancer Research, Technical University of Munich, 81675 Munich, Germany.,German Cancer Consortium (DKTK), 69120 Heidelberg, Germany
| | - Martin Göttlicher
- Institute of Molecular Toxicology and Pharmacology, Helmholtz Zentrum München-German Research Center for Environmental Health, 85764 Neuherberg, Germany.,School of Medicine, Technical University of Munich, Munich, Germany
| | - Mark Kriegsmann
- Institute of Pathology, University Hospital Heidelberg, 69120 Heidelberg, Germany
| | - Ronald Naumann
- Max Planck Institute of Molecular Cell Biology and Genetics, Transgenic Core Facility, 01307 Dresden, Germany
| | - Vigo Heissmeyer
- Institute for Immunology, Biomedical Center Munich, LMU Munich, 82152 Martinsried, Germany.,Research Unit Molecular Immune Regulation, Helmholtz Zentrum München-German Research Center for Environmental Health, 81377 München, Germany
| | - Oliver Plettenburg
- Institute for Medicinal Chemistry, Helmholtz Zentrum München-German Research Center for Environmental Health, 30167 Hannover, Germany.,Centre of Biomolecular Drug Research (BMWZ), Institute of Organic Chemistry, Leibniz Universität Hannover, 30167 Hannover, Germany
| | - Wolfgang Wurst
- Institute for Developmental Genetics, Helmholtz Zentrum München-German Research Center for Environmental Health, 85764 Neuherberg, Germany.,German Center for Neurodegenerative Diseases (DZNE), Site Munich, Munich Cluster for Systems Neurology (SyNergy), Munich, Germany.,Technische Universität München, Lehrstuhl für Entwicklungsgenetik c/o Helmholtz Zentrum München, 85764 Neuherberg, Germany
| | - Daniel Krappmann
- Research Unit Cellular Signal Integration, Institute of Molecular Toxicology and Pharmacology, Helmholtz Zentrum München-German Research Center for Environmental Health, 85764 Neuherberg, Germany
| |
Collapse
|
13
|
Hamp I, O'Neill TJ, Plettenburg O, Krappmann D. A patent review of MALT1 inhibitors (2013-present). Expert Opin Ther Pat 2021; 31:1079-1096. [PMID: 34214002 DOI: 10.1080/13543776.2021.1951703] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
INTRODUCTION MALT1 is the only human paracaspase, a protease with unique cleavage activity and substrate specificity. As a key regulator of immune responses, MALT1 has attracted attention as an immune modulatory target for the treatment of autoimmune/inflammatory diseases. Further, chronic MALT1 protease activation drives survival of lymphomas, suggesting that MALT1 is a suitable drug target for lymphoid malignancies. Recent studies have indicated that MALT1 inhibition impairs immune suppressive function of regulatory T cells in the tumor microenvironment, suggesting that MALT1 inhibitors may boost anti-tumor immunity in the treatment of solid cancers. AREAS COVERED This review summarizes the literature on MALT1 patents and applications. We discuss the potential therapeutic uses for MALT1 inhibitors based on patents and scientific literature. EXPERT OPINION There has been a steep increase in MALT1 inhibitor patents. Compounds with high selectivity and good bioavailability have been developed. An allosteric binding pocket is the preferred site for potent and selective MALT1 targeting. MALT1 inhibitors have moved to early clinical trials, but toxicological studies indicate that long-term MALT1 inhibition can disrupt immune homeostasis and lead to autoimmunity. Even though this poses risks, preventing immune suppression may favor the use of MALT1 inhibitors in cancer immunotherapies.
Collapse
Affiliation(s)
- Isabel Hamp
- Institute for Medicinal Chemistry, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany.,Centre of Biomolecular Drug Research (BMWZ), Institute of Organic Chemistry, Leibniz Universität Hannover, Hannover, Germany
| | - Thomas J O'Neill
- Research Unit Cellular Signal Integration, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| | - Oliver Plettenburg
- Institute for Medicinal Chemistry, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany.,Centre of Biomolecular Drug Research (BMWZ), Institute of Organic Chemistry, Leibniz Universität Hannover, Hannover, Germany
| | - Daniel Krappmann
- Research Unit Cellular Signal Integration, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| |
Collapse
|
14
|
Kim HR, Tagirasa R, Yoo E. Covalent Small Molecule Immunomodulators Targeting the Protease Active Site. J Med Chem 2021; 64:5291-5322. [PMID: 33904753 DOI: 10.1021/acs.jmedchem.1c00172] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Cells of the immune system utilize multiple proteases to regulate cell functions and orchestrate innate and adaptive immune responses. Dysregulated protease activities are implicated in many immune-related disorders; thus, protease inhibitors have been actively investigated for pharmaceutical development. Although historically considered challenging with concerns about toxicity, compounds that covalently modify the protease active site represent an important class of agents, emerging not only as chemical probes but also as approved drugs. Here, we provide an overview of technologies useful for the study of proteases with the focus on recent advances in chemoproteomic methods and screening platforms. By highlighting covalent inhibitors that have been designed to target immunomodulatory proteases, we identify opportunities for the development of small molecule immunomodulators.
Collapse
Affiliation(s)
- Hong-Rae Kim
- Chemical Biology Laboratory, National Cancer Institute, Frederick, Maryland 21702, United States
| | - Ravichandra Tagirasa
- Chemical Biology Laboratory, National Cancer Institute, Frederick, Maryland 21702, United States
| | - Euna Yoo
- Chemical Biology Laboratory, National Cancer Institute, Frederick, Maryland 21702, United States
| |
Collapse
|
15
|
Liang X, Cao Y, Li C, Yu H, Yang C, Liu H. MALT1 as a promising target to treat lymphoma and other diseases related to MALT1 anomalies. Med Res Rev 2021; 41:2388-2422. [PMID: 33763890 DOI: 10.1002/med.21799] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 12/23/2020] [Accepted: 03/03/2021] [Indexed: 12/25/2022]
Abstract
Mucosa-associated lymphoid tissue lymphoma translocation protein 1 (MALT1) is a key adaptor protein that regulates the NF-κB pathway, in which MALT1 functions as a scaffold protein and protease to trigger downstream signals. The abnormal expression of MALT1 is closely associated with lymphomagenesis and other diseases, including solid tumors and autoimmune diseases. MALT1 is the only protease in the underlying pathogenesis of these diseases, and its proteolytic activity can be pharmacologically regulated. Therefore, MALT1 is a potential and promising target for anti-lymphoma and other MALT1-related disease treatments. Currently, the development of MALT1 inhibitors is still in its early stages. This review presents an overview of MALT1, particularly its X-ray structures and biological functions, and elaborates on the pathogenesis of diseases associated with its dysregulation. We then summarize previously reported MALT1 inhibitors, focusing on their molecular structure, biological activity, structure-activity relationship, and limitations. Finally, we propose future research directions to accelerate the discovery of novel MALT1 inhibitors with clinical applications. Overall, this review provides a comprehensive and systematic overview of MALT1-related research advances and serves as a theoretical basis for drug discovery and research.
Collapse
Affiliation(s)
- Xuewu Liang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - YiChun Cao
- School of Pharmacy, Fudan University, Shanghai, China
| | - Chunpu Li
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Haolan Yu
- Department of Urology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Chenghua Yang
- Department of Urology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Hong Liu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| |
Collapse
|
16
|
Wang W, Wei Q, Hao Q, Zhang Y, Li Y, Bi Y, Jin Z, Liu H, Liu X, Yang Z, Xiao S. Cellular CARD11 Inhibits the Fusogenic Activity of Newcastle Disease Virus via CBM Signalosome-Mediated Furin Reduction in Chicken Fibroblasts. Front Microbiol 2021; 12:607451. [PMID: 33603723 PMCID: PMC7884349 DOI: 10.3389/fmicb.2021.607451] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Accepted: 01/07/2021] [Indexed: 12/02/2022] Open
Abstract
Newcastle disease virus (NDV) causes an infectious disease that poses a major threat to poultry health. Our previous study identified a chicken brain-specific caspase recruitment domain-containing protein 11 (CARD11) that was upregulated in chicken neurons and inhibited NDV replication. This raises the question of whether CARD11 plays a role in inhibiting viruses in non-neural cells. Here, chicken fibroblasts were used as a non-neural cell model to investigate the role. CARD11 expression was not significantly upregulated by either velogenic or lentogenic NDV infection in chicken fibroblasts. Viral replication was decreased in DF-1 cells stably overexpressing CARD11, while viral growth was significantly increased in the CARD11-knockdown DF-1 cell line. Moreover, CARD11 colocalized with the viral P protein and aggregated around the fibroblast nucleus, suggesting that an interaction existed between CARD11 and the viral P protein; this interaction was further examined by suppressing viral RNA polymerase activity by using a minigenome assay. Viral replication was inhibited by CARD11 in fibroblasts, and this result was consistent with our previous report in chicken neurons. Importantly, CARD11 was observed to reduce the syncytia induced by either velogenic virus infection or viral haemagglutinin-neuraminidase (HN) and F cotransfection in fibroblasts. We found that CARD11 inhibited the expression of the host protease furin, which is essential for cleavage of the viral F protein to trigger fusogenic activity. Furthermore, the CARD11-Bcl10-MALT1 (CBM) signalosome was found to suppress furin expression, which resulted in a reduction in the cleavage efficiency of the viral F protein to further inhibit viral syncytia. Taken together, our findings mainly demonstrated a novel CARD11 inhibitory mechanism for viral fusogenic activity in chicken fibroblasts, and this mechanism explains the antiviral roles of this molecule in NDV pathogenesis.
Collapse
Affiliation(s)
- Wenbin Wang
- College of Veterinary Medicine, Northwest A&F University, Yangling, China.,Poultry Institute, Shandong Academy of Agricultural Science, Jinan, China
| | - Qiaolin Wei
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Qiqi Hao
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Yajie Zhang
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Yongshan Li
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Youkun Bi
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Zhongyuan Jin
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Haijin Liu
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Xuelan Liu
- Poultry Institute, Shandong Academy of Agricultural Science, Jinan, China
| | - Zengqi Yang
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Sa Xiao
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| |
Collapse
|
17
|
Cheng J, Klei LR, Hubel NE, Zhang M, Schairer R, Maurer LM, Klei HB, Kang H, Concel VJ, Delekta PC, Dang EV, Mintz MA, Baens M, Cyster JG, Parameswaran N, Thome M, Lucas PC, McAllister-Lucas LM. GRK2 suppresses lymphomagenesis by inhibiting the MALT1 proto-oncoprotein. J Clin Invest 2020; 130:1036-1051. [PMID: 31961340 DOI: 10.1172/jci97040] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2017] [Accepted: 11/06/2019] [Indexed: 12/11/2022] Open
Abstract
Antigen receptor-dependent (AgR-dependent) stimulation of the NF-κB transcription factor in lymphocytes is a required event during adaptive immune response, but dysregulated activation of this signaling pathway can lead to lymphoma. AgR stimulation promotes assembly of the CARMA1-BCL10-MALT1 complex, wherein MALT1 acts as (a) a scaffold to recruit components of the canonical NF-κB machinery and (b) a protease to cleave and inactivate specific substrates, including negative regulators of NF-κB. In multiple lymphoma subtypes, malignant B cells hijack AgR signaling pathways to promote their own growth and survival, and inhibiting MALT1 reduces the viability and growth of these tumors. As such, MALT1 has emerged as a potential pharmaceutical target. Here, we identified G protein-coupled receptor kinase 2 (GRK2) as a new MALT1-interacting protein. We demonstrated that GRK2 binds the death domain of MALT1 and inhibits MALT1 scaffolding and proteolytic activities. We found that lower GRK2 levels in activated B cell-type diffuse large B cell lymphoma (ABC-DLBCL) are associated with reduced survival, and that GRK2 knockdown enhances ABC-DLBCL tumor growth in vitro and in vivo. Together, our findings suggest that GRK2 can function as a tumor suppressor by inhibiting MALT1 and provide a roadmap for developing new strategies to inhibit MALT1-dependent lymphomagenesis.
Collapse
Affiliation(s)
| | | | - Nathaniel E Hubel
- Department of Pediatrics and.,Department of Pathology, University of Pittsburgh School of Medicine and UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania, USA
| | - Ming Zhang
- Department of Biochemistry, Center of Immunity and Infection, University of Lausanne, Epalinges, Switzerland
| | - Rebekka Schairer
- Department of Biochemistry, Center of Immunity and Infection, University of Lausanne, Epalinges, Switzerland
| | | | | | - Heejae Kang
- Department of Pathology, University of Pittsburgh School of Medicine and UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania, USA
| | | | - Phillip C Delekta
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, Michigan, USA
| | - Eric V Dang
- Department of Biophysics and Biochemistry, UCSF, San Francisco, California, USA
| | - Michelle A Mintz
- Department of Biophysics and Biochemistry, UCSF, San Francisco, California, USA
| | - Mathijs Baens
- Human Genome Laboratory, VIB Center for the Biology of Disease, and.,Center for Human Genetics, Katholieke Universiteit Leuven, Leuven, Belgium
| | - Jason G Cyster
- Department of Biophysics and Biochemistry, UCSF, San Francisco, California, USA.,Howard Hughes Medical Institute and.,Department of Microbiology and Immunology, UCSF, San Francisco, California, USA
| | | | - Margot Thome
- Department of Biochemistry, Center of Immunity and Infection, University of Lausanne, Epalinges, Switzerland
| | - Peter C Lucas
- Department of Pathology, University of Pittsburgh School of Medicine and UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania, USA
| | | |
Collapse
|
18
|
Cheng J, Maurer LM, Kang H, Lucas PC, McAllister-Lucas LM. Critical protein-protein interactions within the CARMA1-BCL10-MALT1 complex: Take-home points for the cell biologist. Cell Immunol 2020; 355:104158. [PMID: 32721634 DOI: 10.1016/j.cellimm.2020.104158] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 05/25/2020] [Accepted: 07/03/2020] [Indexed: 12/24/2022]
Abstract
The CBM complex, which is composed of the proteins CARMA1, BCL10, and MALT1, serves multiple pivotal roles as a mediator of T-cell receptor and B-cell receptor-dependent NF-κB induction and lymphocyte activation. CARMA1, BCL10, and MALT1 are each proto-oncoproteins and dysregulation of CBM signaling, as a result of somatic gain-of-function mutation or chromosomal translocation, is a hallmark of multiple lymphoid malignancies including Activated B-cell Diffuse Large B-cell Lymphoma. Moreover, loss-of-function as well as gain-of-function germline mutations in CBM complex proteins have been associated with a range of immune dysregulation syndromes. A wealth of detailed structural information has become available over the past decade through meticulous interrogation of the interactions between CBM components. Here, we review key findings regarding the biochemical nature of these protein-protein interactions which have ultimately led the field to a sophisticated understanding of how these proteins assemble into high-order filamentous CBM complexes. To date, approaches to therapeutic inhibition of the CBM complex for the treatment of lymphoid malignancy and/or auto-immunity have focused on blocking MALT1 protease function. We also review key studies relating to the structural impact of MALT1 protease inhibitors on key protein-protein interactions.
Collapse
Affiliation(s)
- Jing Cheng
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittburgh, PA, USA
| | - Lisa M Maurer
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittburgh, PA, USA
| | - Heejae Kang
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Peter C Lucas
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | | |
Collapse
|
19
|
Nicolau CA, Gavard J, Bidère N. TAK1 lessens the activity of the paracaspase MALT1 during T cell receptor signaling. Cell Immunol 2020; 353:104115. [PMID: 32388054 DOI: 10.1016/j.cellimm.2020.104115] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Revised: 03/18/2020] [Accepted: 04/26/2020] [Indexed: 01/03/2023]
Abstract
The CARMA1-BCL10-MALT1 (CBM) complex couples antigen receptors to the activation of Nuclear Factor κB (NF-κB) transcription factors in T/B lymphocytes. Within this signalosome, the MALT1 paracaspase serves dual roles: it is a crucial adaptor for signal transduction to NF-κB signaling, and a protease that shapes NF-κB activity and lymphocyte activation. Although a subtle choreography of ubiquitination and phosphorylation orchestrate the CBM, how precisely this complex and MALT1 enzyme are regulated continue to be elucidated. Here, we report that the chemical inhibition or the siRNA-based silencing of transforming growth factor beta-activated kinase 1 (TAK1), a known partner of the CBM complex required for NF-κB activation, enhanced the processing of MALT1 substrates. We further show that the assembly of the CBM as well as the ubiquitination of MALT1 was augmented when TAK1 was inhibited. Thus, TAK1 may initiate a negative feedback loop to finely tune the CBM complex activity.
Collapse
Affiliation(s)
- Carolina Alves Nicolau
- CRCINA, Team SOAP, INSERM, CNRS, Université de Nantes, Université d'Angers, IRS-UN blg, Room 405, 8 quai Moncousu, Nantes 44007, France; L'Héma-NexT, i-Site NexT, Nantes, France; GDR3697 Micronit, CNRS, Nantes, France
| | - Julie Gavard
- CRCINA, Team SOAP, INSERM, CNRS, Université de Nantes, Université d'Angers, IRS-UN blg, Room 405, 8 quai Moncousu, Nantes 44007, France; L'Héma-NexT, i-Site NexT, Nantes, France; GDR3697 Micronit, CNRS, Nantes, France; Institut de Cancérologie de l'Ouest, Site René Gauducheau, 44800 Saint-Herblain, France
| | - Nicolas Bidère
- CRCINA, Team SOAP, INSERM, CNRS, Université de Nantes, Université d'Angers, IRS-UN blg, Room 405, 8 quai Moncousu, Nantes 44007, France; L'Héma-NexT, i-Site NexT, Nantes, France; GDR3697 Micronit, CNRS, Nantes, France.
| |
Collapse
|
20
|
Abstract
The catalytic activity of the protease MALT1 is required for adaptive immune responses and regulatory T (Treg)-cell development, while dysregulated MALT1 activity can lead to lymphoma. MALT1 activation requires its monoubiquitination on lysine 644 (K644) within the Ig3 domain, localized adjacent to the protease domain. The molecular requirements for MALT1 monoubiquitination and the mechanism by which monoubiquitination activates MALT1 had remained elusive. Here, we show that the Ig3 domain interacts directly with ubiquitin and that an intact Ig3-ubiquitin interaction surface is required for the conjugation of ubiquitin to K644. Moreover, by generating constitutively active MALT1 mutants that overcome the need for monoubiquitination, we reveal an allosteric communication between the ubiquitination site K644, the Ig3-protease interaction surface, and the active site of the protease domain. Finally, we show that MALT1 mutants that alter the Ig3-ubiquitin interface impact the biological response of T cells. Thus, ubiquitin binding by the Ig3 domain promotes MALT1 activation by an allosteric mechanism that is essential for its biological function.
Collapse
|
21
|
Youseff BH, Brewer TG, McNally KL, Izuogu AO, Lubick KJ, Presloid JB, Alqahtani S, Chattopadhyay S, Best SM, Hu X, Taylor RT. TRAF6 Plays a Proviral Role in Tick-Borne Flavivirus Infection through Interaction with the NS3 Protease. iScience 2019; 15:489-501. [PMID: 31129244 PMCID: PMC6536497 DOI: 10.1016/j.isci.2019.05.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Revised: 01/18/2019] [Accepted: 05/08/2019] [Indexed: 02/08/2023] Open
Abstract
Tick-borne flaviviruses (TBFVs) can cause life-threatening encephalitis and hemorrhagic fever. To identify virus-host interactions that may be exploited as therapeutic targets, we analyzed the TBFV polyprotein in silico for antiviral protein-binding motifs. We obtained two putative tumor necrosis factor receptor-associated factor 6 (TRAF6)-binding motifs (TBMs) within the protease domain of the viral nonstructural 3 (NS3) protein. Here, we show that TBFV NS3 interacted with TRAF6 during infection and that TRAF6 supports TBFV replication. The proviral role of TRAF6 was not seen with mosquito-borne flaviviruses, consistent with the lack of conserved TBMs. Mutation of the second TBM within NS3 disrupted TRAF6 binding, coincident with reduced abundance of mature, autocatalytically derived form of the NS3 protease and significant virus attenuation in vitro. Our studies reveal insights into how flaviviruses exploit innate immunity for the purpose of viral replication and identify a potential target for therapeutic design. Langat virus (LGTV) NS3 protease interacts with TRAF6 during infection Tick-borne, unlike mosquito-borne, flaviviruses use TRAF6 for optimal replication E117A mutation of LGTV NS3 reduces TRAF6 binding and mature protease abundance LGTV with a mutated TRAF6-binding motif is attenuated in vitro
Collapse
Affiliation(s)
- Brian H Youseff
- Department of Medical Microbiology and Immunology, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA
| | - Thomas G Brewer
- Department of Medical Microbiology and Immunology, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA
| | - Kristin L McNally
- Innate Immunity and Pathogenesis Unit, Laboratory of Virology, Rocky Mountain Laboratories, DIR, NIAID, NIH, Hamilton, MT 59840, USA
| | - Adaeze O Izuogu
- Department of Medical Microbiology and Immunology, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA
| | - Kirk J Lubick
- Innate Immunity and Pathogenesis Unit, Laboratory of Virology, Rocky Mountain Laboratories, DIR, NIAID, NIH, Hamilton, MT 59840, USA
| | - John B Presloid
- Department of Medical Microbiology and Immunology, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA
| | - Saad Alqahtani
- Department of Chemistry and Biochemistry, University of Toledo, Toledo, OH 43606, USA
| | - Saurabh Chattopadhyay
- Department of Medical Microbiology and Immunology, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA
| | - Sonja M Best
- Innate Immunity and Pathogenesis Unit, Laboratory of Virology, Rocky Mountain Laboratories, DIR, NIAID, NIH, Hamilton, MT 59840, USA
| | - Xiche Hu
- Department of Chemistry and Biochemistry, University of Toledo, Toledo, OH 43606, USA
| | - R Travis Taylor
- Department of Medical Microbiology and Immunology, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA.
| |
Collapse
|
22
|
Lork M, Staal J, Beyaert R. Ubiquitination and phosphorylation of the CARD11-BCL10-MALT1 signalosome in T cells. Cell Immunol 2018; 340:103877. [PMID: 30514565 DOI: 10.1016/j.cellimm.2018.11.001] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Accepted: 11/02/2018] [Indexed: 12/16/2022]
Abstract
Antigen receptor-induced signaling plays an important role in inflammation and immunity. Formation of a CARD11-BCL10-MALT1 (CBM) signaling complex is a key event in T- and B cell receptor-induced gene expression by regulating NF-κB activation and mRNA stability. Deregulated CARD11, BCL10 or MALT1 expression or CBM signaling have been associated with immunodeficiency, autoimmunity and cancer, indicating that CBM formation and function have to be tightly regulated. Over the past years great progress has been made in deciphering the molecular mechanisms of assembly and disassembly of the CBM complex. In this context, several posttranslational modifications play an indispensable role in regulating CBM function and downstream signal transduction. In this review we summarize how the different CBM components as well as their interplay are regulated by protein ubiquitination and phosphorylation in the context of T cell receptor signaling.
Collapse
Affiliation(s)
- Marie Lork
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium; Unit of Molecular Signal Transduction in Inflammation, Center for Inflammation Research, VIB, Ghent, Belgium
| | - Jens Staal
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium; Unit of Molecular Signal Transduction in Inflammation, Center for Inflammation Research, VIB, Ghent, Belgium
| | - Rudi Beyaert
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium; Unit of Molecular Signal Transduction in Inflammation, Center for Inflammation Research, VIB, Ghent, Belgium.
| |
Collapse
|
23
|
Seeholzer T, Kurz S, Schlauderer F, Woods S, Gehring T, Widmann S, Lammens K, Krappmann D. BCL10-CARD11 Fusion Mimics an Active CARD11 Seed That Triggers Constitutive BCL10 Oligomerization and Lymphocyte Activation. Front Immunol 2018; 9:2695. [PMID: 30515170 PMCID: PMC6255920 DOI: 10.3389/fimmu.2018.02695] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Accepted: 10/31/2018] [Indexed: 12/16/2022] Open
Abstract
Assembly of the CARD11/CARMA1-BCL10-MALT1 (CBM) signaling complex upon T or B cell antigen receptor (TCR or BCR) engagement drives lymphocyte activation. Recruitment of pre-assembled BCL10-MALT1 complexes to CARD11 fosters activation of the MALT1 protease and canonical NF-κB signaling. Structural data and in vitro assays have suggested that CARD11 acts as a seed that nucleates the assembly of BCL10 filaments, but the relevance of these findings for CBM complex assembly in cells remains unresolved. To uncouple cellular CARD11 recruitment of BCL10 and BCL10 filament assembly, we generated a BCL10-CARD11 fusion protein that links the C-terminus of BCL10 to the N-terminus of CARD11. When stably expressed in CARD11 KO Jurkat T cells, the BCL10-CARD11 fusion induced constitutive MALT1 activation. Furthermore, in CARD11 KO BJAB B cells, BCL10-CARD11 promoted constitutive NF-κB activation to a similar extent as CARD11 containing oncogenic driver mutations. Using structure-guided destructive mutations in the CARD11-BCL10 (CARD11 R35A) or BCL10-BCL10 (BCL10 R42E) interfaces, we demonstrate that chronic activation by the BCL10-CARD11 fusion protein was independent of the CARD11 CARD. However, activation strictly relied upon the ability of the BCL10 CARD to form oligomers. Thus, by combining distinct CARD mutations in the context of constitutively active BCL10-CARD11 fusion proteins, we provide evidence that BCL10-MALT1 recruitment to CARD11 and BCL10 oligomerization are interconnected processes, which bridge the CARD11 seed to downstream pathways in lymphocytes.
Collapse
Affiliation(s)
- Thomas Seeholzer
- Research Unit Cellular Signal Integration, Institute of Molecular Toxicology and Pharmacology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| | - Susanne Kurz
- Research Unit Cellular Signal Integration, Institute of Molecular Toxicology and Pharmacology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| | | | - Simone Woods
- Research Unit Cellular Signal Integration, Institute of Molecular Toxicology and Pharmacology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| | - Torben Gehring
- Research Unit Cellular Signal Integration, Institute of Molecular Toxicology and Pharmacology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| | - Simon Widmann
- Research Unit Cellular Signal Integration, Institute of Molecular Toxicology and Pharmacology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| | - Katja Lammens
- Gene Center, Ludwig-Maximilians University, Munich, Germany
| | - Daniel Krappmann
- Research Unit Cellular Signal Integration, Institute of Molecular Toxicology and Pharmacology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| |
Collapse
|
24
|
Lu HY, Bauman BM, Arjunaraja S, Dorjbal B, Milner JD, Snow AL, Turvey SE. The CBM-opathies-A Rapidly Expanding Spectrum of Human Inborn Errors of Immunity Caused by Mutations in the CARD11-BCL10-MALT1 Complex. Front Immunol 2018; 9:2078. [PMID: 30283440 PMCID: PMC6156466 DOI: 10.3389/fimmu.2018.02078] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2018] [Accepted: 08/22/2018] [Indexed: 01/06/2023] Open
Abstract
The caspase recruitment domain family member 11 (CARD11 or CARMA1)-B cell CLL/lymphoma 10 (BCL10)-MALT1 paracaspase (MALT1) [CBM] signalosome complex serves as a molecular bridge between cell surface antigen receptor signaling and the activation of the NF-κB, JNK, and mTORC1 signaling axes. This positions the CBM complex as a critical regulator of lymphocyte activation, proliferation, survival, and metabolism. Inborn errors in each of the CBM components have now been linked to a diverse group of human primary immunodeficiency diseases termed "CBM-opathies." Clinical manifestations range from severe combined immunodeficiency to selective B cell lymphocytosis, atopic disease, and specific humoral defects. This surprisingly broad spectrum of phenotypes underscores the importance of "tuning" CBM signaling to preserve immune homeostasis. Here, we review the distinct clinical and immunological phenotypes associated with human CBM complex mutations and introduce new avenues for targeted therapeutic intervention.
Collapse
Affiliation(s)
- Henry Y Lu
- Department of Pediatrics, British Columbia Children's Hospital, The University of British Columbia, Vancouver, BC, Canada.,Experimental Medicine Program, Faculty of Medicine, The University of British Columbia, Vancouver, BC, Canada
| | - Bradly M Bauman
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Swadhinya Arjunaraja
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Batsukh Dorjbal
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Joshua D Milner
- Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Andrew L Snow
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Stuart E Turvey
- Department of Pediatrics, British Columbia Children's Hospital, The University of British Columbia, Vancouver, BC, Canada.,Experimental Medicine Program, Faculty of Medicine, The University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
25
|
Juilland M, Thome M. Holding All the CARDs: How MALT1 Controls CARMA/CARD-Dependent Signaling. Front Immunol 2018; 9:1927. [PMID: 30214442 PMCID: PMC6125328 DOI: 10.3389/fimmu.2018.01927] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Accepted: 08/06/2018] [Indexed: 01/20/2023] Open
Abstract
The scaffold proteins CARMA1-3 (encoded by the genes CARD11, -14 and -10) and CARD9 play major roles in signaling downstream of receptors with immunoreceptor tyrosine activation motifs (ITAMs), G-protein coupled receptors (GPCR) and receptor tyrosine kinases (RTK). These receptors trigger the formation of oligomeric CARMA/CARD-BCL10-MALT1 (CBM) complexes via kinases of the PKC family. The CBM in turn regulates gene expression by the activation of NF-κB and AP-1 transcription factors and controls transcript stability. The paracaspase MALT1 is the only CBM component having an enzymatic (proteolytic) activity and has therefore recently gained attention as a potential drug target. Here we review recent advances in the understanding of the molecular function of the protease MALT1 and summarize how MALT1 scaffold and protease function contribute to the transmission of CBM signals. Finally, we will highlight how dysregulation of MALT1 function can cause pathologies such as immunodeficiency, autoimmunity, psoriasis, and cancer.
Collapse
Affiliation(s)
- Mélanie Juilland
- Department of Biochemistry, University of Lausanne, Epalinges, Switzerland
| | - Margot Thome
- Department of Biochemistry, University of Lausanne, Epalinges, Switzerland
| |
Collapse
|
26
|
Staal J, Driege Y, Haegman M, Borghi A, Hulpiau P, Lievens L, Gul IS, Sundararaman S, Gonçalves A, Dhondt I, Pinzón JH, Braeckman BP, Technau U, Saeys Y, van Roy F, Beyaert R. Ancient Origin of the CARD-Coiled Coil/Bcl10/MALT1-Like Paracaspase Signaling Complex Indicates Unknown Critical Functions. Front Immunol 2018; 9:1136. [PMID: 29881386 PMCID: PMC5978004 DOI: 10.3389/fimmu.2018.01136] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Accepted: 05/07/2018] [Indexed: 12/16/2022] Open
Abstract
The CARD–coiled coil (CC)/Bcl10/MALT1-like paracaspase (CBM) signaling complexes composed of a CARD–CC family member (CARD-9, -10, -11, or -14), Bcl10, and the type 1 paracaspase MALT1 (PCASP1) play a pivotal role in immunity, inflammation, and cancer. Targeting MALT1 proteolytic activity is of potential therapeutic interest. However, little is known about the evolutionary origin and the original functions of the CBM complex. Type 1 paracaspases originated before the last common ancestor of planulozoa (bilaterians and cnidarians). Notably in bilaterians, Ecdysozoa (e.g., nematodes and insects) lacks Bcl10, whereas other lineages have a Bcl10 homolog. A survey of invertebrate CARD–CC homologs revealed such homologs only in species with Bcl10, indicating an ancient common origin of the entire CBM complex. Furthermore, vertebrate-like Syk/Zap70 tyrosine kinase homologs with the ITAM-binding SH2 domain were only found in invertebrate organisms with CARD–CC/Bcl10, indicating that this pathway might be related to the original function of the CBM complex. Moreover, the type 1 paracaspase sequences from invertebrate organisms that have CARD–CC/Bcl10 are more similar to vertebrate paracaspases. Functional analysis of protein–protein interactions, NF-κB signaling, and CYLD cleavage for selected invertebrate type 1 paracaspase and Bcl10 homologs supports this scenario and indicates an ancient origin of the CARD–CC/Bcl10/paracaspase signaling complex. By contrast, many of the known MALT1-associated activities evolved fairly recently, indicating that unknown functions are at the basis of the protein conservation. As a proof-of-concept, we provide initial evidence for a CBM- and NF-κB-independent neuronal function of the Caenorhabditis elegans type 1 paracaspase malt-1. In conclusion, this study shows how evolutionary insights may point at alternative functions of MALT1.
Collapse
Affiliation(s)
- Jens Staal
- Unit of Molecular Signal Transduction in Inflammation, VIB-UGent Center for Inflammation Research (IRC), Ghent, Belgium.,Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Yasmine Driege
- Unit of Molecular Signal Transduction in Inflammation, VIB-UGent Center for Inflammation Research (IRC), Ghent, Belgium.,Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Mira Haegman
- Unit of Molecular Signal Transduction in Inflammation, VIB-UGent Center for Inflammation Research (IRC), Ghent, Belgium.,Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Alice Borghi
- Unit of Molecular Signal Transduction in Inflammation, VIB-UGent Center for Inflammation Research (IRC), Ghent, Belgium.,Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Paco Hulpiau
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium.,Unit of Data Mining and Modeling for Biomedicine, VIB-UGent Center for Inflammation Research (IRC), Ghent, Belgium
| | - Laurens Lievens
- Unit of Molecular Signal Transduction in Inflammation, VIB-UGent Center for Inflammation Research (IRC), Ghent, Belgium.,Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Ismail Sahin Gul
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium.,Unit of Molecular Cell Biology, VIB-UGent Center for Inflammation Research (IRC), Ghent, Belgium
| | - Srividhya Sundararaman
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium.,Unit of Molecular Cell Biology, VIB-UGent Center for Inflammation Research (IRC), Ghent, Belgium
| | - Amanda Gonçalves
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium.,VIB Bio Imaging Core Gent, VIB-UGent Center for Inflammation Research (IRC), Ghent, Belgium
| | - Ineke Dhondt
- Laboratory for Aging Physiology and Molecular Evolution, Biology Department, Ghent University, Ghent, Belgium
| | - Jorge H Pinzón
- Department of Biology, University of Texas Arlington, Arlington, TX, United States
| | - Bart P Braeckman
- Laboratory for Aging Physiology and Molecular Evolution, Biology Department, Ghent University, Ghent, Belgium
| | - Ulrich Technau
- Department of Molecular Evolution and Development, Faculty of Life Sciences, University of Vienna, Vienna, Austria
| | - Yvan Saeys
- Unit of Data Mining and Modeling for Biomedicine, VIB-UGent Center for Inflammation Research (IRC), Ghent, Belgium.,Department of Applied Mathematics, Computer Science and Statistics, Ghent University, Ghent, Belgium
| | - Frans van Roy
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium.,Unit of Molecular Cell Biology, VIB-UGent Center for Inflammation Research (IRC), Ghent, Belgium
| | - Rudi Beyaert
- Unit of Molecular Signal Transduction in Inflammation, VIB-UGent Center for Inflammation Research (IRC), Ghent, Belgium.,Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| |
Collapse
|
27
|
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, Quancard J. N-aryl-piperidine-4-carboxamides as a novel class of potent inhibitors of MALT1 proteolytic activity. Bioorg Med Chem Lett 2018; 28:2153-2158. [PMID: 29759726 DOI: 10.1016/j.bmcl.2018.05.017] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Revised: 04/16/2018] [Accepted: 05/08/2018] [Indexed: 12/11/2022]
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.
Collapse
Affiliation(s)
- Achim Schlapbach
- Novartis Institutes for BioMedical Research, CH-4002 Basel, Switzerland.
| | - Laszlo Revesz
- Novartis Institutes for BioMedical Research, CH-4002 Basel, Switzerland
| | | | - Thomas Zoller
- Novartis Institutes for BioMedical Research, CH-4002 Basel, Switzerland
| | | | | | - Thomas Radimerski
- Novartis Institutes for BioMedical Research, CH-4002 Basel, Switzerland
| | - Jutta Blank
- Novartis Institutes for BioMedical Research, CH-4002 Basel, Switzerland
| | | | - Martin Renatus
- Novartis Institutes for BioMedical Research, CH-4002 Basel, Switzerland
| | - Paulus Erbel
- Novartis Institutes for BioMedical Research, CH-4002 Basel, Switzerland
| | - Samu Melkko
- Novartis Institutes for BioMedical Research, CH-4002 Basel, Switzerland
| | - Richard Heng
- Novartis Institutes for BioMedical Research, CH-4002 Basel, Switzerland
| | - Oliver Simic
- Novartis Institutes for BioMedical Research, CH-4002 Basel, Switzerland
| | - Ralf Endres
- Novartis Institutes for BioMedical Research, CH-4002 Basel, Switzerland
| | - Markus Wartmann
- Novartis Institutes for BioMedical Research, CH-4002 Basel, Switzerland
| | - Jean Quancard
- Novartis Institutes for BioMedical Research, CH-4002 Basel, Switzerland
| |
Collapse
|
28
|
Grondona P, Bucher P, Schulze-Osthoff K, Hailfinger S, Schmitt A. NF-κB Activation in Lymphoid Malignancies: Genetics, Signaling, and Targeted Therapy. Biomedicines 2018; 6:biomedicines6020038. [PMID: 29587428 PMCID: PMC6027339 DOI: 10.3390/biomedicines6020038] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Revised: 03/20/2018] [Accepted: 03/22/2018] [Indexed: 12/12/2022] Open
Abstract
The NF-κB transcription factor family plays a crucial role in lymphocyte proliferation and survival. Consequently, aberrant NF-κB activation has been described in a variety of lymphoid malignancies, including diffuse large B-cell lymphoma, Hodgkin lymphoma, and adult T-cell leukemia. Several factors, such as persistent infections (e.g., with Helicobacter pylori), the pro-inflammatory microenvironment of the cancer, self-reactive immune receptors as well as genetic lesions altering the function of key signaling effectors, contribute to constitutive NF-κB activity in these malignancies. In this review, we will discuss the molecular consequences of recurrent genetic lesions affecting key regulators of NF-κB signaling. We will particularly focus on the oncogenic mechanisms by which these alterations drive deregulated NF-κB activity and thus promote the growth and survival of the malignant cells. As the concept of a targeted therapy based on the mutational status of the malignancy has been supported by several recent preclinical and clinical studies, further insight in the function of NF-κB modulators and in the molecular mechanisms governing aberrant NF-κB activation observed in lymphoid malignancies might lead to the development of additional treatment strategies and thus improve lymphoma therapy.
Collapse
Affiliation(s)
- Paula Grondona
- Interfaculty Institute for Biochemistry, Eberhard Karls University of Tuebingen, Hoppe-Seyler-Str. 4, 72076 Tuebingen, Germany.
| | - Philip Bucher
- Interfaculty Institute for Biochemistry, Eberhard Karls University of Tuebingen, Hoppe-Seyler-Str. 4, 72076 Tuebingen, Germany.
| | - Klaus Schulze-Osthoff
- Interfaculty Institute for Biochemistry, Eberhard Karls University of Tuebingen, Hoppe-Seyler-Str. 4, 72076 Tuebingen, Germany.
| | - Stephan Hailfinger
- Interfaculty Institute for Biochemistry, Eberhard Karls University of Tuebingen, Hoppe-Seyler-Str. 4, 72076 Tuebingen, Germany.
| | - Anja Schmitt
- Interfaculty Institute for Biochemistry, Eberhard Karls University of Tuebingen, Hoppe-Seyler-Str. 4, 72076 Tuebingen, Germany.
| |
Collapse
|
29
|
Gajanayaka N, O'Hara S, Konarski Y, Fernandes J, Muthumani K, Kozlowski M, Angel JB, Kumar A. HIV and HIV-Tat inhibit LPS-induced IL-27 production in human macrophages by distinct intracellular signaling pathways. J Leukoc Biol 2017; 102:925-939. [PMID: 28698313 DOI: 10.1189/jlb.4a0716-332rr] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Revised: 07/09/2017] [Accepted: 07/11/2017] [Indexed: 11/24/2022] Open
Abstract
Monocyte-derived Mϕs (MDMs) from HIV-infected patients and MDM infected in vitro with HIV exhibit a reduced ability to secrete various cytokines, including IL-12. Recently, IL-27, an IL-12 family cytokine, was shown to inhibit HIV replication in Mϕ. Whether HIV infection or HIV accessory protein(s) impact IL-27 production in Mϕs remains unknown. Herein, we show that in vitro HIV infection, as well as intracellular HIV-Tat (Tat) and Tat peptides, inhibit LPS-induced IL-27 production in human MDMs, suggesting impairment of the TLR4 signaling pathway. To understand the signaling pathways governing HIV or Tat-mediated inhibition of LPS-induced IL-27 production, we first demonstrated that p38 MAPK, PI3K, Src-homology region 2 domain-containing tyrosine phosphatase 1 (SHP-1), and Src kinases regulate LPS-induced IL-27 production in MDMs. Tat caused down-regulation of TNFR-associated factor (TRAF)-6 and inhibitor of apoptosis 1 (cIAP-1) and subsequently decreased phosphorylation of downstream PI3K and p38 MAPKs, which were implicated in LPS-induced IL-27 production. Whereas SHP-1 and Src kinases regulated LPS-induced IL-27 production, Tat did not inhibit these kinases, suggesting that they were not involved in Tat-mediated inhibition of LPS-induced IL-27 production. In contrast to Tat, in vitro HIV infection of MDM inhibited LPS-induced IL-27 production via inhibition of p38 MAPK activation. Overall, HIV and Tat inhibit LPS-induced IL-27 production in human macrophages via distinct mechanisms: Tat through the inhibition of cIAP-1-TRAF-6 and subsequent inhibition of PI3K and p38 MAPKs, whereas HIV through the inhibition of p38 MAPK activation.
Collapse
Affiliation(s)
- Niranjala Gajanayaka
- Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, Ontario, Canada.,Research Institute, Children's Hospital of Eastern Ontario, Ottawa, Ontario, Canada
| | - Shifawn O'Hara
- Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, Ontario, Canada.,Research Institute, Children's Hospital of Eastern Ontario, Ottawa, Ontario, Canada
| | - Yulia Konarski
- Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, Ontario, Canada.,Research Institute, Children's Hospital of Eastern Ontario, Ottawa, Ontario, Canada
| | - Jason Fernandes
- Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, Ontario, Canada
| | - Kar Muthumani
- Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA
| | - Maya Kozlowski
- Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, Ontario, Canada
| | - Jonathan B Angel
- Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, Ontario, Canada.,Ottawa Hospital Research Institute, Ottawa, Ontario, Canada; and
| | - Ashok Kumar
- Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, Ontario, Canada; .,Research Institute, Children's Hospital of Eastern Ontario, Ottawa, Ontario, Canada.,Department Pathology and Laboratory Medicine, University of Ottawa, Ottawa, Ontario, Canada
| |
Collapse
|
30
|
Meininger I, Krappmann D. Lymphocyte signaling and activation by the CARMA1-BCL10-MALT1 signalosome. Biol Chem 2017; 397:1315-1333. [PMID: 27420898 DOI: 10.1515/hsz-2016-0216] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Accepted: 07/10/2016] [Indexed: 12/16/2022]
Abstract
The CARMA1-BCL10-MALT1 (CBM) signalosome triggers canonical NF-κB signaling and lymphocyte activation upon antigen-receptor stimulation. Genetic studies in mice and the analysis of human immune pathologies unveiled a critical role of the CBM complex in adaptive immune responses. Great progress has been made in elucidating the fundamental mechanisms that dictate CBM assembly and disassembly. By bridging proximal antigen-receptor signaling to downstream signaling pathways, the CBM complex exerts a crucial scaffolding function. Moreover, the MALT1 subunit confers a unique proteolytic activity that is key for lymphocyte activation. Deregulated 'chronic' CBM signaling drives constitutive NF-κB signaling and MALT1 activation, which contribute to the development of autoimmune and inflammatory diseases as well as lymphomagenesis. Thus, the processes that govern CBM activation and function are promising targets for the treatment of immune disorders. Here, we summarize the current knowledge on the functions and mechanisms of CBM signaling in lymphocytes and how CBM deregulations contribute to aberrant signaling in malignant lymphomas.
Collapse
|
31
|
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, Bornancin F. Two Antagonistic MALT1 Auto-Cleavage Mechanisms Reveal a Role for TRAF6 to Unleash MALT1 Activation. PLoS One 2017; 12:e0169026. [PMID: 28052131 PMCID: PMC5214165 DOI: 10.1371/journal.pone.0169026] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Accepted: 12/09/2016] [Indexed: 11/18/2022] Open
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.
Collapse
Affiliation(s)
- Stefanie Ginster
- Novartis Institutes for BioMedical Research, Novartis Campus, Basel, Switzerland
| | - Maureen Bardet
- Novartis Institutes for BioMedical Research, Novartis Campus, Basel, Switzerland
| | - Adeline Unterreiner
- Novartis Institutes for BioMedical Research, Novartis Campus, Basel, Switzerland
| | - Claire Malinverni
- Novartis Institutes for BioMedical Research, Novartis Campus, Basel, Switzerland
| | - Florian Renner
- Novartis Institutes for BioMedical Research, Novartis Campus, Basel, Switzerland
| | - Stephen Lam
- Novartis Institutes for BioMedical Research, Novartis Campus, Basel, Switzerland
| | - Felix Freuler
- Novartis Institutes for BioMedical Research, Novartis Campus, Basel, Switzerland
| | - Bertran Gerrits
- Novartis Institutes for BioMedical Research, Novartis Campus, Basel, Switzerland
| | - Johannes Voshol
- Novartis Institutes for BioMedical Research, Novartis Campus, Basel, Switzerland
| | - Thomas Calzascia
- Novartis Institutes for BioMedical Research, Novartis Campus, Basel, Switzerland
| | - Catherine H. Régnier
- Novartis Institutes for BioMedical Research, Novartis Campus, Basel, Switzerland
| | - Martin Renatus
- Novartis Institutes for BioMedical Research, Novartis Campus, Basel, Switzerland
| | - Rainer Nikolay
- Novartis Institutes for BioMedical Research, Novartis Campus, Basel, Switzerland
| | - Laura Israël
- Novartis Institutes for BioMedical Research, Novartis Campus, Basel, Switzerland
| | - Frédéric Bornancin
- Novartis Institutes for BioMedical Research, Novartis Campus, Basel, Switzerland
- * E-mail:
| |
Collapse
|
32
|
Krappmann D, Vincendeau M. Mechanisms of NF-κB deregulation in lymphoid malignancies. Semin Cancer Biol 2016; 39:3-14. [DOI: 10.1016/j.semcancer.2016.05.002] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Revised: 05/27/2016] [Accepted: 05/31/2016] [Indexed: 12/17/2022]
|
33
|
Shen W, Du R, Li J, Luo X, Zhao S, Chang A, Zhou W, Gao R, Luo D, Wang J, Hao N, Liu Y, Chen Y, Luo Y, Sun P, Yang S, Luo N, Xiang R. TIFA suppresses hepatocellular carcinoma progression via MALT1-dependent and -independent signaling pathways. Signal Transduct Target Ther 2016; 1:16013. [PMID: 29263897 PMCID: PMC5661659 DOI: 10.1038/sigtrans.2016.13] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Revised: 06/23/2016] [Accepted: 07/04/2016] [Indexed: 02/05/2023] Open
Abstract
TIFA, also called T2BP, was first identified using yeast two-hybrid screening. Our previous work showed that TIFA suppresses hepatocellular carcinoma (HCC) progression via apoptosis and cell cycle arrest. However, the mechanism by which this TIFA suppression occurs remains unclear. Here we demonstrated that TIFA-induced apoptosis demonstrates two distinct time patterns (i.e., at 48 h and >7 days) when TIFA reconstitution occurs. Moreover, we found that MALT1 (a competitor of TIFA) plays a crucial role in short-duration TIFA reconstitution. In this regard, MALT1 silencing with shRNA markedly enhances TIFA-induced apoptosis in vitro and in vivo. In addition, TIFA overexpression triggers JNK and p38 activation in long-duration TIFA reconstitution through TRAF6 binding. In particular, JNK activation leads to TIFA-induced apoptosis while p38 activation governs TIFA-induced cell cycle arrest by p53-p21 signaling in vitro and in vivo. Our data suggest a novel mechanism by which TIFA suppresses HCC progression via both MALT1-dependent and MALT1-independent signaling pathways. This may provide insights into a novel targets where HCC progression may be vulnerable to clinical treatment.
Collapse
Affiliation(s)
- Wenzhi Shen
- Department of Immunology, School of Medicine, Nankai University, Tianjin, China
| | - Renle Du
- Department of Immunology, School of Medicine, Nankai University, Tianjin, China
| | - Jun Li
- Department of Immunology, School of Medicine, Nankai University, Tianjin, China
| | - Xiaohe Luo
- Department of Immunology, School of Medicine, Nankai University, Tianjin, China
| | - Shuangtao Zhao
- Department of Immunology, School of Medicine, Nankai University, Tianjin, China
| | - Antao Chang
- Department of Immunology, School of Medicine, Nankai University, Tianjin, China
| | - Wei Zhou
- Department of Immunology, School of Medicine, Nankai University, Tianjin, China
| | - Ruifang Gao
- Department of Immunology, School of Medicine, Nankai University, Tianjin, China
| | - Dehong Luo
- Department of Immunology, School of Medicine, Nankai University, Tianjin, China
| | - Juan Wang
- Department of Immunology, School of Medicine, Nankai University, Tianjin, China
| | - Na Hao
- Department of Immunology, School of Medicine, Nankai University, Tianjin, China
| | - Yanhua Liu
- International Joint Center for Biomedical Research of the Ministry of Education, Tianjin, China
| | - Yanan Chen
- International Joint Center for Biomedical Research of the Ministry of Education, Tianjin, China
| | - Yunping Luo
- Department of Immunology, Institute of Basic Medical Science, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Peiqing Sun
- Department of Cancer Biology and Comprehensive Cancer Center, Wake Forest University Medical Center, Winston-Salem, North Carolina, USA
| | - Shengyong Yang
- West China Hospital, Molecular Medicine Research Centre, State Key Lab Biotherapy, Sichuan University, Chengdu, China
| | - Na Luo
- Department of Immunology, School of Medicine, Nankai University, Tianjin, China
| | - Rong Xiang
- Department of Immunology, School of Medicine, Nankai University, Tianjin, China
- International Joint Center for Biomedical Research of the Ministry of Education, Tianjin, China
| |
Collapse
|
34
|
Alternative splicing of MALT1 controls signalling and activation of CD4(+) T cells. Nat Commun 2016; 7:11292. [PMID: 27068814 PMCID: PMC4832065 DOI: 10.1038/ncomms11292] [Citation(s) in RCA: 81] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2015] [Accepted: 03/09/2016] [Indexed: 12/25/2022] Open
Abstract
MALT1 channels proximal T-cell receptor (TCR) signalling to downstream signalling pathways. With MALT1A and MALT1B two conserved splice variants exist and we demonstrate here that MALT1 alternative splicing supports optimal T-cell activation. Inclusion of exon7 in MALT1A facilitates the recruitment of TRAF6, which augments MALT1 scaffolding function, but not protease activity. Naive CD4+ T cells express almost exclusively MALT1B and MALT1A expression is induced by TCR stimulation. We identify hnRNP U as a suppressor of exon7 inclusion. Whereas selective depletion of MALT1A impairs T-cell signalling and activation, downregulation of hnRNP U enhances MALT1A expression and T-cell activation. Thus, TCR-induced alternative splicing augments MALT1 scaffolding to enhance downstream signalling and to promote optimal T-cell activation. MALT1 regulates NFκB signalling both as a scaffolding protein and as a protease. Here the authors show that during T cell activation the expression of MALT1 gene switches to an alternatively spliced variant, which increases TCR signal transduction due to enhanced TRAF6 binding.
Collapse
|
35
|
Rosebeck S, Lim MS, Elenitoba-Johnson KSJ, McAllister-Lucas LM, Lucas PC. API2-MALT1 oncoprotein promotes lymphomagenesis via unique program of substrate ubiquitination and proteolysis. World J Biol Chem 2016; 7:128-137. [PMID: 26981201 PMCID: PMC4768116 DOI: 10.4331/wjbc.v7.i1.128] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Revised: 10/16/2015] [Accepted: 12/08/2015] [Indexed: 02/05/2023] Open
Abstract
Lymphoma of mucosa-associated lymphoid tissue (MALT lymphoma) is the most common extranodal B cell tumor and accounts for 8% of non-Hodgkin’s lymphomas. Gastric MALT lymphoma is the best-studied example and is a prototypical neoplasm that occurs in the setting of chronic inflammation brought on by persistent infection or autoimmune disease. Cytogenetic abnormalities are commonly acquired during the course of disease and the most common is chromosomal translocation t(11;18)(q21;q21), which creates the API2-MALT1 fusion oncoprotein. t(11;18)-positive lymphomas can be clinically aggressive and have a higher rate of dissemination than t(11;18)-negative tumors. Many cancers, including MALT lymphomas, characteristically exhibit deregulated over-activation of cellular survival pathways, such as the nuclear factor-κB (NF-κB) pathway. Molecular characterization of API2-MALT1 has revealed it to be a potent activator of NF-κB, which is required for API2-MALT1-induced cellular transformation, however the mechanisms by which API2-MALT1 exerts these effects are only recently becoming apparent. The API2 moiety of the fusion binds tumor necrosis factor (TNF) receptor associated factor (TRAF) 2 and receptor interacting protein 1 (RIP1), two proteins essential for TNF receptor-induced NF-κB activation. By effectively mimicking ligand-bound TNF receptor, API2-MALT1 promotes TRAF2-dependent ubiquitination of RIP1, which then acts as a scaffold for nucleating and activating the canonical NF-κB machinery. Activation occurs, in part, through MALT1 moiety-dependent recruitment of TRAF6, which can directly modify NF-κB essential modulator, the principal downstream regulator of NF-κB. While the intrinsic MALT1 protease catalytic activity is dispensable for this canonical NF-κB signaling, it is critical for non-canonical NF-κB activation. In this regard, API2-MALT1 recognizes NF-κB inducing kinase (NIK), the essential upstream regulator of non-canonical NF-κB, and cleaves it to generate a stable, constitutively active fragment. Thus, API2-MALT1 harnesses multiple unique pathways to achieve deregulated NF-κB activation. Emerging data from our group and others have also detailed additional gain-of-function activities of API2-MALT1 that extend beyond NF-κB activation. Specifically, API2-MALT1 recruits and subverts multiple other signaling factors, including LIM domain and actin-binding protein 1 (LIMA1) and Smac/DIABLO. Like NIK, LIMA1 represents a unique substrate for API2-MALT1 protease activity, but unlike NIK, its cleavage sets in motion a major NF-κB-independent pathway for promoting oncogenesis. In this review, we highlight the most recent results characterizing these unique and diverse gain-of-function activities of API2-MALT1 and how they contribute to lymphomagenesis.
Collapse
|
36
|
Jaworski M, Thome M. The paracaspase MALT1: biological function and potential for therapeutic inhibition. Cell Mol Life Sci 2016; 73:459-73. [PMID: 26507244 PMCID: PMC4713714 DOI: 10.1007/s00018-015-2059-z] [Citation(s) in RCA: 84] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Revised: 09/15/2015] [Accepted: 09/29/2015] [Indexed: 02/07/2023]
Abstract
The paracaspase MALT1 has a central role in the activation of lymphocytes and other immune cells including myeloid cells, mast cells and NK cells. MALT1 activity is required not only for the immune response, but also for the development of natural Treg cells that keep the immune response in check. Exaggerated MALT1 activity has been associated with the development of lymphoid malignancies, and recently developed MALT1 inhibitors show promising anti-tumor effects in xenograft models of diffuse large B cell lymphoma. In this review, we provide an overview of the present understanding of MALT1's function, and discuss possibilities for its therapeutic targeting based on recently developed inhibitors and animal models.
Collapse
Affiliation(s)
- Maike Jaworski
- Department of Biochemistry, University of Lausanne, 1066, Epalinges, Switzerland
| | - Margot Thome
- Department of Biochemistry, University of Lausanne, 1066, Epalinges, Switzerland.
| |
Collapse
|
37
|
Afonina IS, Elton L, Carpentier I, Beyaert R. MALT1--a universal soldier: multiple strategies to ensure NF-κB activation and target gene expression. FEBS J 2015; 282:3286-97. [PMID: 25996250 DOI: 10.1111/febs.13325] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2015] [Accepted: 05/19/2015] [Indexed: 12/27/2022]
Abstract
The paracaspase MALT1 (mucosa associated lymphoid tissue lymphoma translocation gene 1) is an intracellular signaling protein that plays a key role in innate and adaptive immunity. It is essential for nuclear factor κB (NF-κB) activation and proinflammatory gene expression downstream of several cell surface receptors. MALT1 has been most studied in the context of T-cell receptor-induced NF-κB signaling, supporting T-cell activation and proliferation. In addition, MALT1 hyperactivation is associated with specific subtypes of B-cell lymphoma, where it controls tumor cell proliferation and survival. For a long time, MALT1 was believed to function solely as a scaffold protein, providing a platform for the assembly of other NF-κB signaling proteins. However, this view changed dramatically when MALT1 was found to have proteolytic activity that further fine-tunes signaling. MALT1 proteolytic activity is essential for T-cell activation and lymphomagenesis, suggesting that MALT1 is a promising therapeutic target for the treatment of autoimmune diseases and distinct lymphoma entities. However, interference with MALT1 activity may pose a dangerous threat to the normal functioning of the immune system and should be evaluated with great care. Here we discuss the current knowledge on the scaffold and protease functions of MALT1, including an overview of its substrates and the functional implications of their cleavage.
Collapse
Affiliation(s)
- Inna S Afonina
- Inflammation Research Center, Unit of Molecular Signal Transduction in Inflammation, VIB, Department of Biomedical Molecular Biology, Ghent University, Belgium
| | - Lynn Elton
- Inflammation Research Center, Unit of Molecular Signal Transduction in Inflammation, VIB, Department of Biomedical Molecular Biology, Ghent University, Belgium
| | - Isabelle Carpentier
- Inflammation Research Center, Unit of Molecular Signal Transduction in Inflammation, VIB, Department of Biomedical Molecular Biology, Ghent University, Belgium
| | - Rudi Beyaert
- Inflammation Research Center, Unit of Molecular Signal Transduction in Inflammation, VIB, Department of Biomedical Molecular Biology, Ghent University, Belgium
| |
Collapse
|
38
|
Sasaki Y, Iwai K. Roles of the NF-κB Pathway in B-Lymphocyte Biology. Curr Top Microbiol Immunol 2015; 393:177-209. [PMID: 26275874 DOI: 10.1007/82_2015_479] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
NF-κB was originally identified as a family of transcription factors that bind the enhancer of the immunoglobulin κ light-chain gene. Although its function in the regulation of immunoglobulin κ light-chain gene remains unclear, NF-κB plays critical roles in development, survival, and activation of B lymphocytes. In B cells, many receptors, including B-cell antigen receptor (BCR), activate NF-κB pathway, and the molecular mechanism of receptor-mediated activation of IκB kinase (IKK) complex has been partially revealed. In addition to normal B lymphocytes, NF-κB is also involved in the growth of some types of B-cell lymphomas, and many oncogenic mutations involved in constitutive activation of the NF-κB pathway were recently identified in such cancers. In this review, we first summarize the function of NF-κB in B-cell development and activation, and then describe recent progress in understanding the molecular mechanism of receptor-mediated activation of the IKK complex, focusing on the roles of the ubiquitin system. In the last section, we describe oncogenic mutations that induce NF-κB activation in B-cell lymphoma.
Collapse
Affiliation(s)
- Yoshiteru Sasaki
- Department of Molecular and Cellular Physiology, Graduate School of Medicine, Kyoto University, Kyoto, 606-8501, Japan.
| | - Kazuhiro Iwai
- Department of Molecular and Cellular Physiology, Graduate School of Medicine, Kyoto University, Kyoto, 606-8501, Japan
| |
Collapse
|
39
|
Jaworski M, Marsland BJ, Gehrig J, Held W, Favre S, Luther SA, Perroud M, Golshayan D, Gaide O, Thome M. Malt1 protease inactivation efficiently dampens immune responses but causes spontaneous autoimmunity. EMBO J 2014; 33:2765-81. [PMID: 25319413 PMCID: PMC4282555 DOI: 10.15252/embj.201488987] [Citation(s) in RCA: 114] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2014] [Revised: 09/17/2014] [Accepted: 09/17/2014] [Indexed: 01/22/2023] Open
Abstract
The protease activity of the paracaspase Malt1 has recently gained interest as a drug target for immunomodulation and the treatment of diffuse large B-cell lymphomas. To address the consequences of Malt1 protease inactivation on the immune response in vivo, we generated knock-in mice expressing a catalytically inactive C472A mutant of Malt1 that conserves its scaffold function. Like Malt1-deficient mice, knock-in mice had strong defects in the activation of lymphocytes, NK and dendritic cells, and the development of B1 and marginal zone B cells and were completely protected against the induction of autoimmune encephalomyelitis. Malt1 inactivation also protected the mice from experimental induction of colitis. However, Malt1 knock-in mice but not Malt1-deficient mice spontaneously developed signs of autoimmune gastritis that correlated with an absence of Treg cells, an accumulation of T cells with an activated phenotype and high serum levels of IgE and IgG1. Thus, removal of the enzymatic activity of Malt1 efficiently dampens the immune response, but favors autoimmunity through impaired Treg development, which could be relevant for therapeutic Malt1-targeting strategies.
Collapse
Affiliation(s)
- Maike Jaworski
- Department of Biochemistry, Center of Immunity and Infection, University of Lausanne, Epalinges, Switzerland
| | - Ben J Marsland
- Centre Hospitalier Universitaire Vaudois, Service de Pneumologie, Lausanne, Switzerland
| | - Jasmine Gehrig
- Department of Oncology, Ludwig Center for Cancer Research, University of Lausanne, Epalinges, Switzerland
| | - Werner Held
- Department of Oncology, Ludwig Center for Cancer Research, University of Lausanne, Epalinges, Switzerland
| | - Stéphanie Favre
- Department of Biochemistry, Center of Immunity and Infection, University of Lausanne, Epalinges, Switzerland
| | - Sanjiv A Luther
- Department of Biochemistry, Center of Immunity and Infection, University of Lausanne, Epalinges, Switzerland
| | - Mai Perroud
- Department of Biochemistry, Center of Immunity and Infection, University of Lausanne, Epalinges, Switzerland
| | - Déla Golshayan
- Centre Hospitalier Universitaire Vaudois, Transplantation Centre, Lausanne, Switzerland
| | - Olivier Gaide
- Centre Hospitalier Universitaire Vaudois, Service de Dermatologie et Vénéréologie, Lausanne, Switzerland
| | - Margot Thome
- Department of Biochemistry, Center of Immunity and Infection, University of Lausanne, Epalinges, Switzerland
| |
Collapse
|
40
|
Hailfinger S, Lenz G, Thome M. Targeting B-cell lymphomas with inhibitors of the MALT1 paracaspase. Curr Opin Chem Biol 2014; 23:47-55. [PMID: 25285878 DOI: 10.1016/j.cbpa.2014.09.025] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2014] [Revised: 09/15/2014] [Accepted: 09/18/2014] [Indexed: 11/15/2022]
Abstract
The paracaspase MALT1 is an Arg-specific protease that cleaves multiple substrates to promote lymphocyte proliferation and survival. The catalytic activity of MALT1 is normally tightly regulated by antigen receptor triggering, which promotes MALT1 activation by its inducible monoubiquitination-dependent dimerization. Constitutive MALT1 activity is a hallmark of specific subsets of B-cell lymphomas, which are characterized by chromosomal translocations or point mutations that activate MALT1 or its upstream regulators. Recent findings suggest that such lymphomas may be sensitive to treatment with MALT1 inhibitors. Here we review recent progress in the understanding of MALT1 function and regulation, and the development of small molecule MALT1 inhibitors for therapeutic applications.
Collapse
Affiliation(s)
- Stephan Hailfinger
- Interfaculty Institute of Biochemistry, University of Tuebingen, D-72076 Tuebingen, Germany
| | - Georg Lenz
- Department of Hematology, Oncology and Tumor Immunology, Charité Universitätsmedizin Berlin, Germany
| | - Margot Thome
- Department of Biochemistry, University of Lausanne, CH-1066 Epalinges, Switzerland.
| |
Collapse
|
41
|
Baens M, Bonsignore L, Somers R, Vanderheydt C, Weeks SD, Gunnarsson J, Nilsson E, Roth RG, Thome M, Marynen P. MALT1 auto-proteolysis is essential for NF-κB-dependent gene transcription in activated lymphocytes. PLoS One 2014; 9:e103774. [PMID: 25105596 PMCID: PMC4126661 DOI: 10.1371/journal.pone.0103774] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2013] [Accepted: 07/03/2014] [Indexed: 11/30/2022] Open
Abstract
Mucosa-associated lymphoid tissue 1 (MALT1) controls antigen receptor–mediated signalling to nuclear factor κB (NF-κB) through both its adaptor and protease function. Upon antigen stimulation, MALT1 forms a complex with BCL10 and CARMA1, which is essential for initial IκBα phosphorylation and NF-κB nuclear translocation. Parallel induction of MALT1 protease activity serves to inactivate negative regulators of NF-κB signalling, such as A20 and RELB. Here we demonstrate a key role for auto-proteolytic MALT1 cleavage in B- and T-cell receptor signalling. MALT1 cleavage occurred after Arginine 149, between the N-terminal death domain and the first immunoglobulin-like region, and did not affect its proteolytic activity. Jurkat T cells expressing an un-cleavable MALT1-R149A mutant showed unaltered initial IκBα phosphorylation and normal nuclear accumulation of NF-κB subunits. Nevertheless, MALT1 cleavage was required for optimal activation of NF-κB reporter genes and expression of the NF-κB targets IL-2 and CSF2. Transcriptome analysis confirmed that MALT1 cleavage after R149 was required to induce NF-κB transcriptional activity in Jurkat T cells. Collectively, these data demonstrate that auto-proteolytic MALT1 cleavage controls antigen receptor-induced expression of NF-κB target genes downstream of nuclear NF-κB accumulation.
Collapse
Affiliation(s)
- Mathijs Baens
- Human Genome Laboratory, VIB Center for the Biology of Disease, Leuven, Belgium
- Human Genome Laboratory, Center for Human Genetics, KU Leuven, Leuven, Belgium
- * E-mail:
| | - Luca Bonsignore
- Department of Biochemistry, University of Lausanne, Epalinges, Switzerland
| | - Riet Somers
- Human Genome Laboratory, VIB Center for the Biology of Disease, Leuven, Belgium
- Human Genome Laboratory, Center for Human Genetics, KU Leuven, Leuven, Belgium
| | - Charlotte Vanderheydt
- Human Genome Laboratory, VIB Center for the Biology of Disease, Leuven, Belgium
- Human Genome Laboratory, Center for Human Genetics, KU Leuven, Leuven, Belgium
| | - Stephen D. Weeks
- Laboratory for Biocrystallography, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium
| | - Jenny Gunnarsson
- Reagent and Assay Development, Discovery Sciences, Innovative Medicines, AstraZeneca R&D, Mölndal, Sweden
| | - Ewa Nilsson
- Reagent and Assay Development, Discovery Sciences, Innovative Medicines, AstraZeneca R&D, Mölndal, Sweden
| | - Robert G. Roth
- Reagent and Assay Development, Discovery Sciences, Innovative Medicines, AstraZeneca R&D, Mölndal, Sweden
| | - Margot Thome
- Department of Biochemistry, University of Lausanne, Epalinges, Switzerland
| | - Peter Marynen
- Human Genome Laboratory, Center for Human Genetics, KU Leuven, Leuven, Belgium
| |
Collapse
|
42
|
Rosebeck S, Rehman AO, Apel IJ, Kohrt D, Appert A, O’Donnell MA, Ting AT, Du MQ, Baens M, Lucas PC, McAllister-Lucas LM. The API2-MALT1 fusion exploits TNFR pathway-associated RIP1 ubiquitination to promote oncogenic NF-κB signaling. Oncogene 2014; 33:2520-30. [PMID: 23770847 PMCID: PMC4237018 DOI: 10.1038/onc.2013.195] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2012] [Revised: 01/29/2013] [Accepted: 04/03/2013] [Indexed: 02/08/2023]
Abstract
The API2-MALT1 fusion oncoprotein is created by the recurrent t(11;18)(q21;q21) chromosomal translocation in mucosa-associated lymphoid tissue (MALT) lymphoma. We identified receptor interacting protein-1 (RIP1) as a novel API2-MALT1-associated protein, and demonstrate that RIP1 is required for API2-MALT1 to stimulate canonical nuclear factor kappa B (NF-κB). API2-MALT1 promotes ubiquitination of RIP1 at lysine (K) 377, which is necessary for full NF-κB activation. Furthermore, we found that TNF receptor-associated factor 2 (TRAF2) recruitment is required for API2-MALT1 to induce RIP1 ubiquitination, NF-κB activation and cellular transformation. Although both TRAF2 and RIP1 interact with the API2 moiety of API2-MALT1, this moiety alone is insufficient to induce RIP1 ubiquitination or activate NF-κB, indicating that API2-MALT1-dependent RIP1 ubiquitination represents a gain of function requiring the concerted actions of both the API2 and MALT1 moieties of the fusion. Intriguingly, constitutive RIP1 ubiquitination was recently demonstrated in several solid tumors, and now our study implicates RIP1 ubiquitination as a critical component of API2-MALT1-dependent lymphomagenesis.
Collapse
Affiliation(s)
- Shaun Rosebeck
- Department of Pediatrics and Communicable Diseases, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Aasia O. Rehman
- Department of Pediatrics and Communicable Diseases, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Ingrid J. Apel
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Dawn Kohrt
- Department of Pediatrics and Communicable Diseases, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Alex Appert
- Division of Molecular Histopathology, Department of Pathology, University of Cambridge, Laboratory Block, Addenbrooke’s Hospital, Cambridge, UK
| | | | - Adrian T. Ting
- Immunology Institute, Mount Sinai School of Medicine, New York, NY 10029, USA
| | - Ming-Qing Du
- Division of Molecular Histopathology, Department of Pathology, University of Cambridge, Laboratory Block, Addenbrooke’s Hospital, Cambridge, UK
| | - Mathijs Baens
- Human Genome Laboratory, Molecular Genetics, Center for Human Genetics, Catholic University Leuven, Belgium
- Human Genome Laboratory, Department of Molecular and Developmental Genetics, Flanders Institute for Biotechnology (VIB), B-3000 Leuven, Belgium
| | - Peter C. Lucas
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Linda M. McAllister-Lucas
- Department of Pediatrics and Communicable Diseases, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| |
Collapse
|
43
|
Thieblemont C, Bertoni F, Copie-Bergman C, Ferreri AJ, Ponzoni M. Chronic inflammation and extra-nodal marginal-zone lymphomas of MALT-type. Semin Cancer Biol 2014; 24:33-42. [DOI: 10.1016/j.semcancer.2013.11.005] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2013] [Revised: 11/23/2013] [Accepted: 11/29/2013] [Indexed: 12/26/2022]
|
44
|
Mechanisms and consequences of constitutive NF-κB activation in B-cell lymphoid malignancies. Oncogene 2014; 33:5655-65. [DOI: 10.1038/onc.2013.565] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2013] [Revised: 12/09/2013] [Accepted: 12/09/2013] [Indexed: 12/13/2022]
|
45
|
Yang C, David L, Qiao Q, Damko E, Wu H. The CBM signalosome: potential therapeutic target for aggressive lymphoma? Cytokine Growth Factor Rev 2013; 25:175-83. [PMID: 24411492 DOI: 10.1016/j.cytogfr.2013.12.008] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2013] [Accepted: 12/15/2013] [Indexed: 02/02/2023]
Abstract
The CBM signalosome plays a pivotal role in mediating antigen-receptor induced NF-κB signaling to regulate lymphocyte functions. The CBM complex forms filamentous structure and recruits downstream signaling components to activate NF-κB. MALT1, the protease component in the CBM complex, cleaves key proteins in the feedback loop of the NF-κB signaling pathway and enhances NF-κB activation. The aberrant activity of the CBM complex has been linked to aggressive lymphoma. Recent years have witnessed dramatic progresses in understanding the assembly mechanism of the CBM complex, and advances in the development of targeted therapy for aggressive lymphoma. Here, we will highlight these progresses and give an outlook on the potential translation of this knowledge from bench to bedside for aggressive lymphoma patients.
Collapse
Affiliation(s)
- Chenghua Yang
- Joint Center for Translational Research of Chronic Diseases, Changhai Hospital, The Second Military Medical University, Shanghai 2000433, China; Department of Molecular Pharmacology and Chemistry, Sloan-Kettering Institute, New York, NY 10021, USA.
| | - Liron David
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA; Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA 02115, USA
| | - Qi Qiao
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA; Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA 02115, USA
| | - Ermelinda Damko
- Department of Biochemistry, Weill Cornell Medical College, New York, NY 10065, USA
| | - Hao Wu
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA; Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA 02115, USA; Department of Biochemistry, Weill Cornell Medical College, New York, NY 10065, USA
| |
Collapse
|
46
|
Stack J, Hurst TP, Flannery SM, Brennan K, Rupp S, Oda SI, Khan AR, Bowie AG. Poxviral protein A52 stimulates p38 mitogen-activated protein kinase (MAPK) activation by causing tumor necrosis factor receptor-associated factor 6 (TRAF6) self-association leading to transforming growth factor β-activated kinase 1 (TAK1) recruitment. J Biol Chem 2013; 288:33642-33653. [PMID: 24114841 DOI: 10.1074/jbc.m113.485490] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Vaccinia virus encodes a number of proteins that inhibit and manipulate innate immune signaling pathways that also have a role in virulence. These include A52, a protein shown to inhibit IL-1- and Toll-like receptor-stimulated NFκB activation, via interaction with interleukin-1 receptor-associated kinase 2 (IRAK2). Interestingly, A52 was also found to activate p38 MAPK and thus enhance Toll-like receptor-dependent IL-10 induction, which was TRAF6-dependent, but the manner in which A52 manipulates TRAF6 to stimulate p38 activation was unclear. Here, we show that A52 has a non-canonical TRAF6-binding motif that is essential for TRAF6 binding and p38 activation but dispensable for NFκB inhibition and IRAK2 interaction. Wild-type A52, but not a mutant defective in p38 activation and TRAF6 binding (F154A), caused TRAF6 oligomerization and subsequent TRAF6-TAK1 association. The crystal structure of A52 shows that it adopts a Bcl2-like fold and exists as a dimer in solution. Residue Met-65 was identified as being located in the A52 dimer interface, and consistent with that, A52-M65E was impaired in its ability to dimerize. A52-M65E although capable of interacting with TRAF6, was unable to cause either TRAF6 self-association, induce the TRAF6-TAK1 association, or activate p38 MAPK. The results suggest that an A52 dimer causes TRAF6 self-association, leading to TAK1 recruitment and p38 activation. This reveals a molecular mechanism whereby poxviruses manipulate TRAF6 to activate MAPKs (which can be proviral) without stimulating antiviral NFκB activation.
Collapse
Affiliation(s)
- Julianne Stack
- Immunology Research Centre, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland
| | - Tara P Hurst
- Immunology Research Centre, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland
| | - Sinead M Flannery
- Immunology Research Centre, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland
| | - Kiva Brennan
- Immunology Research Centre, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland
| | - Sebastian Rupp
- Immunology Research Centre, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland
| | - Shun-Ichiro Oda
- Immunology Research Centre, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland
| | - Amir R Khan
- Immunology Research Centre, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland
| | - Andrew G Bowie
- Immunology Research Centre, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland.
| |
Collapse
|
47
|
Vincendeau M, Nagel D, Eitelhuber AC, Krappmann D. MALT1 paracaspase: a unique protease involved in B-cell lymphomagenesis. Int J Hematol Oncol 2013. [DOI: 10.2217/ijh.13.45] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
SUMMARY MALT1 is a key regulator of adaptive immunity. MALT1-dependent signaling events control survival, proliferation and differentiation of lymphocytes in response to T- or B-cell receptor stimulation. MALT1 not only regulates physiological lymphocyte activation, but also controls oncogenic signaling in distinct lymphoid malignancies. The fusion protein API2–MALT1 generated by the chromosomal translocation t(11;18) acts as an oncoprotein in the late stages of mucosa-associated lymphoid tissue lymphoma. Moreover, MALT1 is critical for survival and proliferation of the activated B-cell type of diffuse large B-cell lymphomas, one of the most aggressive entities of malignant lymphomas. On the molecular level, MALT1 serves a dual role by functioning as a signaling adaptor and a protease. Both of these functions are critical for triggering the adaptive immune response and for promoting lymphomagenesis. Recent data emphasize that MALT1 is a promising drug target for the treatment of aggressive lymphomas.
Collapse
Affiliation(s)
- Michelle Vincendeau
- Research Unit Cellular Signal Integration, Institute of Molecular Toxicology & Pharmacology, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Ingolstädter Landstraße 1, D-85764 Neuherberg, Germany
| | - Daniel Nagel
- Research Unit Cellular Signal Integration, Institute of Molecular Toxicology & Pharmacology, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Ingolstädter Landstraße 1, D-85764 Neuherberg, Germany
| | - Andrea C Eitelhuber
- Research Unit Cellular Signal Integration, Institute of Molecular Toxicology & Pharmacology, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Ingolstädter Landstraße 1, D-85764 Neuherberg, Germany
| | - Daniel Krappmann
- Research Unit Cellular Signal Integration, Institute of Molecular Toxicology & Pharmacology, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Ingolstädter Landstraße 1, D-85764 Neuherberg, Germany
| |
Collapse
|
48
|
Cabalzar K, Pelzer C, Wolf A, Lenz G, Iwaszkiewicz J, Zoete V, Hailfinger S, Thome M. Monoubiquitination and activity of the paracaspase MALT1 requires glutamate 549 in the dimerization interface. PLoS One 2013; 8:e72051. [PMID: 23977204 PMCID: PMC3747146 DOI: 10.1371/journal.pone.0072051] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2013] [Accepted: 07/06/2013] [Indexed: 12/17/2022] Open
Abstract
The mucosa-associated lymphoid tissue protein-1 (MALT1, also known as paracaspase) is a protease whose activity is essential for the activation of lymphocytes and the growth of cells derived from human diffuse large B-cell lymphomas of the activated B-cell subtype (ABC DLBCL). Crystallographic approaches have shown that MALT1 can form dimers via its protease domain, but why dimerization is relevant for the biological activity of MALT1 remains largely unknown. Using a molecular modeling approach, we predicted Glu 549 (E549) to be localized within the MALT1 dimer interface and thus potentially relevant. Experimental mutation of this residue into alanine (E549A) led to a complete impairment of MALT1 proteolytic activity. This correlated with an impaired capacity of the mutant to form dimers of the protease domain in vitro, and a reduced capacity to promote NF-κB activation and transcription of the growth-promoting cytokine interleukin-2 in antigen receptor-stimulated lymphocytes. Moreover, this mutant could not rescue the growth of ABC DLBCL cell lines upon MALT1 silencing. Interestingly, the MALT1 mutant E549A was unable to undergo monoubiquitination, which we identified previously as a critical step in MALT1 activation. Collectively, these findings suggest a model in which E549 at the dimerization interface is required for the formation of the enzymatically active, monoubiquitinated form of MALT1.
Collapse
Affiliation(s)
- Katrin Cabalzar
- Department of Biochemistry, University of Lausanne, Epalinges, Switzerland
| | - Christiane Pelzer
- Department of Biochemistry, University of Lausanne, Epalinges, Switzerland
| | - Annette Wolf
- Department of Hematology, Oncology and Tumor Immunology, Molecular Cancer Research Center, Charité - Universitätsmedizin Berlin, Germany
| | - Georg Lenz
- Department of Hematology, Oncology and Tumor Immunology, Molecular Cancer Research Center, Charité - Universitätsmedizin Berlin, Germany
| | | | - Vincent Zoete
- Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Stephan Hailfinger
- Department of Biochemistry, University of Lausanne, Epalinges, Switzerland
| | - Margot Thome
- Department of Biochemistry, University of Lausanne, Epalinges, Switzerland
- * E-mail:
| |
Collapse
|
49
|
Jou SY, Chang CC, Wu CH, Chen MR, Tsai CH, Chuang WH, Chen YH, Cheng AL, Doong SL. BCL10GFP fusion protein as a substrate for analysis of determinants required for mucosa-associated lymphoid tissue 1 (MALT1)-mediated cleavage. J Biomed Sci 2012; 19:85. [PMID: 23035874 PMCID: PMC3500650 DOI: 10.1186/1423-0127-19-85] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2012] [Accepted: 10/01/2012] [Indexed: 11/25/2022] Open
Abstract
Background MALT1 belongs to a family of paracaspase and modulates NF-κB signaling pathways through its scaffolding function and proteolytic activity. MALT1 cleaves protein substrates after a positively charged Arginine residue. BCL10, a 233 amino acids polypeptide, is identified as one of the MALT1 proteolytic substrates. MALT1 cleaves BCL10 at the C-terminal end of Arg228. A mere 5 amino acids difference between the substrate and the proteolytic product made it difficult to tell whether the cleavage event took place by using a simple western blot analysis. Here, BCL10GFP was constructed and utilized to examine the specificity and domain determinants for MALT1 cleavage in cells. Methods Various BCL10GFP constructs were transfected into HEK293T cell with MALT1 construct by using calcium phosphate-DNA precipitation method. Lysates of transfectants were resolved by SDS/PAGE and analyzed by western blot analysis. Results BCL10GFP was proteolytically processed by MALT1 as BCL10. The integrity of caspase recruitment domain (CARD) and MALT1-interacting domain on BCL10 were required for MALT1 proteolytic activity. Besides the invariant P1 cleavage site Arg228, P4 Leu225 played a role in defining BCL10 as a good substrate for MALT1. Conclusions We offered a way of monitoring the catalytic activity of MALT1 in HEK293T cells using BCL10GFP as a substrate. BCL10GFP can be utilized as a convenient tool for studying the determinants for efficient MALT1 cleavage in HEK293T cells
Collapse
Affiliation(s)
- Shin-Yi Jou
- Graduate Institute of Microbiology, College of Medicine, National Taiwan University, No.1, Section 1, Jen-Ai Road, Taipei 10051, Taiwan
| | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Beug ST, Cheung HH, LaCasse EC, Korneluk RG. Modulation of immune signalling by inhibitors of apoptosis. Trends Immunol 2012; 33:535-45. [PMID: 22836014 DOI: 10.1016/j.it.2012.06.004] [Citation(s) in RCA: 114] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2012] [Revised: 06/08/2012] [Accepted: 06/21/2012] [Indexed: 12/17/2022]
Abstract
The inhibitor of apoptosis (IAP) genes are critical regulators of multiple pathways that control cell death, proliferation, and differentiation. Several members of the IAP family regulate innate and adaptive immunity through modulation of signal transduction pathways, cytokine production, and cell survival. The regulation of immunity by the IAPs is primarily mediated through the ubiquitin ligase function of cellular IAP (cIAP)1, cIAP2, and X-linked IAP (XIAP), the targets of which impact nuclear factor (NF)-κB and mitogen-activated protein kinase (MAPK) signalling pathways. In addition, neuronal apoptosis inhibitory protein (NAIP), cIAP1, and cIAP2 modulate innate immune responses through control of the inflammasome complex. This review examines the role of mammalian IAPs in regulating immunity and describes the implications of a new class of pan-IAP antagonists for the treatment of immune disorders.
Collapse
Affiliation(s)
- Shawn T Beug
- Apoptosis Research Centre, Children's Hospital of Eastern Ontario Research Institute, Ottawa, Ontario K1H 8L1, Canada
| | | | | | | |
Collapse
|