Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 14.473
Filtrer
1.
Nat Commun ; 15(1): 8505, 2024 Oct 01.
Article de Anglais | MEDLINE | ID: mdl-39353940

RÉSUMÉ

The main forces driving protein complex evolution are currently not well understood, especially in homomers, where quaternary structure might frequently evolve neutrally. Here we examine the factors determining oligomerisation by analysing the evolution of enzymes in circumstances where homomers rarely evolve. We show that 1) In extracellular environments, most enzymes with known structure are monomers, while in the cytoplasm homomers, indicating that the evolution of oligomers is cellular environment dependent; 2) The evolution of quaternary structure within protein orthogroups is more consistent with the predictions of constructive neutral evolution than an adaptive process: quaternary structure is gained easier than it is lost, and most extracellular monomers evolved from proteins that were monomers also in their ancestral state, without the loss of interfaces. Our results indicate that oligomerisation is context-dependent, and even when adaptive, in many cases it is probably not driven by the intrinsic properties of enzymes, like their biochemical function, but rather the properties of the environment where the enzyme is active. These factors might be macromolecular crowding and excluded volume effects facilitating the evolution of interfaces, and the maintenance of cellular homeostasis through shaping cytoplasm fluidity, protein degradation, or diffusion rates.


Sujet(s)
Cytoplasme , Enzymes , Évolution moléculaire , Structure quaternaire des protéines , Enzymes/composition chimique , Enzymes/métabolisme , Enzymes/génétique , Cytoplasme/métabolisme , Multimérisation de protéines
2.
Biomacromolecules ; 25(8): 4715-4727, 2024 Aug 12.
Article de Anglais | MEDLINE | ID: mdl-38959412

RÉSUMÉ

Centromeres are specific segments of chromosomes comprising two types of nucleosomes: canonical nucleosomes containing an octamer of H2A, H2B, H3, and H4 histones and CENP-A nucleosomes in which H3 is replaced with its analogue CENP-A. This modification leads to a difference in DNA wrapping (∼121 bp), considerably less than 147 bp in canonical nucleosomes. We used atomic force microscopy (AFM) and high-speed AFM (HS-AFM) to characterize nanoscale features and dynamics for both types of nucleosomes. For both nucleosomes, spontaneous asymmetric unwrapping of DNA was observed, and this process occurs via a transient state with ∼100 bp DNA wrapped around the core, followed by a rapid dissociation of DNA. Additionally, HS-AFM revealed higher stability of CENP-A nucleosomes compared with H3 nucleosomes in which dissociation of the histone core occurs prior to the nucleosome dissociation. These results help elucidate the differences between these nucleosomes and the potential biological necessity for CENP-A nucleosomes.


Sujet(s)
Centromère , Nucléosomes , Nucléosomes/composition chimique , Nucléosomes/métabolisme , Nucléosomes/ultrastructure , Centromère/composition chimique , Centromère/métabolisme , Structure quaternaire des protéines , ADN/composition chimique , ADN/métabolisme , Histone/composition chimique , Histone/métabolisme , Protéine A du centromère/composition chimique , Protéine A du centromère/métabolisme , Microscopie à force atomique
3.
Protein Sci ; 33(8): e5089, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-39012001

RÉSUMÉ

D-3-phosphoglycerate dehydrogenase (PHGDH) catalyzes the NAD+-dependent conversion of D-3-phospho-glycerate to 3-phosphohydroxypyruvate, the first step in the phosphorylated pathway for L-serine (L-Ser) biosynthesis. L-Ser plays different relevant metabolic roles in eukaryotic cells: alterations in L-Ser metabolism have been linked to serious neurological disorders. The human PHGDH (hPHGDH), showing a homotetrameric state in solution, is made of four domains, among which there are two regulatory domains at the C-terminus: the aspartate kinase-chorismate mutase-tyrA prephenate dehydrogenase (ACT) and allosteric substrate-binding (ASB) domains. The structure of hPHGDH was solved only for a truncated, dimeric form harboring the N-terminal end containing the substrate and the cofactor binding domains. A model ensemble of the tetrameric hPHGDH was generated using AlphaFold coupled with molecular dynamics refinement. By analyzing the inter-subunit interactions at the tetrameric interface, the residues F418, L478, P479, R454, and Y495 were selected and their role was studied by the alanine-scanning mutagenesis approach. The F418A variant modifies the putative ASB, slightly alters the activity, the fraction of protein in the tetrameric state, and the protein stability; it seems relevant in dimers' recognition to yield the tetrameric oligomer. On the contrary, the R454A, L478A, P479A, and Y495A variants (ACT domain) determine a loss of the tetrameric assembly, resulting in low stability and misfolding, triggering the aggregation and hampering the activity. The predicted tetrameric interface seems mediated by residues at the ACT domain, and the tetramer formation seems crucial for proper folding of hPHGDH, which, in turn, is essential for both stability and functionality.


Sujet(s)
Phosphoglycerate dehydrogenase , Phosphoglycerate dehydrogenase/composition chimique , Phosphoglycerate dehydrogenase/métabolisme , Phosphoglycerate dehydrogenase/génétique , Humains , Structure quaternaire des protéines , Modèles moléculaires , Multimérisation de protéines , Simulation de dynamique moléculaire , Domaines protéiques , Cristallographie aux rayons X
4.
Biophys J ; 123(13): 1846-1856, 2024 Jul 02.
Article de Anglais | MEDLINE | ID: mdl-38824390

RÉSUMÉ

Reactions that occur within the lipid membrane involve, at minimum, ternary complexes among the enzyme, substrate, and lipid. For many systems, the impact of the lipid in regulating activity or oligomerization state is poorly understood. Here, we used small-angle neutron scattering (SANS) to structurally characterize an intramembrane aspartyl protease (IAP), a class of membrane-bound enzymes that use membrane-embedded aspartate residues to hydrolyze transmembrane segments of biologically relevant substrates. We focused on an IAP ortholog from the halophilic archaeon Haloferax volcanii (HvoIAP). HvoIAP purified in n-dodecyl-ß-D-maltoside (DDM) fractionates on size-exclusion chromatography (SEC) as two fractions. We show that, in DDM, the smaller SEC fraction is consistent with a compact HvoIAP monomer. Molecular dynamics flexible fitting conducted on an AlphaFold2-generated monomer produces a model in which loops are compact alongside the membrane-embedded helices. In contrast, SANS data collected on the second SEC fraction indicate an oligomer consistent with an elongated assembly of discrete HvoIAP monomers. Analysis of in-line SEC-SANS data of the HvoIAP oligomer, the first such experiment to be conducted on a membrane protein at Oak Ridge National Lab (ORNL), shows a diversity of elongated and spherical species, including one consistent with the tetrameric assembly reported for the Methanoculleus marisnigri JR1 IAP crystal structure not observed previously in solution. Reconstitution of monomeric HvoIAP into bicelles increases enzyme activity and results in the assembly of HvoIAP into a species with similar dimensions as the ensemble of oligomers isolated from DDM. Our study reveals lipid-mediated HvoIAP self-assembly and demonstrates the utility of in-line SEC-SANS in elucidating oligomerization states of small membrane proteins.


Sujet(s)
Aspartic acid proteases , Haloferax volcanii , Diffraction de neutrons , Multimérisation de protéines , Diffusion aux petits angles , Aspartic acid proteases/métabolisme , Aspartic acid proteases/composition chimique , Haloferax volcanii/enzymologie , Membrane cellulaire/métabolisme , Protéines d'archée/composition chimique , Protéines d'archée/métabolisme , Simulation de dynamique moléculaire , Structure quaternaire des protéines
5.
Article de Anglais | MEDLINE | ID: mdl-38862427

RÉSUMÉ

Since its establishment in 2013, BioLiP has become one of the widely used resources for protein-ligand interactions. Nevertheless, several known issues occurred with it over the past decade. For example, the protein-ligand interactions are represented in the form of single chain-based tertiary structures, which may be inappropriate as many interactions involve multiple protein chains (known as quaternary structures). We sought to address these issues, resulting in Q-BioLiP, a comprehensive resource for quaternary structure-based protein-ligand interactions. The major features of Q-BioLiP include: (1) representing protein structures in the form of quaternary structures rather than single chain-based tertiary structures; (2) pairing DNA/RNA chains properly rather than separation; (3) providing both experimental and predicted binding affinities; (4) retaining both biologically relevant and irrelevant interactions to alleviate the wrong justification of ligands' biological relevance; and (5) developing a new quaternary structure-based algorithm for the modelling of protein-ligand complex structure. With these new features, Q-BioLiP is expected to be a valuable resource for studying biomolecule interactions, including protein-small molecule interaction, protein-metal ion interaction, protein-peptide interaction, protein-protein interaction, protein-DNA/RNA interaction, and RNA-small molecule interaction. Q-BioLiP is freely available at https://yanglab.qd.sdu.edu.cn/Q-BioLiP/.


Sujet(s)
Liaison aux protéines , Protéines , Ligands , Protéines/composition chimique , Protéines/métabolisme , Structure quaternaire des protéines , ADN/métabolisme , ADN/composition chimique , Bases de données de protéines , ARN/métabolisme , ARN/composition chimique , Algorithmes
6.
Cell ; 187(13): 3303-3318.e18, 2024 Jun 20.
Article de Anglais | MEDLINE | ID: mdl-38906101

RÉSUMÉ

Gamete formation and subsequent offspring development often involve extended phases of suspended cellular development or even dormancy. How cells adapt to recover and resume growth remains poorly understood. Here, we visualized budding yeast cells undergoing meiosis by cryo-electron tomography (cryoET) and discovered elaborate filamentous assemblies decorating the nucleus, cytoplasm, and mitochondria. To determine filament composition, we developed a "filament identification" (FilamentID) workflow that combines multiscale cryoET/cryo-electron microscopy (cryoEM) analyses of partially lysed cells or organelles. FilamentID identified the mitochondrial filaments as being composed of the conserved aldehyde dehydrogenase Ald4ALDH2 and the nucleoplasmic/cytoplasmic filaments as consisting of acetyl-coenzyme A (CoA) synthetase Acs1ACSS2. Structural characterization further revealed the mechanism underlying polymerization and enabled us to genetically perturb filament formation. Acs1 polymerization facilitates the recovery of chronologically aged spores and, more generally, the cell cycle re-entry of starved cells. FilamentID is broadly applicable to characterize filaments of unknown identity in diverse cellular contexts.


Sujet(s)
Gamétogenèse , Mitochondries , Protéines de Saccharomyces cerevisiae , Saccharomyces cerevisiae , Aldehyde dehydrogenase/métabolisme , Aldehyde dehydrogenase/composition chimique , Noyau de la cellule/métabolisme , Noyau de la cellule/ultrastructure , Coenzyme A ligases/métabolisme , Cryomicroscopie électronique , Cytoplasme/métabolisme , Tomographie en microscopie électronique , Méiose , Mitochondries/métabolisme , Mitochondries/ultrastructure , Saccharomyces cerevisiae/métabolisme , Saccharomyces cerevisiae/ultrastructure , Protéines de Saccharomyces cerevisiae/métabolisme , Protéines de Saccharomyces cerevisiae/composition chimique , Spores fongiques/métabolisme , Modèles moléculaires , Structure quaternaire des protéines
7.
Acta Crystallogr D Struct Biol ; 80(Pt 7): 535-550, 2024 Jul 01.
Article de Anglais | MEDLINE | ID: mdl-38935342

RÉSUMÉ

The formation of a vitrified thin film embedded with randomly oriented macromolecules is an essential prerequisite for cryogenic sample electron microscopy. Most commonly, this is achieved using the plunge-freeze method first described nearly 40 years ago. Although this is a robust method, the behaviour of different macromolecules shows great variation upon freezing and often needs to be optimized to obtain an isotropic, high-resolution reconstruction. For a macromolecule in such a film, the probability of encountering the air-water interface in the time between blotting and freezing and adopting preferred orientations is very high. 3D reconstruction using preferentially oriented particles often leads to anisotropic and uninterpretable maps. Currently, there are no general solutions to this prevalent issue, but several approaches largely focusing on sample preparation with the use of additives and novel grid modifications have been attempted. In this study, the effect of physical and chemical factors on the orientations of macromolecules was investigated through an analysis of selected well studied macromolecules, and important parameters that determine the behaviour of proteins on cryo-EM grids were revealed. These insights highlight the nature of the interactions that cause preferred orientations and can be utilized to systematically address orientation bias for any given macromolecule and to provide a framework to design small-molecule additives to enhance sample stability and behaviour.


Sujet(s)
Cryomicroscopie électronique , Humains , Protéines/composition chimique , Cryomicroscopie électronique/méthodes , Modèles moléculaires , Structure tertiaire des protéines , SARS-CoV-2/composition chimique , Tensioactifs/composition chimique , Structure quaternaire des protéines , Escherichia coli/composition chimique , Température
8.
Biophys J ; 123(13): 1763-1780, 2024 Jul 02.
Article de Anglais | MEDLINE | ID: mdl-38762753

RÉSUMÉ

Cells employ many large macromolecular machines for the execution and regulation of processes that are vital for cell and organismal viability. Interestingly, cells cannot synthesize these machines as functioning units. Instead, cells synthesize the molecular parts that must then assemble into the functional complex. Many important machines, including chaperones such as GroEL and proteases such as the proteasome, comprise protein rings that are stacked on top of one another. While there is some experimental data regarding how stacked-ring complexes such as the proteasome self-assemble, a comprehensive understanding of the dynamics of stacked-ring assembly is currently lacking. Here, we developed a mathematical model of stacked-trimer assembly and performed an analysis of the assembly of the stacked homomeric trimer, which is the simplest stacked-ring architecture. We found that stacked rings are particularly susceptible to a form of kinetic trapping that we term "deadlock," in which the system gets stuck in a state where there are many large intermediates that are not the fully assembled structure but that cannot productively react. When interaction affinities are uniformly strong, deadlock severely limits assembly yield. We thus predicted that stacked rings would avoid situations where all interfaces in the structure have high affinity. Analysis of available crystal structures indicated that indeed the majority-if not all-of stacked trimers do not contain uniformly strong interactions. Finally, to better understand the origins of deadlock, we developed a formal pathway analysis and showed that, when all the binding affinities are strong, many of the possible pathways are utilized. In contrast, optimal assembly strategies utilize only a small number of pathways. Our work suggests that deadlock is a critical factor influencing the evolution of macromolecular machines and provides general principles for understanding the self-assembly efficiency of existing machines.


Sujet(s)
Modèles moléculaires , Multimérisation de protéines , Cinétique , Liaison aux protéines , Structure quaternaire des protéines
9.
Nucleic Acids Res ; 52(10): 5912-5927, 2024 Jun 10.
Article de Anglais | MEDLINE | ID: mdl-38742632

RÉSUMÉ

Mitochondrial single-stranded DNA-binding protein (mtSSB) is essential for mitochondrial DNA (mtDNA) replication. Recently, several mtSSB variants have been associated with autosomal dominant mitochondrial optic atrophy and retinal dystrophy. Here, we have studied at the molecular level the functional consequences of one of the most severe mtSSB variants, R107Q. We first studied the oligomeric state of this variant and observed that the mtSSBR107Q mutant forms stable tetramers in vitro. On the other hand, we showed, using complementary single-molecule approaches, that mtSSBR107Q displays a lower intramolecular ssDNA compaction ability and a higher ssDNA dissociation rate than the WT protein. Real-time competition experiments for ssDNA-binding showed a marked advantage of mtSSBWT over mtSSBR107Q. Combined, these results show that the R107Q mutation significantly impaired the ssDNA-binding and compacting ability of mtSSB, likely by weakening mtSSB ssDNA wrapping efficiency. These features are in line with our molecular modeling of ssDNA on mtSSB showing that the R107Q mutation may destabilize local interactions and results in an electronegative spot that interrupts an ssDNA-interacting-electropositive patch, thus reducing the potential mtSSB-ssDNA interaction sites.


Sujet(s)
ADN simple brin , Protéines de liaison à l'ADN , Mutation , Humains , ADN mitochondrial/génétique , ADN mitochondrial/métabolisme , ADN simple brin/métabolisme , ADN simple brin/génétique , Protéines de liaison à l'ADN/métabolisme , Protéines de liaison à l'ADN/génétique , Protéines de liaison à l'ADN/composition chimique , Protéines mitochondriales/métabolisme , Protéines mitochondriales/génétique , Protéines mitochondriales/composition chimique , Modèles moléculaires , Liaison aux protéines , Structure quaternaire des protéines
10.
Biomolecules ; 14(5)2024 May 13.
Article de Anglais | MEDLINE | ID: mdl-38785981

RÉSUMÉ

The quality prediction of quaternary structure models of a protein complex, in the absence of its true structure, is known as the Estimation of Model Accuracy (EMA). EMA is useful for ranking predicted protein complex structures and using them appropriately in biomedical research, such as protein-protein interaction studies, protein design, and drug discovery. With the advent of more accurate protein complex (multimer) prediction tools, such as AlphaFold2-Multimer and ESMFold, the estimation of the accuracy of protein complex structures has attracted increasing attention. Many deep learning methods have been developed to tackle this problem; however, there is a noticeable absence of a comprehensive overview of these methods to facilitate future development. Addressing this gap, we present a review of deep learning EMA methods for protein complex structures developed in the past several years, analyzing their methodologies, data and feature construction. We also provide a prospective summary of some potential new developments for further improving the accuracy of the EMA methods.


Sujet(s)
Apprentissage profond , Structure quaternaire des protéines , Protéines , Protéines/composition chimique , Modèles moléculaires , Humains
11.
Nat Commun ; 15(1): 3775, 2024 May 06.
Article de Anglais | MEDLINE | ID: mdl-38710701

RÉSUMÉ

SAMHD1 regulates cellular nucleotide homeostasis, controlling dNTP levels by catalysing their hydrolysis into 2'-deoxynucleosides and triphosphate. In differentiated CD4+ macrophage and resting T-cells SAMHD1 activity results in the inhibition of HIV-1 infection through a dNTP blockade. In cancer, SAMHD1 desensitizes cells to nucleoside-analogue chemotherapies. Here we employ time-resolved cryogenic-EM imaging and single-particle analysis to visualise assembly, allostery and catalysis by this multi-subunit enzyme. Our observations reveal how dynamic conformational changes in the SAMHD1 quaternary structure drive the catalytic cycle. We capture five states at high-resolution in a live catalytic reaction, revealing how allosteric activators support assembly of a stable SAMHD1 tetrameric core and how catalysis is driven by the opening and closing of active sites through pairwise coupling of active sites and order-disorder transitions in regulatory domains. This direct visualisation of enzyme catalysis dynamics within an allostery-stabilised platform sets a precedent for mechanistic studies into the regulation of multi-subunit enzymes.


Sujet(s)
Domaine catalytique , Cryomicroscopie électronique , Protéine-1 contenant un domaine SAM et un domaine HD , Protéine-1 contenant un domaine SAM et un domaine HD/métabolisme , Protéine-1 contenant un domaine SAM et un domaine HD/composition chimique , Protéine-1 contenant un domaine SAM et un domaine HD/génétique , Régulation allostérique , Humains , Structure quaternaire des protéines , Catalyse , Biocatalyse , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/métabolisme , Modèles moléculaires
12.
J Biol Chem ; 300(5): 107252, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38569936

RÉSUMÉ

Heterotrimeric GTP-binding protein alpha subunit (Gα) and its cognate regulator of G-protein signaling (RGS) protein transduce signals in eukaryotes spanning protists, amoeba, animals, fungi, and plants. The core catalytic mechanisms of the GTPase activity of Gα and the interaction interface with RGS for the acceleration of GTP hydrolysis seem to be conserved across these groups; however, the RGS gene is under low selective pressure in plants, resulting in its frequent loss. Our current understanding of the structural basis of Gα:RGS regulation in plants has been shaped by Arabidopsis Gα, (AtGPA1), which has a cognate RGS protein. To gain a comprehensive understanding of this regulation beyond Arabidopsis, we obtained the x-ray crystal structures of Oryza sativa Gα, which has no RGS, and Selaginella moellendorffi (a lycophyte) Gα that has low sequence similarity with AtGPA1 but has an RGS. We show that the three-dimensional structure, protein-protein interaction with RGS, and the dynamic features of these Gα are similar to AtGPA1 and metazoan Gα. Molecular dynamic simulation of the Gα-RGS interaction identifies the contacts established by specific residues of the switch regions of GTP-bound Gα, crucial for this interaction, but finds no significant difference due to specific amino acid substitutions. Together, our data provide valuable insights into the regulatory mechanisms of plant G-proteins but do not support the hypothesis of adaptive co-evolution of Gα:RGS proteins in plants.


Sujet(s)
Sous-unités alpha des protéines G , Modèles moléculaires , Protéines végétales , Protéines RGS , Arabidopsis/métabolisme , Arabidopsis/génétique , Protéines d'Arabidopsis/métabolisme , Protéines d'Arabidopsis/composition chimique , Protéines d'Arabidopsis/génétique , Cristallographie aux rayons X , Sous-unités alpha des protéines G/métabolisme , Sous-unités alpha des protéines G/composition chimique , Sous-unités alpha des protéines G/génétique , Oryza/métabolisme , Oryza/génétique , Protéines végétales/métabolisme , Protéines végétales/composition chimique , Protéines végétales/génétique , Liaison aux protéines , Protéines RGS/métabolisme , Protéines RGS/composition chimique , Protéines RGS/génétique , Relation structure-activité , Selaginellaceae/génétique , Selaginellaceae/métabolisme , Structure quaternaire des protéines
13.
J Biol Chem ; 300(5): 107261, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38582450

RÉSUMÉ

Mammalian SLC26 proteins are membrane-based anion transporters that belong to the large SLC26/SulP family, and many of their variants are associated with hereditary diseases. Recent structural studies revealed a strikingly similar homodimeric molecular architecture for several SLC26 members, implying a shared molecular principle. Now a new question emerges as to how these structurally similar proteins execute diverse physiological functions. In this study, we sought to identify the common versus distinct molecular mechanism among the SLC26 proteins using both naturally occurring and artificial missense changes introduced to SLC26A4, SLC26A5, and SLC26A9. We found: (i) the basic residue at the anion binding site is essential for both anion antiport of SLC26A4 and motor functions of SLC26A5, and its conversion to a nonpolar residue is crucial but not sufficient for the fast uncoupled anion transport in SLC26A9; (ii) the conserved polar residues in the N- and C-terminal cytosolic domains are likely involved in dynamic hydrogen-bonding networks and are essential for anion antiport of SLC26A4 but not for motor (SLC26A5) and uncoupled anion transport (SLC26A9) functions; (iii) the hydrophobic interaction between each protomer's last transmembrane helices, TM14, is not of functional significance in SLC26A9 but crucial for the functions of SLC26A4 and SLC26A5, likely contributing to optimally orient the axis of the relative movements of the core domain with respect to the gate domains within the cell membrane. These findings advance our understanding of the molecular mechanisms underlying the diverse physiological roles of the SLC26 family of proteins.


Sujet(s)
Antiports , Transporteurs de sulfate , Animaux , Humains , Antiports/métabolisme , Antiports/génétique , Antiports/composition chimique , Sites de fixation , Cellules HEK293 , Liaison hydrogène , Modèles moléculaires , Mutation faux-sens , Domaines protéiques , Structure quaternaire des protéines , Structure tertiaire des protéines , Transporteurs de sulfate/métabolisme , Transporteurs de sulfate/génétique , Transporteurs de sulfate/composition chimique , Multimérisation de protéines , Structure secondaire des protéines
14.
J Biol Chem ; 300(5): 107245, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38569940

RÉSUMÉ

The IgG-specific endoglycosidases EndoS and EndoS2 from Streptococcus pyogenes can remove conserved N-linked glycans present on the Fc region of host antibodies to inhibit Fc-mediated effector functions. These enzymes are therefore being investigated as therapeutics for suppressing unwanted immune activation, and have additional application as tools for antibody glycan remodeling. EndoS and EndoS2 differ in Fc glycan substrate specificity due to structural differences within their catalytic glycosyl hydrolase domains. However, a chimeric EndoS enzyme with a substituted glycosyl hydrolase from EndoS2 loses catalytic activity, despite high structural homology between the two enzymes, indicating either mechanistic divergence of EndoS and EndoS2, or improperly-formed domain interfaces in the chimeric enzyme. Here, we present the crystal structure of the EndoS2-IgG1 Fc complex determined to 3.0 Å resolution. Comparison of complexed and unliganded EndoS2 reveals relative reorientation of the glycosyl hydrolase, leucine-rich repeat and hybrid immunoglobulin domains. The conformation of the complexed EndoS2 enzyme is also different when compared to the earlier EndoS-IgG1 Fc complex, and results in distinct contact surfaces between the two enzymes and their Fc substrate. These findings indicate mechanistic divergence of EndoS2 and EndoS. It will be important to consider these differences in the design of IgG-specific enzymes, developed to enable customizable antibody glycosylation.


Sujet(s)
Protéines bactériennes , Glycosidases , Immunoglobuline G , Modèles moléculaires , Streptococcus pyogenes , Humains , Protéines bactériennes/composition chimique , Protéines bactériennes/métabolisme , Cristallographie aux rayons X , Glycosidases/composition chimique , Glycosidases/métabolisme , Fragments Fc des immunoglobulines/composition chimique , Fragments Fc des immunoglobulines/métabolisme , Immunoglobuline G/composition chimique , Immunoglobuline G/métabolisme , Streptococcus pyogenes/enzymologie , Spécificité du substrat , Structure quaternaire des protéines
15.
Biochim Biophys Acta Gen Subj ; 1868(6): 130618, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38621595

RÉSUMÉ

The oligomerization of proteins is an important biological control mechanism and has several functions in activity and stability of enzymes, structural proteins, ion channels and transcription factors. The determination of the relevant oligomeric states in terms of geometry (spatial extent), oligomer size (monomer or dimer or oligomer) and affinity (amounts of monomer, dimer and oligomer) is a challenging biophysical problem. Förster resonance energy transfer and fluorescence fluctuation spectroscopy are powerful tools that are sensitive to proximity and oligomerization respectively. Here it is proposed to combine image-based lifetime-detected Forster resonance energy transfer with image correlation spectroscopy and photobleaching to determine distances, oligomer sizes and oligomer distributions. Simulations for simple oligomeric forms illustrate the potential to improve the discrimination between different quaternary states in the cellular milieu.


Sujet(s)
Transfert d'énergie par résonance de fluorescence , Photoblanchiment , Transfert d'énergie par résonance de fluorescence/méthodes , Multimérisation de protéines , Structure quaternaire des protéines , Humains , Simulation numérique
16.
J Biol Chem ; 300(5): 107248, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38556082

RÉSUMÉ

P2X receptors are a family of ligand gated ion channels found in a range of eukaryotic species including humans but are not naturally present in the yeast Saccharomyces cerevisiae. We demonstrate the first recombinant expression and functional gating of the P2X2 receptor in baker's yeast. We leverage the yeast host for facile genetic screens of mutant P2X2 by performing site saturation mutagenesis at residues of interest, including SNPs implicated in deafness and at residues involved in native binding. Deep mutational analysis and rounds of genetic engineering yield mutant P2X2 F303Y A304W, which has altered ligand selectivity toward the ATP analog AMP-PNP. The F303Y A304W variant shows over 100-fold increased intracellular calcium amplitudes with AMP-PNP compared to the WT receptor and has a much lower desensitization rate. Since AMP-PNP does not naturally activate P2X receptors, the F303Y A304W P2X2 may be a starting point for downstream applications in chemogenetic cellular control. Interestingly, the A304W mutation selectively destabilizes the desensitized state, which may provide a mechanistic basis for receptor opening with suboptimal agonists. The yeast system represents an inexpensive, scalable platform for ion channel characterization and engineering by circumventing the more expensive and time-consuming methodologies involving mammalian hosts.


Sujet(s)
Récepteurs purinergiques P2X2 , Saccharomyces cerevisiae , Humains , Substitution d'acide aminé , Ligands , Ingénierie des protéines/méthodes , Récepteurs purinergiques P2X2/métabolisme , Récepteurs purinergiques P2X2/génétique , Saccharomyces cerevisiae/métabolisme , Saccharomyces cerevisiae/génétique , Modèles moléculaires , Structure tertiaire des protéines , Structure quaternaire des protéines , Similitude structurale de protéines , Mutation
17.
Nature ; 628(8008): 657-663, 2024 Apr.
Article de Anglais | MEDLINE | ID: mdl-38509367

RÉSUMÉ

In response to pathogen infection, gasdermin (GSDM) proteins form membrane pores that induce a host cell death process called pyroptosis1-3. Studies of human and mouse GSDM pores have revealed the functions and architectures of assemblies comprising 24 to 33 protomers4-9, but the mechanism and evolutionary origin of membrane targeting and GSDM pore formation remain unknown. Here we determine a structure of a bacterial GSDM (bGSDM) pore and define a conserved mechanism of pore assembly. Engineering a panel of bGSDMs for site-specific proteolytic activation, we demonstrate that diverse bGSDMs form distinct pore sizes that range from smaller mammalian-like assemblies to exceptionally large pores containing more than 50 protomers. We determine a cryo-electron microscopy structure of a Vitiosangium bGSDM in an active 'slinky'-like oligomeric conformation and analyse bGSDM pores in a native lipid environment to create an atomic-level model of a full 52-mer bGSDM pore. Combining our structural analysis with molecular dynamics simulations and cellular assays, our results support a stepwise model of GSDM pore assembly and suggest that a covalently bound palmitoyl can leave a hydrophobic sheath and insert into the membrane before formation of the membrane-spanning ß-strand regions. These results reveal the diversity of GSDM pores found in nature and explain the function of an ancient post-translational modification in enabling programmed host cell death.


Sujet(s)
Gasdermines , Myxococcales , Cryomicroscopie électronique , Gasdermines/composition chimique , Gasdermines/métabolisme , Gasdermines/ultrastructure , Interactions hydrophobes et hydrophiles , Lipides membranaires/composition chimique , Lipides membranaires/métabolisme , Simulation de dynamique moléculaire , Myxococcales/composition chimique , Myxococcales/cytologie , Myxococcales/ultrastructure , Structure quaternaire des protéines , Sous-unités de protéines/composition chimique , Sous-unités de protéines/métabolisme , Protéolyse , Pyroptose
18.
Nature ; 629(8011): 467-473, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38471529

RÉSUMÉ

Prokaryotes have evolved intricate innate immune systems against phage infection1-7. Gabija is a highly widespread prokaryotic defence system that consists of two components, GajA and GajB8. GajA functions as a DNA endonuclease that is inactive in the presence of ATP9. Here, to explore how the Gabija system is activated for anti-phage defence, we report its cryo-electron microscopy structures in five states, including apo GajA, GajA in complex with DNA, GajA bound by ATP, apo GajA-GajB, and GajA-GajB in complex with ATP and Mg2+. GajA is a rhombus-shaped tetramer with its ATPase domain clustered at the centre and the topoisomerase-primase (Toprim) domain located peripherally. ATP binding at the ATPase domain stabilizes the insertion region within the ATPase domain, keeping the Toprim domain in a closed state. Upon ATP depletion by phages, the Toprim domain opens to bind and cleave the DNA substrate. GajB, which docks on GajA, is activated by the cleaved DNA, ultimately leading to prokaryotic cell death. Our study presents a mechanistic landscape of Gabija activation.


Sujet(s)
Bacillus cereus , Protéines bactériennes , Bactériophages , Cryomicroscopie électronique , Immunité innée , Adenosine triphosphatases/métabolisme , Adenosine triphosphatases/composition chimique , Adenosine triphosphatases/ultrastructure , Adénosine triphosphate/composition chimique , Adénosine triphosphate/métabolisme , Apoprotéines/composition chimique , Apoprotéines/immunologie , Apoprotéines/métabolisme , Apoprotéines/ultrastructure , Protéines bactériennes/composition chimique , Protéines bactériennes/immunologie , Protéines bactériennes/métabolisme , Protéines bactériennes/ultrastructure , Bactériophages/immunologie , ADN/métabolisme , ADN/composition chimique , Clivage de l'ADN , Magnésium/composition chimique , Magnésium/métabolisme , Modèles moléculaires , Liaison aux protéines , Domaines protéiques , Viabilité microbienne , Bacillus cereus/composition chimique , Bacillus cereus/immunologie , Bacillus cereus/métabolisme , Bacillus cereus/ultrastructure , Structure quaternaire des protéines , DNA primase/composition chimique , DNA primase/métabolisme , DNA primase/ultrastructure , DNA topoisomerases/composition chimique , DNA topoisomerases/métabolisme , DNA topoisomerases/ultrastructure
19.
J Biol Chem ; 300(4): 107166, 2024 Apr.
Article de Anglais | MEDLINE | ID: mdl-38490435

RÉSUMÉ

Clamp loaders are pentameric ATPases that place circular sliding clamps onto DNA, where they function in DNA replication and genome integrity. The central activity of a clamp loader is the opening of the ring-shaped sliding clamp and the subsequent binding to primer-template (p/t)-junctions. The general architecture of clamp loaders is conserved across all life, suggesting that their mechanism is retained. Recent structural studies of the eukaryotic clamp loader replication factor C (RFC) revealed that it functions using a crab-claw mechanism, where clamp opening is coupled to a massive conformational change in the loader. Here we investigate the clamp loading mechanism of the Escherichia coli clamp loader at high resolution using cryo-electron microscopy. We find that the E. coli clamp loader opens the clamp using a crab-claw motion at a single pivot point, whereas the eukaryotic RFC loader uses motions distributed across the complex. Furthermore, we find clamp opening occurs in multiple steps, starting with a partly open state with a spiral conformation, and proceeding to a wide open clamp in a surprising planar geometry. Finally, our structures in the presence of p/t-junctions illustrate how the clamp closes around p/t-junctions and how the clamp loader initiates release from the loaded clamp. Our results reveal mechanistic distinctions in a macromolecular machine that is conserved across all domains of life.


Sujet(s)
Réplication de l'ADN , Escherichia coli , Cryomicroscopie électronique , Escherichia coli/métabolisme , Escherichia coli/génétique , Protéines Escherichia coli/métabolisme , Protéines Escherichia coli/composition chimique , Protéines Escherichia coli/génétique , Conformation des protéines , Protéine C de réplication/métabolisme , Protéine C de réplication/composition chimique , Protéine C de réplication/génétique , Modèles moléculaires , Structure quaternaire des protéines
20.
J Biol Chem ; 300(4): 107197, 2024 Apr.
Article de Anglais | MEDLINE | ID: mdl-38508314

RÉSUMÉ

Cell polarity oscillations in Myxococcus xanthus motility are driven by a prokaryotic small Ras-like GTPase, mutual gliding protein A (MglA), which switches from one cell pole to the other in response to extracellular signals. MglA dynamics is regulated by MglB, which functions both as a GTPase activating protein (GAP) and a guanine nucleotide exchange factor (GEF) for MglA. With an aim to dissect the asymmetric role of the two MglB protomers in the dual GAP and GEF activities, we generated a functional MglAB complex by coexpressing MglB with a linked construct of MglA and MglB. This strategy enabled us to generate mutations of individual MglB protomers (MglB1 or MglB2 linked to MglA) and delineate their role in GEF and GAP activities. We establish that the C-terminal helix of MglB1, but not MglB2, stimulates nucleotide exchange through a site away from the nucleotide-binding pocket, confirming an allosteric mechanism. Interaction between the N-terminal ß-strand of MglB1 and ß0 of MglA is essential for the optimal GEF activity of MglB. Specific residues of MglB2, which interact with Switch-I of MglA, partially contribute to its GAP activity. Thus, the role of the MglB2 protomer in the GAP activity of MglB is limited to restricting the conformation of MglA active site loops. The direct demonstration of the allosteric mechanism of GEF action provides us new insights into the regulation of small Ras-like GTPases, a feature potentially present in many uncharacterized GEFs.


Sujet(s)
Protéines bactériennes , Protéines d'activation de la GTPase , Myxococcus xanthus , Protéines bactériennes/métabolisme , Protéines bactériennes/composition chimique , Protéines bactériennes/génétique , Activation enzymatique , Protéines d'activation de la GTPase/métabolisme , Protéines d'activation de la GTPase/génétique , Protéines d'activation de la GTPase/composition chimique , Facteurs d'échange de nucléotides guanyliques/métabolisme , Facteurs d'échange de nucléotides guanyliques/composition chimique , Facteurs d'échange de nucléotides guanyliques/génétique , Myxococcus xanthus/métabolisme , Myxococcus xanthus/génétique , Myxococcus xanthus/enzymologie , Multimérisation de protéines , Modèles moléculaires , Structure quaternaire des protéines
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE