Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Nat Commun ; 14(1): 4972, 2023 08 17.
Article in English | MEDLINE | ID: mdl-37591835

ABSTRACT

Th17 cells that produce Interleukin IL-17 are pathogenic in many human diseases, including inflammatory bowel disease, but are, paradoxically, essential for maintaining the integrity of the intestinal barrier in a non-inflammatory state. However, the intracellular mechanisms that regulate distinct transcriptional profiles and functional diversity of Th17 cells remain unclear. Here we show Raftlin1, a lipid raft protein, specifically upregulates and forms a complex with RORγt in pathogenic Th17 cells. Disruption of the RORγt-Raftlin1 complex results in the reduction of pathogenic Th17 cells in response to Citrobacter rodentium; however, there is no effect on nonpathogenic Th17 cells in response to commensal segmented filamentous bacteria. Mechanistically, we show that Raftlin1 recruits distinct phospholipids to RORγt and promotes the pathogenicity of Th17 cells. Thus, we have identified a mechanism that drives the pathogenic function of Th17 cells, which could provide a platform for advanced therapeutic strategies to dampen Th17-mediated inflammatory diseases.


Subject(s)
Nuclear Receptor Subfamily 1, Group F, Member 3 , Th17 Cells , Humans , Nuclear Receptor Subfamily 1, Group F, Member 3/genetics , Virulence , Inflammation , Colon
2.
Mol Cell Proteomics ; 19(9): 1436-1449, 2020 09.
Article in English | MEDLINE | ID: mdl-32541000

ABSTRACT

Kir2.1, a strong inward rectifier potassium channel encoded by the KCNJ2 gene, is a key regulator of the resting membrane potential of the cardiomyocyte and plays an important role in controlling ventricular excitation and action potential duration in the human heart. Mutations in KCNJ2 result in inheritable cardiac diseases in humans, e.g. the type-1 Andersen-Tawil syndrome (ATS1). Understanding the molecular mechanisms that govern the regulation of inward rectifier potassium currents by Kir2.1 in both normal and disease contexts should help uncover novel targets for therapeutic intervention in ATS1 and other Kir2.1-associated channelopathies. The information available to date on protein-protein interactions involving Kir2.1 channels remains limited. Additional efforts are necessary to provide a comprehensive map of the Kir2.1 interactome. Here we describe the generation of a comprehensive map of the Kir2.1 interactome using the proximity-labeling approach BioID. Most of the 218 high-confidence Kir2.1 channel interactions we identified are novel and encompass various molecular mechanisms of Kir2.1 function, ranging from intracellular trafficking to cross-talk with the insulin-like growth factor receptor signaling pathway, as well as lysosomal degradation. Our map also explores the variations in the interactome profiles of Kir2.1WTversus Kir2.1Δ314-315, a trafficking deficient ATS1 mutant, thus uncovering molecular mechanisms whose malfunctions may underlie ATS1 disease. Finally, using patch-clamp analysis, we validate the functional relevance of PKP4, one of our top BioID interactors, to the modulation of Kir2.1-controlled inward rectifier potassium currents. Our results validate the power of our BioID approach in identifying functionally relevant Kir2.1 interactors and underline the value of our Kir2.1 interactome as a repository for numerous novel biological hypotheses on Kir2.1 and Kir2.1-associated diseases.


Subject(s)
Andersen Syndrome/metabolism , Myocytes, Cardiac/metabolism , Plakophilins/metabolism , Potassium Channels, Inwardly Rectifying/metabolism , Potassium/metabolism , Protein Interaction Maps , Action Potentials/drug effects , Action Potentials/physiology , Andersen Syndrome/genetics , Andersen Syndrome/physiopathology , Chromatography, Liquid , Desmosomes/drug effects , Desmosomes/metabolism , HEK293 Cells , Humans , Lysosomes/metabolism , Molecular Chaperones/metabolism , Mutation , Myocytes, Cardiac/drug effects , Patch-Clamp Techniques , Potassium Channels, Inwardly Rectifying/genetics , Protein Interaction Maps/genetics , Protein Interaction Maps/physiology , Protein Transport/genetics , Protein Transport/physiology , Signal Transduction/genetics , Signal Transduction/physiology , Somatomedins/metabolism , Tandem Mass Spectrometry , Utrophin/metabolism
3.
Nat Commun ; 9(1): 4515, 2018 10 30.
Article in English | MEDLINE | ID: mdl-30375383

ABSTRACT

Dysregulated ROR-γt-mediated IL-17 transcription is central to the pathogenesis of several inflammatory disorders, yet the molecular mechanisms that govern the transcription factor activity of ROR-γt in the regulation of IL-17 are not fully defined. Here we show that SUMO-conjugating enzyme Ubc9 interacts with a conserved GKAE motif in ROR-γt to induce SUMOylation of ROR-γt and suppress IL-17 expression. Th17 cells expressing SUMOylation-defective ROR-γt are highly colitogenic upon transfer to Rag1-/- mice. Mechanistically, SUMOylation of ROR-γt facilitates the binding of HDAC2 to the IL-17 promoter and represses IL-17 transcription. Mice with conditional deletion of HDAC2 in CD4+ T cells have elevated IL-17 expression and severe colitis. The identification of the Ubc9/ROR-γt/HDAC2 axis that governs IL-17 expression may open new venues for the development of therapeutic measures for inflammatory disorders.


Subject(s)
Histone Deacetylase 2/metabolism , Interleukin-17/metabolism , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , Sumoylation/immunology , Th17 Cells/immunology , Ubiquitin-Conjugating Enzymes/metabolism , Animals , Colitis/genetics , Colitis/immunology , Colitis/metabolism , Histone Deacetylase 2/immunology , Homeodomain Proteins/genetics , Inflammation , Interleukin-17/immunology , Mice , Mice, Knockout , Nuclear Receptor Subfamily 1, Group F, Member 3/immunology
4.
EMBO J ; 36(21): 3232-3249, 2017 11 02.
Article in English | MEDLINE | ID: mdl-29030483

ABSTRACT

Notch signaling is an evolutionarily conserved signal transduction pathway that is essential for metazoan development. Upon ligand binding, the Notch intracellular domain (NOTCH ICD) translocates into the nucleus and forms a complex with the transcription factor RBPJ (also known as CBF1 or CSL) to activate expression of Notch target genes. In the absence of a Notch signal, RBPJ acts as a transcriptional repressor. Using a proteomic approach, we identified L3MBTL3 (also known as MBT1) as a novel RBPJ interactor. L3MBTL3 competes with NOTCH ICD for binding to RBPJ In the absence of NOTCH ICD, RBPJ recruits L3MBTL3 and the histone demethylase KDM1A (also known as LSD1) to the enhancers of Notch target genes, leading to H3K4me2 demethylation and to transcriptional repression. Importantly, in vivo analyses of the homologs of RBPJ and L3MBTL3 in Drosophila melanogaster and Caenorhabditis elegans demonstrate that the functional link between RBPJ and L3MBTL3 is evolutionarily conserved, thus identifying L3MBTL3 as a universal modulator of Notch signaling in metazoans.


Subject(s)
DNA-Binding Proteins/genetics , Drosophila Proteins/genetics , Histone Demethylases/genetics , Immunoglobulin J Recombination Signal Sequence-Binding Protein/genetics , Neuroglia/metabolism , Receptors, Notch/genetics , Animals , Biological Evolution , Caenorhabditis elegans/genetics , Caenorhabditis elegans/metabolism , Cell Line, Tumor , Conserved Sequence , DNA-Binding Proteins/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Gene Expression Regulation , Histone Demethylases/metabolism , Histones/genetics , Histones/metabolism , Humans , Immunoglobulin J Recombination Signal Sequence-Binding Protein/metabolism , Neuroglia/cytology , Protein Binding , Protein Domains , Receptors, Notch/metabolism , Transcription, Genetic , Two-Hybrid System Techniques
5.
Proc Natl Acad Sci U S A ; 114(25): 6581-6586, 2017 06 20.
Article in English | MEDLINE | ID: mdl-28607076

ABSTRACT

Identification of biomarkers and therapeutic targets is a critical goal of precision medicine. N-glycoproteins are a particularly attractive class of proteins that constitute potential cancer biomarkers and therapeutic targets for small molecules, antibodies, and cellular therapies. Using mass spectrometry (MS), we generated a compendium of 1,091 N-glycoproteins (from 40 human primary lymphomas and cell lines). Hierarchical clustering revealed distinct subtype signatures that included several subtype-specific biomarkers. Orthogonal immunological studies in 671 primary lymphoma tissue biopsies and 32 lymphoma-derived cell lines corroborated MS data. In anaplastic lymphoma kinase-positive (ALK+) anaplastic large cell lymphoma (ALCL), integration of N-glycoproteomics and transcriptome sequencing revealed an ALK-regulated cytokine/receptor signaling network, including vulnerabilities corroborated by a genome-wide clustered regularly interspaced short palindromic screen. Functional targeting of IL-31 receptor ß, an ALCL-enriched and ALK-regulated N-glycoprotein in this network, abrogated ALK+ALCL growth in vitro and in vivo. Our results highlight the utility of functional proteogenomic approaches for discovery of cancer biomarkers and therapeutic targets.


Subject(s)
Biomarkers, Tumor/genetics , Lymphoma/genetics , Transcriptome/genetics , Cell Line, Tumor , Humans , Proteogenomics/methods , Receptor Protein-Tyrosine Kinases/genetics , Signal Transduction/genetics
6.
Mol Cell Proteomics ; 12(10): 2714-23, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23836920

ABSTRACT

Chromosomal translocations encoding chimeric fusion proteins constitute one of the most common mechanisms underlying oncogenic transformation in human cancer. Fusion peptides resulting from such oncogenic chimeric fusions, though unique to specific cancer subtypes, are unexplored as cancer biomarkers. Here we show, using an approach termed fusion peptide multiple reaction monitoring mass spectrometry, the direct identification of different cancer-specific fusion peptides arising from protein chimeras that are generated from the juxtaposition of heterologous genes fused by recurrent chromosomal translocations. Using fusion peptide multiple reaction monitoring mass spectrometry in a clinically relevant scenario, we demonstrate the specific, sensitive, and unambiguous detection of a specific diagnostic fusion peptide in clinical samples of anaplastic large cell lymphoma, but not in a diverse array of benign lymph nodes or other forms of primary malignant lymphomas and cancer-derived cell lines. Our studies highlight the utility of fusion peptides as cancer biomarkers and carry broad implications for the use of protein biomarkers in cancer detection and monitoring.


Subject(s)
Biomarkers, Tumor/metabolism , Lymphoma, Large-Cell, Anaplastic/metabolism , Oncogene Proteins, Fusion/metabolism , Peptides/metabolism , Protein-Tyrosine Kinases/metabolism , Biomarkers, Tumor/genetics , Cell Line, Tumor , Humans , Lymphoma, Large-Cell, Anaplastic/genetics , Mass Spectrometry/methods , Oncogene Proteins, Fusion/genetics , Peptides/genetics , Protein-Tyrosine Kinases/genetics
7.
Blood ; 122(6): 958-68, 2013 Aug 08.
Article in English | MEDLINE | ID: mdl-23814019

ABSTRACT

The mechanisms underlying the pathogenesis of the constitutively active tyrosine kinase nucleophosmin-anaplastic lymphoma kinase (NPM-ALK) expressing anaplastic large cell lymphoma are not completely understood. Here we show using an integrated phosphoproteomic and metabolomic strategy that NPM-ALK induces a metabolic shift toward aerobic glycolysis, increased lactate production, and biomass production. The metabolic shift is mediated through the anaplastic lymphoma kinase (ALK) phosphorylation of the tumor-specific isoform of pyruvate kinase (PKM2) at Y105, resulting in decreased enzymatic activity. Small molecule activation of PKM2 or expression of Y105F PKM2 mutant leads to reversal of the metabolic switch with increased oxidative phosphorylation and reduced lactate production coincident with increased cell death, decreased colony formation, and reduced tumor growth in an in vivo xenograft model. This study provides comprehensive profiling of the phosphoproteomic and metabolomic consequences of NPM-ALK expression and reveals a novel role of ALK in the regulation of multiple components of cellular metabolism. Our studies show that PKM2 is a novel substrate of ALK and plays a critical role in mediating the metabolic shift toward biomass production and tumorigenesis.


Subject(s)
Carrier Proteins/metabolism , Gene Expression Regulation, Neoplastic , Lymphoma, Large-Cell, Anaplastic/metabolism , Membrane Proteins/metabolism , Protein-Tyrosine Kinases/metabolism , Thyroid Hormones/metabolism , Animals , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Proliferation , Humans , Metabolomics , Mice , Mice, SCID , Neoplasm Transplantation , Phosphorylation , Proteomics , Substrate Specificity , Thyroid Hormone-Binding Proteins
8.
J Biol Chem ; 285(50): 39303-13, 2010 Dec 10.
Article in English | MEDLINE | ID: mdl-20943656

ABSTRACT

Deubiquitinating enzymes (DUbs) play important roles in many ubiquitin-dependent pathways, yet how DUbs themselves are regulated is not well understood. Here, we provide insight into the mechanism by which ubiquitination directly enhances the activity of ataxin-3, a DUb implicated in protein quality control and the disease protein in the polyglutamine neurodegenerative disorder, Spinocerebellar Ataxia Type 3. We identify Lys-117, which resides near the catalytic triad, as the primary site of ubiquitination in wild type and pathogenic ataxin-3. Further studies indicate that ubiquitin-dependent activation of ataxin-3 at Lys-117 is important for its ability to reduce high molecular weight ubiquitinated species in cells. Ubiquitination at Lys-117 also facilitates the ability of ataxin-3 to induce aggresome formation in cells. Finally, structure-function studies support a model of activation whereby ubiquitination at Lys-117 enhances ataxin-3 activity independent of the known ubiquitin-binding sites in ataxin-3, most likely through a direct conformational change in or near the catalytic domain.


Subject(s)
Gene Expression Regulation , Lysine/chemistry , Machado-Joseph Disease/metabolism , Nerve Tissue Proteins/chemistry , Neurodegenerative Diseases/metabolism , Nuclear Proteins/chemistry , Repressor Proteins/chemistry , Animals , Ataxin-3 , Binding Sites , Catalysis , Fibroblasts/metabolism , Humans , Mice , Protein Conformation , Structure-Activity Relationship , Transfection , Ubiquitin/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...