Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 11.388
Filter
Add more filters

Publication year range
1.
Cell ; 177(7): 1781-1796.e25, 2019 06 13.
Article in English | MEDLINE | ID: mdl-31104845

ABSTRACT

DNA N6-adenine methylation (6mA) has recently been described in diverse eukaryotes, spanning unicellular organisms to metazoa. Here, we report a DNA 6mA methyltransferase complex in ciliates, termed MTA1c. It consists of two MT-A70 proteins and two homeobox-like DNA-binding proteins and specifically methylates dsDNA. Disruption of the catalytic subunit, MTA1, in the ciliate Oxytricha leads to genome-wide loss of 6mA and abolishment of the consensus ApT dimethylated motif. Mutants fail to complete the sexual cycle, which normally coincides with peak MTA1 expression. We investigate the impact of 6mA on nucleosome occupancy in vitro by reconstructing complete, full-length Oxytricha chromosomes harboring 6mA in native or ectopic positions. We show that 6mA directly disfavors nucleosomes in vitro in a local, quantitative manner, independent of DNA sequence. Furthermore, the chromatin remodeler ACF can overcome this effect. Our study identifies a diverged DNA N6-adenine methyltransferase and defines the role of 6mA in chromatin organization.


Subject(s)
Multienzyme Complexes/metabolism , Nucleosomes/enzymology , Oxytricha/enzymology , Protozoan Proteins/metabolism , Site-Specific DNA-Methyltransferase (Adenine-Specific)/metabolism , Tetrahymena thermophila/enzymology , Multienzyme Complexes/genetics , Nucleosomes/genetics , Oxytricha/genetics , Protozoan Proteins/genetics , Site-Specific DNA-Methyltransferase (Adenine-Specific)/genetics , Tetrahymena thermophila/genetics
2.
Annu Rev Biochem ; 87: 187-216, 2018 06 20.
Article in English | MEDLINE | ID: mdl-29925259

ABSTRACT

How individual enzymes evolved is relatively well understood. However, individual enzymes rarely confer a physiological advantage on their own. Judging by its current state, the emergence of metabolism seemingly demanded the simultaneous emergence of many enzymes. Indeed, how multicomponent interlocked systems, like metabolic pathways, evolved is largely an open question. This complexity can be unlocked if we assume that survival of the fittest applies not only to genes and enzymes but also to the metabolites they produce. This review develops our current knowledge of enzyme evolution into a wider hypothesis of pathway and network evolution. We describe the current models for pathway evolution and offer an integrative metabolite-enzyme coevolution hypothesis. Our hypothesis addresses the origins of new metabolites and of new enzymes and the order of their recruitment. We aim to not only survey established knowledge but also present open questions and potential ways of addressing them.


Subject(s)
Enzymes/genetics , Enzymes/metabolism , Evolution, Molecular , Metabolic Networks and Pathways/genetics , Enzymes/chemistry , Kinetics , Models, Biological , Models, Molecular , Multienzyme Complexes/chemistry , Multienzyme Complexes/genetics , Multienzyme Complexes/metabolism , Phylogeny , Substrate Specificity/genetics
3.
Annu Rev Biochem ; 86: 1-19, 2017 06 20.
Article in English | MEDLINE | ID: mdl-28125288

ABSTRACT

After an undergraduate degree in biology at Harvard, I started graduate school at The Rockefeller Institute for Medical Research in New York City in July 1965. I was attracted to the chemical side of biochemistry and joined Fritz Lipmann's large, hierarchical laboratory to study enzyme mechanisms. That work led to postdoctoral research with Robert Abeles at Brandeis, then a center of what, 30 years later, would be called chemical biology. I spent 15 years on the Massachusetts Institute of Technology faculty, in both the Chemistry and Biology Departments, and then 26 years on the Harvard Medical School Faculty. My research interests have been at the intersection of chemistry, biology, and medicine. One unanticipated major focus has been investigating the chemical logic and enzymatic machinery of natural product biosynthesis, including antibiotics and antitumor agents. In this postgenomic era it is now recognized that there may be from 105 to 106 biosynthetic gene clusters as yet uncharacterized for potential new therapeutic agents.


Subject(s)
Anti-Bacterial Agents/metabolism , Antineoplastic Agents/metabolism , Biochemistry/history , Biological Products/metabolism , Biomedical Research/history , Drug Industry/history , Anti-Bacterial Agents/chemistry , Antineoplastic Agents/chemistry , Biochemistry/trends , Biological Products/chemistry , Biomedical Research/trends , Drug Industry/trends , Gene Expression Regulation , History, 20th Century , History, 21st Century , Humans , Ligases/genetics , Ligases/metabolism , Multienzyme Complexes/genetics , Multienzyme Complexes/metabolism , Vancomycin Resistance/genetics , Workforce
4.
Cell ; 170(6): 1247-1257.e12, 2017 Sep 07.
Article in English | MEDLINE | ID: mdl-28844695

ABSTRACT

The respiratory megacomplex represents the highest-order assembly of respiratory chain complexes, and it allows mitochondria to respond to energy-requiring conditions. To understand its architecture, we examined the human respiratory chain megacomplex-I2III2IV2 (MCI2III2IV2) with 140 subunits and a subset of associated cofactors using cryo-electron microscopy. The MCI2III2IV2 forms a circular structure with the dimeric CIII located in the center, where it is surrounded by two copies each of CI and CIV. Two cytochrome c (Cyt.c) molecules are positioned to accept electrons on the surface of the c1 state CIII dimer. Analyses indicate that CII could insert into the gaps between CI and CIV to form a closed ring, which we termed the electron transport chain supercomplex. The structure not only reveals the precise assignment of individual subunits of human CI and CIII, but also enables future in-depth analysis of the electron transport chain as a whole.


Subject(s)
Electron Transport Chain Complex Proteins/chemistry , Multienzyme Complexes/chemistry , Cryoelectron Microscopy , Electron Transport Chain Complex Proteins/isolation & purification , Electron Transport Chain Complex Proteins/metabolism , Electron Transport Complex I/chemistry , Electron Transport Complex I/isolation & purification , Electron Transport Complex I/metabolism , Electron Transport Complex II/chemistry , Electron Transport Complex II/isolation & purification , Electron Transport Complex II/metabolism , Humans , Mitochondria/chemistry , Mitochondria/metabolism , Models, Molecular , Multienzyme Complexes/isolation & purification , Multienzyme Complexes/metabolism
5.
Nature ; 627(8005): 890-897, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38448592

ABSTRACT

In eukaryotes, DNA compacts into chromatin through nucleosomes1,2. Replication of the eukaryotic genome must be coupled to the transmission of the epigenome encoded in the chromatin3,4. Here we report cryo-electron microscopy structures of yeast (Saccharomyces cerevisiae) replisomes associated with the FACT (facilitates chromatin transactions) complex (comprising Spt16 and Pob3) and an evicted histone hexamer. In these structures, FACT is positioned at the front end of the replisome by engaging with the parental DNA duplex to capture the histones through the middle domain and the acidic carboxyl-terminal domain of Spt16. The H2A-H2B dimer chaperoned by the carboxyl-terminal domain of Spt16 is stably tethered to the H3-H4 tetramer, while the vacant H2A-H2B site is occupied by the histone-binding domain of Mcm2. The Mcm2 histone-binding domain wraps around the DNA-binding surface of one H3-H4 dimer and extends across the tetramerization interface of the H3-H4 tetramer to the binding site of Spt16 middle domain before becoming disordered. This arrangement leaves the remaining DNA-binding surface of the other H3-H4 dimer exposed to additional interactions for further processing. The Mcm2 histone-binding domain and its downstream linker region are nested on top of Tof1, relocating the parental histones to the replisome front for transfer to the newly synthesized lagging-strand DNA. Our findings offer crucial structural insights into the mechanism of replication-coupled histone recycling for maintaining epigenetic inheritance.


Subject(s)
Chromatin , DNA Replication , Epistasis, Genetic , Histones , Saccharomyces cerevisiae , Binding Sites , Chromatin/chemistry , Chromatin/genetics , Chromatin/metabolism , Chromatin/ultrastructure , Cryoelectron Microscopy , DNA Replication/genetics , DNA, Fungal/biosynthesis , DNA, Fungal/chemistry , DNA, Fungal/metabolism , DNA, Fungal/ultrastructure , Epistasis, Genetic/genetics , Histones/chemistry , Histones/metabolism , Histones/ultrastructure , Multienzyme Complexes/chemistry , Multienzyme Complexes/metabolism , Multienzyme Complexes/ultrastructure , Nucleosomes/chemistry , Nucleosomes/metabolism , Nucleosomes/ultrastructure , Protein Binding , Protein Domains , Protein Multimerization , Saccharomyces cerevisiae/cytology , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae/ultrastructure , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae Proteins/ultrastructure
6.
Mol Cell ; 81(18): 3775-3785, 2021 09 16.
Article in English | MEDLINE | ID: mdl-34547238

ABSTRACT

With the elucidation of myriad anabolic and catabolic enzyme-catalyzed cellular pathways crisscrossing each other, an obvious question arose: how could these networks operate with maximal catalytic efficiency and minimal interference? A logical answer was the postulate of metabolic channeling, which in its simplest embodiment assumes that the product generated by one enzyme passes directly to a second without diffusion into the surrounding medium. This tight coupling of activities might increase a pathway's metabolic flux and/or serve to sequester unstable/toxic/reactive intermediates as well as prevent their access to other networks. Here, we present evidence for this concept, commencing with enzymes that feature a physical molecular tunnel, to multi-enzyme complexes that retain pathway substrates through electrostatics or enclosures, and finally to metabolons that feature collections of enzymes assembled into clusters with variable stoichiometric composition. Lastly, we discuss the advantages of reversibly assembled metabolons in the context of the purinosome, the purine biosynthesis metabolon.


Subject(s)
Metabolic Networks and Pathways/physiology , Metabolism/physiology , Metabolome/physiology , Animals , Humans , Multienzyme Complexes/metabolism , Protein Interaction Maps/physiology , Purines/metabolism
7.
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
8.
Cell ; 154(3): 556-68, 2013 Aug 01.
Article in English | MEDLINE | ID: mdl-23911321

ABSTRACT

Skp2 E3 ligase is overexpressed in numerous human cancers and plays a critical role in cell-cycle progression, senescence, metabolism, cancer progression, and metastasis. In the present study, we identified a specific Skp2 inhibitor using high-throughput in silico screening of large and diverse chemical libraries. This Skp2 inhibitor selectively suppresses Skp2 E3 ligase activity, but not activity of other SCF complexes. It also phenocopies the effects observed upon genetic Skp2 deficiency, such as suppressing survival and Akt-mediated glycolysis and triggering p53-independent cellular senescence. Strikingly, we discovered a critical function of Skp2 in positively regulating cancer stem cell populations and self-renewal ability through genetic and pharmacological approaches. Notably, Skp2 inhibitor exhibits potent antitumor activities in multiple animal models and cooperates with chemotherapeutic agents to reduce cancer cell survival. Our study thus provides pharmacological evidence that Skp2 is a promising target for restricting cancer stem cell and cancer progression.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Discovery , Neoplasms/enzymology , Neoplastic Stem Cells/drug effects , S-Phase Kinase-Associated Proteins/antagonists & inhibitors , Ubiquitin-Protein Ligases/antagonists & inhibitors , Animals , Antineoplastic Agents/chemistry , Disease Models, Animal , Drug Screening Assays, Antitumor , Genes, p53 , Glycolysis/drug effects , Humans , Mice , Mice, Nude , Models, Molecular , Multienzyme Complexes/antagonists & inhibitors , Multienzyme Complexes/chemistry , Multienzyme Complexes/metabolism , Neoplasm Transplantation , Neoplasms/drug therapy , Neoplasms/genetics , Neoplastic Stem Cells/metabolism , S-Phase Kinase-Associated Proteins/chemistry , S-Phase Kinase-Associated Proteins/metabolism , Small Molecule Libraries , Structure-Activity Relationship , Transplantation, Heterologous , Ubiquitin-Protein Ligases/chemistry , Ubiquitin-Protein Ligases/metabolism
9.
Nature ; 606(7912): 197-203, 2022 06.
Article in English | MEDLINE | ID: mdl-35585235

ABSTRACT

Eukaryotic genomes are compacted into loops and topologically associating domains (TADs)1-3, which contribute to transcription, recombination and genomic stability4,5. Cohesin extrudes DNA into loops that are thought to lengthen until CTCF boundaries are encountered6-12. Little is known about whether loop extrusion is impeded by DNA-bound machines. Here we show that the minichromosome maintenance (MCM) complex is a barrier that restricts loop extrusion in G1 phase. Single-nucleus Hi-C (high-resolution chromosome conformation capture) of mouse zygotes reveals that MCM loading reduces CTCF-anchored loops and decreases TAD boundary insulation, which suggests that loop extrusion is impeded before reaching CTCF. This effect extends to HCT116 cells, in which MCMs affect the number of CTCF-anchored loops and gene expression. Simulations suggest that MCMs are abundant, randomly positioned and partially permeable barriers. Single-molecule imaging shows that MCMs are physical barriers that frequently constrain cohesin translocation in vitro. Notably, chimeric yeast MCMs that contain a cohesin-interaction motif from human MCM3 induce cohesin pausing, indicating that MCMs are 'active' barriers with binding sites. These findings raise the possibility that cohesin can arrive by loop extrusion at MCMs, which determine the genomic sites at which sister chromatid cohesion is established. On the basis of in vivo, in silico and in vitro data, we conclude that distinct loop extrusion barriers shape the three-dimensional genome.


Subject(s)
Cell Cycle Proteins , Chromosomal Proteins, Non-Histone , DNA , Minichromosome Maintenance Proteins , Animals , CCCTC-Binding Factor/metabolism , Cell Cycle Proteins/metabolism , Chromatids/chemistry , Chromatids/metabolism , Chromosomal Proteins, Non-Histone/metabolism , DNA/chemistry , DNA/metabolism , G1 Phase , HCT116 Cells , Humans , Mice , Minichromosome Maintenance Complex Component 3/chemistry , Minichromosome Maintenance Complex Component 3/metabolism , Minichromosome Maintenance Proteins/chemistry , Minichromosome Maintenance Proteins/metabolism , Multienzyme Complexes/chemistry , Multienzyme Complexes/metabolism , Nucleic Acid Conformation , Saccharomyces cerevisiae , Saccharomyces cerevisiae Proteins/metabolism , Cohesins
10.
Nature ; 611(7935): 399-404, 2022 Nov.
Article in English | MEDLINE | ID: mdl-36289347

ABSTRACT

The SEA complex (SEAC) is a growth regulator that acts as a GTPase-activating protein (GAP) towards Gtr1, a Rag GTPase that relays nutrient status to the Target of Rapamycin Complex 1 (TORC1) in yeast1. Functionally, the SEAC has been divided into two subcomplexes: SEACIT, which has GAP activity and inhibits TORC1, and SEACAT, which regulates SEACIT2. This system is conserved in mammals: the GATOR complex, consisting of GATOR1 (SEACIT) and GATOR2 (SEACAT), transmits amino acid3 and glucose4 signals to mTORC1. Despite its importance, the structure of SEAC/GATOR, and thus molecular understanding of its function, is lacking. Here, we solve the cryo-EM structure of the native eight-subunit SEAC. The SEAC has a modular structure in which a COPII-like cage corresponding to SEACAT binds two flexible wings, which correspond to SEACIT. The wings are tethered to the core via Sea3, which forms part of both modules. The GAP mechanism of GATOR1 is conserved in SEACIT, and GAP activity is unaffected by SEACAT in vitro. In vivo, the wings are essential for recruitment of the SEAC to the vacuole, primarily via the EGO complex. Our results indicate that rather than being a direct inhibitor of SEACIT, SEACAT acts as a scaffold for the binding of TORC1 regulators.


Subject(s)
Cryoelectron Microscopy , GTPase-Activating Proteins , Multienzyme Complexes , Animals , GTP Phosphohydrolases/chemistry , GTP Phosphohydrolases/metabolism , GTP Phosphohydrolases/ultrastructure , GTPase-Activating Proteins/chemistry , GTPase-Activating Proteins/metabolism , GTPase-Activating Proteins/ultrastructure , Mammals , Mechanistic Target of Rapamycin Complex 1/metabolism , Multienzyme Complexes/chemistry , Multienzyme Complexes/metabolism , Multienzyme Complexes/ultrastructure , Saccharomyces cerevisiae/chemistry , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae Proteins/ultrastructure , Protein Subunits/chemistry , Protein Subunits/metabolism , Amino Acids , Glucose , COP-Coated Vesicles/chemistry , COP-Coated Vesicles/metabolism
11.
Nature ; 609(7927): 630-639, 2022 09.
Article in English | MEDLINE | ID: mdl-36002576

ABSTRACT

The Holliday junction is a key intermediate formed during DNA recombination across all kingdoms of life1. In bacteria, the Holliday junction is processed by two homo-hexameric AAA+ ATPase RuvB motors, which assemble together with the RuvA-Holliday junction complex to energize the strand-exchange reaction2. Despite its importance for chromosome maintenance, the structure and mechanism by which this complex facilitates branch migration are unknown. Here, using time-resolved cryo-electron microscopy, we obtained structures of the ATP-hydrolysing RuvAB complex in seven distinct conformational states, captured during assembly and processing of a Holliday junction. Five structures together resolve the complete nucleotide cycle and reveal the spatiotemporal relationship between ATP hydrolysis, nucleotide exchange and context-specific conformational changes in RuvB. Coordinated motions in a converter formed by DNA-disengaged RuvB subunits stimulate hydrolysis and nucleotide exchange. Immobilization of the converter enables RuvB to convert the ATP-contained energy into a lever motion, which generates the pulling force driving the branch migration. We show that RuvB motors rotate together with the DNA substrate, which, together with a progressing nucleotide cycle, forms the mechanistic basis for DNA recombination by continuous branch migration. Together, our data decipher the molecular principles of homologous recombination by the RuvAB complex, elucidate discrete and sequential transition-state intermediates for chemo-mechanical coupling of hexameric AAA+ motors and provide a blueprint for the design of state-specific compounds targeting AAA+ motors.


Subject(s)
ATPases Associated with Diverse Cellular Activities , Bacterial Proteins , DNA Helicases , DNA, Cruciform , ATPases Associated with Diverse Cellular Activities/chemistry , ATPases Associated with Diverse Cellular Activities/metabolism , ATPases Associated with Diverse Cellular Activities/ultrastructure , Adenosine Triphosphate/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Bacterial Proteins/ultrastructure , Cryoelectron Microscopy , DNA Helicases/chemistry , DNA Helicases/metabolism , DNA Helicases/ultrastructure , DNA, Cruciform/chemistry , DNA, Cruciform/metabolism , DNA, Cruciform/ultrastructure , DNA, Single-Stranded/chemistry , DNA, Single-Stranded/metabolism , DNA, Single-Stranded/ultrastructure , Homologous Recombination , Hydrolysis , Multienzyme Complexes/chemistry , Multienzyme Complexes/metabolism , Multienzyme Complexes/ultrastructure , Nucleotides , Protein Conformation , Rotation
12.
Mol Cell ; 80(1): 114-126.e8, 2020 10 01.
Article in English | MEDLINE | ID: mdl-32916094

ABSTRACT

DNA replication is carried out by a multi-protein machine called the replisome. In Saccharomyces cerevisiae, the replisome is composed of over 30 different proteins arranged into multiple subassemblies, each performing distinct activities. Synchrony of these activities is required for efficient replication and preservation of genomic integrity. How this is achieved is particularly puzzling at the lagging strand, where current models of the replisome architecture propose turnover of the canonical lagging strand polymerase, Pol δ, at every cycle of Okazaki fragment synthesis. Here, we established single-molecule fluorescence microscopy protocols to study the binding kinetics of individual replisome subunits in live S. cerevisiae. Our results show long residence times for most subunits at the active replisome, supporting a model where all subassemblies bind tightly and work in a coordinated manner for extended periods, including Pol δ, redefining the architecture of the active eukaryotic replisome.


Subject(s)
DNA Replication , DNA-Directed DNA Polymerase/metabolism , Eukaryotic Cells/metabolism , Multienzyme Complexes/metabolism , Cell Nucleus/metabolism , Kinetics , Models, Biological , Nuclear Proteins/metabolism , Protein Subunits/metabolism , Reproducibility of Results , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Single Molecule Imaging , Time Factors
13.
Mol Cell ; 77(3): 600-617.e4, 2020 02 06.
Article in English | MEDLINE | ID: mdl-31952989

ABSTRACT

Brown adipose tissue (BAT) is highly metabolically active tissue that dissipates energy via UCP1 as heat, and BAT mass is correlated negatively with obesity. The presence of BAT/BAT-like tissue in humans renders BAT as an attractive target against obesity and insulin resistance. Here, we identify Aifm2, a NADH oxidoreductase domain containing flavoprotein, as a lipid droplet (LD)-associated protein highly enriched in BAT. Aifm2 is induced by cold as well as by diet. Upon cold or ß-adrenergic stimulation, Aifm2 associates with the outer side of the mitochondrial inner membrane. As a unique BAT-specific first mammalian NDE (external NADH dehydrogenase)-like enzyme, Aifm2 oxidizes NADH to maintain high cytosolic NAD levels in supporting robust glycolysis and to transfer electrons to the electron transport chain (ETC) for fueling thermogenesis. Aifm2 in BAT and subcutaneous white adipose tissue (WAT) promotes oxygen consumption, uncoupled respiration, and heat production during cold- and diet-induced thermogenesis. Aifm2, thus, can ameliorate diet-induced obesity and insulin resistance.


Subject(s)
Adipose Tissue, Brown/metabolism , Apoptosis Regulatory Proteins/metabolism , Mitochondrial Proteins/metabolism , Thermogenesis/physiology , Adipose Tissue, White/metabolism , Animals , Apoptosis Regulatory Proteins/physiology , Diet , Energy Metabolism , Glucose/metabolism , Glycolysis/physiology , HEK293 Cells , Humans , Insulin Resistance , Lipid Droplets/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout , Mitochondrial Membranes/metabolism , Mitochondrial Proteins/physiology , Multienzyme Complexes/metabolism , NAD/metabolism , NAD/physiology , NADH, NADPH Oxidoreductases/metabolism , Obesity/metabolism , Oxidation-Reduction , Oxygen Consumption , Uncoupling Protein 1/metabolism
14.
Mol Cell ; 78(4): 670-682.e8, 2020 05 21.
Article in English | MEDLINE | ID: mdl-32343944

ABSTRACT

Biomolecular condensates play a key role in organizing RNAs and proteins into membraneless organelles. Bacterial RNP-bodies (BR-bodies) are a biomolecular condensate containing the RNA degradosome mRNA decay machinery, but the biochemical function of such organization remains poorly defined. Here, we define the RNA substrates of BR-bodies through enrichment of the bodies followed by RNA sequencing (RNA-seq). We find that long, poorly translated mRNAs, small RNAs, and antisense RNAs are the main substrates, while rRNA, tRNA, and other conserved non-coding RNAs (ncRNAs) are excluded from these bodies. BR-bodies stimulate the mRNA decay rate of enriched mRNAs, helping to reshape the cellular mRNA pool. We also observe that BR-body formation promotes complete mRNA decay, avoiding the buildup of toxic endo-cleaved mRNA decay intermediates. The combined selective permeability of BR-bodies for both enzymes and substrates together with the stimulation of the sub-steps of mRNA decay provide an effective organization strategy for bacterial mRNA decay.


Subject(s)
Caulobacter crescentus/metabolism , Endoribonucleases/metabolism , Escherichia coli/metabolism , Multienzyme Complexes/metabolism , Organelles/metabolism , Polyribonucleotide Nucleotidyltransferase/metabolism , RNA Helicases/metabolism , RNA Stability , RNA, Messenger/metabolism , Caulobacter crescentus/genetics , Caulobacter crescentus/growth & development , Endoribonucleases/genetics , Escherichia coli/genetics , Escherichia coli/growth & development , Humans , Multienzyme Complexes/genetics , Organelles/genetics , Polyribonucleotide Nucleotidyltransferase/genetics , RNA Helicases/genetics , RNA, Antisense/genetics , RNA, Antisense/metabolism , RNA, Messenger/genetics , RNA, Ribosomal/genetics , RNA, Ribosomal/metabolism , RNA, Small Untranslated/genetics , RNA, Small Untranslated/metabolism , RNA, Transfer/genetics , RNA, Transfer/metabolism , RNA, Untranslated/genetics , RNA, Untranslated/metabolism
15.
Annu Rev Microbiol ; 76: 533-552, 2022 09 08.
Article in English | MEDLINE | ID: mdl-35671533

ABSTRACT

RNA degradosomes are multienzyme complexes composed of ribonucleases, RNA helicases, and metabolic enzymes. RNase E-based degradosomes are widespread in Proteobacteria. The Escherichia coli RNA degradosome is sequestered from transcription in the nucleoid and translation in the cytoplasm by localization to the inner cytoplasmic membrane, where it forms short-lived clusters that are proposed to be sites of mRNA degradation. In Caulobacter crescentus, RNA degradosomes localize to ribonucleoprotein condensates in the interior of the cell [bacterial ribonucleoprotein-bodies (BR-bodies)], which have been proposed to drive the concerted degradation of mRNA to nucleotides. The turnover of mRNA in growing cells is important for maintaining pools of nucleotides for transcription and DNA replication.Membrane attachment of the E. coli RNA degradosome is necessary to avoid wasteful degradation of intermediates in ribosome assembly. Sequestering RNA degradosomes to C. crescentus BR-bodies, which exclude structured RNA, could have a similar role in protecting intermediates in ribosome assembly from degradation.


Subject(s)
Caulobacter crescentus , Endoribonucleases , Escherichia coli , Multienzyme Complexes , Nucleotides , Polyribonucleotide Nucleotidyltransferase , RNA Helicases , RNA Stability , RNA, Messenger , Caulobacter crescentus/enzymology , Caulobacter crescentus/genetics , Endoribonucleases/metabolism , Escherichia coli/enzymology , Escherichia coli/genetics , Multienzyme Complexes/genetics , Multienzyme Complexes/metabolism , Nucleotides/metabolism , Polyribonucleotide Nucleotidyltransferase/genetics , Polyribonucleotide Nucleotidyltransferase/metabolism , RNA Helicases/genetics , RNA Helicases/metabolism , RNA, Bacterial/genetics , RNA, Bacterial/metabolism , RNA, Messenger/metabolism , Ribonucleoproteins/metabolism
16.
Nature ; 598(7880): 364-367, 2021 10.
Article in English | MEDLINE | ID: mdl-34616041

ABSTRACT

The enzymes of the mitochondrial electron transport chain are key players of cell metabolism. Despite being active when isolated, in vivo they associate into supercomplexes1, whose precise role is debated. Supercomplexes CIII2CIV1-2 (refs. 2,3), CICIII2 (ref. 4) and CICIII2CIV (respirasome)5-10 exist in mammals, but in contrast to CICIII2 and the respirasome, to date the only known eukaryotic structures of CIII2CIV1-2 come from Saccharomyces cerevisiae11,12 and plants13, which have different organization. Here we present the first, to our knowledge, structures of mammalian (mouse and ovine) CIII2CIV and its assembly intermediates, in different conformations. We describe the assembly of CIII2CIV from the CIII2 precursor to the final CIII2CIV conformation, driven by the insertion of the N terminus of the assembly factor SCAF1 (ref. 14) deep into CIII2, while its C terminus is integrated into CIV. Our structures (which include CICIII2 and the respirasome) also confirm that SCAF1 is exclusively required for the assembly of CIII2CIV and has no role in the assembly of the respirasome. We show that CIII2 is asymmetric due to the presence of only one copy of subunit 9, which straddles both monomers and prevents the attachment of a second copy of SCAF1 to CIII2, explaining the presence of one copy of CIV in CIII2CIV in mammals. Finally, we show that CIII2 and CIV gain catalytic advantage when assembled into the supercomplex and propose a role for CIII2CIV in fine tuning the efficiency of electron transfer in the electron transport chain.


Subject(s)
Cell Respiration , Mitochondria/enzymology , Multienzyme Complexes/chemistry , Multienzyme Complexes/metabolism , Sheep , Animals , Binding Sites , Datasets as Topic , Electron Transport , Mice , Mitochondria/metabolism , Models, Molecular , NAD/metabolism , Succinic Acid/metabolism
17.
Mol Cell ; 73(4): 749-762.e5, 2019 02 21.
Article in English | MEDLINE | ID: mdl-30661981

ABSTRACT

The introduction of azole heterocycles into a peptide backbone is the principal step in the biosynthesis of numerous compounds with therapeutic potential. One of them is microcin B17, a bacterial topoisomerase inhibitor whose activity depends on the conversion of selected serine and cysteine residues of the precursor peptide to oxazoles and thiazoles by the McbBCD synthetase complex. Crystal structures of McbBCD reveal an octameric B4C2D2 complex with two bound substrate peptides. Each McbB dimer clamps the N-terminal recognition sequence, while the C-terminal heterocycle of the modified peptide is trapped in the active site of McbC. The McbD and McbC active sites are distant from each other, which necessitates alternate shuttling of the peptide substrate between them, while remaining tethered to the McbB dimer. An atomic-level view of the azole synthetase is a starting point for deeper understanding and control of biosynthesis of a large group of ribosomally synthesized natural products.


Subject(s)
Anti-Bacterial Agents/biosynthesis , Bacterial Proteins/metabolism , Bacteriocins/biosynthesis , Escherichia coli Proteins/metabolism , Escherichia coli/enzymology , Multienzyme Complexes/metabolism , Ribosomes/enzymology , Topoisomerase II Inhibitors/metabolism , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacteriocins/chemistry , Bacteriocins/pharmacology , Binding Sites , Crystallography, X-Ray , Escherichia coli/drug effects , Escherichia coli/genetics , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/genetics , Models, Molecular , Multienzyme Complexes/chemistry , Multienzyme Complexes/genetics , Mutation , Protein Binding , Protein Interaction Domains and Motifs , Protein Multimerization , Protein Structure, Quaternary , Ribosomes/drug effects , Ribosomes/genetics , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Structure-Activity Relationship , Topoisomerase II Inhibitors/chemistry , Topoisomerase II Inhibitors/pharmacology , X-Ray Diffraction
18.
PLoS Biol ; 21(1): e3001942, 2023 01.
Article in English | MEDLINE | ID: mdl-36603027

ABSTRACT

RNA processing and degradation shape the transcriptome by generating stable molecules that are necessary for translation (rRNA and tRNA) and by facilitating the turnover of mRNA, which is necessary for the posttranscriptional control of gene expression. In bacteria and the plant chloroplast, RNA degradosomes are multienzyme complexes that process and degrade RNA. In many bacterial species, the endoribonuclease RNase E is the central component of the RNA degradosome. RNase E-based RNA degradosomes are inner membrane proteins in a large family of gram-negative bacteria (ß- and γ-Proteobacteria). Until now, the reason for membrane localization was not understood. Here, we show that a mutant strain of Escherichia coli, in which the RNA degradosome is localized to the interior of the cell, has high levels of 20S and 40S particles that are defective intermediates in ribosome assembly. These particles have aberrant protein composition and contain rRNA precursors that have been cleaved by RNase E. After RNase E cleavage, rRNA fragments are degraded to nucleotides by exoribonucleases. In vitro, rRNA in intact ribosomes is resistant to RNase E cleavage, whereas protein-free rRNA is readily degraded. We conclude that RNA degradosomes in the nucleoid of the mutant strain interfere with cotranscriptional ribosome assembly. We propose that membrane-attached RNA degradosomes in wild-type cells control the quality of ribosome assembly after intermediates are released from the nucleoid. That is, the compact structure of mature ribosomes protects rRNA against cleavage by RNase E. Turnover of a proportion of intermediates in ribosome assembly explains slow growth of the mutant strain. Competition between mRNA and rRNA degradation could be the cause of slower mRNA degradation in the mutant strain. We conclude that attachment of the RNA degradosome to the bacterial inner cytoplasmic membrane prevents wasteful degradation of rRNA precursors, thus explaining the reason for conservation of membrane-attached RNA degradosomes throughout the ß- and γ-Proteobacteria.


Subject(s)
Escherichia coli Proteins , RNA, Ribosomal , RNA, Ribosomal/genetics , RNA, Ribosomal/metabolism , Endoribonucleases/genetics , Endoribonucleases/metabolism , Ribosomes/metabolism , Multienzyme Complexes/metabolism , RNA/metabolism , Escherichia coli/metabolism , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Cell Membrane/metabolism , Bacteria/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Bacterial/genetics
19.
Cell ; 144(3): 353-63, 2011 Feb 04.
Article in English | MEDLINE | ID: mdl-21255825

ABSTRACT

Activation-induced cytidine deaminase (AID) initiates immunoglobulin (Ig) heavy-chain (IgH) class switch recombination (CSR) and Ig variable region somatic hypermutation (SHM) in B lymphocytes by deaminating cytidines on template and nontemplate strands of transcribed DNA substrates. However, the mechanism of AID access to the template DNA strand, particularly when hybridized to a nascent RNA transcript, has been an enigma. We now implicate the RNA exosome, a cellular RNA-processing/degradation complex, in targeting AID to both DNA strands. In B lineage cells activated for CSR, the RNA exosome associates with AID, accumulates on IgH switch regions in an AID-dependent fashion, and is required for optimal CSR. Moreover, both the cellular RNA exosome complex and a recombinant RNA exosome core complex impart robust AID- and transcription-dependent DNA deamination of both strands of transcribed SHM substrates in vitro. Our findings reveal a role for noncoding RNA surveillance machinery in generating antibody diversity.


Subject(s)
B-Lymphocytes/metabolism , Cytidine Deaminase/metabolism , Exoribonucleases/metabolism , Immunoglobulin Class Switching , Immunoglobulin Heavy Chains/genetics , Multienzyme Complexes/metabolism , RNA/metabolism , Animals , B-Lymphocytes/cytology , B-Lymphocytes/enzymology , Cell Line , Cells, Cultured , Humans , Mice , Transcription, Genetic
20.
Nature ; 583(7814): 122-126, 2020 07.
Article in English | MEDLINE | ID: mdl-32461692

ABSTRACT

The cellular NADH/NAD+ ratio is fundamental to biochemistry, but the extent to which it reflects versus drives metabolic physiology in vivo is poorly understood. Here we report the in vivo application of Lactobacillus brevis (Lb)NOX1, a bacterial water-forming NADH oxidase, to assess the metabolic consequences of directly lowering the hepatic cytosolic NADH/NAD+ ratio in mice. By combining this genetic tool with metabolomics, we identify circulating α-hydroxybutyrate levels as a robust marker of an elevated hepatic cytosolic NADH/NAD+ ratio, also known as reductive stress. In humans, elevations in circulating α-hydroxybutyrate levels have previously been associated with impaired glucose tolerance2, insulin resistance3 and mitochondrial disease4, and are associated with a common genetic variant in GCKR5, which has previously been associated with many seemingly disparate metabolic traits. Using LbNOX, we demonstrate that NADH reductive stress mediates the effects of GCKR variation on many metabolic traits, including circulating triglyceride levels, glucose tolerance and FGF21 levels. Our work identifies an elevated hepatic NADH/NAD+ ratio as a latent metabolic parameter that is shaped by human genetic variation and contributes causally to key metabolic traits and diseases. Moreover, it underscores the utility of genetic tools such as LbNOX to empower studies of 'causal metabolism'.


Subject(s)
Liver/metabolism , NAD/metabolism , Stress, Physiological , Adaptor Proteins, Signal Transducing/genetics , Animals , Cytosol/metabolism , Disease Models, Animal , Fibroblast Growth Factors/blood , Genetic Variation , Glucose Tolerance Test , Humans , Insulin Resistance , Levilactobacillus brevis/enzymology , Levilactobacillus brevis/genetics , Male , Mice , Multienzyme Complexes/genetics , Multienzyme Complexes/metabolism , NADH, NADPH Oxidoreductases/genetics , NADH, NADPH Oxidoreductases/metabolism , Oxidation-Reduction , Triglycerides/blood
SELECTION OF CITATIONS
SEARCH DETAIL