Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 268
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Cell ; 184(13): 3519-3527.e10, 2021 06 24.
Article in English | MEDLINE | ID: mdl-34107286

ABSTRACT

Mutations in leucine-rich repeat kinase 2 (LRRK2) are commonly implicated in the pathogenesis of both familial and sporadic Parkinson's disease (PD). LRRK2 regulates critical cellular processes at membranous organelles and forms microtubule-based pathogenic filaments, yet the molecular basis underlying these biological roles of LRRK2 remains largely enigmatic. Here, we determined high-resolution structures of full-length human LRRK2, revealing its architecture and key interdomain scaffolding elements for rationalizing disease-causing mutations. The kinase domain of LRRK2 is captured in an inactive state, a conformation also adopted by the most common PD-associated mutation, LRRK2G2019S. This conformation serves as a framework for structure-guided design of conformational specific inhibitors. We further determined the structure of COR-mediated LRRK2 dimers and found that single-point mutations at the dimer interface abolished pathogenic filamentation in cells. Overall, our study provides mechanistic insights into physiological and pathological roles of LRRK2 and establishes a structural template for future therapeutic intervention in PD.


Subject(s)
Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/chemistry , Amino Acid Sequence , HEK293 Cells , Humans , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/ultrastructure , Models, Molecular , Protein Domains , Protein Multimerization , Protein Structure, Secondary
2.
Nat Immunol ; 24(10): 1735-1747, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37679549

ABSTRACT

Neurodegenerative diseases, including Alzheimer's disease (AD), are characterized by innate immune-mediated inflammation, but functional and mechanistic effects of the adaptive immune system remain unclear. Here we identify brain-resident CD8+ T cells that coexpress CXCR6 and PD-1 and are in proximity to plaque-associated microglia in human and mouse AD brains. We also establish that CD8+ T cells restrict AD pathologies, including ß-amyloid deposition and cognitive decline. Ligand-receptor interaction analysis identifies CXCL16-CXCR6 intercellular communication between microglia and CD8+ T cells. Further, Cxcr6 deficiency impairs accumulation, tissue residency programming and clonal expansion of brain PD-1+CD8+ T cells. Ablation of Cxcr6 or CD8+ T cells ultimately increases proinflammatory cytokine production from microglia, with CXCR6 orchestrating brain CD8+ T cell-microglia colocalization. Collectively, our study reveals protective roles for brain CD8+ T cells and CXCR6 in mouse AD pathogenesis and highlights that microenvironment-specific, intercellular communication orchestrates tissue homeostasis and protection from neuroinflammation.

3.
Nat Immunol ; 21(6): 660-670, 2020 06.
Article in English | MEDLINE | ID: mdl-32341509

ABSTRACT

Within germinal centers (GCs), complex and highly orchestrated molecular programs must balance proliferation, somatic hypermutation and selection to both provide effective humoral immunity and to protect against genomic instability and neoplastic transformation. In contrast to this complexity, GC B cells are canonically divided into two principal populations, dark zone (DZ) and light zone (LZ) cells. We now demonstrate that, following selection in the LZ, B cells migrated to specialized sites within the canonical DZ that contained tingible body macrophages and were sites of ongoing cell division. Proliferating DZ (DZp) cells then transited into the larger DZ to become differentiating DZ (DZd) cells before re-entering the LZ. Multidimensional analysis revealed distinct molecular programs in each population commensurate with observed compartmentalization of noncompatible functions. These data provide a new three-cell population model that both orders critical GC functions and reveals essential molecular programs of humoral adaptive immunity.


Subject(s)
Cellular Microenvironment/genetics , Cellular Microenvironment/immunology , Germinal Center/cytology , Germinal Center/physiology , Animals , Biomarkers , Computational Biology/methods , Fluorescent Antibody Technique , Gene Expression Profiling , Genomics/methods , Mice , Phosphorylation , Proteome , Proteomics/methods , Transcriptome
4.
Cell ; 167(5): 1252-1263.e10, 2016 11 17.
Article in English | MEDLINE | ID: mdl-27863243

ABSTRACT

Many animal tissues/cells are photosensitive, yet only two types of photoreceptors (i.e., opsins and cryptochromes) have been discovered in metazoans. The question arises as to whether unknown types of photoreceptors exist in the animal kingdom. LITE-1, a seven-transmembrane gustatory receptor (GR) homolog, mediates UV-light-induced avoidance behavior in C. elegans. However, it is not known whether LITE-1 functions as a chemoreceptor or photoreceptor. Here, we show that LITE-1 directly absorbs both UVA and UVB light with an extinction coefficient 10-100 times that of opsins and cryptochromes, indicating that LITE-1 is highly efficient in capturing photons. Unlike typical photoreceptors employing a prosthetic chromophore to capture photons, LITE-1 strictly depends on its protein conformation for photon absorption. We have further identified two tryptophan residues critical for LITE-1 function. Interestingly, unlike GPCRs, LITE-1 adopts a reversed membrane topology. Thus, LITE-1, a taste receptor homolog, represents a distinct type of photoreceptor in the animal kingdom.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/metabolism , Membrane Proteins/metabolism , Animals , Caenorhabditis elegans/radiation effects , Caenorhabditis elegans Proteins/chemistry , Caenorhabditis elegans Proteins/isolation & purification , Membrane Proteins/chemistry , Membrane Proteins/isolation & purification , Photons , Protein Conformation , Tryptophan/metabolism , Ultraviolet Rays
5.
Cell ; 167(3): 774-788.e17, 2016 Oct 20.
Article in English | MEDLINE | ID: mdl-27768896

ABSTRACT

Expansion of a hexanucleotide repeat GGGGCC (G4C2) in C9ORF72 is the most common cause of amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD). Transcripts carrying (G4C2) expansions undergo unconventional, non-ATG-dependent translation, generating toxic dipeptide repeat (DPR) proteins thought to contribute to disease. Here, we identify the interactome of all DPRs and find that arginine-containing DPRs, polyGly-Arg (GR) and polyPro-Arg (PR), interact with RNA-binding proteins and proteins with low complexity sequence domains (LCDs) that often mediate the assembly of membrane-less organelles. Indeed, most GR/PR interactors are components of membrane-less organelles such as nucleoli, the nuclear pore complex and stress granules. Genetic analysis in Drosophila demonstrated the functional relevance of these interactions to DPR toxicity. Furthermore, we show that GR and PR altered phase separation of LCD-containing proteins, insinuating into their liquid assemblies and changing their material properties, resulting in perturbed dynamics and/or functions of multiple membrane-less organelles.


Subject(s)
Amyotrophic Lateral Sclerosis/metabolism , Dipeptides/metabolism , Frontotemporal Dementia/metabolism , Proteins/metabolism , RNA-Binding Proteins/metabolism , Amyotrophic Lateral Sclerosis/genetics , Animals , C9orf72 Protein , Cell Nucleolus/metabolism , Cytoplasmic Granules/metabolism , DNA Repeat Expansion , Dipeptides/genetics , Drosophila melanogaster/genetics , Frontotemporal Dementia/genetics , Humans , Intracellular Membranes/metabolism , Nuclear Pore/metabolism , Peptides/genetics , Peptides/metabolism , Proteins/genetics
6.
Nature ; 628(8007): 442-449, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38538798

ABSTRACT

Whereas oncogenes can potentially be inhibited with small molecules, the loss of tumour suppressors is more common and is problematic because the tumour-suppressor proteins are no longer present to be targeted. Notable examples include SMARCB1-mutant cancers, which are highly lethal malignancies driven by the inactivation of a subunit of SWI/SNF (also known as BAF) chromatin-remodelling complexes. Here, to generate mechanistic insights into the consequences of SMARCB1 mutation and to identify vulnerabilities, we contributed 14 SMARCB1-mutant cell lines to a near genome-wide CRISPR screen as part of the Cancer Dependency Map Project1-3. We report that the little-studied gene DDB1-CUL4-associated factor 5 (DCAF5) is required for the survival of SMARCB1-mutant cancers. We show that DCAF5 has a quality-control function for SWI/SNF complexes and promotes the degradation of incompletely assembled SWI/SNF complexes in the absence of SMARCB1. After depletion of DCAF5, SMARCB1-deficient SWI/SNF complexes reaccumulate, bind to target loci and restore SWI/SNF-mediated gene expression to levels that are sufficient to reverse the cancer state, including in vivo. Consequently, cancer results not from the loss of SMARCB1 function per se, but rather from DCAF5-mediated degradation of SWI/SNF complexes. These data indicate that therapeutic targeting of ubiquitin-mediated quality-control factors may effectively reverse the malignant state of some cancers driven by disruption of tumour suppressor complexes.


Subject(s)
Multiprotein Complexes , Mutation , Neoplasms , SMARCB1 Protein , Animals , Female , Humans , Male , Mice , Cell Line, Tumor , CRISPR-Cas Systems , Gene Editing , Neoplasms/genetics , Neoplasms/metabolism , SMARCB1 Protein/deficiency , SMARCB1 Protein/genetics , SMARCB1 Protein/metabolism , Tumor Suppressor Proteins/deficiency , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , Multiprotein Complexes/chemistry , Multiprotein Complexes/metabolism , Proteolysis , Ubiquitin/metabolism
7.
Immunity ; 51(6): 1012-1027.e7, 2019 12 17.
Article in English | MEDLINE | ID: mdl-31668641

ABSTRACT

Regulatory T (Treg) cells are critical mediators of immune tolerance whose activity depends upon T cell receptor (TCR) and mTORC1 kinase signaling, but the mechanisms that dictate functional activation of these pathways are incompletely understood. Here, we showed that amino acids license Treg cell function by priming and sustaining TCR-induced mTORC1 activity. mTORC1 activation was induced by amino acids, especially arginine and leucine, accompanied by the dynamic lysosomal localization of the mTOR and Tsc complexes. Rag and Rheb GTPases were central regulators of amino acid-dependent mTORC1 activation in effector Treg (eTreg) cells. Mice bearing RagA-RagB- or Rheb1-Rheb2-deficient Treg cells developed a fatal autoimmune disease and had reduced eTreg cell accumulation and function. RagA-RagB regulated mitochondrial and lysosomal fitness, while Rheb1-Rheb2 enforced eTreg cell suppressive gene signature. Together, these findings reveal a crucial requirement of amino acid signaling for licensing and sustaining mTORC1 activation and functional programming of Treg cells.


Subject(s)
Arginine/metabolism , Leucine/metabolism , Mechanistic Target of Rapamycin Complex 1/metabolism , Monomeric GTP-Binding Proteins/metabolism , Ras Homolog Enriched in Brain Protein/metabolism , T-Lymphocytes, Regulatory/immunology , Animals , Cell Cycle , Cell Differentiation/physiology , Cell Line , Humans , Immune Tolerance/immunology , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Monomeric GTP-Binding Proteins/genetics , Ras Homolog Enriched in Brain Protein/genetics , Receptors, Antigen, T-Cell/immunology , T-Lymphocytes, Regulatory/cytology
8.
Cell ; 152(1-2): 196-209, 2013 Jan 17.
Article in English | MEDLINE | ID: mdl-23332755

ABSTRACT

In eukaryotic cells a molecular chaperone network associates with translating ribosomes, assisting the maturation of emerging nascent polypeptides. Hsp70 is perhaps the major eukaryotic ribosome-associated chaperone and the first reported to bind cotranslationally to nascent chains. However, little is known about the underlying principles and function of this interaction. Here, we use a sensitive and global approach to define the cotranslational substrate specificity of the yeast Hsp70 SSB. We find that SSB binds to a subset of nascent polypeptides whose intrinsic properties and slow translation rates hinder efficient cotranslational folding. The SSB-ribosome cycle and substrate recognition is modulated by its ribosome-bound cochaperone, RAC. Deletion of SSB leads to widespread aggregation of newly synthesized polypeptides. Thus, cotranslationally acting Hsp70 meets the challenge of folding the eukaryotic proteome by stabilizing its longer, more slowly translated, and aggregation-prone nascent polypeptides.


Subject(s)
Adenosine Triphosphatases/metabolism , HSP70 Heat-Shock Proteins/metabolism , Protein Biosynthesis , Protein Folding , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Ribosomes/metabolism
9.
Mol Cell ; 77(6): 1206-1221.e7, 2020 03 19.
Article in English | MEDLINE | ID: mdl-31980388

ABSTRACT

Alternative polyadenylation (APA) contributes to transcriptome complexity by generating mRNA isoforms with varying 3' UTR lengths. APA leading to 3' UTR shortening (3' US) is a common feature of most cancer cells; however, the molecular mechanisms are not understood. Here, we describe a widespread mechanism promoting 3' US in cancer through ubiquitination of the mRNA 3' end processing complex protein, PCF11, by the cancer-specific MAGE-A11-HUWE1 ubiquitin ligase. MAGE-A11 is normally expressed only in the male germline but is frequently re-activated in cancers. MAGE-A11 is necessary for cancer cell viability and is sufficient to drive tumorigenesis. Screening for targets of MAGE-A11 revealed that it ubiquitinates PCF11, resulting in loss of CFIm25 from the mRNA 3' end processing complex. This leads to APA of many transcripts affecting core oncogenic and tumor suppressors, including cyclin D2 and PTEN. These findings provide insights into the molecular mechanisms driving APA in cancer and suggest therapeutic strategies.


Subject(s)
3' Untranslated Regions/genetics , Antigens, Neoplasm/metabolism , Lung Neoplasms/pathology , Neoplasm Proteins/metabolism , Ovarian Neoplasms/pathology , RNA, Messenger/metabolism , Ubiquitin/metabolism , mRNA Cleavage and Polyadenylation Factors/metabolism , Animals , Antigens, Neoplasm/genetics , Apoptosis , Biomarkers, Tumor , Carcinogenesis , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Cell Proliferation , Cleavage And Polyadenylation Specificity Factor/genetics , Cleavage And Polyadenylation Specificity Factor/metabolism , Female , Gene Expression Regulation, Neoplastic , Humans , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Male , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasm Proteins/genetics , Ovarian Neoplasms/genetics , Ovarian Neoplasms/metabolism , Polyadenylation , RNA Splicing , RNA, Messenger/genetics , Tumor Cells, Cultured , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Ubiquitination , Xenograft Model Antitumor Assays , mRNA Cleavage and Polyadenylation Factors/genetics
10.
Immunity ; 49(5): 899-914.e6, 2018 11 20.
Article in English | MEDLINE | ID: mdl-30413360

ABSTRACT

Interleukin-2 (IL-2) and downstream transcription factor STAT5 are important for maintaining regulatory T (Treg) cell homeostasis and function. Treg cells can respond to low IL-2 levels, but the mechanisms of STAT5 activation during partial IL-2 deficiency remain uncertain. We identified the serine-threonine kinase Mst1 as a signal-dependent amplifier of IL-2-STAT5 activity in Treg cells. High Mst1 and Mst2 (Mst1-Mst2) activity in Treg cells was crucial to prevent tumor resistance and autoimmunity. Mechanistically, Mst1-Mst2 sensed IL-2 signals to promote the STAT5 activation necessary for Treg cell homeostasis and lineage stability and to maintain the highly suppressive phosphorylated-STAT5+ Treg cell subpopulation. Unbiased quantitative proteomics revealed association of Mst1 with the cytoskeletal DOCK8-LRCHs module. Mst1 deficiency limited Treg cell migration and access to IL-2 and activity of the small GTPase Rac, which mediated downstream STAT5 activation. Collectively, IL-2-STAT5 signaling depends upon Mst1-Mst2 functions to maintain a stable Treg cell pool and immune tolerance.


Subject(s)
Hepatocyte Growth Factor/metabolism , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Receptors, Interleukin-2/metabolism , STAT5 Transcription Factor/metabolism , Signal Transduction , T-Lymphocytes, Regulatory/metabolism , Animals , Autoimmunity/genetics , Autoimmunity/immunology , Cell Lineage/genetics , Hepatocyte Growth Factor/genetics , Hippo Signaling Pathway , Interleukin-2/metabolism , Mice , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins/genetics , Serine-Threonine Kinase 3 , T-Lymphocytes, Regulatory/immunology , rac GTP-Binding Proteins/metabolism
12.
Nature ; 600(7888): 308-313, 2021 12.
Article in English | MEDLINE | ID: mdl-34795452

ABSTRACT

Nutrients are emerging regulators of adaptive immunity1. Selective nutrients interplay with immunological signals to activate mechanistic target of rapamycin complex 1 (mTORC1), a key driver of cell metabolism2-4, but how these environmental signals are integrated for immune regulation remains unclear. Here we use genome-wide CRISPR screening combined with protein-protein interaction networks to identify regulatory modules that mediate immune receptor- and nutrient-dependent signalling to mTORC1 in mouse regulatory T (Treg) cells. SEC31A is identified to promote mTORC1 activation by interacting with the GATOR2 component SEC13 to protect it from SKP1-dependent proteasomal degradation. Accordingly, loss of SEC31A impairs T cell priming and Treg suppressive function in mice. In addition, the SWI/SNF complex restricts expression of the amino acid sensor CASTOR1, thereby enhancing mTORC1 activation. Moreover, we reveal that the CCDC101-associated SAGA complex is a potent inhibitor of mTORC1, which limits the expression of glucose and amino acid transporters and maintains T cell quiescence in vivo. Specific deletion of Ccdc101 in mouse Treg cells results in uncontrolled inflammation but improved antitumour immunity. Collectively, our results establish epigenetic and post-translational mechanisms that underpin how nutrient transporters, sensors and transducers interplay with immune signals for three-tiered regulation of mTORC1 activity and identify their pivotal roles in licensing T cell immunity and immune tolerance.


Subject(s)
CRISPR-Cas Systems , Gene Editing , Nutrients , Protein Interaction Maps , T-Lymphocytes, Regulatory , Animals , Female , Male , Mice , Carrier Proteins/metabolism , CRISPR-Cas Systems/genetics , Forkhead Transcription Factors/metabolism , Genome/genetics , Homeostasis , Immune Tolerance , Inflammation/pathology , Mechanistic Target of Rapamycin Complex 1/metabolism , Neoplasms/immunology , Nuclear Proteins/metabolism , Nutrients/metabolism , Proteasome Endopeptidase Complex/metabolism , Proteolysis , S-Phase Kinase-Associated Proteins/metabolism , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Trans-Activators/metabolism
13.
Mol Cell ; 74(4): 742-757.e8, 2019 05 16.
Article in English | MEDLINE | ID: mdl-30979586

ABSTRACT

Disturbances in autophagy and stress granule dynamics have been implicated as potential mechanisms underlying inclusion body myopathy (IBM) and related disorders. Yet the roles of core autophagy proteins in IBM and stress granule dynamics remain poorly characterized. Here, we demonstrate that disrupted expression of the core autophagy proteins ULK1 and ULK2 in mice causes a vacuolar myopathy with ubiquitin and TDP-43-positive inclusions; this myopathy is similar to that caused by VCP/p97 mutations, the most common cause of familial IBM. Mechanistically, we show that ULK1/2 localize to stress granules and phosphorylate VCP, thereby increasing VCP's activity and ability to disassemble stress granules. These data suggest that VCP dysregulation and defective stress granule disassembly contribute to IBM-like disease in Ulk1/2-deficient mice. In addition, stress granule disassembly is accelerated by an ULK1/2 agonist, suggesting ULK1/2 as targets for exploiting the higher-order regulation of stress granules for therapeutic intervention of IBM and related disorders.


Subject(s)
Autophagy-Related Protein-1 Homolog/genetics , Lysosomal Storage Diseases/genetics , Muscular Diseases/genetics , Protein Serine-Threonine Kinases/genetics , Valosin Containing Protein/genetics , Adenosine Triphosphatases/genetics , Animals , Autophagy/genetics , DNA-Binding Proteins/genetics , Disease Models, Animal , Humans , Inclusion Bodies/genetics , Inclusion Bodies/pathology , Lysosomal Storage Diseases/metabolism , Lysosomal Storage Diseases/pathology , Mice , Muscular Diseases/metabolism , Muscular Diseases/pathology , Phosphorylation/genetics , Stress, Physiological/genetics , Ubiquitin/genetics
14.
Immunity ; 46(3): 488-503, 2017 03 21.
Article in English | MEDLINE | ID: mdl-28285833

ABSTRACT

The molecular circuits by which antigens activate quiescent T cells remain poorly understood. We combined temporal profiling of the whole proteome and phosphoproteome via multiplexed isobaric labeling proteomics technology, computational pipelines for integrating multi-omics datasets, and functional perturbation to systemically reconstruct regulatory networks underlying T cell activation. T cell receptors activated the T cell proteome and phosphoproteome with discrete kinetics, marked by early dynamics of phosphorylation and delayed ribosome biogenesis and mitochondrial activation. Systems biology analyses identified multiple functional modules, active kinases, transcription factors and connectivity between them, and mitochondrial pathways including mitoribosomes and complex IV. Genetic perturbation revealed physiological roles for mitochondrial enzyme COX10-mediated oxidative phosphorylation in T cell quiescence exit. Our multi-layer proteomics profiling, integrative network analysis, and functional studies define landscapes of the T cell proteome and phosphoproteome and reveal signaling and bioenergetics pathways that mediate lymphocyte exit from quiescence.


Subject(s)
Lymphocyte Activation/immunology , Signal Transduction/immunology , T-Lymphocytes/immunology , Alkyl and Aryl Transferases/immunology , Animals , Energy Metabolism , Mass Spectrometry , Membrane Proteins/immunology , Mice , Mice, Inbred C57BL , Phosphorylation , Proteomics , Receptors, Antigen, T-Cell/immunology
15.
Nature ; 581(7807): 199-203, 2020 05.
Article in English | MEDLINE | ID: mdl-32404997

ABSTRACT

Recognition of microbe-associated molecular patterns (MAMPs) by pattern recognition receptors (PRRs) triggers the first line of inducible defence against invading pathogens1-3. Receptor-like cytoplasmic kinases (RLCKs) are convergent regulators that associate with multiple PRRs in plants4. The mechanisms that underlie the activation of RLCKs are unclear. Here we show that when MAMPs are detected, the RLCK BOTRYTIS-INDUCED KINASE 1 (BIK1) is monoubiquitinated following phosphorylation, then released from the flagellin receptor FLAGELLIN SENSING 2 (FLS2)-BRASSINOSTEROID INSENSITIVE 1-ASSOCIATED KINASE 1 (BAK1) complex, and internalized dynamically into endocytic compartments. The Arabidopsis E3 ubiquitin ligases RING-H2 FINGER A3A (RHA3A) and RHA3B mediate the monoubiquitination of BIK1, which is essential for the subsequent release of BIK1 from the FLS2-BAK1 complex and activation of immune signalling. Ligand-induced monoubiquitination and endosomal puncta of BIK1 exhibit spatial and temporal dynamics that are distinct from those of the PRR FLS2. Our study reveals the intertwined regulation of PRR-RLCK complex activation by protein phosphorylation and ubiquitination, and shows that ligand-induced monoubiquitination contributes to the release of BIK1 family RLCKs from the PRR complex and activation of PRR signalling.


Subject(s)
Arabidopsis Proteins/chemistry , Arabidopsis Proteins/metabolism , Arabidopsis/immunology , Arabidopsis/metabolism , Plant Immunity/immunology , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/metabolism , Receptors, Pattern Recognition/immunology , Ubiquitin-Protein Ligases/chemistry , Ubiquitin-Protein Ligases/metabolism , Ubiquitination , Arabidopsis/enzymology , Endocytosis , Ligands , Pathogen-Associated Molecular Pattern Molecules/immunology , Phosphorylation , Protein Kinases/metabolism
16.
Nature ; 587(7835): 650-656, 2020 11.
Article in English | MEDLINE | ID: mdl-33149304

ABSTRACT

G-protein-coupled receptors (GPCRs) are membrane proteins that modulate physiology across human tissues in response to extracellular signals. GPCR-mediated signalling can differ because of changes in the sequence1,2 or expression3 of the receptors, leading to signalling bias when comparing diverse physiological systems4. An underexplored source of such bias is the generation of functionally diverse GPCR isoforms with different patterns of expression across different tissues. Here we integrate data from human tissue-level transcriptomes, GPCR sequences and structures, proteomics, single-cell transcriptomics, population-wide genetic association studies and pharmacological experiments. We show how a single GPCR gene can diversify into several isoforms with distinct signalling properties, and how unique isoform combinations expressed in different tissues can generate distinct signalling states. Depending on their structural changes and expression patterns, some of the detected isoforms may influence cellular responses to drugs and represent new targets for developing drugs with improved tissue selectivity. Our findings highlight the need to move from a canonical to a context-specific view of GPCR signalling that considers how combinatorial expression of isoforms in a particular cell type, tissue or organism collectively influences receptor signalling and drug responses.


Subject(s)
Protein Isoforms/chemistry , Protein Isoforms/metabolism , Receptors, G-Protein-Coupled/chemistry , Receptors, G-Protein-Coupled/metabolism , Signal Transduction/drug effects , Transcriptome , Databases, Factual , Gene Expression Profiling , HEK293 Cells , Humans , Molecular Targeted Therapy , Organ Specificity/drug effects , Protein Isoforms/genetics , Proteomics , Receptors, G-Protein-Coupled/antagonists & inhibitors , Receptors, G-Protein-Coupled/genetics , Signal Transduction/genetics , Single-Cell Analysis
17.
Blood ; 142(7): 629-642, 2023 08 17.
Article in English | MEDLINE | ID: mdl-37172201

ABSTRACT

Advancing cure rates for high-risk acute lymphoblastic leukemia (ALL) has been limited by the lack of agents that effectively kill leukemic cells, sparing normal hematopoietic tissue. Molecular glues direct the ubiquitin ligase cellular machinery to target neosubstrates for protein degradation. We developed a novel cereblon modulator, SJ6986, that exhibits potent and selective degradation of GSPT1 and GSPT2 and cytotoxic activity against childhood cancer cell lines. Here, we report in vitro and in vivo testing of the activity of this agent in a panel of ALL cell lines and xenografts. SJ6986 exhibited similar cytotoxicity to the previously described GSPT1 degrader CC-90009 in a panel of leukemia cell lines in vitro, resulting in apoptosis and perturbation of cell cycle progression. SJ6986 was more effective than CC-90009 in suppressing leukemic cell growth in vivo, partly attributable to favorable pharmacokinetic properties, and did not significantly impair differentiation of human CD34+ cells ex vivo. Genome-wide CRISPR/Cas9 screening of ALL cell lines treated with SJ6986 confirmed that components of the CRL4CRBN complex, associated adaptors, regulators, and effectors were integral in mediating the action of SJ6986. SJ6986 is a potent, selective, orally bioavailable GSPT1/2 degrader that shows broad antileukemic activity and has potential for clinical development.


Subject(s)
Antineoplastic Agents , Piperidones , Precursor Cell Lymphoblastic Leukemia-Lymphoma , Humans , Child , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Antineoplastic Agents/chemistry , Piperidones/therapeutic use , Isoindoles/therapeutic use
18.
Mol Psychiatry ; 2024 May 09.
Article in English | MEDLINE | ID: mdl-38724566

ABSTRACT

Psychiatric disorders are highly heritable yet polygenic, potentially involving hundreds of risk genes. Genome-wide association studies have identified hundreds of genomic susceptibility loci with susceptibility to psychiatric disorders; however, the contribution of these loci to the underlying psychopathology and etiology remains elusive. Here we generated deep human brain proteomics data by quantifying 11,608 proteins across 268 subjects using 11-plex tandem mass tag coupled with two-dimensional liquid chromatography-tandem mass spectrometry. Our analysis revealed 788 cis-acting protein quantitative trait loci associated with the expression of 883 proteins at a genome-wide false discovery rate <5%. In contrast to expression at the transcript level and complex diseases that are found to be mainly influenced by noncoding variants, we found protein expression level tends to be regulated by non-synonymous variants. We also provided evidence of 76 shared regulatory signals between gene expression and protein abundance. Mediation analysis revealed that for most (88%) of the colocalized genes, the expression levels of their corresponding proteins are regulated by cis-pQTLs via gene transcription. Using summary data-based Mendelian randomization analysis, we identified 4 proteins and 19 genes that are causally associated with schizophrenia. We further integrated multiple omics data with network analysis to prioritize candidate genes for schizophrenia risk loci. Collectively, our findings underscore the potential of proteome-wide linkage analysis in gaining mechanistic insights into the pathogenesis of psychiatric disorders.

19.
Cell ; 140(4): 567-78, 2010 Feb 19.
Article in English | MEDLINE | ID: mdl-20178748

ABSTRACT

The molecular mechanisms regulating the ubiquitin proteasome system (UPS) at synapses are poorly understood. We report that CaMKIIalpha-an abundant postsynaptic protein kinase-mediates the activity-dependent recruitment of proteasomes to dendritic spines in hippocampal neurons. CaMKIIalpha is biochemically associated with proteasomes in the brain. CaMKIIalpha translocation to synapses is required for activity-induced proteasome accumulation in spines, and is sufficient to redistribute proteasomes to postsynaptic sites. CaMKIIalpha autophosphorylation enhances its binding to proteasomes and promotes proteasome recruitment to spines. In addition to this structural role, CaMKIIalpha stimulates proteasome activity by phosphorylating proteasome subunit Rpt6 on Serine 120. However, CaMKIIalpha translocation, but not its kinase activity, is required for activity-dependent degradation of polyubiquitinated proteins in spines. Our findings reveal a scaffolding role of postsynaptic CaMKIIalpha in activity-dependent proteasome redistribution, which is commensurate with the great abundance of CaMKIIalpha in synapses.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Dendritic Spines/metabolism , Proteasome Endopeptidase Complex/metabolism , Animals , Brain/cytology , Hippocampus/cytology , Neurons/cytology , Phosphorylation , Protein Transport , Rats , Synapses
SELECTION OF CITATIONS
SEARCH DETAIL