Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 5.986
Filter
Add more filters

Publication year range
1.
Cell ; 186(16): 3499-3518.e14, 2023 08 03.
Article in English | MEDLINE | ID: mdl-37437571

ABSTRACT

Chloroplasts are eukaryotic photosynthetic organelles that drive the global carbon cycle. Despite their importance, our understanding of their protein composition, function, and spatial organization remains limited. Here, we determined the localizations of 1,034 candidate chloroplast proteins using fluorescent protein tagging in the model alga Chlamydomonas reinhardtii. The localizations provide insights into the functions of poorly characterized proteins; identify novel components of nucleoids, plastoglobules, and the pyrenoid; and reveal widespread protein targeting to multiple compartments. We discovered and further characterized cellular organizational features, including eleven chloroplast punctate structures, cytosolic crescent structures, and unexpected spatial distributions of enzymes within the chloroplast. We also used machine learning to predict the localizations of other nuclear-encoded Chlamydomonas proteins. The strains and localization atlas developed here will serve as a resource to accelerate studies of chloroplast architecture and functions.


Subject(s)
Biosynthetic Pathways , Chlamydomonas reinhardtii , Chloroplast Proteins , Chlamydomonas reinhardtii/metabolism , Chloroplast Proteins/metabolism , Chloroplasts/metabolism , Photosynthesis
2.
Cell ; 185(11): 1974-1985.e12, 2022 05 26.
Article in English | MEDLINE | ID: mdl-35512704

ABSTRACT

Comprehensive sequencing of patient tumors reveals genomic mutations across tumor types that enable tumorigenesis and progression. A subset of oncogenic driver mutations results in neomorphic activity where the mutant protein mediates functions not engaged by the parental molecule. Here, we identify prevalent variant-enabled neomorph-protein-protein interactions (neoPPI) with a quantitative high-throughput differential screening (qHT-dS) platform. The coupling of highly sensitive BRET biosensors with miniaturized coexpression in an ultra-HTS format allows large-scale monitoring of the interactions of wild-type and mutant variant counterparts with a library of cancer-associated proteins in live cells. The screening of 17,792 interactions with 2,172,864 data points revealed a landscape of gain of interactions encompassing both oncogenic and tumor suppressor mutations. For example, the recurrent BRAF V600E lesion mediates KEAP1 neoPPI, rewiring a BRAFV600E/KEAP1 signaling axis and creating collateral vulnerability to NQO1 substrates, offering a combination therapeutic strategy. Thus, cancer genomic alterations can create neo-interactions, informing variant-directed therapeutic approaches for precision medicine.


Subject(s)
Neoplasms , Proto-Oncogene Proteins B-raf , Carcinogenesis , Humans , Kelch-Like ECH-Associated Protein 1/genetics , Kelch-Like ECH-Associated Protein 1/metabolism , Mutation , NF-E2-Related Factor 2/metabolism , Neoplasms/genetics , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins B-raf/metabolism
3.
Cell ; 185(4): 712-728.e14, 2022 02 17.
Article in English | MEDLINE | ID: mdl-35063084

ABSTRACT

Tau (MAPT) drives neuronal dysfunction in Alzheimer disease (AD) and other tauopathies. To dissect the underlying mechanisms, we combined an engineered ascorbic acid peroxidase (APEX) approach with quantitative affinity purification mass spectrometry (AP-MS) followed by proximity ligation assay (PLA) to characterize Tau interactomes modified by neuronal activity and mutations that cause frontotemporal dementia (FTD) in human induced pluripotent stem cell (iPSC)-derived neurons. We established interactions of Tau with presynaptic vesicle proteins during activity-dependent Tau secretion and mapped the Tau-binding sites to the cytosolic domains of integral synaptic vesicle proteins. We showed that FTD mutations impair bioenergetics and markedly diminished Tau's interaction with mitochondria proteins, which were downregulated in AD brains of multiple cohorts and correlated with disease severity. These multimodal and dynamic Tau interactomes with exquisite spatial resolution shed light on Tau's role in neuronal function and disease and highlight potential therapeutic targets to block Tau-mediated pathogenesis.


Subject(s)
Mitochondria/metabolism , Nerve Degeneration/metabolism , Protein Interaction Maps , Synapses/metabolism , tau Proteins/metabolism , Alzheimer Disease/genetics , Amino Acids/metabolism , Biotinylation , Brain/metabolism , Brain/pathology , Cell Nucleus/metabolism , Disease Progression , Energy Metabolism , Frontotemporal Dementia/genetics , Humans , Induced Pluripotent Stem Cells/metabolism , Mutant Proteins/metabolism , Mutation/genetics , Nerve Degeneration/pathology , Neurons/metabolism , Protein Binding , Protein Domains , Proteomics , Severity of Illness Index , Subcellular Fractions/metabolism , Tauopathies/genetics , tau Proteins/chemistry
4.
Cell ; 182(4): 1027-1043.e17, 2020 08 20.
Article in English | MEDLINE | ID: mdl-32822567

ABSTRACT

Cell-surface protein-protein interactions (PPIs) mediate cell-cell communication, recognition, and responses. We executed an interactome screen of 564 human cell-surface and secreted proteins, most of which are immunoglobulin superfamily (IgSF) proteins, using a high-throughput, automated ELISA-based screening platform employing a pooled-protein strategy to test all 318,096 PPI combinations. Screen results, augmented by phylogenetic homology analysis, revealed ∼380 previously unreported PPIs. We validated a subset using surface plasmon resonance and cell binding assays. Observed PPIs reveal a large and complex network of interactions both within and across biological systems. We identified new PPIs for receptors with well-characterized ligands and binding partners for "orphan" receptors. New PPIs include proteins expressed on multiple cell types and involved in diverse processes including immune and nervous system development and function, differentiation/proliferation, metabolism, vascularization, and reproduction. These PPIs provide a resource for further biological investigation into their functional relevance and may offer new therapeutic drug targets.


Subject(s)
Ligands , Protein Interaction Maps/physiology , Receptors, Cell Surface/metabolism , DCC Receptor/chemistry , DCC Receptor/metabolism , Humans , Phylogeny , Receptor-Like Protein Tyrosine Phosphatases, Class 2/chemistry , Receptor-Like Protein Tyrosine Phosphatases, Class 2/metabolism , Receptors, Cell Surface/chemistry , Receptors, Cell Surface/classification , Receptors, Interleukin-1/chemistry , Receptors, Interleukin-1/metabolism , Signaling Lymphocytic Activation Molecule Family/chemistry , Signaling Lymphocytic Activation Molecule Family/metabolism , Surface Plasmon Resonance
5.
Cell ; 183(2): 503-521.e19, 2020 10 15.
Article in English | MEDLINE | ID: mdl-33007266

ABSTRACT

The control over the extent and timing of G protein signaling is provided by the regulator of G protein signaling (RGS) proteins that deactivate G protein α subunits (Gα). Mammalian genomes encode 20 canonical RGS and 16 Gα genes with key roles in physiology and disease. To understand the principles governing the selectivity of Gα regulation by RGS, we examine the catalytic activity of all canonical human RGS proteins and their selectivity for a complete set of Gα substrates using real-time kinetic measurements in living cells. The data reveal rules governing RGS-Gα recognition, the structural basis of its selectivity, and provide principles for engineering RGS proteins with defined selectivity. The study also explores the evolution of RGS-Gα selectivity through ancestral reconstruction and demonstrates how naturally occurring non-synonymous variants in RGS alter signaling. These results provide a blueprint for decoding signaling selectivity and advance our understanding of molecular recognition principles.


Subject(s)
GTP-Binding Protein alpha Subunits/metabolism , GTP-Binding Protein alpha Subunits/physiology , RGS Proteins/genetics , Animals , Female , GTP-Binding Protein Regulators/metabolism , GTP-Binding Protein alpha Subunits/genetics , HEK293 Cells , Humans , Kinetics , Male , Mice , Mice, Inbred C57BL , Neurons/metabolism , Primary Cell Culture , Protein Binding , RGS Proteins/metabolism , RGS Proteins/physiology , Signal Transduction/genetics
6.
Cell ; 175(1): 239-253.e17, 2018 09 20.
Article in English | MEDLINE | ID: mdl-30197081

ABSTRACT

Many disease-causing missense mutations affect intrinsically disordered regions (IDRs) of proteins, but the molecular mechanism of their pathogenicity is enigmatic. Here, we employ a peptide-based proteomic screen to investigate the impact of mutations in IDRs on protein-protein interactions. We find that mutations in disordered cytosolic regions of three transmembrane proteins (GLUT1, ITPR1, and CACNA1H) lead to an increased clathrin binding. All three mutations create dileucine motifs known to mediate clathrin-dependent trafficking. Follow-up experiments on GLUT1 (SLC2A1), the glucose transporter causative of GLUT1 deficiency syndrome, revealed that the mutated protein mislocalizes to intracellular compartments. Mutant GLUT1 interacts with adaptor proteins (APs) in vitro, and knocking down AP-2 reverts the cellular mislocalization and restores glucose transport. A systematic analysis of other known disease-causing variants revealed a significant and specific overrepresentation of gained dileucine motifs in structurally disordered cytosolic domains of transmembrane proteins. Thus, several mutations in disordered regions appear to cause "dileucineopathies."


Subject(s)
Glucose Transporter Type 1/physiology , Intrinsically Disordered Proteins/genetics , Intrinsically Disordered Proteins/physiology , Amino Acid Motifs/genetics , Amino Acid Sequence , Animals , Binding Sites , Calcium Channels, T-Type/genetics , Calcium Channels, T-Type/physiology , Carbohydrate Metabolism, Inborn Errors , Clathrin/metabolism , Cytoplasm/metabolism , Glucose Transporter Type 1/genetics , Glucose Transporter Type 1/metabolism , Humans , Inositol 1,4,5-Trisphosphate Receptors/genetics , Inositol 1,4,5-Trisphosphate Receptors/physiology , Intrinsically Disordered Proteins/metabolism , Leucine/metabolism , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Monosaccharide Transport Proteins/deficiency , Mutation/genetics , Peptides , Protein Binding , Proteomics/methods
7.
Cell ; 167(4): 985-1000.e21, 2016 11 03.
Article in English | MEDLINE | ID: mdl-27881304

ABSTRACT

Mitochondrial sirtuins, SIRT3-5, are NAD+-dependent deacylases and ADP-ribosyltransferases that are critical for stress responses. However, a comprehensive understanding of sirtuin targets, regulation of sirtuin activity, and the relationships between sirtuins remains a key challenge in mitochondrial physiology. Here, we employ systematic interaction proteomics to elucidate the mitochondrial sirtuin protein interaction landscape. This work reveals sirtuin interactions with numerous functional modules within mitochondria, identifies candidate sirtuin substrates, and uncovers a fundamental role for sequestration of SIRT3 by ATP synthase in mitochondrial homeostasis. In healthy mitochondria, a pool of SIRT3 binds ATP synthase, but upon matrix pH reduction with concomitant loss of mitochondrial membrane potential, SIRT3 dissociates. This release correlates with rapid deacetylation of matrix proteins, and SIRT3 is required for recovery of membrane potential. In vitro reconstitution experiments, as well as analysis of CRISPR/Cas9-engineered cells, indicate that pH-dependent SIRT3 release requires H135 in the ATP5O subunit of ATP synthase. Our SIRT3-5 interaction network provides a framework for discovering novel biological functions regulated by mitochondrial sirtuins.


Subject(s)
Mitochondria/metabolism , Protein Interaction Maps , Sirtuin 3/metabolism , Acetylation , Adenosine Triphosphatases/metabolism , Animals , Carrier Proteins/metabolism , HeLa Cells , Humans , Immunoprecipitation , Membrane Proteins/metabolism , Mice , Mitochondrial Proteins/metabolism , Mitochondrial Proton-Translocating ATPases , Sirtuins/classification , Sirtuins/metabolism
8.
Mol Cell ; 83(14): 2595-2611.e11, 2023 07 20.
Article in English | MEDLINE | ID: mdl-37421941

ABSTRACT

RNA-binding proteins (RBPs) control RNA metabolism to orchestrate gene expression and, when dysfunctional, underlie human diseases. Proteome-wide discovery efforts predict thousands of RBP candidates, many of which lack canonical RNA-binding domains (RBDs). Here, we present a hybrid ensemble RBP classifier (HydRA), which leverages information from both intermolecular protein interactions and internal protein sequence patterns to predict RNA-binding capacity with unparalleled specificity and sensitivity using support vector machines (SVMs), convolutional neural networks (CNNs), and Transformer-based protein language models. Occlusion mapping by HydRA robustly detects known RBDs and predicts hundreds of uncharacterized RNA-binding associated domains. Enhanced CLIP (eCLIP) for HydRA-predicted RBP candidates reveals transcriptome-wide RNA targets and confirms RNA-binding activity for HydRA-predicted RNA-binding associated domains. HydRA accelerates construction of a comprehensive RBP catalog and expands the diversity of RNA-binding associated domains.


Subject(s)
Deep Learning , Hydra , Animals , Humans , RNA/metabolism , Protein Binding , Binding Sites/genetics , Hydra/genetics , Hydra/metabolism
9.
Mol Cell ; 83(5): 770-786.e9, 2023 03 02.
Article in English | MEDLINE | ID: mdl-36805027

ABSTRACT

E3 ligase recruitment of proteins containing terminal destabilizing motifs (degrons) is emerging as a major form of regulation. How those E3s discriminate bona fide substrates from other proteins with terminal degron-like sequences remains unclear. Here, we report that human KLHDC2, a CRL2 substrate receptor targeting C-terminal Gly-Gly degrons, is regulated through interconversion between two assemblies. In the self-inactivated homotetramer, KLHDC2's C-terminal Gly-Ser motif mimics a degron and engages the substrate-binding domain of another protomer. True substrates capture the monomeric CRL2KLHDC2, driving E3 activation by neddylation and subsequent substrate ubiquitylation. Non-substrates such as NEDD8 bind KLHDC2 with high affinity, but its slow on rate prevents productive association with CRL2KLHDC2. Without substrate, neddylated CRL2KLHDC2 assemblies are deactivated via distinct mechanisms: the monomer by deneddylation and the tetramer by auto-ubiquitylation. Thus, substrate specificity is amplified by KLHDC2 self-assembly acting like a molecular timer, where only bona fide substrates may bind before E3 ligase inactivation.


Subject(s)
Proteins , Ubiquitin-Protein Ligases , Humans , Carrier Proteins , Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism
10.
Immunity ; 54(9): 2042-2056.e8, 2021 09 14.
Article in English | MEDLINE | ID: mdl-34407391

ABSTRACT

Recruitment of immune cells to the site of inflammation by the chemokine CCL1 is important in the pathology of inflammatory diseases. Here, we examined the role of CCL1 in pulmonary fibrosis (PF). Bronchoalveolar lavage fluid from PF mouse models contained high amounts of CCL1, as did lung biopsies from PF patients. Immunofluorescence analyses revealed that alveolar macrophages and CD4+ T cells were major producers of CCL1 and targeted deletion of Ccl1 in these cells blunted pathology. Deletion of the CCL1 receptor Ccr8 in fibroblasts limited migration, but not activation, in response to CCL1. Mass spectrometry analyses of CCL1 complexes identified AMFR as a CCL1 receptor, and deletion of Amfr impaired fibroblast activation. Mechanistically, CCL1 binding triggered ubiquitination of the ERK inhibitor Spry1 by AMFR, thus activating Ras-mediated profibrotic protein synthesis. Antibody blockade of CCL1 ameliorated PF pathology, supporting the therapeutic potential of targeting this pathway for treating fibroproliferative lung diseases.


Subject(s)
Chemokine CCL1/metabolism , Fibroblasts/metabolism , Membrane Proteins/metabolism , Myofibroblasts/metabolism , Phosphoproteins/metabolism , Pulmonary Fibrosis/metabolism , Receptors, Autocrine Motility Factor/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cell Differentiation/physiology , Fibroblasts/pathology , Humans , Mice , Myofibroblasts/pathology , Pulmonary Fibrosis/pathology , Signal Transduction/physiology
11.
Mol Cell ; 81(19): 4091-4103.e9, 2021 10 07.
Article in English | MEDLINE | ID: mdl-34348091

ABSTRACT

We describe PROPER-seq (protein-protein interaction sequencing) to map protein-protein interactions (PPIs) en masse. PROPER-seq first converts transcriptomes of input cells into RNA-barcoded protein libraries, in which all interacting protein pairs are captured through nucleotide barcode ligation, recorded as chimeric DNA sequences, and decoded at once by sequencing and mapping. We applied PROPER-seq to human embryonic kidney cells, T lymphocytes, and endothelial cells and identified 210,518 human PPIs (collected in the PROPER v.1.0 database). Among these, 1,365 and 2,480 PPIs are supported by published co-immunoprecipitation (coIP) and affinity purification-mass spectrometry (AP-MS) data, 17,638 PPIs are predicted by the prePPI algorithm without previous experimental validation, and 100 PPIs overlap human synthetic lethal gene pairs. In addition, four previously uncharacterized interaction partners with poly(ADP-ribose) polymerase 1 (PARP1) (a critical protein in DNA repair) known as XPO1, MATR3, IPO5, and LEO1 are validated in vivo. PROPER-seq presents a time-effective technology to map PPIs at the transcriptome scale, and PROPER v.1.0 provides a rich resource for studying PPIs.


Subject(s)
Computational Biology , Gene Expression Profiling , Protein Interaction Mapping , Protein Interaction Maps , Proteins/genetics , Proteins/metabolism , RNA-Seq , Transcriptome , Databases, Genetic , Female , Genes, Lethal , HEK293 Cells , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Jurkat Cells , Karyopherins/genetics , Karyopherins/metabolism , Kidney/metabolism , Male , Nuclear Matrix-Associated Proteins/genetics , Nuclear Matrix-Associated Proteins/metabolism , Poly (ADP-Ribose) Polymerase-1/genetics , Poly (ADP-Ribose) Polymerase-1/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Cytoplasmic and Nuclear/metabolism , Software , T-Lymphocytes/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , beta Karyopherins/genetics , beta Karyopherins/metabolism , Exportin 1 Protein
12.
Trends Biochem Sci ; 49(3): 257-276, 2024 03.
Article in English | MEDLINE | ID: mdl-38233282

ABSTRACT

Histone lysine demethylases (KDMs) regulate eukaryotic gene transcription by catalysing the removal of methyl groups from histone proteins. These enzymes are intricately regulated by the kinase signalling system in response to internal and external stimuli. Here, we review the mechanisms by which kinase-mediated phosphorylation influence human histone KDM function. These include the changing of histone KDM subcellular localisation or chromatin binding, the altering of protein half-life, changes to histone KDM complex formation that result in histone demethylation, non-histone demethylation or demethylase-independent effects, and effects on histone KDM complex dissociation. We also explore the structural context of phospho-sites on histone KDMs and evaluate how this relates to function.


Subject(s)
Histone Demethylases , Histones , Humans , Histone Demethylases/metabolism , Histones/metabolism , Jumonji Domain-Containing Histone Demethylases/chemistry , Jumonji Domain-Containing Histone Demethylases/genetics , Jumonji Domain-Containing Histone Demethylases/metabolism , Phosphorylation , Demethylation
13.
Trends Biochem Sci ; 49(2): 169-182, 2024 02.
Article in English | MEDLINE | ID: mdl-38103971

ABSTRACT

The α-kinase eukaryotic elongation factor 2 kinase (eEF-2K) regulates translational elongation by phosphorylating its ribosome-associated substrate, the GTPase eEF-2. eEF-2K is activated by calmodulin (CaM) through a distinctive mechanism unlike that in other CaM-dependent kinases (CAMK). We describe recent structural insights into this unique activation process and examine the effects of specific regulatory signals on this mechanism. We also highlight key unanswered questions to guide future structure-function studies. These include structural mechanisms which enable eEF-2K to interact with upstream/downstream partners and facilitate its integration of diverse inputs, including Ca2+ transients, phosphorylation mediated by energy/nutrient-sensing pathways, pH changes, and metabolites. Answering these questions is key to establishing how eEF-2K harmonizes translation with cellular requirements within the boundaries of its molecular landscape.


Subject(s)
Elongation Factor 2 Kinase , Protein Biosynthesis , Elongation Factor 2 Kinase/chemistry , Elongation Factor 2 Kinase/genetics , Elongation Factor 2 Kinase/metabolism , Phosphorylation , Calmodulin/chemistry , Calmodulin/genetics , Calmodulin/metabolism
14.
Immunity ; 51(3): 522-534.e7, 2019 09 17.
Article in English | MEDLINE | ID: mdl-31471107

ABSTRACT

Although recent progress provides mechanistic insights into the pathogenesis of pulmonary fibrosis (PF), rare anti-PF therapeutics show definitive promise for treating this disease. Repeated lung epithelial injury results in injury-repairing response and inflammation, which drive the development of PF. Here, we report that chronic lung injury inactivated the ubiquitin-editing enzyme A20, causing progressive accumulation of the transcription factor C/EBPß in alveolar macrophages (AMs) from PF patients and mice, which upregulated a number of immunosuppressive and profibrotic factors promoting PF development. In response to chronic lung injury, elevated glycogen synthase kinase-3ß (GSK-3ß) interacted with and phosphorylated A20 to suppress C/EBPß degradation. Ectopic expression of A20 or pharmacological restoration of A20 activity by disturbing the A20-GSK-3ß interaction accelerated C/EBPß degradation and showed potent therapeutic efficacy against experimental PF. Our study indicates that a regulatory mechanism of the GSK-3ß-A20-C/EBPß axis in AMs may be a potential target for treating PF and fibroproliferative lung diseases.


Subject(s)
CCAAT-Enhancer-Binding Protein-beta/metabolism , Macrophages/metabolism , Pulmonary Fibrosis/metabolism , Transcription Factors/metabolism , Ubiquitin/metabolism , Animals , Cell Line , Glycogen Synthase Kinase 3 beta/metabolism , HEK293 Cells , Humans , Inflammation/metabolism , Mice , Mice, Inbred C57BL , Phosphorylation/physiology , Signal Transduction/physiology , Ubiquitin-Protein Ligases/metabolism , Ubiquitination/physiology , Up-Regulation/physiology
15.
Trends Biochem Sci ; 48(6): 527-538, 2023 06.
Article in English | MEDLINE | ID: mdl-37061423

ABSTRACT

Protein-protein interactions (PPIs) drive biological processes, and disruption of PPIs can cause disease. With recent breakthroughs in structure prediction and a deluge of genomic sequence data, computational methods to predict PPIs and model spatial structures of protein complexes are now approaching the accuracy of experimental approaches for permanent interactions and show promise for elucidating transient interactions. As we describe here, the key to this success is rich evolutionary information deciphered from thousands of homologous sequences that coevolve in interacting partners. This covariation signal, revealed by sophisticated statistical and machine learning (ML) algorithms, predicts physiological interactions. Accurate artificial intelligence (AI)-based modeling of protein structures promises to provide accurate 3D models of PPIs at a proteome-wide scale.


Subject(s)
Artificial Intelligence , Protein Interaction Mapping , Protein Interaction Mapping/methods , Algorithms , Machine Learning , Proteome , Computational Biology/methods
16.
EMBO J ; 42(8): e112304, 2023 04 17.
Article in English | MEDLINE | ID: mdl-36825429

ABSTRACT

The tumor suppressor p53 is critical for tumor suppression, but the regulatory role of p53 in alcohol-induced fatty liver remains unclear. Here, we show a role for p53 in regulating ethanol metabolism via acetaldehyde dehydrogenase 2 (ALDH2), a key enzyme responsible for the oxidization of alcohol. By repressing ethanol oxidization, p53 suppresses intracellular levels of acetyl-CoA and histone acetylation, leading to the inhibition of the stearoyl-CoA desaturase-1 (SCD1) gene expression. Mechanistically, p53 directly binds to ALDH2 and prevents the formation of its active tetramer and indirectly limits the production of pyruvate that promotes the activity of ALDH2. Notably, p53-deficient mice exhibit increased lipid accumulation, which can be reversed by ALDH2 depletion. Moreover, liver-specific knockdown of SCD1 alleviates ethanol-induced hepatic steatosis caused by p53 loss. By contrast, overexpression of SCD1 in liver promotes ethanol-induced fatty liver development in wild-type mice, while it has a mild effect on p53-/- or ALDH2-/- mice. Overall, our findings reveal a previously unrecognized function of p53 in alcohol-induced fatty liver and uncover pyruvate as a natural regulator of ALDH2.


Subject(s)
Aldehyde Dehydrogenase, Mitochondrial , Fatty Liver, Alcoholic , Fatty Liver , Tumor Suppressor Protein p53 , Animals , Mice , Aldehyde Dehydrogenase, Mitochondrial/genetics , Aldehyde Dehydrogenase, Mitochondrial/metabolism , Ethanol/toxicity , Ethanol/metabolism , Fatty Liver/genetics , Fatty Liver/metabolism , Fatty Liver, Alcoholic/genetics , Fatty Liver, Alcoholic/metabolism , Liver/metabolism , Pyruvates/metabolism , Pyruvates/pharmacology , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
17.
Plant Cell ; 2024 Apr 18.
Article in English | MEDLINE | ID: mdl-38635962

ABSTRACT

Protein S-acylation catalyzed by protein S-acyl transferases (PATs) is a reversible lipid modification regulating protein targeting, stability, and interaction profiles. PATs are encoded by large gene families in plants, and many proteins including receptor-like cytoplasmic kinases (RLCKs) and receptor-like kinases (RLKs) are subject to S-acylation. However, few PATs have been assigned substrates, and few S-acylated proteins have known upstream enzymes. We report that Arabidopsis (Arabidopsis thaliana) class A PATs redundantly mediate pollen tube guidance and participate in the S-acylation of POLLEN RECEPTOR KINASE1 (PRK1) and LOST IN POLLEN TUBE GUIDANCE1 (LIP1), a critical RLK or RLCK for pollen tube guidance, respectively. PAT1, PAT2, PAT3, PAT4, and PAT8, collectively named PENTAPAT for simplicity, are enriched in pollen and show similar subcellular distribution. Functional loss of PENTAPAT reduces seed set due to male gametophytic defects. Specifically, pentapat pollen tubes are compromised in directional growth. We determine that PRK1 and LIP1 interact with PENTAPAT, and their S-acylation is reduced in pentapat pollen. The plasma membrane (PM) association of LIP1 is reduced in pentapat pollen, whereas point mutations reducing PRK1 S-acylation affect its affinity with its interacting proteins. Our results suggest a key role of S-acylation in pollen tube guidance through modulating PM receptor complexes.

18.
Mol Cell ; 74(5): 996-1009.e7, 2019 06 06.
Article in English | MEDLINE | ID: mdl-30975460

ABSTRACT

Nucleotide-binding site leucine-rich repeat (NLR) receptors perceive pathogen effectors and trigger plant immunity. However, the mechanisms underlying NLR-triggered defense responses remain obscure. The recently discovered Pigm locus in rice encodes a cluster of NLRs, including PigmR, which confers broad-spectrum resistance to blast fungus. Here, we identify PIBP1 (PigmR-INTERACTING and BLAST RESISTANCE PROTEIN 1), an RRM (RNA-recognition motif) protein that specifically interacts with PigmR and other similar NLRs to trigger blast resistance. PigmR-promoted nuclear accumulation of PIBP1 ensures full blast resistance. We find that PIBP1 and a homolog, Os06 g02240, bind DNA and function as unconventional transcription factors at the promoters of the defense genes OsWAK14 and OsPAL1, activating their expression. Knockout of PIBP1 and Os06 g02240 greatly attenuated blast resistance. Collectively, our study discovers previously unappreciated RRM transcription factors that directly interact with NLRs to activate plant defense, establishing a direct link between transcriptional activation of immune responses with NLR-mediated pathogen perception.


Subject(s)
Disease Resistance/genetics , NLR Proteins/genetics , Oryza/genetics , Plant Diseases/genetics , Plant Proteins/genetics , Transcription Factors/genetics , Binding Sites , Fungi/pathogenicity , Gene Expression Regulation, Plant , Oryza/microbiology , Plant Diseases/microbiology , Plant Immunity/genetics , Promoter Regions, Genetic , Protein Binding/genetics , Signal Transduction/genetics
19.
Brief Bioinform ; 25(2)2024 Jan 22.
Article in English | MEDLINE | ID: mdl-38446741

ABSTRACT

Identifying protein-protein interactions (PPIs) is crucial for deciphering biological pathways. Numerous prediction methods have been developed as cheap alternatives to biological experiments, reporting surprisingly high accuracy estimates. We systematically investigated how much reproducible deep learning models depend on data leakage, sequence similarities and node degree information, and compared them with basic machine learning models. We found that overlaps between training and test sets resulting from random splitting lead to strongly overestimated performances. In this setting, models learn solely from sequence similarities and node degrees. When data leakage is avoided by minimizing sequence similarities between training and test set, performances become random. Moreover, baseline models directly leveraging sequence similarity and network topology show good performances at a fraction of the computational cost. Thus, we advocate that any improvements should be reported relative to baseline methods in the future. Our findings suggest that predicting PPIs remains an unsolved task for proteins showing little sequence similarity to previously studied proteins, highlighting that further experimental research into the 'dark' protein interactome and better computational methods are needed.


Subject(s)
Machine Learning
20.
Brief Bioinform ; 25(3)2024 Mar 27.
Article in English | MEDLINE | ID: mdl-38609331

ABSTRACT

Natural language processing (NLP) has become an essential technique in various fields, offering a wide range of possibilities for analyzing data and developing diverse NLP tasks. In the biomedical domain, understanding the complex relationships between compounds and proteins is critical, especially in the context of signal transduction and biochemical pathways. Among these relationships, protein-protein interactions (PPIs) are of particular interest, given their potential to trigger a variety of biological reactions. To improve the ability to predict PPI events, we propose the protein event detection dataset (PEDD), which comprises 6823 abstracts, 39 488 sentences and 182 937 gene pairs. Our PEDD dataset has been utilized in the AI CUP Biomedical Paper Analysis competition, where systems are challenged to predict 12 different relation types. In this paper, we review the state-of-the-art relation extraction research and provide an overview of the PEDD's compilation process. Furthermore, we present the results of the PPI extraction competition and evaluate several language models' performances on the PEDD. This paper's outcomes will provide a valuable roadmap for future studies on protein event detection in NLP. By addressing this critical challenge, we hope to enable breakthroughs in drug discovery and enhance our understanding of the molecular mechanisms underlying various diseases.


Subject(s)
Drug Discovery , Natural Language Processing , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL