1
|
Bauman BM, Stinson JR, Kallarakal MA, Huang LH, Frank AM, Sukumar G, Saucier N, Dalgard CL, Chan AY, Milner JD, Cooper MA, Snow AL. Dominant interfering CARD11 variants disrupt JNK signaling to promote GATA3 expression in T cells. J Exp Med 2025; 222:e20240272. [PMID: 40111223 PMCID: PMC11924952 DOI: 10.1084/jem.20240272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2024] [Revised: 12/06/2024] [Accepted: 02/07/2025] [Indexed: 03/22/2025] Open
Abstract
Several "primary atopic disorders" are linked to monogenic defects that attenuate TCR signaling, favoring T helper type 2 (TH2) cell differentiation. Patients with CARD11-associated atopy with dominant interference of NF-κB signaling (CADINS) disease suffer from severe atopy, caused by germline loss-of-function/dominant interfering (LOF/DI) CARD11 variants. The CARD11 scaffold enables TCR-induced activation of NF-κB, mTORC1, and JNK signaling, yet the function of CARD11-dependent JNK signaling in T cells remains nebulous. Here we show that CARD11 is critical for TCR-induced activation of JNK1 and JNK2, as well as canonical JUN/FOS AP-1 family members. Patient-derived CARD11 DI variants attenuated WT CARD11 JNK signaling, mirroring effects on NF-κB. Transcriptome profiling revealed JNK inhibition upregulated TCR-induced expression of GATA3 and NFATC1, key transcription factors for TH2 cell development. Further, impaired CARD11-JNK signaling was linked to enhanced GATA3 expression in CADINS patient T cells. Our findings reveal a novel intrinsic mechanism connecting impaired CARD11-dependent JNK signaling to enhanced GATA3/NFAT2 induction and TH2 cell differentiation in CADINS patients.
Collapse
Affiliation(s)
- Bradly M. Bauman
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA
| | - Jeffrey R. Stinson
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA
| | - Melissa A. Kallarakal
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Lei Haley Huang
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY, USA
- Department of Pediatrics, Columbia University Irving Medical Center, New York, NY, USA
| | - Andrew M. Frank
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA
- Student Bioinformatics Initiative, Center for Military Precision Health, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Gauthaman Sukumar
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA
- Student Bioinformatics Initiative, Center for Military Precision Health, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
- The American Genome Center, Center for Military Precision Health, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Nermina Saucier
- Division of Rheumatology/Immunology, Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, USA
| | - Clifton L. Dalgard
- The American Genome Center, Center for Military Precision Health, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
- Department of Anatomy, Physiology and Genetics, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Alice Y. Chan
- Department of Pediatrics, University of California San Francisco, San Francisco, CA, USA
| | - Joshua D. Milner
- Department of Pediatrics, Columbia University Irving Medical Center, New York, NY, USA
| | - Megan A. Cooper
- Division of Rheumatology/Immunology, Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, USA
| | - Andrew L. Snow
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| |
Collapse
|
2
|
Kang H, Maurer LM, Cheng J, Smyers M, Klei LR, Hu D, Hofstatter Azambuja J, Murai MJ, Mady A, Ahmad E, Trotta M, Klei HB, Liu M, Ekambaram P, Nikolovska-Coleska Z, Chen BB, McAllister-Lucas LM, Lucas PC. A small-molecule inhibitor of BCL10-MALT1 interaction abrogates progression of diffuse large B cell lymphoma. J Clin Invest 2025; 135:e164573. [PMID: 40231473 PMCID: PMC11996864 DOI: 10.1172/jci164573] [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: 08/25/2022] [Accepted: 02/11/2025] [Indexed: 04/16/2025] Open
Abstract
Diffuse large B cell lymphoma (DLBCL) is the most common type of non-Hodgkin lymphoma, and the activated B cell-like subtype (ABC-DLBCL) is associated with particularly poor outcome. Many ABC-DLBCLs harbor gain-of-function mutations that cause inappropriate assembly of the CARMA1-BCL10-MALT1 (CBM) signalosome, a cytoplasmic complex that drives downstream NF-κB signaling. MALT1 is the effector protein of the CBM signalosome such that its recruitment to the signalosome via interaction with BCL10 allows it to exert both protease and scaffolding activities that together synergize in driving NF-κB. Here, we demonstrate that a molecular groove located between two adjacent immunoglobulin-like domains within MALT1 represents a binding pocket for BCL10. Leveraging this discovery, we performed an in silico screen to identify small molecules that dock within this MALT1 groove and act as BCL10-MALT1 protein-protein interaction (PPI) inhibitors. We report the identification of M1i-124 as a first-in-class compound that blocks BCL10-MALT1 interaction, abrogates MALT1 scaffolding and protease activities, promotes degradation of BCL10 and MALT1 proteins, and specifically targets ABC-DLBCLs characterized by dysregulated MALT1. Our findings demonstrate that small-molecule inhibitors of BCL10-MALT1 interaction can function as potent agents to block MALT1 signaling in selected lymphomas, and provide a road map for clinical development of a new class of precision-medicine therapeutics.
Collapse
Affiliation(s)
| | - Lisa M. Maurer
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Jing Cheng
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota, USA
| | - Mei Smyers
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Linda R. Klei
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Dong Hu
- Department of Pathology and
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota, USA
| | - Juliana Hofstatter Azambuja
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota, USA
| | - Marcelo J. Murai
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Ahmed Mady
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Ejaz Ahmad
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Matthew Trotta
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Hanna B. Klei
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Minda Liu
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Prasanna Ekambaram
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | | | - Bill B. Chen
- Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Linda M. McAllister-Lucas
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania, USA
- Department of Pediatrics and Adolescent Medicine, Mayo Clinic, Rochester, Minnesota, USA
- Mayo Clinic Comprehensive Cancer Center, Rochester, Minnesota, USA
| | - Peter C. Lucas
- Department of Pathology and
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota, USA
- UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania, USA
- Mayo Clinic Comprehensive Cancer Center, Rochester, Minnesota, USA
| |
Collapse
|
3
|
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
|
4
|
Tao Y, Tian C, Qi S, Jia Z, Xu Z, Meng J, Xu G, Hu H, Wang X, Zhang T, You H, Lan X, Lin X, Yu G, Zhou H, Liu J, Zheng H. Targeting both death and paracaspase domains of MALT1 with antisense oligonucleotides overcomes resistance to immune-checkpoint inhibitors. NATURE CANCER 2025; 6:702-717. [PMID: 40075237 DOI: 10.1038/s43018-025-00930-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 02/13/2025] [Indexed: 03/14/2025]
Abstract
Targeting MALT1's paracaspase activity has been explored for B cell lymphoma and solid tumors. While the role of MALT1 in promoting cancer cell proliferation has been investigated, its involvement in immune evasion is unclear. Here we report that MALT1 promotes immune evasion through its paracaspase and death domain. In a paracaspase-dependent manner, MALT1 protects CD274 mRNA from degradation by its cleavage of ROQUIN1 and ROQUIN2. In a death-domain-dependent manner, MALT1 promotes the proliferation and polarization of tumor-associated macrophages to generate an immunosuppressive tumor microenvironment. Targeting MALT1 with antisense oligonucleotides inhibits PD-L1 expression in patient-derived tumor cells and suppresses the proliferation and M2-like polarization of tumor-associated macrophages isolated from patients with cancer. In preclinical models of solid tumors in female mice, treatment with MALT1 antisense oligonucleotides overcomes resistance to immune-checkpoint inhibitors. Together, our study demonstrates that targeting MALT1 is a potential strategy to overcome immune-checkpoint inhibitor resistance.
Collapse
Affiliation(s)
- Yuwei Tao
- Center for Cancer Biology, School of Basic Medical Sciences, Tsinghua University, Beijing, China
| | - Chen Tian
- Center for Cancer Biology, School of Basic Medical Sciences, Tsinghua University, Beijing, China
| | - Shaolong Qi
- Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Department of Chemistry, Tsinghua University, Beijing, China
| | - Ziqi Jia
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zheng Xu
- Department of Colorectal Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jingjing Meng
- Center for Cancer Biology, School of Basic Medical Sciences, Tsinghua University, Beijing, China
| | - Guoyuan Xu
- Center for Cancer Biology, School of Basic Medical Sciences, Tsinghua University, Beijing, China
| | - Haitian Hu
- Center for Cancer Biology, School of Basic Medical Sciences, Tsinghua University, Beijing, China
| | - Xuxiang Wang
- Center for Cancer Biology, School of Basic Medical Sciences, Tsinghua University, Beijing, China
| | - Tengjiang Zhang
- Center for Cancer Biology, School of Basic Medical Sciences, Tsinghua University, Beijing, China
| | - Huiwen You
- Center for Cancer Biology, School of Basic Medical Sciences, Tsinghua University, Beijing, China
| | - Xun Lan
- State Key Laboratory of Molecular Oncology and Center for Cancer Biology, School of Basic Medical Sciences, Tsinghua University, Beijing, China
| | - Xin Lin
- State Key Laboratory of Molecular Oncology and Center for Cancer Biology, School of Basic Medical Sciences, Tsinghua University, Beijing, China
| | - Guocan Yu
- Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Department of Chemistry, Tsinghua University, Beijing, China
| | - Haitao Zhou
- Department of Colorectal Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jiaqi Liu
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Hanqiu Zheng
- State Key Laboratory of Molecular Oncology and Center for Cancer Biology, School of Basic Medical Sciences, Tsinghua University, Beijing, China.
- SXMU-Tsinghua Collaborative Innovation Center for Frontier Medicine, Shanxi Medical University, Taiyuan, China.
| |
Collapse
|
5
|
Zhang RY, Wang ZX, Zhang MY, Wang YF, Zhou SL, Xu JL, Lin WX, Ji TR, Chen YD, Lu T, Li NG, Shi ZH. MALT1 Inhibitors and Degraders: Strategies for NF-κB-Driven Malignancies. J Med Chem 2025; 68:5075-5096. [PMID: 39999563 DOI: 10.1021/acs.jmedchem.4c02873] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/27/2025]
Abstract
Mucosa-associated lymphoid tissue protein 1 (MALT1), a cysteine protease and the sole paracaspase in humans, plays a pivotal role in the survival and proliferation of NF-κB-dependent malignant cancers, particularly MALT lymphoma and diffuse large B-cell lymphoma (DLBCL). Dysregulated MALT1 activity is implicated in various malignancies, highlighting its importance as a therapeutic target. This Perspective provides an overview of MALT1's structural and functional characteristics, summarizes recent advancements in small-molecule inhibitors and degraders targeting this protein, and discusses compound structures, structure-activity relationship (SAR) analyses, and biological activities. We aim to inform future research efforts to enhance the activity, selectivity, and pharmacological properties of MALT1-targeting compounds, establishing a foundational framework for drug development in this critical area of cancer therapy.
Collapse
Affiliation(s)
- Ru-Yue Zhang
- Laboratory of Molecular Design and Drug Discovery, School of Science, China Pharmaceutical University, 639 Longmian Avenue, Nanjing, Jiangsu 211198, China
| | - Zi-Xuan Wang
- National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, Jiangsu 210023, China
| | - Meng-Yuan Zhang
- National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, Jiangsu 210023, China
| | - Yu-Fan Wang
- Laboratory of Molecular Design and Drug Discovery, School of Science, China Pharmaceutical University, 639 Longmian Avenue, Nanjing, Jiangsu 211198, China
| | - Si-Li Zhou
- Laboratory of Molecular Design and Drug Discovery, School of Science, China Pharmaceutical University, 639 Longmian Avenue, Nanjing, Jiangsu 211198, China
| | - Jia-Lu Xu
- Laboratory of Molecular Design and Drug Discovery, School of Science, China Pharmaceutical University, 639 Longmian Avenue, Nanjing, Jiangsu 211198, China
| | - Wen-Xuan Lin
- Laboratory of Molecular Design and Drug Discovery, School of Science, China Pharmaceutical University, 639 Longmian Avenue, Nanjing, Jiangsu 211198, China
| | - Tian-Rui Ji
- Laboratory of Molecular Design and Drug Discovery, School of Science, China Pharmaceutical University, 639 Longmian Avenue, Nanjing, Jiangsu 211198, China
| | - Ya-Dong Chen
- Laboratory of Molecular Design and Drug Discovery, School of Science, China Pharmaceutical University, 639 Longmian Avenue, Nanjing, Jiangsu 211198, China
| | - Tao Lu
- Laboratory of Molecular Design and Drug Discovery, School of Science, China Pharmaceutical University, 639 Longmian Avenue, Nanjing, Jiangsu 211198, China
| | - Nian-Guang Li
- National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, Jiangsu 210023, China
| | - Zhi-Hao Shi
- Laboratory of Molecular Design and Drug Discovery, School of Science, China Pharmaceutical University, 639 Longmian Avenue, Nanjing, Jiangsu 211198, China
| |
Collapse
|
6
|
Wu EJ, Kandalkar AT, Ehrmann JF, Tong AB, Zhang J, Cong Q, Wu H. A structural atlas of death domain fold proteins reveals their versatile roles in biology and function. Proc Natl Acad Sci U S A 2025; 122:e2426986122. [PMID: 39977327 PMCID: PMC11874512 DOI: 10.1073/pnas.2426986122] [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: 12/24/2024] [Accepted: 01/23/2025] [Indexed: 02/22/2025] Open
Abstract
Death domain fold (DDF) superfamily proteins are critically important players in pathways of cell death and inflammation. DDFs are often essential scaffolding domains in receptors, adaptors, or effectors of these pathways by mediating homo- and hetero-oligomerization including helical filament assembly. At the downstream ends of these pathways, effector oligomerization by DDFs brings the enzyme domains into proximity for their dimerization and activation. Hundreds of structures of these domains have been solved. However, a comprehensive understanding of DDFs is lacking. In this article, we report the curation of a DDF structural atlas as a public website (deathdomain.org) and deduce the common and distinct principles of DDF-mediated oligomerization among the four families (death domain or DD, death effector domain or DED, caspase recruitment domain or CARD, and pyrin domain or PYD). We further annotate DDFs genome-wide based on AlphaFold-predicted models and protein sequences. These studies reveal mechanistic rules for this widely distributed domain superfamily.
Collapse
Affiliation(s)
- Emily J. Wu
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA02115
- Program in Cellular and Molecular Medicine, Boston Children’s Hospital, Boston, MA02115
- Saratoga High School, Saratoga, CA95070
| | - Ankita T. Kandalkar
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA02115
- Program in Cellular and Molecular Medicine, Boston Children’s Hospital, Boston, MA02115
- Department of Biology, College of Science, Northeastern University, Boston, MA02115
| | - Julian F. Ehrmann
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA02115
- Program in Cellular and Molecular Medicine, Boston Children’s Hospital, Boston, MA02115
| | - Alexander B. Tong
- Jason L. Choy Laboratory of Single-Molecule Biophysics, Institute for Quantitative Biosciences, Chemistry Graduate Group, University of California, Berkeley, CA94720
| | - Jing Zhang
- Department of Biophysics, University of Texas Southwestern Medical Center, Dallas, TX75390
- Eugene McDermott Center for Human Growth and Development, University of Texas, Southwestern Medical Center, Dallas, TX75390
- Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX75390
| | - Qian Cong
- Department of Biophysics, University of Texas Southwestern Medical Center, Dallas, TX75390
- Eugene McDermott Center for Human Growth and Development, University of Texas, Southwestern Medical Center, Dallas, TX75390
- Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX75390
| | - Hao Wu
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA02115
- Program in Cellular and Molecular Medicine, Boston Children’s Hospital, Boston, MA02115
- Department of Biology, College of Science, Northeastern University, Boston, MA02115
| |
Collapse
|
7
|
Shen G, Wang G, Chen J, Guo Y, Zhang W, Xu C, Chen L, Wang Q. MALT1 promotes the antibacterial immune response by activating NF-κB signaling and enhancing hemocyte phagocytosis in the Chinese mitten crab. FISH & SHELLFISH IMMUNOLOGY 2025; 157:110100. [PMID: 39733914 DOI: 10.1016/j.fsi.2024.110100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 12/19/2024] [Accepted: 12/25/2024] [Indexed: 12/31/2024]
Abstract
Mucosa-associated lymphoid tissue lymphoma translocation protein 1 (MALT1), a scaffold protein, plays a pivotal role in the NF-κB pathway downstream of T-cell receptors (TCRs) and B-cell receptors (BCRs). As a key signaling hub, MALT1 integrates various pathways, making it essential for both innate and adaptive immunity. However, its role in the antibacterial immune responses of crustaceans remains unclear. Here, we characterized MALT1 from the Chinese mitten crab (Eriocheir sinensis), denoted as EsMALT1, and compared its sequence and domain conservation with MALT1 from other species. Furthermore, Vibrio parahaemolyticus infection upregulated EsMALT1 expression markedly. Knockdown of EsMALT1 in hemocytes inhibits the translocation of the NF-κB-like transcription factors EsRelish and EsDorsal from the cytoplasm to the nucleus in response to Vibrio parahaemolyticus stimulation, thereby reducing the expression of the antimicrobial peptides anti-lipopolysaccharide factor (ALF), and Crustins. At the cellular level, silencing of EsMALT1 expression significantly inhibited the phagocytic capacity of crab hemocytes against Vibrio parahaemolyticus. In vivo, silencing of EsMALT1 rendered crabs susceptible to bacterial infection and impaired their bacterial clearance. In conclusion, EsMALT1 promotes both humoral and cellular immunity in E. sinensis, making it essential for the induction of antibacterial immune responses.
Collapse
Affiliation(s)
- Guoqing Shen
- School of Life Sciences, East China Normal University, Shanghai, China
| | - Guangyu Wang
- School of Aquatic and Life Sciences, Shanghai Ocean University, Shanghai, China
| | - Jinming Chen
- School of Aquatic and Life Sciences, Shanghai Ocean University, Shanghai, China
| | - Yanan Guo
- School of Life Sciences, East China Normal University, Shanghai, China
| | - Wen Zhang
- School of Life Sciences, East China Normal University, Shanghai, China
| | - Chaohui Xu
- School of Aquatic and Life Sciences, Shanghai Ocean University, Shanghai, China
| | - Liqiao Chen
- School of Life Sciences, East China Normal University, Shanghai, China.
| | - Qun Wang
- School of Aquatic and Life Sciences, Shanghai Ocean University, Shanghai, China.
| |
Collapse
|
8
|
Singh S, Pradhan D, Puri P, Sharma S, Jain AK. Profiling CARD14 gene expression in Indian Psoriasis patients. Sci Rep 2024; 14:28798. [PMID: 39567556 PMCID: PMC11579362 DOI: 10.1038/s41598-024-78267-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Accepted: 10/29/2024] [Indexed: 11/22/2024] Open
Abstract
Several Genome Wide linkage Studies on psoriasis performed to gain insight of genetic architecture of the disease. Caspase Recruitment Domain-containing family 14 (CARD14) also known as CARMA2 or BIMP2; cytogenic location: 17q25.3, is a scaffold protein that primarily controls the skin epidermis's nuclear factor kB (NF-kB) signaling pathway activity in skin epidermis, a master gene for inflammation, has been shown to be linked with rare, heritable form of psoriasis. CARD14 is predominantly expressed in keratinocytes and epithelial cells, but also in unidentified dermal cells. For better understanding of molecular processes involved in CARD14 underlying Indian psoriatic patients, we analyzed gene expression of 42 moderates to severe cases of plaque psoriasis and same number of controls using qPCR and its validation through Immunohistochemistry (IHC). This study identifies that the expression of CARD14 in dermal endothelial cells among patients with psoriasis and explores the potential functional consequences associated with an overactive CARD14 gene. Furthermore, the expression data from the western population was consistent with the results of the qPCR validation of the candidate gene. There is a significant correlation between Indian psoriasis vulgaris patients and CARD14 up-regulation, as evidenced by a roughly two-fold shift in lesional tissue expression. This provides insights into the pathways and genes linked to the pathogenesis of psoriasis.
Collapse
Affiliation(s)
- S Singh
- ICMR-National Institute of Pathology, New Delhi, 110029, India
| | - D Pradhan
- Bioinformatics Centralized Core Research Facility-AIIMS, New Delhi, 110029, India
- Bioinformatics Centralized Core Research Facility-AIIMS, New Delhi, 110029, India
| | - P Puri
- Department of Dermatology, VMMC & Safdarjang Hospital, New Delhi, 110029, India
- Dermatology Department, VMMC & Safdarjang Hospital, New Delhi, 110029, India
| | - Shruti Sharma
- ICMR-National Institute of Pathology, New Delhi, 110029, India.
- ICMR-National Institute of Pathology, SriRamachari Bhawan Safdarjang Hospital Campus, New Delhi, 110029, India.
| | - A K Jain
- ICMR-National Institute of Pathology, New Delhi, 110029, India.
| |
Collapse
|
9
|
Yang CY, Tseng YC, Tu YF, Kuo BJ, Hsu LC, Lien CI, Lin YS, Wang YT, Lu YC, Su TW, Lo YC, Lin SC. Reverse hierarchical DED assembly in the cFLIP-procaspase-8 and cFLIP-procaspase-8-FADD complexes. Nat Commun 2024; 15:8974. [PMID: 39419969 PMCID: PMC11487272 DOI: 10.1038/s41467-024-53306-1] [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: 06/07/2024] [Accepted: 10/09/2024] [Indexed: 10/19/2024] Open
Abstract
cFLIP, a master anti-apoptotic regulator, targets the FADD-induced DED complexes of procaspase-8 in death receptor and ripoptosome signaling pathways. Several tumor cells maintain relatively high levels of cFLIP in achieving their immortality. However, understanding the three-dimensional regulatory mechanism initiated or mediated by elevated levels of cFLIP has been limited by the absence of the atomic coordinates for cFLIP-induced DED complexes. Here we report the crystal plus cryo-EM structures to uncover an unconventional mechanism where cFLIP and procaspase-8 autonomously form a binary tandem DED complex, independent of FADD. This complex gains the ability to recruit FADD, thereby allosterically modulating cFLIP assembly and partially activating caspase-8 for RIPK1 cleavage. Our structure-guided mutagenesis experiments provide critical insights into these regulatory mechanisms, elucidating the resistance to apoptosis and necroptosis in achieving immortality. Finally, this research offers a unified model for the intricate bidirectional hierarchy-based processes using multiprotein helical assembly to govern cell fate decisions.
Collapse
Affiliation(s)
- Chao-Yu Yang
- Genomics Research Center, Academia Sinica, Taipei, 11529, Taiwan
| | - Yi-Chun Tseng
- Genomics Research Center, Academia Sinica, Taipei, 11529, Taiwan
- Department of Biotechnology and Bioindustry Sciences, College of Bioscience and Biotechnology, National Cheng Kung University, Tainan, 70101, Taiwan
| | - Yi-Fan Tu
- Department of Biotechnology and Bioindustry Sciences, College of Bioscience and Biotechnology, National Cheng Kung University, Tainan, 70101, Taiwan
| | - Bai-Jiun Kuo
- Department of Biotechnology and Bioindustry Sciences, College of Bioscience and Biotechnology, National Cheng Kung University, Tainan, 70101, Taiwan
| | - Li-Chung Hsu
- Institute of Molecular Medicine, College of Medicine, National Taiwan University, Taipei, 10002, Taiwan
- Center of Precision Medicine, College of Medicine, National Taiwan University, Taipei, 10002, Taiwan
| | - Chia-I Lien
- Institute of Molecular Medicine, College of Medicine, National Taiwan University, Taipei, 10002, Taiwan
| | - You-Sheng Lin
- Institute of Molecular Medicine, College of Medicine, National Taiwan University, Taipei, 10002, Taiwan
| | - Yin-Ting Wang
- Genomics Research Center, Academia Sinica, Taipei, 11529, Taiwan
| | - Yen-Chen Lu
- Department of Biotechnology and Bioindustry Sciences, College of Bioscience and Biotechnology, National Cheng Kung University, Tainan, 70101, Taiwan
| | - Tsung-Wei Su
- Genomics Research Center, Academia Sinica, Taipei, 11529, Taiwan
| | - Yu-Chih Lo
- Department of Biotechnology and Bioindustry Sciences, College of Bioscience and Biotechnology, National Cheng Kung University, Tainan, 70101, Taiwan.
| | - Su-Chang Lin
- Genomics Research Center, Academia Sinica, Taipei, 11529, Taiwan.
| |
Collapse
|
10
|
Iliaki S, Kreike M, Ferreras Moreno N, De Meyer F, Aidarova A, Braun H, Libert C, Afonina IS, Beyaert R. Polo-like kinase 1 (PLK1) is a novel CARD14-binding protein in keratinocytes. Biochem Pharmacol 2024; 228:116316. [PMID: 38797267 DOI: 10.1016/j.bcp.2024.116316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2024] [Revised: 05/08/2024] [Accepted: 05/23/2024] [Indexed: 05/29/2024]
Abstract
Caspase recruitment domain (CARD)-containing protein 14 (CARD14) is an intracellular protein that mediates nuclear factor-kappa B (NF-ĸB) signaling and proinflammatory gene expression in skin keratinocytes. Several hyperactivating CARD14 mutations have been associated with psoriasis and other inflammatory skin diseases. CARD14-induced NF-ĸB signaling is dependent on the formation of a CARD14-BCL10-MALT1 (CBM) signaling complex, but upstream receptors and molecular mechanisms that activate and regulate CARD14 signaling are still largely unclear. Using unbiased affinity purification and mass spectrometry (AP-MS) screening, we discover polo-like kinase 1 (PLK1) as a novel CARD14-binding protein. CARD14-PLK1 binding is independent of the CARD14 CARD domain but involves a consensus phospho-dependent PLK1-binding motif in the CARD14 linker region (LR). Expression of the psoriasis-associated CARD14(E138A) variant in human keratinocytes induces the recruitment of PLK1 to CARD14-containing signalosomes in interphase cells, but does not affect the specific location of PLK1 in mitotic cells. Finally, disruption of the PLK1-binding motif in CARD14(E138A) increases CARD14-induced proinflammatory signaling and gene expression. Together, our data identify PLK1 as a novel CARD14-binding protein and indicate a negative regulatory role for PLK1 in CARD14 signaling.
Collapse
Affiliation(s)
- Styliani Iliaki
- Center for Inflammation Research, Unit of Molecular Signal Transduction in Inflammation, VIB, B-9052 Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, B-9052 Ghent, Belgium
| | - Marja Kreike
- Center for Inflammation Research, Unit of Molecular Signal Transduction in Inflammation, VIB, B-9052 Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, B-9052 Ghent, Belgium
| | - Natalia Ferreras Moreno
- Center for Inflammation Research, Unit of Molecular Signal Transduction in Inflammation, VIB, B-9052 Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, B-9052 Ghent, Belgium; Center for Inflammation Research, Unit of Mouse Genetics and Inflammation, VIB, B-9052 Ghent, Belgium
| | - Femke De Meyer
- Center for Inflammation Research, Unit of Molecular Signal Transduction in Inflammation, VIB, B-9052 Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, B-9052 Ghent, Belgium
| | - Aigerim Aidarova
- Center for Inflammation Research, Unit of Molecular Signal Transduction in Inflammation, VIB, B-9052 Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, B-9052 Ghent, Belgium
| | - Harald Braun
- Center for Inflammation Research, Unit of Molecular Signal Transduction in Inflammation, VIB, B-9052 Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, B-9052 Ghent, Belgium
| | - Claude Libert
- Department of Biomedical Molecular Biology, Ghent University, B-9052 Ghent, Belgium; Center for Inflammation Research, Unit of Mouse Genetics and Inflammation, VIB, B-9052 Ghent, Belgium
| | - Inna S Afonina
- Center for Inflammation Research, Unit of Molecular Signal Transduction in Inflammation, VIB, B-9052 Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, B-9052 Ghent, Belgium
| | - Rudi Beyaert
- Center for Inflammation Research, Unit of Molecular Signal Transduction in Inflammation, VIB, B-9052 Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, B-9052 Ghent, Belgium.
| |
Collapse
|
11
|
Wallerstein J, Han X, Levkovets M, Lesovoy D, Malmodin D, Mirabello C, Wallner B, Sun R, Sandalova T, Agback P, Karlsson G, Achour A, Agback T, Orekhov V. Insights into mechanisms of MALT1 allostery from NMR and AlphaFold dynamic analyses. Commun Biol 2024; 7:868. [PMID: 39014105 PMCID: PMC11252132 DOI: 10.1038/s42003-024-06558-y] [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: 02/16/2024] [Accepted: 07/05/2024] [Indexed: 07/18/2024] Open
Abstract
Mucosa-associated lymphoid tissue lymphoma-translocation protein 1 (MALT1) is an attractive target for the development of modulatory compounds in the treatment of lymphoma and other cancers. While the three-dimensional structure of MALT1 has been previously determined through X-ray analysis, its dynamic behaviour in solution has remained unexplored. We present here dynamic analyses of the apo MALT1 form along with the E549A mutation. This investigation used NMR 15N relaxation and NOE measurements between side-chain methyl groups. Our findings confirm that MALT1 exists as a monomer in solution, and demonstrate that the domains display semi-independent movements in relation to each other. Our dynamic study, covering multiple time scales, along with the assessment of conformational populations by Molecular Dynamic simulations, Alpha Fold modelling and PCA analysis, put the side chain of residue W580 in an inward position, shedding light at potential mechanisms underlying the allosteric regulation of this enzyme.
Collapse
Affiliation(s)
- Johan Wallerstein
- Department of Chemistry and Molecular Biology, University of Gothenburg, Box 465, SE-40530, Gothenburg, Sweden
| | - Xiao Han
- Science for Life Laboratory, Department of Medicine, Solna, Karolinska Institute, SE-17165, Solna, Sweden
- Division of Infectious Diseases, Karolinska University Hospital, SE‑171 76, Stockholm, Sweden
| | - Maria Levkovets
- Swedish NMR Centre, University of Gothenburg, Box 465, SE-40530, Gothenburg, Sweden
| | - Dmitry Lesovoy
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry RAS, 117997, Moscow, Russia
| | - Daniel Malmodin
- Swedish NMR Centre, University of Gothenburg, Box 465, SE-40530, Gothenburg, Sweden
| | - Claudio Mirabello
- Dept of Physics, Chemistry and Biology, Linköping University, 581 83, Linköping, Sweden
- National Bioinformatics Infrastructure Sweden, Science for Life Laboratory, Solna, Sweden
| | - Björn Wallner
- National Bioinformatics Infrastructure Sweden, Science for Life Laboratory, Solna, Sweden
| | - Renhua Sun
- Science for Life Laboratory, Department of Medicine, Solna, Karolinska Institute, SE-17165, Solna, Sweden
- Division of Infectious Diseases, Karolinska University Hospital, SE‑171 76, Stockholm, Sweden
| | - Tatyana Sandalova
- Science for Life Laboratory, Department of Medicine, Solna, Karolinska Institute, SE-17165, Solna, Sweden
- Division of Infectious Diseases, Karolinska University Hospital, SE‑171 76, Stockholm, Sweden
| | - Peter Agback
- Department of Molecular Sciences, Swedish University of Agricultural Sciences, PO Box 7015, SE-750 07, Uppsala, Sweden
| | - Göran Karlsson
- Department of Chemistry and Molecular Biology, University of Gothenburg, Box 465, SE-40530, Gothenburg, Sweden
- Swedish NMR Centre, University of Gothenburg, Box 465, SE-40530, Gothenburg, Sweden
| | - Adnane Achour
- Science for Life Laboratory, Department of Medicine, Solna, Karolinska Institute, SE-17165, Solna, Sweden
- Division of Infectious Diseases, Karolinska University Hospital, SE‑171 76, Stockholm, Sweden
| | - Tatiana Agback
- Department of Molecular Sciences, Swedish University of Agricultural Sciences, PO Box 7015, SE-750 07, Uppsala, Sweden.
| | - Vladislav Orekhov
- Department of Chemistry and Molecular Biology, University of Gothenburg, Box 465, SE-40530, Gothenburg, Sweden.
- Swedish NMR Centre, University of Gothenburg, Box 465, SE-40530, Gothenburg, Sweden.
| |
Collapse
|
12
|
Kuo BJ, Lin SC, Tu YF, Huang PH, Lo YC. Study of individual domains contributing to MALT1 dimerization in BCL10-independent and dependent assembly. Biochem Biophys Res Commun 2024; 717:150029. [PMID: 38714015 DOI: 10.1016/j.bbrc.2024.150029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 04/17/2024] [Accepted: 04/28/2024] [Indexed: 05/09/2024]
Abstract
The CARMA-BCL10-MALT1 (CBM) signalosome functions as a pivotal supramolecular module, integrating diverse receptor-induced signaling pathways to regulate BCL10-dependent NF-kB activation in innate and adaptive immunity. Conversely, the API2-MALT1 fusion protein in t(11; 18)(q21; q21) MALT lymphoma constitutively induces BCL10-independent NF-kB activation. MALT1 dimer formation is indispensable for the requisite proteolytic activity and is critical for NF-kB activation regulation in both scenarios. However, the molecular assembly of MALT1 individual domains in CBM activation remains elusive. Here we report the crystal structure of the MALT1 death domain (DD) at a resolution of 2.1 Å, incorporating reconstructed residues in previously disordered loops 1 and 2. Additionally, we observe a conformational regulation element (CRE) regulating stem-helix formation in NLRPs pyrin (PYD) within the MALT1 DD structure. The structure reveals a stem-helix-mediated dimer further corroborated in solution. To elucidate how the BCL10 filament facilitates MALT1 dimerization, we reconstitute a BCL10-CARD-MALT1-DD-IG1-IG2 complex model. We propose a N+7 rule for BCL10-dependent MALT1 dimerization via the IG1-IG2 domain and for MALT1-dependent cleavage in trans. Biochemical data further indicates concentration-dependent dimerization of the MALT1 IG1-IG2 domain, facilitating MALT1 dimerization in BCL10-independent manner. Our findings provide a structural and biochemical foundation for understanding MALT1 dimeric mechanisms, shedding light on potential BCL10-independent MALT1 dimer formation and high-order BCL10-MALT1 assembly.
Collapse
Affiliation(s)
- Bai-Jiun Kuo
- Department of Biotechnology and Bioindustry Sciences, College of Bioscience and Biotechnology, National Cheng Kung University, Tainan, 70101, Taiwan
| | - Su-Chang Lin
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | - Yi-Fan Tu
- Department of Biotechnology and Bioindustry Sciences, College of Bioscience and Biotechnology, National Cheng Kung University, Tainan, 70101, Taiwan; Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | - Po-Hui Huang
- Department of Biotechnology and Bioindustry Sciences, College of Bioscience and Biotechnology, National Cheng Kung University, Tainan, 70101, Taiwan
| | - Yu-Chih Lo
- Department of Biotechnology and Bioindustry Sciences, College of Bioscience and Biotechnology, National Cheng Kung University, Tainan, 70101, Taiwan.
| |
Collapse
|
13
|
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
|
14
|
Staal J, Driege Y, Van Gaever F, Steels J, Beyaert R. Chimeric and mutant CARD9 constructs enable analyses of conserved and diverged autoinhibition mechanisms in the CARD-CC protein family. FEBS J 2024; 291:1220-1245. [PMID: 38098267 DOI: 10.1111/febs.17035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 11/09/2023] [Accepted: 12/13/2023] [Indexed: 12/27/2023]
Abstract
Caspase recruitment domain-containing protein (CARD)9, CARD10, CARD11, and CARD14 all belong to the CARD-coiled coil (CC) protein family and originated from a single common ancestral protein early in vertebrate evolution. All four proteins form CARD-CC/BCL10/MALT1 (CBM) complexes leading to nuclear factor-kappa-B (NF-κB) activation after upstream phosphorylation by various protein kinase C (PKC) isoforms. CBM complex signaling is critical for innate and adaptive immunity, but aberrant activation can cause autoimmune or autoinflammatory diseases, or be oncogenic. CARD9 shows a superior auto-inhibition compared with other CARD-CC family proteins, with very low spontaneous activity when overexpressed in HEK293T cells. In contrast, the poor auto-inhibition of other CARD-CC family proteins, especially CARD10 (CARMA3) and CARD14 (CARMA2), is hampering characterization of upstream activators or activating mutations in overexpression studies. We grafted different domains from CARD10, 11, and 14 on CARD9 to generate chimeric CARD9 backbones for functional characterization of activating mutants using NF-κB reporter gene activation in HEK293T cells as readout. CARD11 (CARMA1) activity was not further reduced by grafting on CARD9 backbones. The chimeric CARD9 approach was subsequently validated by using several known disease-associated mutations in CARD10 and CARD14, and additional screening allowed us to identify several previously unknown activating natural variants in human CARD9 and CARD10. Using Genebass as a resource of exome-based disease association statistics, we found that activated alleles of CARD9 correlate with irritable bowel syndrome (IBS), constipation, osteoarthritis, fibromyalgia, insomnia, anxiety, and depression, which can occur as comorbidities.
Collapse
Affiliation(s)
- Jens Staal
- Unit of Molecular Signal Transduction in Inflammation, VIB-UGent Center for Inflammation Research, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Belgium
- Department of Biochemistry and Microbiology, Ghent University, Belgium
| | - Yasmine Driege
- Unit of Molecular Signal Transduction in Inflammation, VIB-UGent Center for Inflammation Research, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Belgium
| | - Femke Van Gaever
- Unit of Molecular Signal Transduction in Inflammation, VIB-UGent Center for Inflammation Research, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Belgium
| | - Jill Steels
- Unit of Molecular Signal Transduction in Inflammation, VIB-UGent Center for Inflammation Research, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Belgium
| | - Rudi Beyaert
- Unit of Molecular Signal Transduction in Inflammation, VIB-UGent Center for Inflammation Research, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Belgium
| |
Collapse
|
15
|
Bacher S, Schmitz ML. Open questions in the NF-κB field. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2024; 1871:119469. [PMID: 37951506 DOI: 10.1016/j.bbamcr.2023.119469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 03/23/2023] [Accepted: 03/23/2023] [Indexed: 11/14/2023]
Abstract
A variety of stress signals leads to activation of the inducible transcription factor NF-κB, one of the master regulators of the innate immune response. Despite a wealth of information available on the NF-κB core components and its control by different activation pathways and negative feedback loops, several levels of complexity hamper our understanding of the system. This has also contributed to the limited success of NF-κB inhibitors in the clinic and explains some of their unexpected effects. Here we consider the molecular and cellular events generating this complexity at all levels and point to a number of unresolved questions in the field. We also discuss potential future experimental and computational strategies to provide a deeper understanding of NF-κB and its coregulatory signaling networks.
Collapse
Affiliation(s)
- Susanne Bacher
- Institute of Biochemistry, Justus Liebig University Giessen (Germany), Member of the German Center for Lung Research (DZL), Germany
| | - M Lienhard Schmitz
- Institute of Biochemistry, Justus Liebig University Giessen (Germany), Member of the German Center for Lung Research (DZL), Germany.
| |
Collapse
|
16
|
Sezer A, Mahmutović L, Akçeşme B. In silico study of polyphenols as potential inhibitors of MALT1 protein in non-Hodgkin lymphoma. Med Oncol 2023; 41:37. [PMID: 38155268 DOI: 10.1007/s12032-023-02261-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Accepted: 11/16/2023] [Indexed: 12/30/2023]
Abstract
Non-Hodgkin lymphoma (NHL) is one of the most common cancer types. Deregulated signaling pathways can trigger certain NHL subtypes, including Diffuse Large B-cell lymphoma. NF-ĸB signaling pathway, which is responsible for the proliferation, growth, and survival of cells, has an essential role in lymphoma development. Although different signals control NF-ĸB activation in various lymphoid malignancies, the characteristic one is the CARD11-BCL10-MALT1 (CBM) complex. The CBM complex is responsible for the initiation of adaptive immune response. Our study is focused on the molecular docking of ten polyphenols as potential CARD11-BCL10-MALT1 complex inhibitors, essentially through MALT1 inhibition. Molecular docking was performed by Auto Dock Tools and AutoDock Vina tool, while SwissADME was used for drug-likeness and absorption, distribution, metabolism, excretion, and toxicity (ADMET) analysis of the ligands. Out of 66 ligands that were used in this study, we selected and visualized five. Selection criteria were based on the binding energy score and position of the ligands on the used protein. 2D and 3D visualizations showed interactions of ligands with the protein. Five ligands are considered potential inhibitors of MALT1, thus affecting NF-ĸB signaling pathway. However, additional in vivo and in vitro studies are required to confirm their mechanism of inhibition.
Collapse
Affiliation(s)
- Abas Sezer
- Department of Genetics and Bioengineering, Faculty of Engineering and Natural Sciences, International University of Sarajevo, Hrasnička Cesta 15, 71000, Sarajevo, Bosnia and Herzegovina
| | - Lejla Mahmutović
- Department of Genetics and Bioengineering, Faculty of Engineering and Natural Sciences, International University of Sarajevo, Hrasnička Cesta 15, 71000, Sarajevo, Bosnia and Herzegovina
| | - Betül Akçeşme
- Department of Genetics and Bioengineering, Faculty of Engineering and Natural Sciences, International University of Sarajevo, Hrasnička Cesta 15, 71000, Sarajevo, Bosnia and Herzegovina.
- Department of Basic Medical Sciences, Division of Medical Biology, University of Health Sciences, 34000, Istanbul, Turkey.
| |
Collapse
|
17
|
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
|
18
|
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
|
19
|
Bcl10 phosphorylation-dependent droplet-like condensation positively regulates DNA virus-induced innate immune signaling. SCIENCE CHINA. LIFE SCIENCES 2023; 66:283-297. [PMID: 36115893 DOI: 10.1007/s11427-022-2169-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Accepted: 07/20/2022] [Indexed: 10/14/2022]
Abstract
B-cell lymphoma 10 (Bcl10) is a scaffolding protein that functions as an upstream regulator of NF-κB signaling by forming a complex with Mucosa-associated lymphoid tissue lymphoma translocation protein 1 (Malt1) and CARD-coiled coil protein family. This study showed that Bcl10 was involved in type I interferon (IFN) expression in response to DNA virus infection and that Bcl10-deficient mice were more susceptible to Herpes simplex virus 1 (HSV-1) infection than control mice. Mechanistically, DNA virus infection can trigger Bcl10 recruitment to the STING-TBK1 complex, leading to Bcl10 phosphorylation by TBK1. The phosphorylated Bcl10 undergoes droplet-like condensation and forms oligomers, which induce TBK1 phosphorylation and translocation to the perinuclear region. The activated TBK1 phosphorylates IRF3, which induces the expression of type I IFNs. This study elucidates that Bcl10 induces an innate immune response by undergoing droplet-like condensation and participating in signalosome formation downstream of the cGAS-STING pathway.
Collapse
|
20
|
Pomerantz JL, Milner JD, Snow AL. Elevated IgE from attenuated CARD11 signaling: lessons from atopic mice and humans. Curr Opin Immunol 2022; 79:102255. [PMID: 36334349 PMCID: PMC10424059 DOI: 10.1016/j.coi.2022.102255] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Revised: 08/17/2022] [Accepted: 09/30/2022] [Indexed: 11/07/2022]
Abstract
CARD11 encodes a large scaffold protein responsible for integrating antigen-receptor engagement with downstream signaling to NF-kB and other outputs in lymphocytes. Over the past 10 years, several human-inborn errors of immunity have been linked to pathogenic CARD11 mutations. Most recently, severe atopic patients were discovered that carried heterozygous dominant-negative CARD11 mutations. Here, we review the mechanistic connections between attenuated CARD11 signaling, elevated IgE, and atopy, comparing and contrasting key insights from both human patients and murine models. Continued investigation of abnormal CARD11 signaling in both contexts should inform novel therapeutic strategies to combat allergic pathogenesis.
Collapse
Affiliation(s)
- Joel L Pomerantz
- Department of Biological Chemistry, Institute for Cell Engineering, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Joshua D Milner
- Department of Pediatrics, Columbia University Irving Medical Center, New York, NY, USA
| | - Andrew L Snow
- Department of Pharmacology & Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, MD, USA.
| |
Collapse
|
21
|
Feng S, Zhang H, Tang Z, Peng X, Yang M, Wei X, Zhong W. Direct Construction of Chiral Ether via Highly Efficient Heck Reaction and N-Directed Asymmetric Hydrogenation for Large-Scale Synthesis of MALT1 Inhibitor RGT-068A. Org Process Res Dev 2022. [DOI: 10.1021/acs.oprd.2c00229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Song Feng
- Regor Therapeutics Group, 1206 Zhangjiang Road, Building C, Zhangjiang Hi-Tech Park, Shanghai 201203, China
| | - Haishen Zhang
- Pharmaron (Ningbo) Technology Development Co., No. 800 Bin-Hai 4th Road, Hangzhou Bay New Zone, Ningbo 315366, Zhejiang, China
| | - Zhiyong Tang
- Pharmaron (Ningbo) Technology Development Co., No. 800 Bin-Hai 4th Road, Hangzhou Bay New Zone, Ningbo 315366, Zhejiang, China
| | - Xingao Peng
- BioDuro-Sundia, 388 Jialilue Road, Zhangjiang Hi-Tech Park, Shanghai 201203, China
| | - Min Yang
- Regor Therapeutics Group, 1206 Zhangjiang Road, Building C, Zhangjiang Hi-Tech Park, Shanghai 201203, China
| | - Xudong Wei
- Pharmaron (Ningbo) Technology Development Co., No. 800 Bin-Hai 4th Road, Hangzhou Bay New Zone, Ningbo 315366, Zhejiang, China
| | - Wenge Zhong
- Regor Therapeutics Group, 1206 Zhangjiang Road, Building C, Zhangjiang Hi-Tech Park, Shanghai 201203, China
| |
Collapse
|
22
|
Mempel TR, Krappmann D. Combining precision oncology and immunotherapy by targeting the MALT1 protease. J Immunother Cancer 2022; 10:e005442. [PMID: 36270731 PMCID: PMC9594517 DOI: 10.1136/jitc-2022-005442] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/17/2022] [Indexed: 11/30/2022] Open
Abstract
An innovative strategy for cancer therapy is to combine the inhibition of cancer cell-intrinsic oncogenic signaling with cancer cell-extrinsic immunological activation of the tumor microenvironment (TME). In general, such approaches will focus on two or more distinct molecular targets in the malignant cells and in cells of the surrounding TME. In contrast, the protease Mucosa-associated lymphoid tissue protein 1 (MALT1) represents a candidate to enable such a dual approach by engaging only a single target. Originally identified and now in clinical trials as a lymphoma drug target based on its role in the survival and proliferation of malignant lymphomas addicted to chronic B cell receptor signaling, MALT1 proteolytic activity has recently gained additional attention through reports describing its tumor-promoting roles in several types of non-hematological solid cancer, such as breast cancer and glioblastoma. Besides cancer cells, regulatory T (Treg) cells in the TME are particularly dependent on MALT1 to sustain their immune-suppressive functions, and MALT1 inhibition can selectively reprogram tumor-infiltrating Treg cells into Foxp3-expressing proinflammatory antitumor effector cells. Thereby, MALT1 inhibition induces local inflammation in the TME and synergizes with anti-PD-1 checkpoint blockade to induce antitumor immunity and facilitate tumor control or rejection. This new concept of boosting tumor immunotherapy in solid cancer by MALT1 precision targeting in the TME has now entered clinical evaluation. The dual effects of MALT1 inhibitors on cancer cells and immune cells therefore offer a unique opportunity for combining precision oncology and immunotherapy to simultaneously impair cancer cell growth and neutralize immunosuppression in the TME. Further, MALT1 targeting may provide a proof of concept that modulation of Treg cell function in the TME represents a feasible strategy to augment the efficacy of cancer immunotherapy. Here, we review the role of MALT1 protease in physiological and oncogenic signaling, summarize the landscape of tumor indications for which MALT1 is emerging as a therapeutic target, and consider strategies to increase the chances for safe and successful use of MALT1 inhibitors in cancer therapy.
Collapse
Affiliation(s)
- Thorsten R Mempel
- Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| | - Daniel Krappmann
- Research Unit Cellular Signal Integration, Molecular Targets and Therapeutics Center, Helmholtz Center Munich - German Research Center for Environmental Health, Neuherberg, Germany
| |
Collapse
|
23
|
Han X, Levkovets M, Lesovoy D, Sun R, Wallerstein J, Sandalova T, Agback T, Achour A, Agback P, Orekhov VY. Assignment of IVL-Methyl side chain of the ligand-free monomeric human MALT1 paracaspase-IgL 3 domain in solution. BIOMOLECULAR NMR ASSIGNMENTS 2022; 16:363-371. [PMID: 36094731 PMCID: PMC9510110 DOI: 10.1007/s12104-022-10105-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/03/2022] [Accepted: 08/31/2022] [Indexed: 06/15/2023]
Abstract
Mucosa-associated lymphoid tissue protein 1 (MALT1) plays a key role in adaptive immune responses by modulating specific intracellular signalling pathways that control the development and proliferation of both T and B cells. Dysfunction of these pathways is coupled to the progress of highly aggressive lymphoma as well as to potential development of an array of different immune disorders. In contrast to other signalling mediators, MALT1 is not only activated through the formation of the CBM complex together with the proteins CARMA1 and Bcl10, but also by acting as a protease that cleaves multiple substrates to promote lymphocyte proliferation and survival via the NF-κB signalling pathway. Herein, we present the partial 1H, 13C Ile/Val/Leu-Methyl resonance assignment of the monomeric apo form of the paracaspase-IgL3 domain of human MALT1. Our results provide a solid ground for future elucidation of both the three-dimensional structure and the dynamics of MALT1, key for adequate development of inhibitors, and a thorough molecular understanding of its function(s).
Collapse
Affiliation(s)
- Xiao Han
- Science for Life Laboratory, Department of Medicine, Karolinska Institute, and, Division of Infectious Diseases, Karolinska University Hospital, 171 76, Stockholm, Sweden
| | - Maria Levkovets
- Department of Chemistry and Molecular Biology, University of Gothenburg, Box 465, 40530, Gothenburg, Sweden
| | - Dmitry Lesovoy
- Department of Structural Biology, Shemyakin-Ovchinnikov, Institute of Bioorganic Chemistry RAS, Moscow, Russia, 117997
| | - Renhua Sun
- Science for Life Laboratory, Department of Medicine, Karolinska Institute, and, Division of Infectious Diseases, Karolinska University Hospital, 171 76, Stockholm, Sweden
| | - Johan Wallerstein
- Department of Chemistry and Molecular Biology, University of Gothenburg, Box 465, 40530, Gothenburg, Sweden
| | - Tatyana Sandalova
- Science for Life Laboratory, Department of Medicine, Karolinska Institute, and, Division of Infectious Diseases, Karolinska University Hospital, 171 76, Stockholm, Sweden
| | - Tatiana Agback
- Department of Molecular Sciences, Swedish University of Agricultural Sciences, Box 7015, 750 07, Uppsala, Sweden
| | - Adnane Achour
- Science for Life Laboratory, Department of Medicine, Karolinska Institute, and, Division of Infectious Diseases, Karolinska University Hospital, 171 76, Stockholm, Sweden
| | - Peter Agback
- Department of Molecular Sciences, Swedish University of Agricultural Sciences, Box 7015, 750 07, Uppsala, Sweden.
| | - Vladislav Yu Orekhov
- Department of Chemistry and Molecular Biology, University of Gothenburg, Box 465, 40530, Gothenburg, Sweden.
- Swedish NMR Centre, University of Gothenburg, Box 465, 40530, Gothenburg, Sweden.
| |
Collapse
|
24
|
Xia M, David L, Teater M, Gutierrez J, Wang X, Meydan C, Lytle A, Slack GW, Scott DW, Morin RD, Onder O, Elenitoba-Johnson KS, Zamponi N, Cerchietti L, Lu T, Philippar U, Fontan L, Wu H, Melnick AM. BCL10 Mutations Define Distinct Dependencies Guiding Precision Therapy for DLBCL. Cancer Discov 2022; 12:1922-1941. [PMID: 35658124 PMCID: PMC9357155 DOI: 10.1158/2159-8290.cd-21-1566] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Revised: 05/03/2022] [Accepted: 06/01/2022] [Indexed: 01/09/2023]
Abstract
Activated B cell-like diffuse large B-cell lymphomas (ABC-DLBCL) have unfavorable outcomes and chronic activation of CARD11-BCL10-MALT1 (CBM) signal amplification complexes that form due to polymerization of BCL10 subunits, which is affected by recurrent somatic mutations in ABC-DLBCLs. Herein, we show that BCL10 mutants fall into at least two functionally distinct classes: missense mutations of the BCL10 CARD domain and truncation of its C-terminal tail. Truncating mutations abrogated a motif through which MALT1 inhibits BCL10 polymerization, trapping MALT1 in its activated filament-bound state. CARD missense mutations enhanced BCL10 filament formation, forming glutamine network structures that stabilize BCL10 filaments. Mutant forms of BCL10 were less dependent on upstream CARD11 activation and thus manifested resistance to BTK inhibitors, whereas BCL10 truncating but not CARD mutants were hypersensitive to MALT1 inhibitors. Therefore, BCL10 mutations are potential biomarkers for BTK inhibitor resistance in ABC-DLBCL, and further precision can be achieved by selecting therapy based on specific biochemical effects of distinct mutation classes. SIGNIFICANCE ABC-DLBCLs feature frequent mutations of signaling mediators that converge on the CBM complex. We use structure-function approaches to reveal that BCL10 mutations fall into two distinct biochemical classes. Both classes confer resistance to BTK inhibitors, whereas BCL10 truncations confer hyperresponsiveness to MALT1 inhibitors, providing a road map for precision therapies in ABC-DLBCLs. See related commentary by Phelan and Oellerich, p. 1844. This article is highlighted in the In This Issue feature, p. 1825.
Collapse
Affiliation(s)
- Min Xia
- Division of Hematology/Oncology, Department of Medicine, Weill Cornell Medicine, New York, New York
| | - Liron David
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, Massachusetts
| | - Matt Teater
- Division of Hematology/Oncology, Department of Medicine, Weill Cornell Medicine, New York, New York
| | - Johana Gutierrez
- Division of Hematology/Oncology, Department of Medicine, Weill Cornell Medicine, New York, New York
| | - Xiang Wang
- Division of Hematology/Oncology, Department of Medicine, Weill Cornell Medicine, New York, New York
| | - Cem Meydan
- Institute for Computational Biomedicine, Department of Physiology and Biophysics, Weill Cornell Medicine, New York, New York
| | - Andrew Lytle
- Centre for Lymphoid Cancer, BC Cancer Research, Vancouver, British Columbia, Canada
| | - Graham W. Slack
- Centre for Lymphoid Cancer, BC Cancer Research, Vancouver, British Columbia, Canada
| | - David W. Scott
- Centre for Lymphoid Cancer, BC Cancer Research, Vancouver, British Columbia, Canada
| | - Ryan D. Morin
- Genome Sciences Centre, British Columbia Cancer Agency, Vancouver, British Columbia, Canada
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Ozlem Onder
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Kojo S.J. Elenitoba-Johnson
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Nahuel Zamponi
- Division of Hematology/Oncology, Department of Medicine, Weill Cornell Medicine, New York, New York
| | - Leandro Cerchietti
- Division of Hematology/Oncology, Department of Medicine, Weill Cornell Medicine, New York, New York
| | - Tianbao Lu
- Janssen Research & Development, Springhouse, Pennsylvania
| | | | - Lorena Fontan
- Division of Hematology/Oncology, Department of Medicine, Weill Cornell Medicine, New York, New York
| | - Hao Wu
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, Massachusetts
| | - Ari M. Melnick
- Division of Hematology/Oncology, Department of Medicine, Weill Cornell Medicine, New York, New York
| |
Collapse
|
25
|
Cunha C, Koike T, Seki Y, Yamamoto M, Iwashima M. Schnurri 3 promotes Th2 cytokine production during the late phase of T-cell antigen stimulation. Eur J Immunol 2022; 52:1077-1094. [PMID: 35490426 PMCID: PMC9276650 DOI: 10.1002/eji.202149633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 03/22/2022] [Accepted: 04/28/2022] [Indexed: 11/29/2022]
Abstract
Th1 and Th2 polarization is determined by the coordination of numerous factors including the affinity and strength of the antigen-receptor interaction, predominant cytokine environment, and costimulatory molecules present. Here, we show that Schnurri (SHN) proteins have distinct roles in Th1 and Th2 polarization. SHN2 was previously found to block the induction of GATA3 and Th2 differentiation. We found that, in contrast to SHN2, SHN3 is critical for IL-4 production and Th2 polarization. Strength of stimulation controls SHN2 and SHN3 expression patterns, where higher doses of antigen receptor stimulation promoted SHN3 expression and IL-4 production, along with repression of SHN2 expression. SHN3-deficient T cells showed a substantial defect in IL-4 production and expression of AP-1 components, particularly c-Jun and Jun B. This loss of early IL-4 production led to reduced GATA3 expression and impaired Th2 differentiation. Together, these findings uncover SHN3 as a novel, critical regulator of Th2 development.
Collapse
Affiliation(s)
- Christina Cunha
- Department of Microbiology and ImmunologyLoyola UniversityChicagoIllinoisUSA
| | - Toru Koike
- Department of Biology, Faculty of ScienceShizuoka UniversityShizuokaJapan
| | - Yoichi Seki
- Department of Microbiology and ImmunologyLoyola UniversityChicagoIllinoisUSA
- Van Kampen Cardiovascular Research Laboratory, Department of Thoracic and Cardiovascular Surgery, Stritch School of MedicineLoyola UniversityChicagoIllinoisUSA
| | - Mutsumi Yamamoto
- Department of Microbiology and ImmunologyLoyola UniversityChicagoIllinoisUSA
- Van Kampen Cardiovascular Research Laboratory, Department of Thoracic and Cardiovascular Surgery, Stritch School of MedicineLoyola UniversityChicagoIllinoisUSA
| | - Makio Iwashima
- Department of Microbiology and ImmunologyLoyola UniversityChicagoIllinoisUSA
- Van Kampen Cardiovascular Research Laboratory, Department of Thoracic and Cardiovascular Surgery, Stritch School of MedicineLoyola UniversityChicagoIllinoisUSA
| |
Collapse
|
26
|
Kutzner K, Woods S, Karayel O, Gehring T, Yin H, Flatley A, Graß C, Wimberger N, Tofaute MJ, Seeholzer T, Feederle R, Mann M, Krappmann D. Phosphorylation of serine-893 in CARD11 suppresses the formation and activity of the CARD11-BCL10-MALT1 complex in T and B cells. Sci Signal 2022; 15:eabk3083. [PMID: 35230873 DOI: 10.1126/scisignal.abk3083] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
CARD11 acts as a gatekeeper for adaptive immune responses after T cell or B cell antigen receptor (TCR/BCR) ligation on lymphocytes. PKCθ/β-catalyzed phosphorylation of CARD11 promotes the assembly of the CARD11-BCL10-MALT1 (CBM) complex and lymphocyte activation. Here, we demonstrated that PKCθ/β-dependent CARD11 phosphorylation also suppressed CARD11 functions in T or B cells. Through mass spectrometry-based proteomics analysis, we identified multiple constitutive and inducible CARD11 phosphorylation sites in T cells. We demonstrated that a single TCR- or BCR-inducible phosphorylation on Ser893 in the carboxyl terminus of CARD11 prevented the activation of the transcription factor NF-κB, the kinase JNK, and the protease MALT1. Moreover, CARD11 Ser893 phosphorylation sensitized BCR-addicted lymphoma cells to toxicity induced by Bruton's tyrosine kinase (BTK) inhibitors. Phosphorylation of Ser893 in CARD11 by PKCθ controlled the strength of CARD11 scaffolding by impairing the formation of the CBM complex. Thus, PKCθ simultaneously catalyzes both stimulatory and inhibitory CARD11 phosphorylation events, which shape the strength of CARD11 signaling in lymphocytes.
Collapse
Affiliation(s)
- Kerstin Kutzner
- Research Unit Cellular Signal Integration, Helmholtz Zentrum München-German Research Center for Environmental Health. Ingolstaedter Landstr. 1, 85764 Neuherberg, Germany
| | - Simone Woods
- Research Unit Cellular Signal Integration, Helmholtz Zentrum München-German Research Center for Environmental Health. Ingolstaedter Landstr. 1, 85764 Neuherberg, Germany
| | - Ozge Karayel
- Department of Proteomics and Signal Transduction, Max Planck Institute of Biochemistry, Am Klopferspitz 18, 82152 Planegg, Germany
| | - Torben Gehring
- Research Unit Cellular Signal Integration, Helmholtz Zentrum München-German Research Center for Environmental Health. Ingolstaedter Landstr. 1, 85764 Neuherberg, Germany
| | - Hongli Yin
- Research Unit Cellular Signal Integration, Helmholtz Zentrum München-German Research Center for Environmental Health. Ingolstaedter Landstr. 1, 85764 Neuherberg, Germany
| | - Andrew Flatley
- Monoclonal Antibody Core Facility, Institute for Diabetes and Obesity, Helmholtz Zentrum München-German Research Center for Environmental Health, Ingolstaedter Landstr. 1, 85764 Neuherberg, Germany
| | - Carina Graß
- Research Unit Cellular Signal Integration, Helmholtz Zentrum München-German Research Center for Environmental Health. Ingolstaedter Landstr. 1, 85764 Neuherberg, Germany
| | - Nicole Wimberger
- Research Unit Cellular Signal Integration, Helmholtz Zentrum München-German Research Center for Environmental Health. Ingolstaedter Landstr. 1, 85764 Neuherberg, Germany
| | - Marie J Tofaute
- Research Unit Cellular Signal Integration, Helmholtz Zentrum München-German Research Center for Environmental Health. Ingolstaedter Landstr. 1, 85764 Neuherberg, Germany
| | - Thomas Seeholzer
- Research Unit Cellular Signal Integration, Helmholtz Zentrum München-German Research Center for Environmental Health. Ingolstaedter Landstr. 1, 85764 Neuherberg, Germany
| | - Regina Feederle
- Monoclonal Antibody Core Facility, Institute for Diabetes and Obesity, Helmholtz Zentrum München-German Research Center for Environmental Health, Ingolstaedter Landstr. 1, 85764 Neuherberg, Germany
| | - Matthias Mann
- Department of Proteomics and Signal Transduction, Max Planck Institute of Biochemistry, Am Klopferspitz 18, 82152 Planegg, Germany
| | - Daniel Krappmann
- Research Unit Cellular Signal Integration, Helmholtz Zentrum München-German Research Center for Environmental Health. Ingolstaedter Landstr. 1, 85764 Neuherberg, Germany
| |
Collapse
|
27
|
Zhang YY, Peng J, Luo XJ. Post-translational modification of MALT1 and its role in B cell- and T cell-related diseases. Biochem Pharmacol 2022; 198:114977. [PMID: 35218741 DOI: 10.1016/j.bcp.2022.114977] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Revised: 02/18/2022] [Accepted: 02/18/2022] [Indexed: 02/06/2023]
Abstract
Mucosa-associated lymphoid tissue lymphoma translocation protein 1 (MALT1) is a multifunctional protein. MALT1 functions as an adaptor protein to assemble and recruit proteins such as B-cell lymphoma 10 (BCL10) and caspase-recruitment domain (CARD)-containing coiled-coil protein 11 (CARD11). Conversely it also acts as a paracaspase to cleave specified substrates. Because of its involvement in immunity, inflammation and cancer through its dual functions of scaffolding and catalytic activity, MALT1 is becoming a promising therapeutic target in B cell- and T cell-related diseases. There is growing evidence that the function of MALT1 is subtly modulated via post-translational modifications. This review summarized recent progress in relevant studies regarding the physiological and pathophysiological functions of MALT1, post-translational modifications of MALT1 and its role in B cell- and T cell- related diseases. In addition, the current available MALT1 inhibitors were also discussed.
Collapse
Affiliation(s)
- Yi-Yue Zhang
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410078, China
| | - Jun Peng
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410078, China; Hunan Provincial Key Laboratory of Cardiovascular Research, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410078, China.
| | - Xiu-Ju Luo
- Department of Laboratory Medicine, The Third Xiangya Hospital of Central South University, Changsha 410013, China.
| |
Collapse
|
28
|
Bedsaul JR, Shah N, Hutcherson SM, Pomerantz JL. Mechanistic impact of oligomer poisoning by dominant-negative CARD11 variants. iScience 2022; 25:103810. [PMID: 35198875 PMCID: PMC8844825 DOI: 10.1016/j.isci.2022.103810] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 12/10/2021] [Accepted: 01/19/2022] [Indexed: 11/25/2022] Open
Abstract
The CARD11 scaffold controls antigen receptor signaling to NF-κB, JNK, and mTOR. Three classes of germline mutations in CARD11 cause Primary Immunodeficiency, including homozygous loss-of-function (LOF) mutations in CARD11 deficiency, heterozygous gain-of-function (GOF) mutations in BENTA disease, and heterozygous dominant-negative LOF mutations in CADINS. Here, we characterize LOF CARD11 mutants with a range of dominant-negative activities to identify the mechanistic properties that cause these variants to exert dominant effects when heterozygous. We find that strong dominant negatives can poison signaling from mixed wild-type:mutant oligomers at two steps in the CARD11 signaling cycle, at the Opening Step and at the Cofactor Association Step. Our findings provide evidence that CARD11 oligomer subunits cooperate in at least two steps during antigen receptor signaling and reveal how different LOF mutations in the same oligomeric signaling hub may cause disease with different inheritance patterns.
Collapse
Affiliation(s)
- Jacquelyn R. Bedsaul
- Department of Biological Chemistry and Institute for Cell Engineering, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Neha Shah
- Department of Biological Chemistry and Institute for Cell Engineering, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Shelby M. Hutcherson
- Department of Biological Chemistry and Institute for Cell Engineering, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Joel L. Pomerantz
- Department of Biological Chemistry and Institute for Cell Engineering, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| |
Collapse
|
29
|
Yin H, Karayel O, Chao YY, Seeholzer T, Hamp I, Plettenburg O, Gehring T, Zielinski C, Mann M, Krappmann D. A20 and ABIN-1 cooperate in balancing CBM complex-triggered NF-κB signaling in activated T cells. Cell Mol Life Sci 2022; 79:112. [PMID: 35099607 PMCID: PMC8803816 DOI: 10.1007/s00018-022-04154-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 01/06/2022] [Accepted: 01/15/2022] [Indexed: 11/03/2022]
Abstract
T cell activation initiates protective adaptive immunity, but counterbalancing mechanisms are critical to prevent overshooting responses and to maintain immune homeostasis. The CARD11-BCL10-MALT1 (CBM) complex bridges T cell receptor engagement to NF-κB signaling and MALT1 protease activation. Here, we show that ABIN-1 is modulating the suppressive function of A20 in T cells. Using quantitative mass spectrometry, we identified ABIN-1 as an interactor of the CBM signalosome in activated T cells. A20 and ABIN-1 counteract inducible activation of human primary CD4 and Jurkat T cells. While A20 overexpression is able to silence CBM complex-triggered NF-κB and MALT1 protease activation independent of ABIN-1, the negative regulatory function of ABIN-1 depends on A20. The suppressive function of A20 in T cells relies on ubiquitin binding through the C-terminal zinc finger (ZnF)4/7 motifs, but does not involve the deubiquitinating activity of the OTU domain. Our mechanistic studies reveal that the A20/ABIN-1 module is recruited to the CBM complex via A20 ZnF4/7 and that proteasomal degradation of A20 and ABIN-1 releases the CBM complex from the negative impact of both regulators. Ubiquitin binding to A20 ZnF4/7 promotes destructive K48-polyubiquitination to itself and to ABIN-1. Further, after prolonged T cell stimulation, ABIN-1 antagonizes MALT1-catalyzed cleavage of re-synthesized A20 and thereby diminishes sustained CBM complex signaling. Taken together, interdependent post-translational mechanisms are tightly controlling expression and activity of the A20/ABIN-1 silencing module and the cooperative action of both negative regulators is critical to balance CBM complex signaling and T cell activation.
Collapse
Affiliation(s)
- Hongli Yin
- Research Unit Cellular Signal Integration, Molecular Targets and Therapeutics Center, Helmholtz Zentrum München-German Research Center for Environmental Health, Ingolstaedter Landstr. 1, 85764, Neuherberg, Germany
| | - Ozge Karayel
- Department of Proteomics and Signal Transduction, Max-Planck Institute of Biochemistry, Martinsried, Germany
| | - Ying-Yin Chao
- Department of Infection Immunology, Leibniz Institute for Natural Product Research and Infection Biology, Hans-Knöll-Institute and Friedrich Schiller University Jena, Jena, Germany.,Central Institute for Translational Cancer Research (TranslaTUM), Technical University of Munich, Munich, Germany
| | - Thomas Seeholzer
- Research Unit Cellular Signal Integration, Molecular Targets and Therapeutics Center, Helmholtz Zentrum München-German Research Center for Environmental Health, Ingolstaedter Landstr. 1, 85764, Neuherberg, Germany
| | - Isabel Hamp
- Institute for Medicinal Chemistry, Molecular Targets and Therapeutics Center, 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
| | - Oliver Plettenburg
- Institute for Medicinal Chemistry, Molecular Targets and Therapeutics Center, 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
| | - Torben Gehring
- Research Unit Cellular Signal Integration, Molecular Targets and Therapeutics Center, Helmholtz Zentrum München-German Research Center for Environmental Health, Ingolstaedter Landstr. 1, 85764, Neuherberg, Germany
| | - Christina Zielinski
- Department of Infection Immunology, Leibniz Institute for Natural Product Research and Infection Biology, Hans-Knöll-Institute and Friedrich Schiller University Jena, Jena, Germany
| | - Matthias Mann
- Department of Proteomics and Signal Transduction, Max-Planck Institute of Biochemistry, Martinsried, Germany
| | - Daniel Krappmann
- Research Unit Cellular Signal Integration, Molecular Targets and Therapeutics Center, Helmholtz Zentrum München-German Research Center for Environmental Health, Ingolstaedter Landstr. 1, 85764, Neuherberg, Germany.
| |
Collapse
|
30
|
Lu HY, Sharma M, Sharma AA, Lacson A, Szpurko A, Luider J, Dharmani-Khan P, Shameli A, Bell PA, Guilcher GMT, Lewis VA, Vasquez MR, Desai S, McGonigle L, Murguia-Favela L, Wright NAM, Sergi C, Wine E, Overall CM, Suresh S, Turvey SE. Mechanistic understanding of the combined immunodeficiency in complete human CARD11 deficiency. J Allergy Clin Immunol 2021; 148:1559-1574.e13. [PMID: 33872653 DOI: 10.1016/j.jaci.2021.04.006] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2020] [Revised: 04/01/2021] [Accepted: 04/07/2021] [Indexed: 01/04/2023]
Abstract
BACKGROUND Germline pathogenic variants impairing the caspase recruitment domain family member 11 (CARD11)-B cell chronic lymphocytic leukemia/lymphoma 10 (BCL10)-MALT1 paracaspase (MALT1) (CBM) complex are associated with diverse human diseases including combined immunodeficiency (CID), atopy, and lymphoproliferation. However, the impact of CARD11 deficiency on human B-cell development, signaling, and function is incompletely understood. OBJECTIVES This study sought to determine the cellular, immunological, and biochemical basis of disease for 2 unrelated patients who presented with profound CID associated with viral and fungal respiratory infections, interstitial lung disease, and severe colitis. METHODS Patients underwent next-generation sequencing, immunophenotyping by flow cytometry, signaling assays by immunoblot, and transcriptome profiling by RNA-sequencing. RESULTS Both patients carried identical novel pathogenic biallelic loss-of-function variants in CARD11 (c.2509C>T; p.Arg837∗) leading to undetectable protein expression. This variant prevented CBM complex formation, severely impairing the activation of nuclear factor-κB, c-Jun N-terminal kinase, and MALT1 paracaspase activity in B and T cells. This functional defect resulted in a developmental block in B cells at the naive and type 1 transitional B-cell stage and impaired circulating T follicular helper cell (cTFH) development, which was associated with impaired antibody responses and absent germinal center structures on lymph node histology. Transcriptomics indicated that CARD11-dependent signaling is essential for immune signaling pathways involved in the development of these cells. Both patients underwent hematopoietic stem cell transplantations, which led to functional normalization. CONCLUSIONS Complete human CARD11 deficiency causes profound CID by impairing naive/type 1 B-cell and cTFH cell development and abolishing activation of MALT1 paracaspase, NF-κB, and JNK activity. Hematopoietic stem cell transplantation functionally restores impaired signaling pathways.
Collapse
Affiliation(s)
- Henry Y Lu
- Department of Pediatrics, British Columbia Children's Hospital, The University of British Columbia, Vancouver, British Columbia, Canada; Experimental Medicine Program, Faculty of Medicine, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Mehul Sharma
- Department of Pediatrics, British Columbia Children's Hospital, The University of British Columbia, Vancouver, British Columbia, Canada; Experimental Medicine Program, Faculty of Medicine, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Ashish A Sharma
- Department of Pathology, Case Western Reserve University, Cleveland, Ohio; Department of Pathology, Emory University, Atlanta, Ga
| | - Atilano Lacson
- Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, Alberta, Canada
| | - Ashley Szpurko
- Section of Oncology/Bone Marrow Therapy, Departments of Oncology and Pediatrics, Alberta Children's Hospital, University of Calgary, Calgary, Alberta, Canada
| | - Joanne Luider
- Department of Pathology and Laboratory Medicine, University of Calgary, Alberta Precision Laboratories, Calgary, Alberta, Canada
| | - Poonam Dharmani-Khan
- Department of Pathology and Laboratory Medicine, University of Calgary, Alberta Precision Laboratories, Calgary, Alberta, Canada
| | - Afshin Shameli
- Department of Pathology and Laboratory Medicine, University of Calgary, Alberta Precision Laboratories, Calgary, Alberta, Canada
| | - Peter A Bell
- Centre for Blood Research, The University of British Columbia, Vancouver, British Columbia, Canada; Department of Oral Biological and Medical Sciences, Faculty of Dentistry, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Gregory M T Guilcher
- Section of Oncology/Bone Marrow Therapy, Departments of Oncology and Pediatrics, Alberta Children's Hospital, University of Calgary, Calgary, Alberta, Canada
| | - Victor A Lewis
- Section of Oncology/Bone Marrow Therapy, Departments of Oncology and Pediatrics, Alberta Children's Hospital, University of Calgary, Calgary, Alberta, Canada
| | - Marta Rojas Vasquez
- Department of Pediatrics, Division of Immunology, Hematology, Oncology and Palliative Care (iHOPE), University of Alberta, Edmonton, Alberta, Canada
| | - Sunil Desai
- Department of Pediatrics, Division of Immunology, Hematology, Oncology and Palliative Care (iHOPE), University of Alberta, Edmonton, Alberta, Canada
| | - Lyle McGonigle
- Department of Pediatrics, Division of General and Community Pediatrics, University of Alberta, Edmonton, Alberta, Canada
| | - Luis Murguia-Favela
- Section of Pediatric Hematology-Immunology, Department of Pediatrics, Alberta Children's Hospital, University of Calgary, Calgary, Alberta, Canada
| | - Nicola A M Wright
- Section of Pediatric Hematology-Immunology, Department of Pediatrics, Alberta Children's Hospital, University of Calgary, Calgary, Alberta, Canada
| | - Consolato Sergi
- Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, Alberta, Canada
| | - Eytan Wine
- Division of Pediatric Gastroenterology and Nutrition, Department of Pediatrics, University of Alberta, Edmonton, Alberta, Canada
| | - Christopher M Overall
- Centre for Blood Research, The University of British Columbia, Vancouver, British Columbia, Canada; Department of Oral Biological and Medical Sciences, Faculty of Dentistry, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Sneha Suresh
- Department of Pediatrics, Division of Immunology, Hematology, Oncology and Palliative Care (iHOPE), University of Alberta, Edmonton, Alberta, Canada
| | - Stuart E Turvey
- Department of Pediatrics, British Columbia Children's Hospital, The University of British Columbia, Vancouver, British Columbia, Canada; Experimental Medicine Program, Faculty of Medicine, The University of British Columbia, Vancouver, British Columbia, Canada.
| |
Collapse
|
31
|
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
|
32
|
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
|
33
|
WITHDRAWN: Inhibition of Malt1 protease induces apoptosis and cell death in cardiomyocytes. BBA ADVANCES 2021. [DOI: 10.1016/j.bbadva.2021.100017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|
34
|
Campanello L, Traver MK, Shroff H, Schaefer BC, Losert W. Signaling through polymerization and degradation: Analysis and simulations of T cell activation mediated by Bcl10. PLoS Comput Biol 2021; 17:e1007986. [PMID: 34014917 PMCID: PMC8184007 DOI: 10.1371/journal.pcbi.1007986] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 06/07/2021] [Accepted: 04/28/2021] [Indexed: 12/05/2022] Open
Abstract
The adaptive immune system serves as a potent and highly specific defense mechanism against pathogen infection. One component of this system, the effector T cell, facilitates pathogen clearance upon detection of specific antigens by the T cell receptor (TCR). A critical process in effector T cell activation is transmission of signals from the TCR to a key transcriptional regulator, NF-κB. The transmission of this signal involves a highly dynamic process in which helical filaments of Bcl10, a key protein constituent of the TCR signaling cascade, undergo competing processes of polymeric assembly and macroautophagy-dependent degradation. Through computational analysis of three-dimensional, super-resolution optical micrographs, we quantitatively characterize TCR-stimulated Bcl10 filament assembly and length dynamics, and demonstrate that filaments become shorter over time. Additionally, we develop an image-based, bootstrap-like resampling method that demonstrates the preferred association between autophagosomes and both Bcl10-filament ends and punctate-Bcl10 structures, implying that autophagosome-driven macroautophagy is directly responsible for Bcl10 filament shortening. We probe Bcl10 polymerization-depolymerization dynamics with a stochastic Monte-Carlo simulation of nucleation-limited filament assembly and degradation, and we show that high probabilities of filament nucleation in response to TCR engagement could provide the observed robust, homogeneous, and tunable response dynamic. Furthermore, we demonstrate that the speed of filament disassembly preferentially at filament ends provides effective regulatory control. Taken together, these data suggest that Bcl10 filament growth and degradation act as an excitable system that provides a digital response mechanism and the reliable timing critical for T cell activation and regulatory processes. The immune system serves to protect organisms against pathogen-mediated disease. While a strong immune response is needed to eliminate pathogens in host organisms, immune responses that are too robust or too persistent can trigger autoimmune disorders, cancer, and a variety of additional serious human pathologies. Thus, a careful balance of activating and inhibitory mechanisms is necessary to prevent detrimental health outcomes of immune responses. For example, activated effector T cells marshal the immune response and direct killing of pathogen-infected cells; however, effector T cells that are chronically activated can damage and destroy healthy tissue. Here, we study an important internal activation pathway in effector T cells that involves the growth and counterbalancing disassembly (involving a process called macroautophagy) of filamentous cytoplasmic signaling structures. We utilize image analysis of 3-D super-resolution images and Monte Carlo simulations to study a key signal-transduction protein, Bcl10. We found that the speed of filament disassembly has the greatest effect on the magnitude and duration of the response, implying that pharmaceutical interventions aimed at macroautophagy may have substantial impact on effector T cell function. Given that filamentous structures are utilized in numerous immune signaling pathways, our analysis methods could have broad applicability in the signal transduction field.
Collapse
Affiliation(s)
- Leonard Campanello
- Department of Physics, University of Maryland College Park, College Park, Maryland, United States of America
- Institute for Physical Science and Technology, University of Maryland College Park, College Park, Maryland, United States of America
| | - Maria K. Traver
- Department of Microbiology and Immunology, Uniformed Services University of the Health Sciences, Bethesda, Maryland, United States of America
| | - Hari Shroff
- Department of Physics, University of Maryland College Park, College Park, Maryland, United States of America
- Laboratory of High-Resolution Optical Imaging, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Brian C. Schaefer
- Department of Microbiology and Immunology, Uniformed Services University of the Health Sciences, Bethesda, Maryland, United States of America
- * E-mail: (BCS); (WL)
| | - Wolfgang Losert
- Department of Physics, University of Maryland College Park, College Park, Maryland, United States of America
- Institute for Physical Science and Technology, University of Maryland College Park, College Park, Maryland, United States of America
- * E-mail: (BCS); (WL)
| |
Collapse
|
35
|
Huoh YS, Hur S. Death domain fold proteins in immune signaling and transcriptional regulation. FEBS J 2021; 289:4082-4097. [PMID: 33905163 DOI: 10.1111/febs.15901] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 04/07/2021] [Accepted: 04/23/2021] [Indexed: 01/02/2023]
Abstract
Death domain fold (DDF) superfamily comprises of the death domain (DD), death effector domain (DED), caspase activation recruitment domain (CARD), and pyrin domain (PYD). By utilizing a conserved mode of interaction involving six distinct surfaces, a DDF serves as a building block that can densely pack into homomultimers or filaments. Studies of immune signaling components have revealed that DDF-mediated filament formation plays a central role in mediating signal transduction and amplification. The unique ability of DDFs to self-oligomerize upon external signals and induce oligomerization of partner molecules underlies key processes in many innate immune signaling pathways, as exemplified by RIG-I-like receptor signalosome and inflammasome assembly. Recent studies showed that DDFs are not only limited to immune signaling pathways, but also are involved with transcriptional regulation and other biological processes. Considering that DDF annotation still remains a challenge, the current list of DDFs and their functions may represent just the tip of the iceberg within the full spectrum of DDF biology. In this review, we discuss recent advances in our understanding of DDF functions, structures, and assembly architectures with a focus on CARD- and PYD-containing proteins. We also discuss areas of future research and the potential relationship of DDFs with biomolecular condensates formed by liquid-liquid phase separation (LLPS).
Collapse
Affiliation(s)
- Yu-San Huoh
- Department of Biological Chemistry and Molecular Pharmacology, Blavatnik Institute at Harvard Medical School, Boston, MA, USA.,Program in Cellular and Molecular Medicine, Boston Children's Hospital, MA, USA
| | - Sun Hur
- Department of Biological Chemistry and Molecular Pharmacology, Blavatnik Institute at Harvard Medical School, Boston, MA, USA.,Program in Cellular and Molecular Medicine, Boston Children's Hospital, MA, USA
| |
Collapse
|
36
|
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
|
37
|
Rodríguez Gama A, Miller T, Halfmann R. Mechanics of a molecular mousetrap-nucleation-limited innate immune signaling. Biophys J 2021; 120:1150-1160. [PMID: 33460595 PMCID: PMC8059202 DOI: 10.1016/j.bpj.2021.01.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 01/05/2021] [Accepted: 01/07/2021] [Indexed: 12/17/2022] Open
Abstract
Innate immune responses, such as cell death and inflammatory signaling, are typically switch-like in nature. They also involve "prion-like" self-templating polymerization of one or more signaling proteins into massive macromolecular assemblies known as signalosomes. Despite the wealth of atomic-resolution structural information on signalosomes, how the constituent polymers nucleate and whether the switch-like nature of that event at the molecular scale relates to the digital nature of innate immune signaling at the cellular scale remains unknown. In this perspective, we review current knowledge of innate immune signalosome assembly, with an emphasis on structural constraints that allow the proteins to accumulate in inactive soluble forms poised for abrupt polymerization. We propose that structurally encoded nucleation barriers to protein polymerization kinetically regulate the corresponding pathways, which allows for extremely sensitive, rapid, and decisive signaling upon pathogen detection. We discuss how nucleation barriers satisfy the rigorous on-demand functions of the innate immune system but also predispose the system to precocious activation that may contribute to progressive age-associated inflammation.
Collapse
Affiliation(s)
| | - Tayla Miller
- Stowers Institute for Medical Research, Kansas City, Missouri
| | - Randal Halfmann
- Stowers Institute for Medical Research, Kansas City, Missouri; Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, Kansas.
| |
Collapse
|
38
|
Methods to Study CARD11-BCL10-MALT1 Dependent Canonical NF-κB Activation in Jurkat T Cells. Methods Mol Biol 2021; 2366:125-143. [PMID: 34236636 DOI: 10.1007/978-1-0716-1669-7_8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Jurkat T cells have been of central importance for the discovery of signalling mediators driving NF-κB activation in response to T cell antigen receptor (TCR)/CD28 co-stimulation. The critical function of the key regulators identified in Jurkat T cells has subsequently been verified in primary murine and human T cells. CRISPR/Cas9-mediated genomic editing techniques in combination with viral reconstitution are powerful tools that now enable the investigation of the exact molecular mechanisms that govern T cell signalling, especially the impact of protein-protein interactions, protein modifications, or cancer-associated gain- or loss-of-function mutations. As exemplified by the CARD11 gene encoding a key regulator of NF-κB signalling in T cells, we describe here the detailed workflow for the generation of CRISPR/Cas9 knockout (KO) Jurkat T cells and the subsequent reconstitution using a lentiviral transduction protocol. In addition, we explain the use of a stable NF-κB-dependent EGFP reporter system that enables a reliable quantification of NF-κB transcriptional activation in the reconstituted KO Jurkat T cells.
Collapse
|
39
|
Moon CS, Reglero C, Cortes JR, Quinn SA, Alvarez S, Zhao J, Lin WHW, Cooke AJ, Abate F, Soderquist CR, Fiñana C, Inghirami G, Campo E, Bhagat G, Rabadan R, Palomero T, Ferrando AA. FYN-TRAF3IP2 induces NF-κB signaling-driven peripheral T cell lymphoma. NATURE CANCER 2021; 2:98-113. [PMID: 33928261 PMCID: PMC8081346 DOI: 10.1038/s43018-020-00161-w] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Accepted: 12/01/2020] [Indexed: 12/11/2022]
Abstract
Angioimmunoblastic T cell lymphoma (AITL) and peripheral T cell lymphoma not-otherwise-specified (PTCL, NOS) have poor prognosis and lack driver actionable targets for directed therapies in most cases. Here we identify FYN-TRAF3IP2 as a recurrent oncogenic gene fusion in AITL and PTCL, NOS tumors. Mechanistically, we show that FYN-TRAF3IP2 leads to aberrant NF-κB signaling downstream of T cell receptor activation. Consistent with a driver oncogenic role, FYN-TRAF3IP2 expression in hematopoietic progenitors induces NF-κB-driven T cell transformation in mice and cooperates with loss of the Tet2 tumor suppressor in PTCL development. Moreover, abrogation of NF-κB signaling in FYN-TRAF3IP2-induced tumors with IκB kinase inhibitors delivers strong anti-lymphoma effects in vitro and in vivo. These results demonstrate an oncogenic and pharmacologically targetable role for FYN-TRAF3IP2 in PTCLs and call for the clinical testing of anti-NF-κB targeted therapies in these diseases.
Collapse
Affiliation(s)
- Christine S Moon
- Institute for Cancer Genetics, Columbia University, New York, NY, USA
| | - Clara Reglero
- Institute for Cancer Genetics, Columbia University, New York, NY, USA
| | - Jose R Cortes
- Institute for Cancer Genetics, Columbia University, New York, NY, USA
| | - S Aidan Quinn
- Institute for Cancer Genetics, Columbia University, New York, NY, USA
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY, USA
| | - Silvia Alvarez
- Institute for Cancer Genetics, Columbia University, New York, NY, USA
| | - Junfei Zhao
- Department of Systems Biology, Columbia University, New York, NY, USA
| | - Wen-Hsuan W Lin
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY, USA
| | - Anisha J Cooke
- Institute for Cancer Genetics, Columbia University, New York, NY, USA
| | - Francesco Abate
- Department of Systems Biology, Columbia University, New York, NY, USA
| | - Craig R Soderquist
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY, USA
| | - Claudia Fiñana
- Institute for Cancer Genetics, Columbia University, New York, NY, USA
| | - Giorgio Inghirami
- Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Elias Campo
- Department of Pathology, Hospital Clinic of Barcelona, Institute of Biomedical Research August Pi i Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain
| | - Govind Bhagat
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY, USA
| | - Raul Rabadan
- Department of Systems Biology, Columbia University, New York, NY, USA
- Department of Biomedical Informatics, Columbia University, New York, NY, USA
| | - Teresa Palomero
- Institute for Cancer Genetics, Columbia University, New York, NY, USA.
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY, USA.
| | - Adolfo A Ferrando
- Institute for Cancer Genetics, Columbia University, New York, NY, USA.
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY, USA.
- Department of Systems Biology, Columbia University, New York, NY, USA.
- Department of Pediatrics, Columbia University Medical Center, New York, NY, USA.
| |
Collapse
|
40
|
Meitlis I, Allenspach EJ, Bauman BM, Phan IQ, Dabbah G, Schmitt EG, Camp ND, Torgerson TR, Nickerson DA, Bamshad MJ, Hagin D, Luthers CR, Stinson JR, Gray J, Lundgren I, Church JA, Butte MJ, Jordan MB, Aceves SS, Schwartz DM, Milner JD, Schuval S, Skoda-Smith S, Cooper MA, Starita LM, Rawlings DJ, Snow AL, James RG. Multiplexed Functional Assessment of Genetic Variants in CARD11. Am J Hum Genet 2020; 107:1029-1043. [PMID: 33202260 PMCID: PMC7820631 DOI: 10.1016/j.ajhg.2020.10.015] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Accepted: 10/27/2020] [Indexed: 12/28/2022] Open
Abstract
Genetic testing has increased the number of variants identified in disease genes, but the diagnostic utility is limited by lack of understanding variant function. CARD11 encodes an adaptor protein that expresses dominant-negative and gain-of-function variants associated with distinct immunodeficiencies. Here, we used a "cloning-free" saturation genome editing approach in a diploid cell line to simultaneously score 2,542 variants for decreased or increased function in the region of CARD11 associated with immunodeficiency. We also described an exon-skipping mechanism for CARD11 dominant-negative activity. The classification of reported clinical variants was sensitive (94.6%) and specific (88.9%), which rendered the data immediately useful for interpretation of seven coding and splicing variants implicated in immunodeficiency found in our clinic. This approach is generalizable for variant interpretation in many other clinically actionable genes, in any relevant cell type.
Collapse
Affiliation(s)
- Iana Meitlis
- Seattle Children's Research Institute, Seattle, WA 98101, USA
| | - Eric J Allenspach
- Seattle Children's Research Institute, Seattle, WA 98101, USA; Department of Pediatrics, University of Washington, Seattle, WA 98195, USA; Brotman-Baty Institute for Precision Medicine, Seattle, WA 98195, USA
| | - Bradly M Bauman
- Department of Pharmacology & Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
| | - Isabelle Q Phan
- Seattle Children's Research Institute, Seattle, WA 98101, USA
| | - Gina Dabbah
- Department of Pharmacology & Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
| | - Erica G Schmitt
- Department of Pediatrics, Division of Rheumatology/Immunology, Washington University in St. Louis, MO 63130, USA
| | - Nathan D Camp
- Seattle Children's Research Institute, Seattle, WA 98101, USA
| | | | - Deborah A Nickerson
- Seattle Children's Research Institute, Seattle, WA 98101, USA; Department of Genome Sciences, University of Washington, Seattle, WA 98195, USA; Brotman-Baty Institute for Precision Medicine, Seattle, WA 98195, USA
| | - Michael J Bamshad
- Department of Pediatrics, University of Washington, Seattle, WA 98195, USA; Department of Genome Sciences, University of Washington, Seattle, WA 98195, USA; Brotman-Baty Institute for Precision Medicine, Seattle, WA 98195, USA
| | - David Hagin
- Allergy and Clinical Immunology Unit, Department of Medicine, Tel Aviv Sourasky Medical Center and Sackler Faculty of Medicine, University of Tel Aviv, Tel Aviv 62919, Israel
| | - Christopher R Luthers
- Department of Pharmacology & Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
| | - Jeffrey R Stinson
- Department of Pharmacology & Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
| | - Jessica Gray
- Divisions of Immunobiology, and Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| | | | - Joseph A Church
- Department of Pediatrics, Keck School of Medicine, University of Southern California and Children's Hospital Los Angeles, Los Angeles, CA 90033, USA
| | - Manish J Butte
- Division of Immunology, Allergy, and Rheumatology, Department of Pediatrics, University of California Los Angeles, Los Angeles, CA 90404, USA
| | - Mike B Jordan
- Divisions of Immunobiology, and Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| | - Seema S Aceves
- Division of Allergy Immunology, Departments of Pediatrics and Medicine, University of California, San Diego, and Rady Children's Hospital, San Diego, CA 92123, USA
| | | | - Joshua D Milner
- Department of Pediatrics, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Susan Schuval
- Department of Pediatrics, Stonybrook University, Stony Brook, NY 11794, USA
| | - Suzanne Skoda-Smith
- Seattle Children's Research Institute, Seattle, WA 98101, USA; Department of Pediatrics, University of Washington, Seattle, WA 98195, USA
| | - Megan A Cooper
- Department of Pediatrics, Division of Rheumatology/Immunology, Washington University in St. Louis, MO 63130, USA
| | - Lea M Starita
- Department of Genome Sciences, University of Washington, Seattle, WA 98195, USA; Brotman-Baty Institute for Precision Medicine, Seattle, WA 98195, USA
| | - David J Rawlings
- Seattle Children's Research Institute, Seattle, WA 98101, USA; Department of Pediatrics, University of Washington, Seattle, WA 98195, USA; Department of Immunology, University of Washington, Seattle, WA 98195, USA
| | - Andrew L Snow
- Department of Pharmacology & Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
| | - Richard G James
- Seattle Children's Research Institute, Seattle, WA 98101, USA; Department of Pediatrics, University of Washington, Seattle, WA 98195, USA; Department of Pharmacology, University of Washington, Seattle, WA 98195, USA; Brotman-Baty Institute for Precision Medicine, Seattle, WA 98195, USA.
| |
Collapse
|
41
|
Bao W, Sun C, Sun X, He M, Yu H, Yan W, Wen F, Zhang L, Yang C. Targeting BCL10 by small peptides for the treatment of B cell lymphoma. Am J Cancer Res 2020; 10:11622-11636. [PMID: 33052237 PMCID: PMC7546004 DOI: 10.7150/thno.47533] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Accepted: 09/08/2020] [Indexed: 12/13/2022] Open
Abstract
Rationale: Constitutive activation of the NF-κB signalling pathway plays a pivotal role in the pathogenesis of activated B cell-like diffuse large B-cell lymphomas (ABC-DLBCLs), the most aggressive and chemoresistant form of DLBCL. In ABC-DLBCLs, the CARMA1-BCL10 (CB) complex forms a filamentous structure and functions as a supramolecular organizing centre (CB-SMOC) that is required for constitutive NF-κB activation, making it an attractive drug target for ABC-DLBCL treatment. However, a pharmaceutical approach targeting CB-SMOC has been lacking. Here, we developed Bcl10 peptide inhibitors (BPIs) that specifically target the BCL10 filamentation process. Methods: Electron microscopy and immunofluorescence imaging were used to visualize the effect of the BPIs on the BCL10 filamentation process. The cytotoxicity of the tested BPIs was evaluated in DLBCL cell lines according to cell proliferation assays. Different in vitro experiments (pharmacokinetics, immunoprecipitation, western blotting, annexin V and PI staining) were conducted to determine the functional mechanisms of the BPIs. The in vivo therapeutic effect of the BPIs was examined in different xenograft DLBCL mouse models. Finally, Ki67 and TUNEL staining and histopathology analysis were used to evaluate the antineoplastic mechanisms and systemic toxicity of the BPIs. Results: We showed that these BPIs can effectively disrupt the BCL10 filamentation process, destabilize BCL10 and suppress NF-κB signalling in ABC-DLBCL cells. By examining a panel of DLBCL cell lines, we found that these BPIs selectively repressed the growth of CB-SMOC-dependent DLBCL cells by inducing apoptosis and cell cycle arrest. Moreover, by converting the BPIs to acquire a D-retro inverso (DRI) configuration, we developed DRI-BPIs with significantly improved intracellular stability and unimpaired BPI activity. These DRI-BPIs selectively repressed the growth of CB-SMOC-dependent DLBCL tumors in mouse xenograft models without eliciting discernible adverse effects. Conclusion: We developed novel BPIs to target the BCL10 filamentation process and demonstrated that targeting BCL10 by BPIs is a potentially safe and effective pharmaceutical approach for the treatment of ABC-DLBCL and other CB-SMOC-dependent malignancies.
Collapse
|
42
|
Gehring T, Erdmann T, Rahm M, Graß C, Flatley A, O'Neill TJ, Woods S, Meininger I, Karayel O, Kutzner K, Grau M, Shinohara H, Lammens K, Feederle R, Hauck SM, Lenz G, Krappmann D. MALT1 Phosphorylation Controls Activation of T Lymphocytes and Survival of ABC-DLBCL Tumor Cells. Cell Rep 2020; 29:873-888.e10. [PMID: 31644910 DOI: 10.1016/j.celrep.2019.09.040] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Revised: 06/24/2019] [Accepted: 09/12/2019] [Indexed: 02/06/2023] Open
Abstract
The CARMA1/CARD11-BCL10-MALT1 (CBM) complex bridges T and B cell antigen receptor (TCR/BCR) ligation to MALT1 protease activation and canonical nuclear factor κB (NF-κB) signaling. Using unbiased mass spectrometry, we discover multiple serine phosphorylation sites in the MALT1 C terminus after T cell activation. Phospho-specific antibodies reveal that CBM-associated MALT1 is transiently hyper-phosphorylated upon TCR/CD28 co-stimulation. We identify a dual role for CK1α as a kinase that is essential for CBM signalosome assembly as well as MALT1 phosphorylation. Although MALT1 phosphorylation is largely dispensable for protease activity, it fosters canonical NF-κB signaling in Jurkat and murine CD4 T cells. Moreover, constitutive MALT1 phosphorylation promotes survival of activated B cell-type diffuse large B cell lymphoma (ABC-DLBCL) cells addicted to chronic BCR signaling. Thus, MALT1 phosphorylation triggers optimal NF-κB activation in lymphocytes and survival of lymphoma cells.
Collapse
Affiliation(s)
- Torben Gehring
- Research Unit Cellular Signal Integration, Institute of Molecular Toxicology and Pharmacology, Helmholtz-Zentrum München, German Research Center for Environmental Health (GmbH), Ingolstädter Landstraße 1, 85764 Neuherberg, Germany
| | - Tabea Erdmann
- Department of Medicine A, Hematology, Oncology and Pneumology, University Hospital Münster, 48149 Münster, Germany
| | - Marco Rahm
- Research Unit Protein Science, Helmholtz-Zentrum München, German Research Center for Environmental Health (GmbH), Ingolstädter Landstraße 1, 85764 Neuherberg, Germany
| | - Carina Graß
- Research Unit Cellular Signal Integration, Institute of Molecular Toxicology and Pharmacology, Helmholtz-Zentrum München, German Research Center for Environmental Health (GmbH), Ingolstädter Landstraße 1, 85764 Neuherberg, Germany
| | - Andrew Flatley
- Monoclonal Antibody Core Facility and Research Group, Institute for Diabetes and Obesity, Helmholtz-Zentrum München, German Research Center for Environmental Health (GmbH) Ingolstädter Landstraße 1, 85764 Neuherberg, Germany
| | - Thomas J O'Neill
- Research Unit Cellular Signal Integration, Institute of Molecular Toxicology and Pharmacology, Helmholtz-Zentrum München, German Research Center for Environmental Health (GmbH), Ingolstädter Landstraße 1, 85764 Neuherberg, Germany
| | - Simone Woods
- Research Unit Cellular Signal Integration, Institute of Molecular Toxicology and Pharmacology, Helmholtz-Zentrum München, German Research Center for Environmental Health (GmbH), Ingolstädter Landstraße 1, 85764 Neuherberg, Germany
| | - Isabel Meininger
- Research Unit Cellular Signal Integration, Institute of Molecular Toxicology and Pharmacology, Helmholtz-Zentrum München, German Research Center for Environmental Health (GmbH), Ingolstädter Landstraße 1, 85764 Neuherberg, Germany
| | - Ozge Karayel
- Department of Proteomics and Signal Transduction, Max-Planck Institute of Biochemistry, Am Klopferspitz 18, 82152 Planegg, Germany
| | - Kerstin Kutzner
- Research Unit Cellular Signal Integration, Institute of Molecular Toxicology and Pharmacology, Helmholtz-Zentrum München, German Research Center for Environmental Health (GmbH), Ingolstädter Landstraße 1, 85764 Neuherberg, Germany
| | - Michael Grau
- Department of Medicine A, Hematology, Oncology and Pneumology, University Hospital Münster, 48149 Münster, Germany
| | - Hisaaki Shinohara
- Laboratory for Systems Immunology, Faculty of Pharmaceutical Sciences, Sanyo-Onoda City University.1-1-1, Daigakudori, Sanyo-onoda City, Yamaguchi 756-0884, Japan
| | - Katja Lammens
- Gene Center, Ludwig-Maximilians University, Feodor-Lynen-Str. 25, 81377 München, Germany
| | - Regina Feederle
- Monoclonal Antibody Core Facility and Research Group, Institute for Diabetes and Obesity, Helmholtz-Zentrum München, German Research Center for Environmental Health (GmbH) Ingolstädter Landstraße 1, 85764 Neuherberg, Germany
| | - Stefanie M Hauck
- Research Unit Protein Science, Helmholtz-Zentrum München, German Research Center for Environmental Health (GmbH), Ingolstädter Landstraße 1, 85764 Neuherberg, Germany
| | - Georg Lenz
- Department of Medicine A, Hematology, Oncology and Pneumology, University Hospital Münster, 48149 Münster, Germany
| | - Daniel Krappmann
- Research Unit Cellular Signal Integration, Institute of Molecular Toxicology and Pharmacology, Helmholtz-Zentrum München, German Research Center for Environmental Health (GmbH), Ingolstädter Landstraße 1, 85764 Neuherberg, Germany.
| |
Collapse
|
43
|
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
|
44
|
Staal J, Driege Y, Haegman M, Kreike M, Iliaki S, Vanneste D, Lork M, Afonina IS, Braun H, Beyaert R. Defining the combinatorial space of PKC::CARD‐CC signal transduction nodes. FEBS J 2020; 288:1630-1647. [DOI: 10.1111/febs.15522] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Revised: 07/12/2020] [Accepted: 07/30/2020] [Indexed: 12/14/2022]
Affiliation(s)
- Jens Staal
- Department of Biomedical Molecular Biology Ghent University Ghent Belgium
- Center for Inflammation Research Unit of Molecular Signal Transduction in Inflammation VIB Ghent Belgium
| | - Yasmine Driege
- Department of Biomedical Molecular Biology Ghent University Ghent Belgium
- Center for Inflammation Research Unit of Molecular Signal Transduction in Inflammation VIB Ghent Belgium
| | - Mira Haegman
- Department of Biomedical Molecular Biology Ghent University Ghent Belgium
- Center for Inflammation Research Unit of Molecular Signal Transduction in Inflammation VIB Ghent Belgium
| | - Marja Kreike
- Department of Biomedical Molecular Biology Ghent University Ghent Belgium
- Center for Inflammation Research Unit of Molecular Signal Transduction in Inflammation VIB Ghent Belgium
| | - Styliani Iliaki
- Department of Biomedical Molecular Biology Ghent University Ghent Belgium
- Center for Inflammation Research Unit of Molecular Signal Transduction in Inflammation VIB Ghent Belgium
| | - Domien Vanneste
- Department of Biomedical Molecular Biology Ghent University Ghent Belgium
- Center for Inflammation Research Unit of Molecular Signal Transduction in Inflammation VIB Ghent Belgium
| | - Marie Lork
- Department of Biomedical Molecular Biology Ghent University Ghent Belgium
- Center for Inflammation Research Unit of Molecular Signal Transduction in Inflammation VIB Ghent Belgium
| | - Inna S. Afonina
- Department of Biomedical Molecular Biology Ghent University Ghent Belgium
- Center for Inflammation Research Unit of Molecular Signal Transduction in Inflammation VIB Ghent Belgium
| | - Harald Braun
- Department of Biomedical Molecular Biology Ghent University Ghent Belgium
- Center for Inflammation Research Unit of Molecular Signal Transduction in Inflammation VIB Ghent Belgium
| | - Rudi Beyaert
- Department of Biomedical Molecular Biology Ghent University Ghent Belgium
- Center for Inflammation Research Unit of Molecular Signal Transduction in Inflammation VIB Ghent Belgium
| |
Collapse
|
45
|
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
|
46
|
Hughes N, Erbel P, Bornancin F, Wiesmann C, Schiering N, Villard F, Decock A, Rubi B, Melkko S, Spanka C, Buschmann N, Pissot‐Soldermann C, Simic O, Beerli R, Sorge M, Tintelnot‐Blomley M, Beltz K, Régnier CH, Quancard J, Schlapbach A, Langlois J, Renatus M. Stabilizing Inactive Conformations of MALT1 as an Effective Approach to Inhibit Its Protease Activity. ADVANCED THERAPEUTICS 2020. [DOI: 10.1002/adtp.202000078] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Nicola Hughes
- Novartis Institutes for Biomedical Reseach (NIBR) Novartis Campus Basel CH‐4002 Switzerland
| | - Paul Erbel
- Novartis Institutes for Biomedical Reseach (NIBR) Novartis Campus Basel CH‐4002 Switzerland
| | - Frédéric Bornancin
- Novartis Institutes for Biomedical Reseach (NIBR) Novartis Campus Basel CH‐4002 Switzerland
| | - Christian Wiesmann
- Novartis Institutes for Biomedical Reseach (NIBR) Novartis Campus Basel CH‐4002 Switzerland
| | - Nikolaus Schiering
- Novartis Institutes for Biomedical Reseach (NIBR) Novartis Campus Basel CH‐4002 Switzerland
| | - Frédéric Villard
- Novartis Institutes for Biomedical Reseach (NIBR) Novartis Campus Basel CH‐4002 Switzerland
| | - Arnaud Decock
- Novartis Institutes for Biomedical Reseach (NIBR) Novartis Campus Basel CH‐4002 Switzerland
| | - Bertran Rubi
- Laboratorium für Organische Chemie Zürich CH‐8093 Switzerland
| | - Samu Melkko
- Novartis Institutes for Biomedical Reseach (NIBR) Novartis Campus Basel CH‐4002 Switzerland
| | - Carsten Spanka
- Novartis Institutes for Biomedical Reseach (NIBR) Novartis Campus Basel CH‐4002 Switzerland
| | - Nicole Buschmann
- Novartis Institutes for Biomedical Reseach (NIBR) Novartis Campus Basel CH‐4002 Switzerland
| | | | - Oliver Simic
- Novartis Institutes for Biomedical Reseach (NIBR) Novartis Campus Basel CH‐4002 Switzerland
| | - René Beerli
- Novartis Institutes for Biomedical Reseach (NIBR) Novartis Campus Basel CH‐4002 Switzerland
| | - Mickael Sorge
- Novartis Institutes for Biomedical Reseach (NIBR) Novartis Campus Basel CH‐4002 Switzerland
| | | | - Karen Beltz
- Novartis Institutes for Biomedical Reseach (NIBR) Novartis Campus Basel CH‐4002 Switzerland
| | - Catherine H. Régnier
- Novartis Institutes for Biomedical Reseach (NIBR) Novartis Campus Basel CH‐4002 Switzerland
| | - Jean Quancard
- Novartis Institutes for Biomedical Reseach (NIBR) Novartis Campus Basel CH‐4002 Switzerland
| | - Achim Schlapbach
- Novartis Institutes for Biomedical Reseach (NIBR) Novartis Campus Basel CH‐4002 Switzerland
| | - Jean‐Baptiste Langlois
- Novartis Institutes for Biomedical Reseach (NIBR) Novartis Campus Basel CH‐4002 Switzerland
| | - Martin Renatus
- Novartis Institutes for Biomedical Reseach (NIBR) Novartis Campus Basel CH‐4002 Switzerland
| |
Collapse
|
47
|
Assembly of platforms for signal transduction in the new era: dimerization, helical filament assembly, and beyond. Exp Mol Med 2020; 52:356-366. [PMID: 32139779 PMCID: PMC7156525 DOI: 10.1038/s12276-020-0391-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2020] [Accepted: 01/31/2020] [Indexed: 11/08/2022] Open
Abstract
Supramolecular organizing center (SMOC)-mediated signal transduction is an emerging concept in the field of signal transduction that is ushering in a new era. The formation of location-specific, higher-order SMOCs is particularly important for cell death and innate immune signaling processes. Several protein interaction domains, including the death domain (DD) superfamily and the CIDE domain, are representative mediators of SMOC assembly in cell death and innate immune signaling pathways. DD superfamily- and CIDE domain-containing proteins form SMOCs that activate various caspases and provide signaling scaffold platforms. These assemblies can lead to signal transduction and amplification during signaling events. In this review, we summarize recent findings on the molecular basis of DD superfamily- and CIDE domain-mediated SMOC formation. Improved understanding of large molecular signaling complexes that form during innate (nonspecific) immune responses could help develop treatments for multiple diseases including cancer. Correct cell signaling requires precise protein interactions and binding, which are mediated by specific sites on the surface of the protein molecules involved. Innate immune responses and cell death mechanisms rely on such protein interactions, and defects can cause signaling abnormalities and trigger disease. Hyun Ho Park and co-workers at Chung-Ang University in Seoul, South Korea, reviewed recent insights into the presence of supramolecular organizing centers (SMOCs), localized complexes of signaling proteins that form during immune responses. The researchers highlight existing understanding of SMOC assembly processes. A better understanding of SMOCs will help to explain enzyme activation, signal amplification and cell signaling control mechanisms.
Collapse
|
48
|
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
|
49
|
Park HH. Domain swapping of death domain superfamily: Alternative strategy for dimerization. Int J Biol Macromol 2019; 138:565-572. [DOI: 10.1016/j.ijbiomac.2019.07.139] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Revised: 07/22/2019] [Accepted: 07/23/2019] [Indexed: 01/20/2023]
|
50
|
Gentle IE. Supramolecular Complexes in Cell Death and Inflammation and Their Regulation by Autophagy. Front Cell Dev Biol 2019; 7:73. [PMID: 31131275 PMCID: PMC6509160 DOI: 10.3389/fcell.2019.00073] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Accepted: 04/10/2019] [Indexed: 12/23/2022] Open
Abstract
Signaling activation is a tightly regulated process involving myriad posttranslational modifications such as phosphorylation/dephosphorylation, ubiquitylation/deubiquitylation, proteolytical cleavage events as well as translocation of proteins to new compartments within the cell. In addition to each of these events potentially regulating individual proteins, the assembly of very large supramolecular complexes has emerged as a common theme in signal transduction and is now known to regulate many signaling events. This is particularly evident in pathways regulating both inflammation and cell death/survival. Regulation of the assembly and silencing of these complexes plays important roles in immune signaling and inflammation and the fate of cells to either die or survive. Here we will give a summary of some of the better studied supramolecular complexes involved in inflammation and cell death, particularly with a focus on diseases caused by their autoactivation and the role autophagy either plays or may be playing in their regulation.
Collapse
Affiliation(s)
- Ian E Gentle
- Faculty of Medicine, Institute of Medical Microbiology and Hygiene, Medical Center, University of Freiburg, Freiburg im Breisgau, Germany
| |
Collapse
|