Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 10 de 10
Filtrer
Plus de filtres










Base de données
Gamme d'année
1.
Nat Commun ; 15(1): 5830, 2024 Jul 11.
Article de Anglais | MEDLINE | ID: mdl-38992057

RÉSUMÉ

Impaired ion channels regulating Golgi pH lead to structural alterations in the Golgi apparatus, such as fragmentation, which is found, along with cognitive impairment, in Alzheimer's disease. However, the causal relationship between altered Golgi structure and cognitive impairment remains elusive due to the lack of understanding of ion channels in the Golgi apparatus of brain cells. Here, we identify that a transmembrane protein TMEM87A, renamed Golgi-pH-regulating cation channel (GolpHCat), expressed in astrocytes and neurons that contributes to hippocampus-dependent memory. We find that GolpHCat displays unique voltage-dependent currents, which is potently inhibited by gluconate. Additionally, we gain structural insights into the ion conduction through GolpHCat at the molecular level by determining three high-resolution cryogenic-electron microscopy structures of human GolpHCat. GolpHCat-knockout mice show fragmented Golgi morphology and altered protein glycosylation and functions in the hippocampus, leading to impaired spatial memory. These findings suggest a molecular target for Golgi-related diseases and cognitive impairment.


Sujet(s)
Appareil de Golgi , Hippocampe , Souris knockout , Neurones , Animaux , Humains , Mâle , Souris , Astrocytes/métabolisme , Dysfonctionnement cognitif/métabolisme , Dysfonctionnement cognitif/physiopathologie , Dysfonctionnement cognitif/anatomopathologie , Cryomicroscopie électronique , Glycosylation , Appareil de Golgi/métabolisme , Cellules HEK293 , Hippocampe/métabolisme , Concentration en ions d'hydrogène , Canaux ioniques/métabolisme , Canaux ioniques/génétique , Protéines membranaires/métabolisme , Protéines membranaires/génétique , Mémoire/physiologie , Souris de lignée C57BL , Neurones/métabolisme , Mémoire spatiale/physiologie
2.
Elife ; 122023 09 01.
Article de Anglais | MEDLINE | ID: mdl-37656635

RÉSUMÉ

G-protein coupled receptors (GPCRs) present specific activation pathways and signaling among receptor subtypes. Hence, an extensive knowledge of the structural dynamics of the receptor is critical for the development of therapeutics. Here, we target the adenosine A1 receptor (A1R), for which a negligible number of drugs have been approved. We combine molecular dynamics simulations, enhanced sampling techniques, network theory and pocket detection to decipher the activation pathway of A1R, decode the allosteric networks and identify transient pockets. The A1R activation pathway reveal hidden intermediate and pre-active states together with the inactive and fully-active states observed experimentally. The protein energy networks computed throughout these conformational states successfully unravel the extra and intracellular allosteric centers and the communication pathways that couples them. We observe that the allosteric networks are dynamic, being increased along activation and fine-tuned in presence of the trimeric G-proteins. Overlap of transient pockets and energy networks uncover how the allosteric coupling between pockets and distinct functional regions of the receptor is altered along activation. By an in-depth analysis of the bridge between activation pathway, energy networks and transient pockets, we provide a further understanding of A1R. This information can be useful to ease the design of allosteric modulators for A1R.


Sujet(s)
Protéines G , Transduction du signal , Adénosine , Communication , Savoir
3.
J Am Chem Soc ; 144(16): 7146-7159, 2022 04 27.
Article de Anglais | MEDLINE | ID: mdl-35412310

RÉSUMÉ

Deciphering the molecular mechanisms of enzymatic allosteric regulation requires the structural characterization of functional states and also their time evolution toward the formation of the allosterically activated ternary complex. The transient nature and usually slow millisecond time scale interconversion between these functional states hamper their experimental and computational characterization. Here, we combine extensive molecular dynamics simulations, enhanced sampling techniques, and dynamical networks to describe the allosteric activation of imidazole glycerol phosphate synthase (IGPS) from the substrate-free form to the active ternary complex. IGPS is a heterodimeric bienzyme complex whose HisH subunit is responsible for hydrolyzing glutamine and delivering ammonia for the cyclase activity in HisF. Despite significant advances in understanding the underlying allosteric mechanism, essential molecular details of the long-range millisecond allosteric activation of IGPS remain hidden. Without using a priori information of the active state, our simulations uncover how IGPS, with the allosteric effector bound in HisF, spontaneously captures glutamine in a catalytically inactive HisH conformation, subsequently attains a closed HisF:HisH interface, and finally forms the oxyanion hole in HisH for efficient glutamine hydrolysis. We show that the combined effector and substrate binding dramatically decreases the conformational barrier associated with oxyanion hole formation, in line with the experimentally observed 4500-fold activity increase in glutamine hydrolysis. The allosteric activation is controlled by correlated time-evolving dynamic networks connecting the effector and substrate binding sites. This computational strategy tailored to describe millisecond events can be used to rationalize the effect of mutations on the allosteric regulation and guide IGPS engineering efforts.


Sujet(s)
Aminohydrolases , Glutamine , Régulation allostérique , Aminohydrolases/composition chimique , Aminohydrolases/génétique , Aminohydrolases/métabolisme , Sites de fixation , Glutamine/métabolisme
4.
Biochem Soc Trans ; 50(1): 241-252, 2022 02 28.
Article de Anglais | MEDLINE | ID: mdl-35076690

RÉSUMÉ

There have been numerous advances in the development of computational and statistical methods and applications of big data and artificial intelligence (AI) techniques for computer-aided drug design (CADD). Drug design is a costly and laborious process considering the biological complexity of diseases. To effectively and efficiently design and develop a new drug, CADD can be used to apply cutting-edge techniques to various limitations in the drug design field. Data pre-processing approaches, which clean the raw data for consistent and reproducible applications of big data and AI methods are introduced. We include the current status of the applicability of big data and AI methods to drug design areas such as the identification of binding sites in target proteins, structure-based virtual screening (SBVS), and absorption, distribution, metabolism, excretion and toxicity (ADMET) property prediction. Data pre-processing and applications of big data and AI methods enable the accurate and comprehensive analysis of massive biomedical data and the development of predictive models in the field of drug design. Understanding and analyzing biological, chemical, or pharmaceutical architectures of biomedical entities related to drug design will provide beneficial information in the biomedical big data era.


Sujet(s)
Intelligence artificielle , Mégadonnées , Conception de médicament , Découverte de médicament/méthodes , Protéines
5.
ACS Catal ; 11(21): 13733-13743, 2021 Nov 05.
Article de Anglais | MEDLINE | ID: mdl-34777912

RÉSUMÉ

Allostery is a central mechanism for the regulation of multi-enzyme complexes. The mechanistic basis that drives allosteric regulation is poorly understood but harbors key information for enzyme engineering. In the present study, we focus on the tryptophan synthase complex that is composed of TrpA and TrpB subunits, which allosterically activate each other. Specifically, we develop a rational approach for identifying key amino acid residues of TrpB distal from the active site. Those residues are predicted to be crucial for shifting the inefficient conformational ensemble of the isolated TrpB to a productive ensemble through intra-subunit allosteric effects. The experimental validation of the conformationally driven TrpB design demonstrates its superior stand-alone activity in the absence of TrpA, comparable to those enhancements obtained after multiple rounds of experimental laboratory evolution. Our work evidences that the current challenge of distal active site prediction for enhanced function in computational enzyme design has become within reach.

6.
Chembiochem ; 22(5): 904-914, 2021 03 02.
Article de Anglais | MEDLINE | ID: mdl-33094545

RÉSUMÉ

Machine learning (ML) has pervaded most areas of protein engineering, including stability and stereoselectivity. Using limonene epoxide hydrolase as the model enzyme and innov'SAR as the ML platform, comprising a digital signal process, we achieved high protein robustness that can resist unfolding with concomitant detrimental aggregation. Fourier transform (FT) allows us to take into account the order of the protein sequence and the nonlinear interactions between positions, and thus to grasp epistatic phenomena. The innov'SAR approach is interpolative, extrapolative and makes outside-the-box, predictions not found in other state-of-the-art ML or deep learning approaches. Equally significant is the finding that our approach to ML in the present context, flanked by advanced molecular dynamics simulations, uncovers the connection between epistatic mutational interactions and protein robustness.


Sujet(s)
Epoxide hydrolase/composition chimique , Epoxide hydrolase/métabolisme , Apprentissage machine , Mutation , Pliage des protéines , Multimérisation de protéines , Rhodococcus/enzymologie , Epoxide hydrolase/génétique , Limonène/composition chimique , Limonène/métabolisme , Simulation de dynamique moléculaire , Ingénierie des protéines
7.
J Am Chem Soc ; 141(33): 13049-13056, 2019 08 21.
Article de Anglais | MEDLINE | ID: mdl-31356074

RÉSUMÉ

Multimeric enzyme complexes are ubiquitous in nature and catalyze a broad range of useful biological transformations. They are often characterized by a tight allosteric coupling between subunits, making them highly inefficient when isolated. A good example is Tryptophan synthase (TrpS), an allosteric heterodimeric enzyme in the form of an αßßα complex that catalyzes the biosynthesis of L-tryptophan. In this study, we decipher the allosteric regulation existing in TrpS from Pyrococcus furiosus (PfTrpS), and how the allosteric conformational ensemble is recovered in laboratory-evolved stand-alone ß-subunit variants. We find that recovering the conformational ensemble of a subdomain of TrpS affecting the relative stabilities of open, partially closed, and closed conformations is a prerequisite for enhancing the catalytic efficiency of the ß-subunit in the absence of its binding partner. The distal mutations resuscitate the allosterically driven conformational regulation and alter the populations and rates of exchange between these multiple conformational states, which are essential for the multistep reaction pathway of the enzyme. Interestingly, these distal mutations can be a priori predicted by careful analysis of the conformational ensemble of the TrpS enzyme through computational methods. Our study provides the enzyme design field with a rational approach for evolving allosteric enzymes toward improved stand-alone function for biosynthetic applications.


Sujet(s)
Pyrococcus furiosus/enzymologie , Tryptophan synthase/composition chimique , Régulation allostérique , Domaine catalytique , Cristallographie aux rayons X , Modèles moléculaires , Conformation des protéines , Multimérisation de protéines , Pyrococcus furiosus/composition chimique , Pyrococcus furiosus/métabolisme , Tryptophane/métabolisme , Tryptophan synthase/métabolisme
8.
Chem Commun (Camb) ; 54(50): 6622-6634, 2018 Jun 19.
Article de Anglais | MEDLINE | ID: mdl-29780987

RÉSUMÉ

The free energy landscape concept that describes enzymes as an ensemble of differently populated conformational sub-states in dynamic equilibrium is key for evaluating enzyme activity, enantioselectivity, and specificity. Mutations introduced in the enzyme sequence can alter the populations of the pre-existing conformational states, thus strongly modifying the enzyme ability to accommodate alternative substrates, revert its enantiopreferences, and even increase the activity for some residual promiscuous reactions. In this feature article, we present an overview of the current experimental and computational strategies to explore the conformational free energy landscape of enzymes. We provide a series of recent publications that highlight the key role of conformational dynamics for the enzyme evolution towards new functions and substrates, and provide some perspectives on how conformational dynamism should be considered in future computational enzyme design protocols.


Sujet(s)
Enzymes/composition chimique , Biocatalyse , Domaine catalytique , Enzymes/génétique , Simulation de dynamique moléculaire , Mutation , Conformation des protéines , Thermodynamique
9.
Chem Commun (Camb) ; 53(68): 9454-9457, 2017 Aug 22.
Article de Anglais | MEDLINE | ID: mdl-28795696

RÉSUMÉ

The long-standing problem of achieving high activity of a thermophilic enzyme at low temperatures and short reaction times with little tradeoff in thermostability has been solved by directed evolution, an alcohol dehydrogenase found in hot springs serving as the catalyst in enantioselective ketone reductions.


Sujet(s)
Alcohol dehydrogenase/métabolisme , Évolution moléculaire dirigée , Température , Alcohol dehydrogenase/composition chimique , Biocatalyse , Stabilité enzymatique , Cétones/composition chimique , Cétones/métabolisme , Stéréoisomérie
10.
Org Biomol Chem ; 15(19): 4122-4129, 2017 May 16.
Article de Anglais | MEDLINE | ID: mdl-28436515

RÉSUMÉ

Alcohol Dehydrogenase (ADH) enzymes catalyse the reversible reduction of prochiral ketones to the corresponding alcohols. These enzymes present two differently shaped active site pockets, which dictate their substrate scope and selectivity. In this study, we computationally evaluate the effect of two commonly reported active site mutations (I86A, and W110T) on a secondary alcohol dehydrogenase from Thermoanaerobacter brockii (TbSADH) through Molecular Dynamics simulations. Our results indicate that the introduced mutations induce dramatic changes in the shape of the active site, but most importantly they impact the substrate-enzyme interactions. We demonstrate that the combination of Molecular Dynamics simulations with the tools POVME and NCIplot corresponds to a powerful strategy for rationalising and engineering the stereoselectivity of ADH variants.


Sujet(s)
Alcohol dehydrogenase/métabolisme , Zinc/métabolisme , Alcohol dehydrogenase/composition chimique , Alcohol dehydrogenase/génétique , Domaine catalytique , Simulation de dynamique moléculaire , Mutation , Stéréoisomérie , Spécificité du substrat , Thermoanaerobacter/enzymologie
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE