Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
J Biol Chem ; 294(40): 14648-14660, 2019 10 04.
Article in English | MEDLINE | ID: mdl-31391255

ABSTRACT

The activation of key signaling pathways downstream of antigen receptor engagement is critically required for normal lymphocyte activation during the adaptive immune response. CARD11 is a multidomain signaling scaffold protein required for antigen receptor signaling to NF-κB, c-Jun N-terminal kinase, and mTOR. Germline mutations in the CARD11 gene result in at least four types of primary immunodeficiency, and somatic CARD11 gain-of-function mutations drive constitutive NF-κB activity in diffuse large B cell lymphoma and other lymphoid cancers. In response to antigen receptor triggering, CARD11 transitions from a closed, inactive state to an open, active scaffold that recruits multiple signaling partners into a complex to relay downstream signaling. However, how this signal-induced CARD11 conversion occurs remains poorly understood. Here we investigate the role of Inducible Element 1 (IE1), a short regulatory element in the CARD11 Inhibitory Domain, in the CARD11 signaling cycle. We find that IE1 controls the signal-dependent Opening Step that makes CARD11 accessible to the binding of cofactors, including Bcl10, MALT1, and the HOIP catalytic subunit of the linear ubiquitin chain assembly complex. Surprisingly, we find that IE1 is also required at an independent step for the maximal activation of HOIP and MALT1 enzymatic activity after cofactor recruitment to CARD11. This role of IE1 reveals that there is an Enzymatic Activation Step in the CARD11 signaling cycle that is distinct from the Cofactor Association Step. Our results indicate that CARD11 has evolved to actively coordinate scaffold opening and the induction of enzymatic activity among recruited cofactors during antigen receptor signaling.


Subject(s)
Adaptive Immunity/genetics , CARD Signaling Adaptor Proteins/chemistry , Guanylate Cyclase/chemistry , Multiprotein Complexes/chemistry , Receptors, Antigen/genetics , B-Cell CLL-Lymphoma 10 Protein/genetics , CARD Signaling Adaptor Proteins/genetics , CARD Signaling Adaptor Proteins/ultrastructure , Germ-Line Mutation/genetics , Guanylate Cyclase/genetics , Guanylate Cyclase/ultrastructure , Humans , JNK Mitogen-Activated Protein Kinases/genetics , Jurkat Cells , Lymphocyte Activation/genetics , Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein/genetics , Multiprotein Complexes/genetics , Multiprotein Complexes/ultrastructure , NF-kappa B/genetics , Protein Binding/genetics , Protein Conformation , Receptors, Antigen/chemistry , Signal Transduction/genetics , TOR Serine-Threonine Kinases/genetics , Ubiquitin-Protein Ligases/genetics
2.
J Clin Pathol ; 71(4): 372-378, 2018 Apr.
Article in English | MEDLINE | ID: mdl-29197855

ABSTRACT

AIMS: Genetic abnormalities, including copy number variants (CNV), copy number neutral loss of heterozygosity (CN-LOH) and gene mutations, underlie the pathogenesis of myeloid malignancies and serve as important diagnostic, prognostic and/or therapeutic markers. Currently, multiple testing strategies are required for comprehensive genetic testing in myeloid malignancies. The aim of this proof-of-principle study was to investigate the feasibility of combining detection of genome-wide large CNVs, CN-LOH and targeted gene mutations into a single assay using next-generation sequencing (NGS). METHODS: For genome-wide CNV detection, we designed a single nucleotide polymorphism (SNP) sequencing backbone with 22 762 SNP regions evenly distributed across the entire genome. For targeted mutation detection, 62 frequently mutated genes in myeloid malignancies were targeted. We combined this SNP sequencing backbone with a targeted mutation panel, and sequenced 9 healthy individuals and 16 patients with myeloid malignancies using NGS. RESULTS: We detected 52 somatic CNVs, 11 instances of CN-LOH and 39 oncogenic mutations in the 16 patients with myeloid malignancies, and none in the 9 healthy individuals. All CNVs and CN-LOH were confirmed by SNP microarray analysis. CONCLUSIONS: We describe a genome-wide SNP sequencing backbone which allows for sensitive detection of genome-wide CNVs and CN-LOH using NGS. This proof-of-principle study has demonstrated that this strategy can provide more comprehensive genetic profiling for patients with myeloid malignancies using a single assay.


Subject(s)
DNA Copy Number Variations/genetics , High-Throughput Nucleotide Sequencing/methods , Myeloproliferative Disorders/genetics , Female , Humans , Male , Polymorphism, Single Nucleotide/genetics
4.
J Biol Chem ; 291(16): 8324-36, 2016 Apr 15.
Article in English | MEDLINE | ID: mdl-26884335

ABSTRACT

Several classes of signaling proteins contain autoinhibitory domains that prevent unwarranted signaling and coordinate the induction of activity in response to external cues. CARD11, a scaffold protein critical for antigen receptor signaling to NF-κB, undergoes autoregulation by a poorly understood inhibitory domain (ID), which keeps CARD11 inactive in the absence of receptor triggering through inhibitory intramolecular interactions. This autoinhibitory strategy makes CARD11 highly susceptible to gain-of-function mutations that are frequently observed in diffuse large B cell lymphoma (DLBCL) and that disrupt ID-mediated autoinhibition, leading to constitutive NF-κB activity, which can promote lymphoma proliferation. Although DLBCL-associated CARD11 mutations in the caspase recruitment domain (CARD), LATCH domain, and coiled coil have been shown to disrupt intramolecular ID binding, surprisingly, no gain-of-function mutations in the ID itself have been reported and validated. In this study, we solve this paradox and report that the CARD11 ID contains an unusual array of four repressive elements that function cooperatively with redundancy to prevent spontaneous NF-κB activation. Our quantitative analysis suggests that potent oncogenic CARD11 mutations must perturb autoinhibition by at least three repressive elements. Our results explain the lack of ID mutations in DLBCL and reveal an unusual autoinhibitory domain structure and strategy for preventing unwarranted scaffold signaling to NF-κB.


Subject(s)
CARD Signaling Adaptor Proteins/metabolism , Guanylate Cyclase/metabolism , Lymphoma, Large B-Cell, Diffuse/metabolism , Neoplasm Proteins/metabolism , Signal Transduction , CARD Signaling Adaptor Proteins/genetics , Cell Proliferation , Guanylate Cyclase/genetics , HEK293 Cells , Humans , Jurkat Cells , Lymphoma, Large B-Cell, Diffuse/genetics , Lymphoma, Large B-Cell, Diffuse/pathology , NF-kappa B/genetics , NF-kappa B/metabolism , Neoplasm Proteins/genetics , Protein Structure, Tertiary
5.
J Biol Chem ; 291(16): 8338-48, 2016 Apr 15.
Article in English | MEDLINE | ID: mdl-26884334

ABSTRACT

The CARD11 signaling scaffold transmits signaling between antigen receptors on B and T lymphocytes and the transcription factor NF-κB during the adaptive immune response. CARD11 activity is controlled by an inhibitory domain (ID), which participates in intramolecular interactions and prevents cofactor binding prior to receptor triggering. Oncogenic CARD11 mutations associated with the activated B cell-like subtype of diffuse large B cell lymphoma somehow perturb ID-mediated autoinhibition to confer CARD11 with the dysregulated spontaneous signaling to NF-κB that is required for the proliferation and survival of the lymphoma. Here, we investigate how the four repressive elements (REs) we have discovered in the CARD11 ID function to inhibit CARD11 activity with cooperativity and redundancy. We find that each RE contributes to the maintenance of the closed inactive state of CARD11 that predominates in the absence of receptor engagement. Each RE also contributes to the prevention of Bcl10 binding in the basal unstimulated state. RE1, RE2, and RE3 participate in intramolecular interactions with other CARD11 domains and share domain targets for binding. Remarkably, diffuse large B cell lymphoma-associated gain-of-function mutations in the caspase recruitment domain, LATCH, or coiled coil can perturb intramolecular interactions mediated by multiple REs, suggesting how single amino acid oncogenic CARD11 mutations can perturb or bypass the action of redundant inhibitory REs to achieve the level of hyperactive CARD11 signaling required to support lymphoma growth.


Subject(s)
CARD Signaling Adaptor Proteins/metabolism , Guanylate Cyclase/metabolism , Lymphoma, Large B-Cell, Diffuse/metabolism , Neoplasm Proteins/metabolism , Signal Transduction , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Amino Acid Substitution , B-Cell CLL-Lymphoma 10 Protein , CARD Signaling Adaptor Proteins/genetics , Guanylate Cyclase/genetics , HEK293 Cells , Humans , Jurkat Cells , Lymphoma, Large B-Cell, Diffuse/genetics , Mutation , NF-kappa B/genetics , NF-kappa B/metabolism , Neoplasm Proteins/genetics , Protein Structure, Tertiary
6.
Mol Cell Biol ; 36(5): 794-808, 2015 Dec 28.
Article in English | MEDLINE | ID: mdl-26711259

ABSTRACT

NF-κB activation downstream of antigen receptor engagement is a highly regulated event required for lymphocyte activation during the adaptive immune response. The pathway is often dysregulated in lymphoma, leading to constitutive NF-κB activity that supports the aberrant proliferation of transformed lymphocytes. To identify novel regulators of antigen receptor signaling to NF-κB, we developed bioluminescence resonance energy transfer-based interaction cloning (BRIC), a screening strategy that can detect protein-protein interactions in live mammalian cells in a high-throughput manner. Using this strategy, we identified the RING finger protein RNF181 as an interactor of CARD11, a key signaling scaffold in the antigen receptor pathway. We present evidence that RNF181 functions as an E3 ubiquitin ligase to inhibit antigen receptor signaling to NF-κB downstream of CARD11. The levels of the obligate signaling protein Bcl10 are reduced by RNF181 even prior to signaling, and Bcl10 can serve as a substrate for RNF181 E3 ligase activity in vitro. Furthermore, RNF181 limits the proliferation of human diffuse large B cell lymphoma cells that depend upon aberrant CARD11 signaling to NF-κB for growth and survival in culture. Our results define a new regulatory checkpoint that can modulate the output of CARD11 signaling to NF-κB in both normal and transformed lymphocytes.


Subject(s)
CARD Signaling Adaptor Proteins/metabolism , Guanylate Cyclase/metabolism , Lymphoma/metabolism , Receptors, Antigen/metabolism , Signal Transduction , Ubiquitin-Protein Ligases/metabolism , Adaptor Proteins, Signal Transducing/metabolism , B-Cell CLL-Lymphoma 10 Protein , CARD Signaling Adaptor Proteins/chemistry , Cell Survival , Guanylate Cyclase/chemistry , HEK293 Cells , Humans , Jurkat Cells , NF-kappa B/metabolism , Protein Interaction Maps , Protein Structure, Tertiary , Ubiquitination
SELECTION OF CITATIONS
SEARCH DETAIL
...