Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 720
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Cell ; 186(9): 1895-1911.e21, 2023 04 27.
Article in English | MEDLINE | ID: mdl-37028429

ABSTRACT

Cells respond to environmental cues by remodeling their inventories of multiprotein complexes. Cellular repertoires of SCF (SKP1-CUL1-F box protein) ubiquitin ligase complexes, which mediate much protein degradation, require CAND1 to distribute the limiting CUL1 subunit across the family of ∼70 different F box proteins. Yet, how a single factor coordinately assembles numerous distinct multiprotein complexes remains unknown. We obtained cryo-EM structures of CAND1-bound SCF complexes in multiple states and correlated mutational effects on structures, biochemistry, and cellular assays. The data suggest that CAND1 clasps idling catalytic domains of an inactive SCF, rolls around, and allosterically rocks and destabilizes the SCF. New SCF production proceeds in reverse, through SKP1-F box allosterically destabilizing CAND1. The CAND1-SCF conformational ensemble recycles CUL1 from inactive complexes, fueling mixing and matching of SCF parts for E3 activation in response to substrate availability. Our data reveal biogenesis of a predominant family of E3 ligases, and the molecular basis for systemwide multiprotein complex assembly.


Subject(s)
Cullin Proteins , F-Box Proteins , SKP Cullin F-Box Protein Ligases , Transcription Factors , Humans , Cullin Proteins/chemistry , Cullin Proteins/metabolism , F-Box Proteins/metabolism , Molecular Conformation , SKP Cullin F-Box Protein Ligases/chemistry , SKP Cullin F-Box Protein Ligases/metabolism , Transcription Factors/metabolism , Ubiquitin-Protein Ligases/metabolism
2.
Cell ; 185(26): 5040-5058.e19, 2022 12 22.
Article in English | MEDLINE | ID: mdl-36563667

ABSTRACT

Spatial molecular profiling of complex tissues is essential to investigate cellular function in physiological and pathological states. However, methods for molecular analysis of large biological specimens imaged in 3D are lacking. Here, we present DISCO-MS, a technology that combines whole-organ/whole-organism clearing and imaging, deep-learning-based image analysis, robotic tissue extraction, and ultra-high-sensitivity mass spectrometry. DISCO-MS yielded proteome data indistinguishable from uncleared samples in both rodent and human tissues. We used DISCO-MS to investigate microglia activation along axonal tracts after brain injury and characterized early- and late-stage individual amyloid-beta plaques in a mouse model of Alzheimer's disease. DISCO-bot robotic sample extraction enabled us to study the regional heterogeneity of immune cells in intact mouse bodies and aortic plaques in a complete human heart. DISCO-MS enables unbiased proteome analysis of preclinical and clinical tissues after unbiased imaging of entire specimens in 3D, identifying diagnostic and therapeutic opportunities for complex diseases. VIDEO ABSTRACT.


Subject(s)
Alzheimer Disease , Proteome , Mice , Humans , Animals , Proteome/analysis , Proteomics/methods , Alzheimer Disease/pathology , Amyloid beta-Peptides , Mass Spectrometry , Plaque, Amyloid
3.
Cell ; 184(18): 4840-4840.e1, 2021 09 02.
Article in English | MEDLINE | ID: mdl-34478659

ABSTRACT

In-depth quantitative proteomic analysis of clinical specimens offers unique and invaluable insights into their protein composition and biochemical state in physiological and pathological conditions. A plethora of methodologies have been developed and customized. Here, we summarize the most common and emerging mass spectrometry (MS)-based workflows. Due to space limitations the overview cannot be complete. To view this SnapShot, open or download the PDF.


Subject(s)
Proteomics , Humans , Mass Spectrometry , Proteins/metabolism
4.
Cell ; 176(1-2): 167-181.e21, 2019 01 10.
Article in English | MEDLINE | ID: mdl-30595447

ABSTRACT

Covalent DNA-protein cross-links (DPCs) impede replication fork progression and threaten genome integrity. Using Xenopus egg extracts, we previously showed that replication fork collision with DPCs causes their proteolysis, followed by translesion DNA synthesis. We show here that when DPC proteolysis is blocked, the replicative DNA helicase CMG (CDC45, MCM2-7, GINS), which travels on the leading strand template, bypasses an intact leading strand DPC. Single-molecule imaging reveals that GINS does not dissociate from CMG during bypass and that CMG slows dramatically after bypass, likely due to uncoupling from the stalled leading strand. The DNA helicase RTEL1 facilitates bypass, apparently by generating single-stranded DNA beyond the DPC. The absence of RTEL1 impairs DPC proteolysis, suggesting that CMG must bypass the DPC to enable proteolysis. Our results suggest a mechanism that prevents inadvertent CMG destruction by DPC proteases, and they reveal CMG's remarkable capacity to overcome obstacles on its translocation strand.


Subject(s)
DNA Helicases/metabolism , DNA Helicases/physiology , DNA Repair/physiology , Animals , Cell Cycle Proteins/metabolism , DNA/metabolism , DNA Replication , DNA, Single-Stranded , DNA-Binding Proteins/physiology , Female , Male , Proteolysis , Single Molecule Imaging/methods , Xenopus laevis/metabolism
5.
Nat Immunol ; 22(4): 485-496, 2021 04.
Article in English | MEDLINE | ID: mdl-33767426

ABSTRACT

Evasion of host immunity is a hallmark of cancer; however, mechanisms linking oncogenic mutations and immune escape are incompletely understood. Through loss-of-function screening of 1,001 tumor suppressor genes, we identified death-associated protein kinase 3 (DAPK3) as a previously unrecognized driver of anti-tumor immunity through the stimulator of interferon genes (STING) pathway of cytosolic DNA sensing. Loss of DAPK3 expression or kinase activity impaired STING activation and interferon (IFN)-ß-stimulated gene induction. DAPK3 deficiency in IFN-ß-producing tumors drove rapid growth and reduced infiltration of CD103+CD8α+ dendritic cells and cytotoxic lymphocytes, attenuating the response to cancer chemo-immunotherapy. Mechanistically, DAPK3 coordinated post-translational modification of STING. In unstimulated cells, DAPK3 inhibited STING K48-linked poly-ubiquitination and proteasome-mediated degradation. After cGAMP stimulation, DAPK3 was required for STING K63-linked poly-ubiquitination and STING-TANK-binding kinase 1 interaction. Comprehensive phospho-proteomics uncovered a DAPK3-specific phospho-site on the E3 ligase LMO7, critical for LMO7-STING interaction and STING K63-linked poly-ubiquitination. Thus, DAPK3 is an essential kinase for STING activation that drives tumor-intrinsic innate immunity and tumor immune surveillance.


Subject(s)
Death-Associated Protein Kinases/metabolism , Human Umbilical Vein Endothelial Cells/enzymology , Immunity, Innate , Interferon-beta/metabolism , Membrane Proteins/metabolism , Neoplasms/enzymology , Tumor Escape , Animals , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Cell Line, Tumor , Death-Associated Protein Kinases/genetics , Female , Gene Expression Regulation, Neoplastic , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Immune Checkpoint Inhibitors/pharmacology , Immunity, Innate/drug effects , Interferon-beta/genetics , LIM Domain Proteins/genetics , LIM Domain Proteins/metabolism , Membrane Proteins/genetics , Mice, Inbred C57BL , Mice, Knockout , Neoplasms/drug therapy , Neoplasms/genetics , Neoplasms/immunology , Phosphorylation , Signal Transduction , Transcription Factors/genetics , Transcription Factors/metabolism , Tumor Escape/drug effects , Ubiquitination
7.
Cell ; 175(1): 159-170.e16, 2018 09 20.
Article in English | MEDLINE | ID: mdl-30241606

ABSTRACT

Most high-grade serous ovarian cancer (HGSOC) patients develop resistance to platinum-based chemotherapy and recur, but 15% remain disease free over a decade. To discover drivers of long-term survival, we quantitatively analyzed the proteomes of platinum-resistant and -sensitive HGSOC patients from minute amounts of formalin-fixed, paraffin-embedded tumors. This revealed cancer/testis antigen 45 (CT45) as an independent prognostic factor associated with a doubling of disease-free survival in advanced-stage HGSOC. Phospho- and interaction proteomics tied CT45 to DNA damage pathways through direct interaction with the PP4 phosphatase complex. In vitro, CT45 regulated PP4 activity, and its high expression led to increased DNA damage and platinum sensitivity. CT45-derived HLA class I peptides, identified by immunopeptidomics, activate patient-derived cytotoxic T cells and promote tumor cell killing. This study highlights the power of clinical cancer proteomics to identify targets for chemo- and immunotherapy and illuminate their biological roles.


Subject(s)
Antigens, Neoplasm/physiology , Drug Resistance, Neoplasm/genetics , Proteomics/methods , Aged , Amino Acid Sequence/genetics , Antineoplastic Agents/therapeutic use , DNA Methylation/drug effects , Female , Gene Expression Regulation, Neoplastic/genetics , Humans , Immunotherapy/methods , Kaplan-Meier Estimate , Middle Aged , Neoplasm Recurrence, Local/drug therapy , Ovarian Neoplasms/drug therapy , Phosphoprotein Phosphatases/metabolism , Phosphoprotein Phosphatases/physiology , Prognosis
8.
Nat Immunol ; 21(8): 927-937, 2020 08.
Article in English | MEDLINE | ID: mdl-32632289

ABSTRACT

In response to pathogenic threats, naive T cells rapidly transition from a quiescent to an activated state, yet the underlying mechanisms are incompletely understood. Using a pulsed SILAC approach, we investigated the dynamics of mRNA translation kinetics and protein turnover in human naive and activated T cells. Our datasets uncovered that transcription factors maintaining T cell quiescence had constitutively high turnover, which facilitated their depletion following activation. Furthermore, naive T cells maintained a surprisingly large number of idling ribosomes as well as 242 repressed mRNA species and a reservoir of glycolytic enzymes. These components were rapidly engaged following stimulation, promoting an immediate translational and glycolytic switch to ramp up the T cell activation program. Our data elucidate new insights into how T cells maintain a prepared state to mount a rapid immune response, and provide a resource of protein turnover, absolute translation kinetics and protein synthesis rates in T cells ( https://www.immunomics.ch ).


Subject(s)
Lymphocyte Activation/physiology , Protein Biosynthesis/immunology , T-Lymphocytes/immunology , Humans , RNA, Messenger/immunology , RNA, Messenger/metabolism , Transcription Factors/immunology , Transcription Factors/metabolism
9.
Mol Cell ; 84(2): 293-308.e14, 2024 Jan 18.
Article in English | MEDLINE | ID: mdl-38113892

ABSTRACT

Ubiquitylation is catalyzed by coordinated actions of E3 and E2 enzymes. Molecular principles governing many important E3-E2 partnerships remain unknown, including those for RING-family GID/CTLH E3 ubiquitin ligases and their dedicated E2, Ubc8/UBE2H (yeast/human nomenclature). GID/CTLH-Ubc8/UBE2H-mediated ubiquitylation regulates biological processes ranging from yeast metabolic signaling to human development. Here, cryoelectron microscopy (cryo-EM), biochemistry, and cell biology reveal this exquisitely specific E3-E2 pairing through an unconventional catalytic assembly and auxiliary interactions 70-100 Å away, mediated by E2 multisite phosphorylation. Rather than dynamic polyelectrostatic interactions reported for other ubiquitylation complexes, multiple Ubc8/UBE2H phosphorylation sites within acidic CK2-targeted sequences specifically anchor the E2 C termini to E3 basic patches. Positions of phospho-dependent interactions relative to the catalytic domains correlate across evolution. Overall, our data show that phosphorylation-dependent multivalency establishes a specific E3-E2 partnership, is antagonistic with dephosphorylation, rigidifies the catalytic centers within a flexing GID E3-substrate assembly, and facilitates substrate collision with ubiquitylation active sites.


Subject(s)
Saccharomyces cerevisiae , Ubiquitin-Conjugating Enzymes , Humans , Ubiquitin-Conjugating Enzymes/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Phosphorylation , Cryoelectron Microscopy , Ubiquitin-Protein Ligases/metabolism , Ubiquitination
10.
Mol Cell ; 84(10): 1948-1963.e11, 2024 May 16.
Article in English | MEDLINE | ID: mdl-38759627

ABSTRACT

The yeast glucose-induced degradation-deficient (GID) E3 ubiquitin ligase forms a suite of complexes with interchangeable receptors that selectively recruit N-terminal degron motifs of metabolic enzyme substrates. The orthologous higher eukaryotic C-terminal to LisH (CTLH) E3 complex has been proposed to also recognize substrates through an alternative subunit, WDR26, which promotes the formation of supramolecular CTLH E3 assemblies. Here, we discover that human WDR26 binds the metabolic enzyme nicotinamide/nicotinic-acid-mononucleotide-adenylyltransferase 1 (NMNAT1) and mediates its CTLH E3-dependent ubiquitylation independently of canonical GID/CTLH E3-family substrate receptors. The CTLH subunit YPEL5 inhibits NMNAT1 ubiquitylation and cellular turnover by WDR26-CTLH E3, thereby affecting NMNAT1-mediated metabolic activation and cytotoxicity of the prodrug tiazofurin. Cryoelectron microscopy (cryo-EM) structures of NMNAT1- and YPEL5-bound WDR26-CTLH E3 complexes reveal an internal basic degron motif of NMNAT1 essential for targeting by WDR26-CTLH E3 and degron mimicry by YPEL5's N terminus antagonizing substrate binding. Thus, our data provide a mechanistic understanding of how YPEL5-WDR26-CTLH E3 acts as a modulator of NMNAT1-dependent metabolism.


Subject(s)
Nicotinamide-Nucleotide Adenylyltransferase , Prodrugs , Ubiquitin-Protein Ligases , Ubiquitination , Humans , HEK293 Cells , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Protein Ligases/genetics , Prodrugs/metabolism , Nicotinamide-Nucleotide Adenylyltransferase/metabolism , Nicotinamide-Nucleotide Adenylyltransferase/genetics , Substrate Specificity , Cryoelectron Microscopy , Protein Binding
11.
Nat Immunol ; 20(4): 493-502, 2019 04.
Article in English | MEDLINE | ID: mdl-30833792

ABSTRACT

Interferon-stimulated genes (ISGs) form the backbone of the innate immune system and are important for limiting intra- and intercellular viral replication and spread. We conducted a mass-spectrometry-based survey to understand the fundamental organization of the innate immune system and to explore the molecular functions of individual ISGs. We identified interactions between 104 ISGs and 1,401 cellular binding partners engaging in 2,734 high-confidence interactions. 90% of these interactions are unreported so far, and our survey therefore illuminates a far wider activity spectrum of ISGs than is currently known. Integration of the resulting ISG-interaction network with published datasets and functional studies allowed us to identify regulators of immunity and processes related to the immune system. Given the extraordinary robustness of the innate immune system, this ISG network may serve as a blueprint for therapeutic targeting of cellular systems to efficiently fight viral infections.


Subject(s)
Immunity, Innate , Interferons/physiology , Protein Interaction Mapping , Antigens, Neoplasm/metabolism , Biomarkers, Tumor/metabolism , Carrier Proteins/metabolism , Cell Line , Gene Expression , Glycoproteins/metabolism , HEK293 Cells , HeLa Cells , Humans , Immunity, Innate/genetics , Mass Spectrometry , Receptors, CCR4/metabolism , Receptors, Peptide/metabolism , Ribonucleoproteins/metabolism , Viral Proteins/metabolism
12.
Immunity ; 55(12): 2271-2284.e7, 2022 12 13.
Article in English | MEDLINE | ID: mdl-36384135

ABSTRACT

The NLRP3 inflammasome plays a central role in antimicrobial defense as well as in the context of sterile inflammatory conditions. NLRP3 activity is governed by two independent signals: the first signal primes NLRP3, rendering it responsive to the second signal, which then triggers inflammasome formation. Our understanding of how NLRP3 priming contributes to inflammasome activation remains limited. Here, we show that IKKß, a kinase activated during priming, induces recruitment of NLRP3 to phosphatidylinositol-4-phosphate (PI4P), a phospholipid enriched on the trans-Golgi network. NEK7, a mitotic spindle kinase that had previously been thought to be indispensable for NLRP3 activation, was redundant for inflammasome formation when IKKß recruited NLRP3 to PI4P. Studying iPSC-derived human macrophages revealed that the IKKß-mediated NEK7-independent pathway constitutes the predominant NLRP3 priming mechanism in human myeloid cells. Our results suggest that PI4P binding represents a primed state into which NLRP3 is brought by IKKß activity.


Subject(s)
Inflammasomes , NLR Family, Pyrin Domain-Containing 3 Protein , Humans , I-kappa B Kinase , Inflammasomes/metabolism , Mice, Inbred C57BL , NIMA-Related Kinases/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Protein Serine-Threonine Kinases/metabolism , trans-Golgi Network/metabolism
13.
Cell ; 167(3): 829-842.e13, 2016 Oct 20.
Article in English | MEDLINE | ID: mdl-27745970

ABSTRACT

Metabolic activity is intimately linked to T cell fate and function. Using high-resolution mass spectrometry, we generated dynamic metabolome and proteome profiles of human primary naive T cells following activation. We discovered critical changes in the arginine metabolism that led to a drop in intracellular L-arginine concentration. Elevating L-arginine levels induced global metabolic changes including a shift from glycolysis to oxidative phosphorylation in activated T cells and promoted the generation of central memory-like cells endowed with higher survival capacity and, in a mouse model, anti-tumor activity. Proteome-wide probing of structural alterations, validated by the analysis of knockout T cell clones, identified three transcriptional regulators (BAZ1B, PSIP1, and TSN) that sensed L-arginine levels and promoted T cell survival. Thus, intracellular L-arginine concentrations directly impact the metabolic fitness and survival capacity of T cells that are crucial for anti-tumor responses.


Subject(s)
Arginine/metabolism , CD4-Positive T-Lymphocytes/immunology , Immunomodulation , Lymphocyte Activation , Melanoma, Experimental/immunology , Skin Neoplasms/immunology , Adaptor Proteins, Signal Transducing/metabolism , Animals , CD4-Positive T-Lymphocytes/metabolism , DNA-Binding Proteins/metabolism , Gene Knockout Techniques , Glycolysis , Humans , Immunologic Memory , Metabolome , Mice , Mice, Inbred BALB C , Oxidative Phosphorylation , Proteome , Transcription Factors/metabolism , Transcription, Genetic
14.
Cell ; 163(3): 712-23, 2015 Oct 22.
Article in English | MEDLINE | ID: mdl-26496610

ABSTRACT

The organization of a cell emerges from the interactions in protein networks. The interactome is critically dependent on the strengths of interactions and the cellular abundances of the connected proteins, both of which span orders of magnitude. However, these aspects have not yet been analyzed globally. Here, we have generated a library of HeLa cell lines expressing 1,125 GFP-tagged proteins under near-endogenous control, which we used as input for a next-generation interaction survey. Using quantitative proteomics, we detect specific interactions, estimate interaction stoichiometries, and measure cellular abundances of interacting proteins. These three quantitative dimensions reveal that the protein network is dominated by weak, substoichiometric interactions that play a pivotal role in defining network topology. The minority of stable complexes can be identified by their unique stoichiometry signature. This study provides a rich interaction dataset connecting thousands of proteins and introduces a framework for quantitative network analysis.


Subject(s)
Protein Interaction Mapping , Proteomics/methods , Cell Line , Chromosomes, Artificial, Bacterial/genetics , Humans
15.
Cell ; 161(4): 919-32, 2015 May 07.
Article in English | MEDLINE | ID: mdl-25957690

ABSTRACT

Aging has been associated with a progressive decline of proteostasis, but how this process affects proteome composition remains largely unexplored. Here, we profiled more than 5,000 proteins along the lifespan of the nematode C. elegans. We find that one-third of proteins change in abundance at least 2-fold during aging, resulting in a severe proteome imbalance. These changes are reduced in the long-lived daf-2 mutant but are enhanced in the short-lived daf-16 mutant. While ribosomal proteins decline and lose normal stoichiometry, proteasome complexes increase. Proteome imbalance is accompanied by widespread protein aggregation, with abundant proteins that exceed solubility contributing most to aggregate load. Notably, the properties by which proteins are selected for aggregation differ in the daf-2 mutant, and an increased formation of aggregates associated with small heat-shock proteins is observed. We suggest that sequestering proteins into chaperone-enriched aggregates is a protective strategy to slow proteostasis decline during nematode aging.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/growth & development , Caenorhabditis elegans/metabolism , Proteome/metabolism , Aging , Animals , Caenorhabditis elegans Proteins/genetics , Mutation , Protein Aggregates
16.
Mol Cell ; 82(12): 2335-2349, 2022 06 16.
Article in English | MEDLINE | ID: mdl-35714588

ABSTRACT

Mass spectrometry (MS)-based proteomics has become a powerful technology to quantify the entire complement of proteins in cells or tissues. Here, we review challenges and recent advances in the LC-MS-based analysis of minute protein amounts, down to the level of single cells. Application of this technology revealed that single-cell transcriptomes are dominated by stochastic noise due to the very low number of transcripts per cell, whereas the single-cell proteome appears to be complete. The spatial organization of cells in tissues can be studied by emerging technologies, including multiplexed imaging and spatial transcriptomics, which can now be combined with ultra-sensitive proteomics. Combined with high-content imaging, artificial intelligence and single-cell laser microdissection, MS-based proteomics provides an unbiased molecular readout close to the functional level. Potential applications range from basic biological questions to precision medicine.


Subject(s)
Artificial Intelligence , Proteomics , Mass Spectrometry/methods , Proteome/metabolism , Proteomics/methods
17.
Nat Immunol ; 18(5): 583-593, 2017 05.
Article in English | MEDLINE | ID: mdl-28263321

ABSTRACT

The immune system is unique in its dynamic interplay between numerous cell types. However, a system-wide view of how immune cells communicate to protect against disease has not yet been established. We applied high-resolution mass-spectrometry-based proteomics to characterize 28 primary human hematopoietic cell populations in steady and activated states at a depth of >10,000 proteins in total. Protein copy numbers revealed a specialization of immune cells for ligand and receptor expression, thereby connecting distinct immune functions. By integrating total and secreted proteomes, we discovered fundamental intercellular communication structures and previously unknown connections between cell types. Our publicly accessible (http://www.immprot.org/) proteomic resource provides a framework for the orchestration of cellular interplay and a reference for altered communication associated with pathology.


Subject(s)
Blood Cells/physiology , Immunity, Cellular , Protein Interaction Maps , Proteome , Proteomics , Animals , Bodily Secretions , Cell Communication , Computer Simulation , Humans , Mass Spectrometry , Social Support
18.
Nature ; 624(7990): 192-200, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37968396

ABSTRACT

Cellular functions are mediated by protein-protein interactions, and mapping the interactome provides fundamental insights into biological systems. Affinity purification coupled to mass spectrometry is an ideal tool for such mapping, but it has been difficult to identify low copy number complexes, membrane complexes and complexes that are disrupted by protein tagging. As a result, our current knowledge of the interactome is far from complete, and assessing the reliability of reported interactions is challenging. Here we develop a sensitive high-throughput method using highly reproducible affinity enrichment coupled to mass spectrometry combined with a quantitative two-dimensional analysis strategy to comprehensively map the interactome of Saccharomyces cerevisiae. Thousand-fold reduced volumes in 96-well format enabled replicate analysis of the endogenous GFP-tagged library covering the entire expressed yeast proteome1. The 4,159 pull-downs generated a highly structured network of 3,927 proteins connected by 31,004 interactions, doubling the number of proteins and tripling the number of reliable interactions compared with existing interactome maps2. This includes very-low-abundance epigenetic complexes, organellar membrane complexes and non-taggable complexes inferred by abundance correlation. This nearly saturated interactome reveals that the vast majority of yeast proteins are highly connected, with an average of 16 interactors. Similar to social networks between humans, the average shortest distance between proteins is 4.2 interactions. AlphaFold-Multimer provided novel insights into the functional roles of previously uncharacterized proteins in complexes. Our web portal ( www.yeast-interactome.org ) enables extensive exploration of the interactome dataset.


Subject(s)
Protein Interaction Mapping , Protein Interaction Maps , Proteome , Saccharomyces cerevisiae Proteins , Saccharomyces cerevisiae , Mass Spectrometry , Protein Interaction Mapping/methods , Proteome/chemistry , Proteome/metabolism , Reproducibility of Results , Saccharomyces cerevisiae/chemistry , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/metabolism , Epigenesis, Genetic , Databases, Factual
19.
Nature ; 617(7961): 581-591, 2023 May.
Article in English | MEDLINE | ID: mdl-37165188

ABSTRACT

The spatiotemporal structure of the human microbiome1,2, proteome3 and metabolome4,5 reflects and determines regional intestinal physiology and may have implications for disease6. Yet, little is known about the distribution of microorganisms, their environment and their biochemical activity in the gut because of reliance on stool samples and limited access to only some regions of the gut using endoscopy in fasting or sedated individuals7. To address these deficiencies, we developed an ingestible device that collects samples from multiple regions of the human intestinal tract during normal digestion. Collection of 240 intestinal samples from 15 healthy individuals using the device and subsequent multi-omics analyses identified significant differences between bacteria, phages, host proteins and metabolites in the intestines versus stool. Certain microbial taxa were differentially enriched and prophage induction was more prevalent in the intestines than in stool. The host proteome and bile acid profiles varied along the intestines and were highly distinct from those of stool. Correlations between gradients in bile acid concentrations and microbial abundance predicted species that altered the bile acid pool through deconjugation. Furthermore, microbially conjugated bile acid concentrations exhibited amino acid-dependent trends that were not apparent in stool. Overall, non-invasive, longitudinal profiling of microorganisms, proteins and bile acids along the intestinal tract under physiological conditions can help elucidate the roles of the gut microbiome and metabolome in human physiology and disease.


Subject(s)
Bile Acids and Salts , Gastrointestinal Microbiome , Intestines , Metabolome , Proteome , Humans , Bile Acids and Salts/metabolism , Gastrointestinal Microbiome/physiology , Proteome/metabolism , Bacteria/classification , Bacteria/isolation & purification , Bacteriophages/isolation & purification , Bacteriophages/physiology , Feces/chemistry , Feces/microbiology , Feces/virology , Intestines/chemistry , Intestines/metabolism , Intestines/microbiology , Intestines/physiology , Intestines/virology , Digestion/physiology
20.
Mol Cell ; 81(11): 2445-2459.e13, 2021 06 03.
Article in English | MEDLINE | ID: mdl-33905682

ABSTRACT

How are E3 ubiquitin ligases configured to match substrate quaternary structures? Here, by studying the yeast GID complex (mutation of which causes deficiency in glucose-induced degradation of gluconeogenic enzymes), we discover supramolecular chelate assembly as an E3 ligase strategy for targeting an oligomeric substrate. Cryoelectron microscopy (cryo-EM) structures show that, to bind the tetrameric substrate fructose-1,6-bisphosphatase (Fbp1), two minimally functional GID E3s assemble into the 20-protein Chelator-GIDSR4, which resembles an organometallic supramolecular chelate. The Chelator-GIDSR4 assembly avidly binds multiple Fbp1 degrons so that multiple Fbp1 protomers are simultaneously ubiquitylated at lysines near the allosteric and substrate binding sites. Importantly, key structural and biochemical features, including capacity for supramolecular assembly, are preserved in the human ortholog, the CTLH E3. Based on our integrative structural, biochemical, and cell biological data, we propose that higher-order E3 ligase assembly generally enables multipronged targeting, capable of simultaneously incapacitating multiple protomers and functionalities of oligomeric substrates.


Subject(s)
Adaptor Proteins, Signal Transducing/chemistry , Cell Adhesion Molecules/chemistry , Fructose-Bisphosphatase/chemistry , Intracellular Signaling Peptides and Proteins/chemistry , Multienzyme Complexes/chemistry , Saccharomyces cerevisiae Proteins/chemistry , Ubiquitin-Conjugating Enzymes/chemistry , Ubiquitin/chemistry , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Binding Sites , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/metabolism , Cryoelectron Microscopy , Fructose-Bisphosphatase/genetics , Fructose-Bisphosphatase/metabolism , Gene Expression , Gluconeogenesis/genetics , Humans , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , K562 Cells , Kinetics , Models, Molecular , Multienzyme Complexes/genetics , Multienzyme Complexes/metabolism , Promoter Regions, Genetic , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Saccharomyces cerevisiae , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Sf9 Cells , Spodoptera , Structural Homology, Protein , Substrate Specificity , Ubiquitin/genetics , Ubiquitin/metabolism , Ubiquitin-Conjugating Enzymes/genetics , Ubiquitin-Conjugating Enzymes/metabolism , Ubiquitination
SELECTION OF CITATIONS
SEARCH DETAIL