Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 506.170
Filter
Add more filters

Publication year range
1.
Annu Rev Biochem ; 91: 423-447, 2022 06 21.
Article in English | MEDLINE | ID: mdl-35363508

ABSTRACT

Biochemistry and molecular biology rely on the recognition of structural complementarity between molecules. Molecular interactions must be both quickly reversible, i.e., tenuous, and specific. How the cell reconciles these conflicting demands is the subject of this article. The problem and its theoretical solution are discussed within the wider theoretical context of the thermodynamics of stochastic processes (stochastic thermodynamics). The solution-an irreversible reaction cycle that decreases internal error at the expense of entropy export into the environment-is shown to be widely employed by biological processes that transmit genetic and regulatory information.


Subject(s)
Kinetics , Stochastic Processes , Thermodynamics
2.
Cell ; 185(4): 690-711.e45, 2022 02 17.
Article in English | MEDLINE | ID: mdl-35108499

ABSTRACT

Single-cell (sc)RNA-seq, together with RNA velocity and metabolic labeling, reveals cellular states and transitions at unprecedented resolution. Fully exploiting these data, however, requires kinetic models capable of unveiling governing regulatory functions. Here, we introduce an analytical framework dynamo (https://github.com/aristoteleo/dynamo-release), which infers absolute RNA velocity, reconstructs continuous vector fields that predict cell fates, employs differential geometry to extract underlying regulations, and ultimately predicts optimal reprogramming paths and perturbation outcomes. We highlight dynamo's power to overcome fundamental limitations of conventional splicing-based RNA velocity analyses to enable accurate velocity estimations on a metabolically labeled human hematopoiesis scRNA-seq dataset. Furthermore, differential geometry analyses reveal mechanisms driving early megakaryocyte appearance and elucidate asymmetrical regulation within the PU.1-GATA1 circuit. Leveraging the least-action-path method, dynamo accurately predicts drivers of numerous hematopoietic transitions. Finally, in silico perturbations predict cell-fate diversions induced by gene perturbations. Dynamo, thus, represents an important step in advancing quantitative and predictive theories of cell-state transitions.


Subject(s)
Single-Cell Analysis , Transcriptome/genetics , Algorithms , Female , Gene Expression Regulation , HL-60 Cells , Hematopoiesis/genetics , Hematopoietic Stem Cells/metabolism , Humans , Kinetics , Models, Biological , RNA, Messenger/metabolism , Staining and Labeling
3.
Cell ; 185(2): 345-360.e28, 2022 01 20.
Article in English | MEDLINE | ID: mdl-35063075

ABSTRACT

We present a whole-cell fully dynamical kinetic model (WCM) of JCVI-syn3A, a minimal cell with a reduced genome of 493 genes that has retained few regulatory proteins or small RNAs. Cryo-electron tomograms provide the cell geometry and ribosome distributions. Time-dependent behaviors of concentrations and reaction fluxes from stochastic-deterministic simulations over a cell cycle reveal how the cell balances demands of its metabolism, genetic information processes, and growth, and offer insight into the principles of life for this minimal cell. The energy economy of each process including active transport of amino acids, nucleosides, and ions is analyzed. WCM reveals how emergent imbalances lead to slowdowns in the rates of transcription and translation. Integration of experimental data is critical in building a kinetic model from which emerges a genome-wide distribution of mRNA half-lives, multiple DNA replication events that can be compared to qPCR results, and the experimentally observed doubling behavior.


Subject(s)
Cells/cytology , Computer Simulation , Adenosine Triphosphate/metabolism , Cell Cycle/genetics , Cell Proliferation/genetics , Cells/metabolism , DNA Replication/genetics , Gene Expression Regulation , Imaging, Three-Dimensional , Kinetics , Lipids/chemistry , Metabolic Networks and Pathways , Metabolome , Molecular Sequence Annotation , Nucleotides/metabolism , Thermodynamics , Time Factors
4.
Cell ; 185(1): 145-157.e13, 2022 01 06.
Article in English | MEDLINE | ID: mdl-34995513

ABSTRACT

Contrary to multicellular organisms that display segmentation during development, communities of unicellular organisms are believed to be devoid of such sophisticated patterning. Unexpectedly, we find that the gene expression underlying the nitrogen stress response of a developing Bacillus subtilis biofilm becomes organized into a ring-like pattern. Mathematical modeling and genetic probing of the underlying circuit indicate that this patterning is generated by a clock and wavefront mechanism, similar to that driving vertebrate somitogenesis. We experimentally validated this hypothesis by showing that predicted nutrient conditions can even lead to multiple concentric rings, resembling segments. We additionally confirmed that this patterning mechanism is driven by cell-autonomous oscillations. Importantly, we show that the clock and wavefront process also spatially patterns sporulation within the biofilm. Together, these findings reveal a biofilm segmentation clock that organizes cellular differentiation in space and time, thereby challenging the paradigm that such patterning mechanisms are exclusive to plant and animal development.


Subject(s)
Bacillus subtilis/growth & development , Bacillus subtilis/genetics , Biofilms/growth & development , Body Patterning/genetics , Bacillus subtilis/metabolism , Gene Expression , Gene Expression Regulation, Developmental , Kinetics , Models, Biological , Nitrogen/metabolism , Signal Transduction/genetics , Somites/growth & development , Spores, Bacterial/growth & development , Stress, Physiological/genetics , Time Factors
5.
Cell ; 184(16): 4115-4136, 2021 08 05.
Article in English | MEDLINE | ID: mdl-34358468

ABSTRACT

Emerging tissue transformation technologies provide an unprecedented opportunity to investigate system-level molecular and anatomical features in situ. Hydrogel-based methods engineer physicochemical tissue properties to render intact organs optically transparent and size and shape adjustable while preserving biomolecules at their physiological locations. When combined with advanced molecular tools, labeling, and imaging techniques, tissue transformation enables three-dimensional (3D) mapping of molecules, cells, and their interrelationships at increasing speeds and resolutions. In this review, we discuss the basic engineering principles of tissue transformation and labeling techniques as well as their broad applications, current challenges, and future potential.


Subject(s)
Hydrogels/chemistry , Tissue Engineering , Animals , Diffusion , Humans , Hydrogels/chemical synthesis , Kinetics , Permeability , Transcriptome/genetics
6.
Cell ; 184(22): 5670-5685.e23, 2021 10 28.
Article in English | MEDLINE | ID: mdl-34637702

ABSTRACT

We describe an approach to study the conformation of individual proteins during single particle tracking (SPT) in living cells. "Binder/tag" is based on incorporation of a 7-mer peptide (the tag) into a protein where its solvent exposure is controlled by protein conformation. Only upon exposure can the peptide specifically interact with a reporter protein (the binder). Thus, simple fluorescence localization reflects protein conformation. Through direct excitation of bright dyes, the trajectory and conformation of individual proteins can be followed. Simple protein engineering provides highly specific biosensors suitable for SPT and FRET. We describe tagSrc, tagFyn, tagSyk, tagFAK, and an orthogonal binder/tag pair. SPT showed slowly diffusing islands of activated Src within Src clusters and dynamics of activation in adhesions. Quantitative analysis and stochastic modeling revealed in vivo Src kinetics. The simplicity of binder/tag can provide access to diverse proteins.


Subject(s)
Biosensing Techniques , Peptides/chemistry , Single Molecule Imaging , Animals , Cell Adhesion , Cell Line , Cell Survival , Embryo, Mammalian/cytology , Enzyme Activation , Fibroblasts/metabolism , Fluorescence Resonance Energy Transfer , Humans , Kinetics , Mice , Nanoparticles/chemistry , Protein Conformation , src-Family Kinases/metabolism
7.
Cell ; 184(21): 5448-5464.e22, 2021 10 14.
Article in English | MEDLINE | ID: mdl-34624221

ABSTRACT

Structural maintenance of chromosomes (SMC) complexes organize genome topology in all kingdoms of life and have been proposed to perform this function by DNA loop extrusion. How this process works is unknown. Here, we have analyzed how loop extrusion is mediated by human cohesin-NIPBL complexes, which enable chromatin folding in interphase cells. We have identified DNA binding sites and large-scale conformational changes that are required for loop extrusion and have determined how these are coordinated. Our results suggest that DNA is translocated by a spontaneous 50 nm-swing of cohesin's hinge, which hands DNA over to the ATPase head of SMC3, where upon binding of ATP, DNA is clamped by NIPBL. During this process, NIPBL "jumps ship" from the hinge toward the SMC3 head and might thereby couple the spontaneous hinge swing to ATP-dependent DNA clamping. These results reveal mechanistic principles of how cohesin-NIPBL and possibly other SMC complexes mediate loop extrusion.


Subject(s)
Cell Cycle Proteins/metabolism , Chromosomal Proteins, Non-Histone/metabolism , DNA/chemistry , Nucleic Acid Conformation , Adenosine Triphosphatases/metabolism , Adenosine Triphosphate/metabolism , Binding Sites , Cell Cycle Proteins/chemistry , DNA/metabolism , Fluorescence Resonance Energy Transfer , HeLa Cells , Humans , Hydrolysis , Kinetics , Microscopy, Atomic Force , Models, Molecular , Nuclear Proteins/metabolism , Protein Conformation , Cohesins
8.
Cell ; 184(7): 1884-1894.e14, 2021 04 01.
Article in English | MEDLINE | ID: mdl-33743210

ABSTRACT

G-protein-coupled receptors (GPCRs) represent a ubiquitous membrane protein family and are important drug targets. Their diverse signaling pathways are driven by complex pharmacology arising from a conformational ensemble rarely captured by structural methods. Here, fluorine nuclear magnetic resonance spectroscopy (19F NMR) is used to delineate key functional states of the adenosine A2A receptor (A2AR) complexed with heterotrimeric G protein (Gαsß1γ2) in a phospholipid membrane milieu. Analysis of A2AR spectra as a function of ligand, G protein, and nucleotide identifies an ensemble represented by inactive states, a G-protein-bound activation intermediate, and distinct nucleotide-free states associated with either partial- or full-agonist-driven activation. The Gßγ subunit is found to be critical in facilitating ligand-dependent allosteric transmission, as shown by 19F NMR, biochemical, and computational studies. The results provide a mechanistic basis for understanding basal signaling, efficacy, precoupling, and allostery in GPCRs.


Subject(s)
Heterotrimeric GTP-Binding Proteins/chemistry , Receptor, Adenosine A2A/chemistry , Allosteric Regulation , Binding Sites , Heterotrimeric GTP-Binding Proteins/genetics , Heterotrimeric GTP-Binding Proteins/metabolism , Humans , Kinetics , Ligands , Lipid Bilayers/chemistry , Lipid Bilayers/metabolism , Magnetic Resonance Spectroscopy , Molecular Dynamics Simulation , Nanostructures/chemistry , Protein Binding , Protein Conformation , Protein Subunits/chemistry , Protein Subunits/genetics , Protein Subunits/metabolism , Receptor, Adenosine A2A/genetics , Receptor, Adenosine A2A/metabolism , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification , Signal Transduction
9.
Cell ; 184(15): 3962-3980.e17, 2021 07 22.
Article in English | MEDLINE | ID: mdl-34171305

ABSTRACT

T cell-mediated immunity plays an important role in controlling SARS-CoV-2 infection, but the repertoire of naturally processed and presented viral epitopes on class I human leukocyte antigen (HLA-I) remains uncharacterized. Here, we report the first HLA-I immunopeptidome of SARS-CoV-2 in two cell lines at different times post infection using mass spectrometry. We found HLA-I peptides derived not only from canonical open reading frames (ORFs) but also from internal out-of-frame ORFs in spike and nucleocapsid not captured by current vaccines. Some peptides from out-of-frame ORFs elicited T cell responses in a humanized mouse model and individuals with COVID-19 that exceeded responses to canonical peptides, including some of the strongest epitopes reported to date. Whole-proteome analysis of infected cells revealed that early expressed viral proteins contribute more to HLA-I presentation and immunogenicity. These biological insights, as well as the discovery of out-of-frame ORF epitopes, will facilitate selection of peptides for immune monitoring and vaccine development.


Subject(s)
Epitopes, T-Lymphocyte/immunology , Histocompatibility Antigens Class I/immunology , Open Reading Frames/genetics , Peptides/immunology , Proteome/immunology , SARS-CoV-2/immunology , A549 Cells , Alleles , Amino Acid Sequence , Animals , Antigen Presentation/immunology , COVID-19/immunology , COVID-19/virology , Female , HEK293 Cells , Humans , Kinetics , Male , Mice , Peptides/chemistry , T-Lymphocytes/immunology
10.
Cell ; 184(9): 2384-2393.e12, 2021 04 29.
Article in English | MEDLINE | ID: mdl-33794143

ABSTRACT

The global spread of SARS-CoV-2/COVID-19 is devastating health systems and economies worldwide. Recombinant or vaccine-induced neutralizing antibodies are used to combat the COVID-19 pandemic. However, the recently emerged SARS-CoV-2 variants B.1.1.7 (UK), B.1.351 (South Africa), and P.1 (Brazil) harbor mutations in the viral spike (S) protein that may alter virus-host cell interactions and confer resistance to inhibitors and antibodies. Here, using pseudoparticles, we show that entry of all variants into human cells is susceptible to blockade by the entry inhibitors soluble ACE2, Camostat, EK-1, and EK-1-C4. In contrast, entry of the B.1.351 and P.1 variant was partially (Casirivimab) or fully (Bamlanivimab) resistant to antibodies used for COVID-19 treatment. Moreover, entry of these variants was less efficiently inhibited by plasma from convalescent COVID-19 patients and sera from BNT162b2-vaccinated individuals. These results suggest that SARS-CoV-2 may escape neutralizing antibody responses, which has important implications for efforts to contain the pandemic.


Subject(s)
Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , SARS-CoV-2/immunology , Animals , COVID-19/immunology , COVID-19/therapy , COVID-19/virology , Cell Line , Drug Resistance, Viral , Humans , Immunization, Passive , Kinetics , Membrane Fusion , Models, Molecular , Neutralization Tests , Serine Endopeptidases/metabolism , Solubility , Spike Glycoprotein, Coronavirus/immunology , Vaccination , Virus Internalization , COVID-19 Serotherapy
11.
Cell ; 184(17): 4480-4494.e15, 2021 08 19.
Article in English | MEDLINE | ID: mdl-34320407

ABSTRACT

In neutrophils, nicotinamide adenine dinucleotide phosphate (NADPH) generated via the pentose phosphate pathway fuels NADPH oxidase NOX2 to produce reactive oxygen species for killing invading pathogens. However, excessive NOX2 activity can exacerbate inflammation, as in acute respiratory distress syndrome (ARDS). Here, we use two unbiased chemical proteomic strategies to show that small-molecule LDC7559, or a more potent designed analog NA-11, inhibits the NOX2-dependent oxidative burst in neutrophils by activating the glycolytic enzyme phosphofructokinase-1 liver type (PFKL) and dampening flux through the pentose phosphate pathway. Accordingly, neutrophils treated with NA-11 had reduced NOX2-dependent outputs, including neutrophil cell death (NETosis) and tissue damage. A high-resolution structure of PFKL confirmed binding of NA-11 to the AMP/ADP allosteric activation site and explained why NA-11 failed to agonize phosphofructokinase-1 platelet type (PFKP) or muscle type (PFKM). Thus, NA-11 represents a tool for selective activation of PFKL, the main phosphofructokinase-1 isoform expressed in immune cells.


Subject(s)
Phagocytosis , Phosphofructokinase-1, Liver Type/metabolism , Respiratory Burst , Adenosine Diphosphate/metabolism , Adenosine Monophosphate/metabolism , Allosteric Regulation/drug effects , Enzyme Activation/drug effects , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Glycolysis/drug effects , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Kinetics , Microbial Viability/drug effects , Models, Molecular , NADPH Oxidases/metabolism , Neutrophils/drug effects , Neutrophils/metabolism , Phagocytosis/drug effects , Phosphate-Binding Proteins/metabolism , Phosphofructokinase-1, Liver Type/antagonists & inhibitors , Phosphofructokinase-1, Liver Type/ultrastructure , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Recombinant Proteins/isolation & purification , Respiratory Burst/drug effects , Tetradecanoylphorbol Acetate/pharmacology
12.
Annu Rev Biochem ; 89: 557-581, 2020 06 20.
Article in English | MEDLINE | ID: mdl-32208767

ABSTRACT

The binding affinity and kinetics of target engagement are fundamental to establishing structure-activity relationships (SARs) for prospective therapeutic agents. Enhancing these binding parameters for operative targets, while minimizing binding to off-target sites, can translate to improved drug efficacy and a widened therapeutic window. Compound activity is typically assessed through modulation of an observed phenotype in cultured cells. Quantifying the corresponding binding properties under common cellular conditions can provide more meaningful interpretation of the cellular SAR analysis. Consequently, methods for assessing drug binding in living cells have advanced and are now integral to medicinal chemistry workflows. In this review, we survey key technological advancements that support quantitative assessments of target occupancy in cultured cells, emphasizing generalizable methodologies able to deliver analytical precision that heretofore required reductionist biochemical approaches.


Subject(s)
Chemistry, Pharmaceutical/methods , Fluorescent Dyes/chemistry , High-Throughput Screening Assays , Molecular Probe Techniques , Molecular Targeted Therapy/methods , Bioluminescence Resonance Energy Transfer Techniques , Cell Survival/drug effects , Cells, Cultured , Genes, Reporter , Humans , Kinetics , Optical Imaging/methods , Small Molecule Libraries/chemical synthesis , Small Molecule Libraries/pharmacology , Structure-Activity Relationship
13.
Annu Rev Biochem ; 89: 443-470, 2020 06 20.
Article in English | MEDLINE | ID: mdl-32569525

ABSTRACT

Manipulation of individual molecules with optical tweezers provides a powerful means of interrogating the structure and folding of proteins. Mechanical force is not only a relevant quantity in cellular protein folding and function, but also a convenient parameter for biophysical folding studies. Optical tweezers offer precise control in the force range relevant for protein folding and unfolding, from which single-molecule kinetic and thermodynamic information about these processes can be extracted. In this review, we describe both physical principles and practical aspects of optical tweezers measurements and discuss recent advances in the use of this technique for the study of protein folding. In particular, we describe the characterization of folding energy landscapes at high resolution, studies of structurally complex multidomain proteins, folding in the presence of chaperones, and the ability to investigate real-time cotranslational folding of a polypeptide.


Subject(s)
Escherichia coli/genetics , Molecular Chaperones/genetics , Optical Tweezers , Protein Biosynthesis , Proteome/chemistry , Ribosomes/genetics , Escherichia coli/metabolism , Humans , Kinetics , Microscopy, Atomic Force , Models, Molecular , Molecular Chaperones/chemistry , Molecular Chaperones/metabolism , Protein Binding , Protein Folding , Protein Interaction Domains and Motifs , Proteome/biosynthesis , Proteome/genetics , Proteostasis/genetics , Ribosomes/metabolism , Ribosomes/ultrastructure , Thermodynamics
14.
Annu Rev Biochem ; 89: 605-636, 2020 06 20.
Article in English | MEDLINE | ID: mdl-32569521

ABSTRACT

ATP-binding cassette (ABC) transporters constitute one of the largest and most ancient protein superfamilies found in all living organisms. They function as molecular machines by coupling ATP binding, hydrolysis, and phosphate release to translocation of diverse substrates across membranes. The substrates range from vitamins, steroids, lipids, and ions to peptides, proteins, polysaccharides, and xenobiotics. ABC transporters undergo substantial conformational changes during substrate translocation. A comprehensive understanding of their inner workings thus requires linking these structural rearrangements to the different functional state transitions. Recent advances in single-particle cryogenic electron microscopy have not only delivered crucial information on the architecture of several medically relevant ABC transporters and their supramolecular assemblies, including the ATP-sensitive potassium channel and the peptide-loading complex, but also made it possible to explore the entire conformational space of these nanomachines under turnover conditions and thereby gain detailed mechanistic insights into their mode of action.


Subject(s)
ATP-Binding Cassette Transporters/chemistry , Adenosine Triphosphate/chemistry , Bacteria/metabolism , Cell Membrane/metabolism , Drug Resistance, Multiple/genetics , Mitochondria/metabolism , ATP-Binding Cassette Transporters/classification , ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/metabolism , Adenosine Triphosphate/metabolism , Bacteria/drug effects , Bacteria/genetics , Binding Sites , Biological Transport , Biomechanical Phenomena , Cell Membrane/drug effects , Humans , Kinetics , Mitochondria/drug effects , Models, Molecular , Protein Binding , Protein Interaction Domains and Motifs , Protein Structure, Secondary , Substrate Specificity , Xenobiotics/metabolism , Xenobiotics/pharmacology
15.
Cell ; 183(7): 1785-1800.e26, 2020 12 23.
Article in English | MEDLINE | ID: mdl-33333025

ABSTRACT

All proteins interact with other cellular components to fulfill their function. While tremendous progress has been made in the identification of protein complexes, their assembly and dynamics remain difficult to characterize. Here, we present a high-throughput strategy to analyze the native assembly kinetics of protein complexes. We apply our approach to characterize the co-assembly for 320 pairs of nucleoporins (NUPs) constituting the ≈50 MDa nuclear pore complex (NPC) in yeast. Some NUPs co-assemble fast via rapid exchange whereas others require lengthy maturation steps. This reveals a hierarchical principle of NPC biogenesis where individual subcomplexes form on a minute timescale and then co-assemble from center to periphery in a ∼1 h-long maturation process. Intriguingly, the NUP Mlp1 stands out as joining very late and associating preferentially with aged NPCs. Our approach is readily applicable beyond the NPC, making it possible to analyze the intracellular dynamics of a variety of multiprotein assemblies.


Subject(s)
Macromolecular Substances/metabolism , Multiprotein Complexes/metabolism , Saccharomyces cerevisiae/metabolism , Staining and Labeling , Biological Assay , Kinetics , Models, Biological , Nuclear Pore/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Time Factors
16.
Cell ; 183(7): 2003-2019.e16, 2020 12 23.
Article in English | MEDLINE | ID: mdl-33308478

ABSTRACT

The ability to record transient cellular events in the DNA or RNA of cells would enable precise, large-scale analysis, selection, and reprogramming of heterogeneous cell populations. Here, we report a molecular technology for stable genetic tagging of cells that exhibit activity-related increases in intracellular calcium concentration (FLiCRE). We used FLiCRE to transcriptionally label activated neural ensembles in the nucleus accumbens of the mouse brain during brief stimulation of aversive inputs. Using single-cell RNA sequencing, we detected FLiCRE transcripts among the endogenous transcriptome, providing simultaneous readout of both cell-type and calcium activation history. We identified a cell type in the nucleus accumbens activated downstream of long-range excitatory projections. Taking advantage of FLiCRE's modular design, we expressed an optogenetic channel selectively in this cell type and showed that direct recruitment of this otherwise genetically inaccessible population elicits behavioral aversion. The specificity and minute resolution of FLiCRE enables molecularly informed characterization, manipulation, and reprogramming of activated cellular ensembles.


Subject(s)
Behavior, Animal , Calcium/metabolism , Corpus Striatum/metabolism , Animals , Female , HEK293 Cells , Humans , Kinetics , Male , Mice, Inbred C57BL , Neurons/metabolism , Optogenetics , Rats , Single-Cell Analysis , Transcriptome/genetics
17.
Cell ; 183(7): 1986-2002.e26, 2020 12 23.
Article in English | MEDLINE | ID: mdl-33333022

ABSTRACT

Serotonin plays a central role in cognition and is the target of most pharmaceuticals for psychiatric disorders. Existing drugs have limited efficacy; creation of improved versions will require better understanding of serotonergic circuitry, which has been hampered by our inability to monitor serotonin release and transport with high spatial and temporal resolution. We developed and applied a binding-pocket redesign strategy, guided by machine learning, to create a high-performance, soluble, fluorescent serotonin sensor (iSeroSnFR), enabling optical detection of millisecond-scale serotonin transients. We demonstrate that iSeroSnFR can be used to detect serotonin release in freely behaving mice during fear conditioning, social interaction, and sleep/wake transitions. We also developed a robust assay of serotonin transporter function and modulation by drugs. We expect that both machine-learning-guided binding-pocket redesign and iSeroSnFR will have broad utility for the development of other sensors and in vitro and in vivo serotonin detection, respectively.


Subject(s)
Directed Molecular Evolution , Machine Learning , Serotonin/metabolism , Algorithms , Amino Acid Sequence , Amygdala/physiology , Animals , Behavior, Animal , Binding Sites , Brain/metabolism , HEK293 Cells , Humans , Kinetics , Linear Models , Mice , Mice, Inbred C57BL , Photons , Protein Binding , Serotonin Plasma Membrane Transport Proteins/metabolism , Sleep/physiology , Wakefulness/physiology
18.
Cell ; 183(3): 752-770.e22, 2020 10 29.
Article in English | MEDLINE | ID: mdl-33125891

ABSTRACT

A greater understanding of hematopoietic stem cell (HSC) regulation is required for dissecting protective versus detrimental immunity to pathogens that cause chronic infections such as Mycobacterium tuberculosis (Mtb). We have shown that systemic administration of Bacille Calmette-Guérin (BCG) or ß-glucan reprograms HSCs in the bone marrow (BM) via a type II interferon (IFN-II) or interleukin-1 (IL1) response, respectively, which confers protective trained immunity against Mtb. Here, we demonstrate that, unlike BCG or ß-glucan, Mtb reprograms HSCs via an IFN-I response that suppresses myelopoiesis and impairs development of protective trained immunity to Mtb. Mechanistically, IFN-I signaling dysregulates iron metabolism, depolarizes mitochondrial membrane potential, and induces cell death specifically in myeloid progenitors. Additionally, activation of the IFN-I/iron axis in HSCs impairs trained immunity to Mtb infection. These results identify an unanticipated immune evasion strategy of Mtb in the BM that controls the magnitude and intrinsic anti-microbial capacity of innate immunity to infection.


Subject(s)
Hematopoietic Stem Cells/microbiology , Immunity , Mycobacterium tuberculosis/physiology , Myelopoiesis , Animals , Bone Marrow Cells/metabolism , Cell Proliferation , Disease Susceptibility , Homeostasis , Interferon Type I/metabolism , Iron/metabolism , Kinetics , Lung/microbiology , Lung/pathology , Macrophages/immunology , Mice, Inbred C57BL , Myeloid Cells/metabolism , Necrosis , Signal Transduction , Transcription, Genetic , Tuberculosis/immunology , Tuberculosis/microbiology , Tuberculosis/pathology
19.
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
20.
Cell ; 183(7): 1848-1866.e26, 2020 12 23.
Article in English | MEDLINE | ID: mdl-33301708

ABSTRACT

Obesity is a major cancer risk factor, but how differences in systemic metabolism change the tumor microenvironment (TME) and impact anti-tumor immunity is not understood. Here, we demonstrate that high-fat diet (HFD)-induced obesity impairs CD8+ T cell function in the murine TME, accelerating tumor growth. We generate a single-cell resolution atlas of cellular metabolism in the TME, detailing how it changes with diet-induced obesity. We find that tumor and CD8+ T cells display distinct metabolic adaptations to obesity. Tumor cells increase fat uptake with HFD, whereas tumor-infiltrating CD8+ T cells do not. These differential adaptations lead to altered fatty acid partitioning in HFD tumors, impairing CD8+ T cell infiltration and function. Blocking metabolic reprogramming by tumor cells in obese mice improves anti-tumor immunity. Analysis of human cancers reveals similar transcriptional changes in CD8+ T cell markers, suggesting interventions that exploit metabolism to improve cancer immunotherapy.


Subject(s)
Immunity , Neoplasms/immunology , Neoplasms/metabolism , Obesity/metabolism , Tumor Microenvironment , Adiposity , Animals , CD8-Positive T-Lymphocytes/immunology , Cell Line, Tumor , Cell Proliferation , Diet, High-Fat , Fatty Acids/metabolism , HEK293 Cells , Humans , Hypoxia-Inducible Factor-Proline Dioxygenases/metabolism , Kinetics , Lymphocytes, Tumor-Infiltrating , Mice, Inbred C57BL , Mice, Knockout , Oxidation-Reduction , Principal Component Analysis , Procollagen-Proline Dioxygenase/metabolism , Proteomics
SELECTION OF CITATIONS
SEARCH DETAIL