Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 332
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Cell ; 185(2): 361-378.e25, 2022 01 20.
Article in English | MEDLINE | ID: mdl-34982960

ABSTRACT

Nuclear pore complexes (NPCs) mediate the nucleocytoplasmic transport of macromolecules. Here we provide a structure of the isolated yeast NPC in which the inner ring is resolved by cryo-EM at sub-nanometer resolution to show how flexible connectors tie together different structural and functional layers. These connectors may be targets for phosphorylation and regulated disassembly in cells with an open mitosis. Moreover, some nucleoporin pairs and transport factors have similar interaction motifs, which suggests an evolutionary and mechanistic link between assembly and transport. We provide evidence for three major NPC variants that may foreshadow functional specializations at the nuclear periphery. Cryo-electron tomography extended these studies, providing a model of the in situ NPC with a radially expanded inner ring. Our comprehensive model reveals features of the nuclear basket and central transporter, suggests a role for the lumenal Pom152 ring in restricting dilation, and highlights structural plasticity that may be required for transport.


Subject(s)
Adaptation, Physiological , Nuclear Pore/metabolism , Saccharomyces cerevisiae/physiology , Amino Acid Motifs , Amino Acid Sequence , Fluorescence , Molecular Docking Simulation , Nuclear Envelope/metabolism , Nuclear Pore/chemistry , Nuclear Pore Complex Proteins/chemistry , Nuclear Pore Complex Proteins/metabolism , Protein Domains , Reproducibility of Results , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/metabolism
2.
Cell ; 177(6): 1384-1403, 2019 05 30.
Article in English | MEDLINE | ID: mdl-31150619

ABSTRACT

Integrative structure determination is a powerful approach to modeling the structures of biological systems based on data produced by multiple experimental and theoretical methods, with implications for our understanding of cellular biology and drug discovery. This Primer introduces the theory and methods of integrative approaches, emphasizing the kinds of data that can be effectively included in developing models and using the nuclear pore complex as an example to illustrate the practice and challenges involved. These guidelines are intended to aid the researcher in understanding and applying integrative structural methods to systems of their interest and thus take advantage of this rapidly evolving field.


Subject(s)
Computational Biology/methods , Systems Biology/methods , Algorithms , Animals , Humans , Models, Molecular , Molecular Biology , Nuclear Pore/physiology , Software , Systems Analysis , Systems Integration
3.
Cell ; 173(1): 11-19, 2018 03 22.
Article in English | MEDLINE | ID: mdl-29570991

ABSTRACT

The construction of a predictive model of an entire eukaryotic cell that describes its dynamic structure from atomic to cellular scales is a grand challenge at the intersection of biology, chemistry, physics, and computer science. Having such a model will open new dimensions in biological research and accelerate healthcare advancements. Developing the necessary experimental and modeling methods presents abundant opportunities for a community effort to realize this goal. Here, we present a vision for creation of a spatiotemporal multi-scale model of the pancreatic ß-cell, a relevant target for understanding and modulating the pathogenesis of diabetes.


Subject(s)
Insulin-Secreting Cells/metabolism , Models, Biological , Computational Biology , Drug Discovery , Humans , Insulin-Secreting Cells/cytology , Proteins/chemistry , Proteins/metabolism
4.
Cell ; 173(3): 693-705.e22, 2018 04 19.
Article in English | MEDLINE | ID: mdl-29677513

ABSTRACT

Liquid-liquid phase separation (LLPS) is believed to underlie formation of biomolecular condensates, cellular compartments that concentrate macromolecules without surrounding membranes. Physical mechanisms that control condensate formation/dissolution are poorly understood. The RNA-binding protein fused in sarcoma (FUS) undergoes LLPS in vitro and associates with condensates in cells. We show that the importin karyopherin-ß2/transportin-1 inhibits LLPS of FUS. This activity depends on tight binding of karyopherin-ß2 to the C-terminal proline-tyrosine nuclear localization signal (PY-NLS) of FUS. Nuclear magnetic resonance (NMR) analyses reveal weak interactions of karyopherin-ß2 with sequence elements and structural domains distributed throughout the entirety of FUS. Biochemical analyses demonstrate that most of these same regions also contribute to LLPS of FUS. The data lead to a model where high-affinity binding of karyopherin-ß2 to the FUS PY-NLS tethers the proteins together, allowing multiple, distributed weak intermolecular contacts to disrupt FUS self-association, blocking LLPS. Karyopherin-ß2 may act analogously to control condensates in diverse cellular contexts.


Subject(s)
Active Transport, Cell Nucleus , Nuclear Localization Signals , RNA-Binding Protein FUS/chemistry , beta Karyopherins/chemistry , Binding Sites , Frontotemporal Lobar Degeneration/metabolism , Humans , Karyopherins/metabolism , Light , Liquid-Liquid Extraction , Macromolecular Substances , Magnetic Resonance Spectroscopy , Mutation , Nephelometry and Turbidimetry , Protein Binding , Protein Domains , RNA/chemistry , Scattering, Radiation , Temperature
5.
Cell ; 167(5): 1215-1228.e25, 2016 11 17.
Article in English | MEDLINE | ID: mdl-27839866

ABSTRACT

The last steps in mRNA export and remodeling are performed by the Nup82 complex, a large conserved assembly at the cytoplasmic face of the nuclear pore complex (NPC). By integrating diverse structural data, we have determined the molecular architecture of the native Nup82 complex at subnanometer precision. The complex consists of two compositionally identical multiprotein subunits that adopt different configurations. The Nup82 complex fits into the NPC through the outer ring Nup84 complex. Our map shows that this entire 14-MDa Nup82-Nup84 complex assembly positions the cytoplasmic mRNA export factor docking sites and messenger ribonucleoprotein (mRNP) remodeling machinery right over the NPC's central channel rather than on distal cytoplasmic filaments, as previously supposed. We suggest that this configuration efficiently captures and remodels exporting mRNP particles immediately upon reaching the cytoplasmic side of the NPC.


Subject(s)
Nuclear Pore Complex Proteins/chemistry , Nuclear Pore/chemistry , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Yeasts/metabolism , Active Transport, Cell Nucleus , Fungal Proteins , Nuclear Pore Complex Proteins/ultrastructure , RNA, Messenger , Saccharomyces cerevisiae/cytology , Saccharomyces cerevisiae Proteins/ultrastructure
6.
Mol Cell ; 83(18): 3283-3302.e5, 2023 09 21.
Article in English | MEDLINE | ID: mdl-37738963

ABSTRACT

Nuclear pore complexes (NPCs) direct the nucleocytoplasmic transport of macromolecules. Here, we provide a composite multiscale structure of the yeast NPC, based on improved 3D density maps from cryogenic electron microscopy and AlphaFold2 models. Key features of the inner and outer rings were integrated into a comprehensive model. We resolved flexible connectors that tie together the core scaffold, along with equatorial transmembrane complexes and a lumenal ring that anchor this channel within the pore membrane. The organization of the nuclear double outer ring reveals an architecture that may be shared with ancestral NPCs. Additional connections between the core scaffold and the central transporter suggest that under certain conditions, a degree of local organization is present at the periphery of the transport machinery. These connectors may couple conformational changes in the scaffold to the central transporter to modulate transport. Collectively, this analysis provides insights into assembly, transport, and NPC evolution.


Subject(s)
Nuclear Pore , Saccharomyces cerevisiae , Saccharomyces cerevisiae/genetics , Membrane Transport Proteins
7.
Nature ; 626(7997): 194-206, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38096902

ABSTRACT

The LINE-1 (L1) retrotransposon is an ancient genetic parasite that has written around one-third of the human genome through a 'copy and paste' mechanism catalysed by its multifunctional enzyme, open reading frame 2 protein (ORF2p)1. ORF2p reverse transcriptase (RT) and endonuclease activities have been implicated in the pathophysiology of cancer2,3, autoimmunity4,5 and ageing6,7, making ORF2p a potential therapeutic target. However, a lack of structural and mechanistic knowledge has hampered efforts to rationally exploit it. We report structures of the human ORF2p 'core' (residues 238-1061, including the RT domain) by X-ray crystallography and cryo-electron microscopy in several conformational states. Our analyses identified two previously undescribed folded domains, extensive contacts to RNA templates and associated adaptations that contribute to unique aspects of the L1 replication cycle. Computed integrative structural models of full-length ORF2p show a dynamic closed-ring conformation that appears to open during retrotransposition. We characterize ORF2p RT inhibition and reveal its underlying structural basis. Imaging and biochemistry show that non-canonical cytosolic ORF2p RT activity can produce RNA:DNA hybrids, activating innate immune signalling through cGAS/STING and resulting in interferon production6-8. In contrast to retroviral RTs, L1 RT is efficiently primed by short RNAs and hairpins, which probably explains cytosolic priming. Other biochemical activities including processivity, DNA-directed polymerization, non-templated base addition and template switching together allow us to propose a revised L1 insertion model. Finally, our evolutionary analysis demonstrates structural conservation between ORF2p and other RNA- and DNA-dependent polymerases. We therefore provide key mechanistic insights into L1 polymerization and insertion, shed light on the evolutionary history of L1 and enable rational drug development targeting L1.


Subject(s)
Endonucleases , Long Interspersed Nucleotide Elements , RNA-Directed DNA Polymerase , Reverse Transcription , Humans , Cryoelectron Microscopy , Endonucleases/chemistry , Endonucleases/genetics , Endonucleases/metabolism , Long Interspersed Nucleotide Elements/genetics , RNA/genetics , RNA-Directed DNA Polymerase/chemistry , RNA-Directed DNA Polymerase/genetics , RNA-Directed DNA Polymerase/metabolism , Crystallography, X-Ray , DNA/biosynthesis , DNA/genetics , Immunity, Innate , Interferons/biosynthesis
8.
Cell ; 158(5): 1123-1135, 2014 08 28.
Article in English | MEDLINE | ID: mdl-25171412

ABSTRACT

Eukaryotic translation initiation requires the recruitment of the large, multiprotein eIF3 complex to the 40S ribosomal subunit. We present X-ray structures of all major components of the minimal, six-subunit Saccharomyces cerevisiae eIF3 core. These structures, together with electron microscopy reconstructions, cross-linking coupled to mass spectrometry, and integrative structure modeling, allowed us to position and orient all eIF3 components on the 40S⋅eIF1 complex, revealing an extended, modular arrangement of eIF3 subunits. Yeast eIF3 engages 40S in a clamp-like manner, fully encircling 40S to position key initiation factors on opposite ends of the mRNA channel, providing a platform for the recruitment, assembly, and regulation of the translation initiation machinery. The structures of eIF3 components reported here also have implications for understanding the architecture of the mammalian 43S preinitiation complex and the complex of eIF3, 40S, and the hepatitis C internal ribosomal entry site RNA.


Subject(s)
Eukaryotic Initiation Factor-1/chemistry , Eukaryotic Initiation Factor-3/chemistry , Peptide Chain Initiation, Translational , Ribosome Subunits, Small, Eukaryotic/chemistry , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae/chemistry , Amino Acid Sequence , Animals , Crystallography, X-Ray , Dimerization , Eukaryotic Initiation Factor-1/metabolism , Eukaryotic Initiation Factor-3/metabolism , Hepacivirus/chemistry , Humans , Mammals/metabolism , Microscopy, Electron , Models, Molecular , Molecular Sequence Data , Ribonucleoproteins/chemistry , Ribosome Subunits, Small, Eukaryotic/metabolism , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Sequence Alignment
9.
Cell ; 158(2): 412-421, 2014 Jul 17.
Article in English | MEDLINE | ID: mdl-25036635

ABSTRACT

Although biosynthetic gene clusters (BGCs) have been discovered for hundreds of bacterial metabolites, our knowledge of their diversity remains limited. Here, we used a novel algorithm to systematically identify BGCs in the extensive extant microbial sequencing data. Network analysis of the predicted BGCs revealed large gene cluster families, the vast majority uncharacterized. We experimentally characterized the most prominent family, consisting of two subfamilies of hundreds of BGCs distributed throughout the Proteobacteria; their products are aryl polyenes, lipids with an aryl head group conjugated to a polyene tail. We identified a distant relationship to a third subfamily of aryl polyene BGCs, and together the three subfamilies represent the largest known family of biosynthetic gene clusters, with more than 1,000 members. Although these clusters are widely divergent in sequence, their small molecule products are remarkably conserved, indicating for the first time the important roles these compounds play in Gram-negative cell biology.


Subject(s)
Algorithms , Bacteria/genetics , Bacteria/metabolism , Bacteria/chemistry , Bacteria/classification , Mutation , Oxidative Stress , Phylogeny , Secondary Metabolism
10.
Nature ; 613(7944): 575-581, 2023 01.
Article in English | MEDLINE | ID: mdl-36599981

ABSTRACT

Understanding how the nuclear pore complex (NPC) is assembled is of fundamental importance to grasp the mechanisms behind its essential function and understand its role during the evolution of eukaryotes1-4. There are at least two NPC assembly pathways-one during the exit from mitosis and one during nuclear growth in interphase-but we currently lack a quantitative map of these events. Here we use fluorescence correlation spectroscopy calibrated live imaging of endogenously fluorescently tagged nucleoporins to map the changes in the composition and stoichiometry of seven major modules of the human NPC during its assembly in single dividing cells. This systematic quantitative map reveals that the two assembly pathways have distinct molecular mechanisms, in which the order of addition of two large structural components, the central ring complex and nuclear filaments are inverted. The dynamic stoichiometry data was integrated to create a spatiotemporal model of the NPC assembly pathway and predict the structures of postmitotic NPC assembly intermediates.


Subject(s)
Nuclear Pore Complex Proteins , Nuclear Pore , Humans , Interphase , Mitosis , Nuclear Pore/chemistry , Nuclear Pore/metabolism , Nuclear Pore Complex Proteins/chemistry , Nuclear Pore Complex Proteins/metabolism , Spectrometry, Fluorescence
11.
Nat Rev Genet ; 23(6): 342-354, 2022 06.
Article in English | MEDLINE | ID: mdl-35013567

ABSTRACT

Understanding the effects of genetic variation is a fundamental problem in biology that requires methods to analyse both physical and functional consequences of sequence changes at systems-wide and mechanistic scales. To achieve a systems view, protein interaction networks map which proteins physically interact, while genetic interaction networks inform on the phenotypic consequences of perturbing these protein interactions. Until recently, understanding the molecular mechanisms that underlie these interactions often required biophysical methods to determine the structures of the proteins involved. The past decade has seen the emergence of new approaches based on coevolution, deep mutational scanning and genome-scale genetic or chemical-genetic interaction mapping that enable modelling of the structures of individual proteins or protein complexes. Here, we review the emerging use of large-scale genetic datasets and deep learning approaches to model protein structures and their interactions, and discuss the integration of structural data from different sources.


Subject(s)
Protein Interaction Maps , Proteins , Epistasis, Genetic , Gene Regulatory Networks , Mutation , Protein Interaction Mapping , Proteins/genetics , Proteins/metabolism
12.
Nucleic Acids Res ; 51(D1): D488-D508, 2023 01 06.
Article in English | MEDLINE | ID: mdl-36420884

ABSTRACT

The Research Collaboratory for Structural Bioinformatics Protein Data Bank (RCSB PDB), founding member of the Worldwide Protein Data Bank (wwPDB), is the US data center for the open-access PDB archive. As wwPDB-designated Archive Keeper, RCSB PDB is also responsible for PDB data security. Annually, RCSB PDB serves >10 000 depositors of three-dimensional (3D) biostructures working on all permanently inhabited continents. RCSB PDB delivers data from its research-focused RCSB.org web portal to many millions of PDB data consumers based in virtually every United Nations-recognized country, territory, etc. This Database Issue contribution describes upgrades to the research-focused RCSB.org web portal that created a one-stop-shop for open access to ∼200 000 experimentally-determined PDB structures of biological macromolecules alongside >1 000 000 incorporated Computed Structure Models (CSMs) predicted using artificial intelligence/machine learning methods. RCSB.org is a 'living data resource.' Every PDB structure and CSM is integrated weekly with related functional annotations from external biodata resources, providing up-to-date information for the entire corpus of 3D biostructure data freely available from RCSB.org with no usage limitations. Within RCSB.org, PDB structures and the CSMs are clearly identified as to their provenance and reliability. Both are fully searchable, and can be analyzed and visualized using the full complement of RCSB.org web portal capabilities.


Subject(s)
Artificial Intelligence , Databases, Protein , Proteins , Machine Learning , Protein Conformation , Proteins/chemistry , Reproducibility of Results
13.
Proc Natl Acad Sci U S A ; 119(32): e2202695119, 2022 08 09.
Article in English | MEDLINE | ID: mdl-35921440

ABSTRACT

Characterizing relationships between Zn2+, insulin, and insulin vesicles is of vital importance to the study of pancreatic beta cells. However, the precise content of Zn2+ and the specific location of insulin inside insulin vesicles are not clear, which hinders a thorough understanding of the insulin secretion process and diseases caused by blood sugar dysregulation. Here, we demonstrated the colocalization of Zn2+ and insulin in both single extracellular insulin vesicles and pancreatic beta cells by using an X-ray scanning coherent diffraction imaging (ptychography) technique. We also analyzed the elemental Zn2+ and Ca2+ contents of insulin vesicles using electron microscopy and energy dispersive spectroscopy (EDS) mapping. We found that the presence of Zn2+ is an important characteristic that can be used to distinguish insulin vesicles from other types of vesicles in pancreatic beta cells and that the content of Zn2+ is proportional to the size of insulin vesicles. By using dual-energy contrast X-ray microscopy and scanning transmission X-ray microscopy (STXM) image stacks, we observed that insulin accumulates in the off-center position of extracellular insulin vesicles. Furthermore, the spatial distribution of insulin vesicles and their colocalization with other organelles inside pancreatic beta cells were demonstrated using three-dimensional (3D) imaging by combining X-ray ptychography and an equally sloped tomography (EST) algorithm. This study describes a powerful method to univocally describe the location and quantitative analysis of intracellular insulin, which will be of great significance to the study of diabetes and other blood sugar diseases.


Subject(s)
Insulin-Secreting Cells , Insulin , Secretory Vesicles , Zinc , Animals , Blood Glucose , Cell Line , Insulin/analysis , Insulin/metabolism , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/ultrastructure , Rats , Secretory Vesicles/chemistry , Secretory Vesicles/metabolism , Spectrometry, X-Ray Emission , X-Ray Diffraction , Zinc/analysis
14.
Proc Natl Acad Sci U S A ; 119(5)2022 02 01.
Article in English | MEDLINE | ID: mdl-35082148

ABSTRACT

Triggering receptor expressed on myeloid cells 2 (TREM2) is a single-pass transmembrane receptor of the immunoglobulin superfamily that is secreted in a soluble (sTREM2) form. Mutations in TREM2 have been linked to increased risk of Alzheimer's disease (AD). A prominent neuropathological component of AD is deposition of the amyloid-ß (Aß) into plaques, particularly Aß40 and Aß42. While the membrane-bound form of TREM2 is known to facilitate uptake of Aß fibrils and the polarization of microglial processes toward amyloid plaques, the role of its soluble ectodomain, particularly in interactions with monomeric or fibrillar Aß, has been less clear. Our results demonstrate that sTREM2 does not bind to monomeric Aß40 and Aß42, even at a high micromolar concentration, while it does bind to fibrillar Aß42 and Aß40 with equal affinities (2.6 ± 0.3 µM and 2.3 ± 0.4 µM). Kinetic analysis shows that sTREM2 inhibits the secondary nucleation step in the fibrillization of Aß, while having little effect on the primary nucleation pathway. Furthermore, binding of sTREM2 to fibrils markedly enhanced uptake of fibrils into human microglial and neuroglioma derived cell lines. The disease-associated sTREM2 mutant, R47H, displayed little to no effect on fibril nucleation and binding, but it decreased uptake and functional responses markedly. We also probed the structure of the WT sTREM2-Aß fibril complex using integrative molecular modeling based primarily on the cross-linking mass spectrometry data. The model shows that sTREM2 binds fibrils along one face of the structure, leaving a second, mutation-sensitive site free to mediate cellular binding and uptake.


Subject(s)
Amyloid beta-Peptides/metabolism , Amyloid/metabolism , Membrane Glycoproteins/metabolism , Receptors, Immunologic/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Amyloid/genetics , Amyloid beta-Peptides/genetics , Animals , Humans , Kinetics , Membrane Glycoproteins/genetics , Mice , Microglia/metabolism , Mutation/genetics , Peptide Fragments/genetics , Peptide Fragments/metabolism , Plaque, Amyloid/genetics , Plaque, Amyloid/metabolism , Receptors, Immunologic/genetics , tau Proteins/genetics , tau Proteins/metabolism
16.
Nature ; 555(7697): 475-482, 2018 03 22.
Article in English | MEDLINE | ID: mdl-29539637

ABSTRACT

Nuclear pore complexes play central roles as gatekeepers of RNA and protein transport between the cytoplasm and nucleoplasm. However, their large size and dynamic nature have impeded a full structural and functional elucidation. Here we determined the structure of the entire 552-protein nuclear pore complex of the yeast Saccharomyces cerevisiae at sub-nanometre precision by satisfying a wide range of data relating to the molecular arrangement of its constituents. The nuclear pore complex incorporates sturdy diagonal columns and connector cables attached to these columns, imbuing the structure with strength and flexibility. These cables also tie together all other elements of the nuclear pore complex, including membrane-interacting regions, outer rings and RNA-processing platforms. Inwardly directed anchors create a high density of transport factor-docking Phe-Gly repeats in the central channel, organized into distinct functional units. This integrative structure enables us to rationalize the architecture, transport mechanism and evolutionary origins of the nuclear pore complex.


Subject(s)
Nuclear Pore Complex Proteins/chemistry , Nuclear Pore Complex Proteins/metabolism , Nuclear Pore/chemistry , Nuclear Pore/metabolism , Saccharomyces cerevisiae/chemistry , Cross-Linking Reagents/chemistry , Mass Spectrometry , Models, Molecular , Protein Stability , Protein Transport , RNA Transport
17.
Proc Natl Acad Sci U S A ; 118(19)2021 05 11.
Article in English | MEDLINE | ID: mdl-33941673

ABSTRACT

Structural maintenance of chromosomes (SMC) complexes are critical chromatin modulators. In eukaryotes, the cohesin and condensin SMC complexes organize chromatin, while the Smc5/6 complex directly regulates DNA replication and repair. The molecular basis for the distinct functions of Smc5/6 is poorly understood. Here, we report an integrative structural study of the budding yeast Smc5/6 holo-complex using electron microscopy, cross-linking mass spectrometry, and computational modeling. We show that the Smc5/6 complex possesses several unique features, while sharing some architectural characteristics with other SMC complexes. In contrast to arm-folded structures of cohesin and condensin, Smc5 and Smc6 arm regions do not fold back on themselves. Instead, these long filamentous regions interact with subunits uniquely acquired by the Smc5/6 complex, namely the Nse2 SUMO ligase and the Nse5/Nse6 subcomplex, with the latter also serving as a linchpin connecting distal parts of the complex. Our 3.0-Å resolution cryoelectron microscopy structure of the Nse5/Nse6 core further reveals a clasped-hand topology and a dimeric interface important for cell growth. Finally, we provide evidence that Nse5/Nse6 uses its SUMO-binding motifs to contribute to Nse2-mediated sumoylation. Collectively, our integrative study identifies distinct structural features of the Smc5/6 complex and functional cooperation among its coevolved unique subunits.


Subject(s)
Cell Cycle Proteins/chemistry , Multiprotein Complexes/chemistry , Protein Domains , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae/metabolism , Binding Sites , Cell Cycle Proteins/metabolism , Chromosomal Proteins, Non-Histone/chemistry , Chromosomal Proteins, Non-Histone/metabolism , Cryoelectron Microscopy/methods , Mass Spectrometry/methods , Models, Molecular , Multiprotein Complexes/metabolism , Multiprotein Complexes/ultrastructure , Protein Binding , Saccharomyces cerevisiae Proteins/metabolism , Sumoylation
18.
Proc Natl Acad Sci U S A ; 118(35)2021 08 31.
Article in English | MEDLINE | ID: mdl-34453000

ABSTRACT

Comprehensive modeling of a whole cell requires an integration of vast amounts of information on various aspects of the cell and its parts. To divide and conquer this task, we introduce Bayesian metamodeling, a general approach to modeling complex systems by integrating a collection of heterogeneous input models. Each input model can in principle be based on any type of data and can describe a different aspect of the modeled system using any mathematical representation, scale, and level of granularity. These input models are 1) converted to a standardized statistical representation relying on probabilistic graphical models, 2) coupled by modeling their mutual relations with the physical world, and 3) finally harmonized with respect to each other. To illustrate Bayesian metamodeling, we provide a proof-of-principle metamodel of glucose-stimulated insulin secretion by human pancreatic ß-cells. The input models include a coarse-grained spatiotemporal simulation of insulin vesicle trafficking, docking, and exocytosis; a molecular network model of glucose-stimulated insulin secretion signaling; a network model of insulin metabolism; a structural model of glucagon-like peptide-1 receptor activation; a linear model of a pancreatic cell population; and ordinary differential equations for systemic postprandial insulin response. Metamodeling benefits from decentralized computing, while often producing a more accurate, precise, and complete model that contextualizes input models as well as resolves conflicting information. We anticipate Bayesian metamodeling will facilitate collaborative science by providing a framework for sharing expertise, resources, data, and models, as exemplified by the Pancreatic ß-Cell Consortium.


Subject(s)
Models, Biological , Bayes Theorem , Computer Simulation , Humans , Linear Models
19.
Cell ; 134(5): 866-76, 2008 Sep 05.
Article in English | MEDLINE | ID: mdl-18722006

ABSTRACT

The nearly 600 proteases in the human genome regulate a diversity of biological processes, including programmed cell death. Comprehensive characterization of protease signaling in complex biological samples is limited by available proteomic methods. We have developed a general approach for global identification of proteolytic cleavage sites using an engineered enzyme to selectively biotinylate free protein N termini for positive enrichment of corresponding N-terminal peptides. Using this method to study apoptosis, we have sequenced 333 caspase-like cleavage sites distributed among 292 protein substrates. These sites are generally not predicted by in vitro caspase substrate specificity but can be used to predict other physiological caspase cleavage sites. Structural bioinformatic studies show that caspase cleavage sites often appear in surface-accessible loops and even occasionally in helical regions. Strikingly, we also find that a disproportionate number of caspase substrates physically interact, suggesting that these dimeric proteases target protein complexes and networks to elicit apoptosis.


Subject(s)
Apoptosis/drug effects , Caspases/metabolism , Proteins/analysis , Proteins/metabolism , Proteomics , Antineoplastic Agents, Phytogenic/pharmacology , Caspases/chemistry , Etoposide/pharmacology , Humans , Jurkat Cells , Protein Binding , Substrate Specificity
20.
Mol Cell ; 59(5): 794-806, 2015 Sep 03.
Article in English | MEDLINE | ID: mdl-26340423

ABSTRACT

TFIIH is essential for both RNA polymerase II transcription and DNA repair, and mutations in TFIIH can result in human disease. Here, we determine the molecular architecture of human and yeast TFIIH by an integrative approach using chemical crosslinking/mass spectrometry (CXMS) data, biochemical analyses, and previously published electron microscopy maps. We identified four new conserved "topological regions" that function as hubs for TFIIH assembly and more than 35 conserved topological features within TFIIH, illuminating a network of interactions involved in TFIIH assembly and regulation of its activities. We show that one of these conserved regions, the p62/Tfb1 Anchor region, directly interacts with the DNA helicase subunit XPD/Rad3 in native TFIIH and is required for the integrity and function of TFIIH. We also reveal the structural basis for defects in patients with xeroderma pigmentosum and trichothiodystrophy, with mutations found at the interface between the p62 Anchor region and the XPD subunit.


Subject(s)
Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/metabolism , Transcription Factor TFIIH/chemistry , Transcription Factor TFIIH/metabolism , Cross-Linking Reagents , DNA Helicases/chemistry , DNA Helicases/genetics , DNA Helicases/metabolism , DNA Repair , Humans , Mass Spectrometry , Models, Molecular , Mutation , Protein Interaction Domains and Motifs , Protein Subunits , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics , Transcription Factor TFIIH/genetics , Transcription Factors, TFII/chemistry , Transcription Factors, TFII/genetics , Transcription Factors, TFII/metabolism , Transcription, Genetic , Xeroderma Pigmentosum/genetics , Xeroderma Pigmentosum/metabolism , Xeroderma Pigmentosum Group D Protein/chemistry , Xeroderma Pigmentosum Group D Protein/genetics , Xeroderma Pigmentosum Group D Protein/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL