Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 148
Filtrar
1.
J Chem Inf Model ; 62(17): 4032-4048, 2022 09 12.
Artículo en Inglés | MEDLINE | ID: mdl-35960209

RESUMEN

Automatic design of molecules with specific chemical and biochemical properties is an important process in material informatics and computational drug discovery. In this study, we designed a novel coarse-grained tree representation of molecules (Reversible Junction Tree; "RJT") for the aforementioned purposes, which is reversely convertible to the original molecule without external information. By leveraging this representation, we further formulated the molecular design and optimization problem as a tree-structure construction using deep reinforcement learning ("RJT-RL"). In this method, all of the intermediate and final states of reinforcement learning are convertible to valid molecules, which could efficiently guide the optimization process in simple benchmark tasks. We further examined the multiobjective optimization and fine-tuning of the reinforcement learning models using RJT-RL, demonstrating the applicability of our method to more realistic tasks in drug discovery.


Asunto(s)
Aprendizaje Profundo , Refuerzo en Psicología , Descubrimiento de Drogas , Aprendizaje
2.
Nat Commun ; 13(1): 2991, 2022 05 30.
Artículo en Inglés | MEDLINE | ID: mdl-35637178

RESUMEN

Computational material discovery is under intense study owing to its ability to explore the vast space of chemical systems. Neural network potentials (NNPs) have been shown to be particularly effective in conducting atomistic simulations for such purposes. However, existing NNPs are generally designed for narrow target materials, making them unsuitable for broader applications in material discovery. Here we report a development of universal NNP called PreFerred Potential (PFP), which is able to handle any combination of 45 elements. Particular emphasis is placed on the datasets, which include a diverse set of virtual structures used to attain the universality. We demonstrated the applicability of PFP in selected domains: lithium diffusion in LiFeSO4F, molecular adsorption in metal-organic frameworks, an order-disorder transition of Cu-Au alloys, and material discovery for a Fischer-Tropsch catalyst. They showcase the power of PFP, and this technology provides a highly useful tool for material discovery.


Asunto(s)
Estructuras Metalorgánicas , Redes Neurales de la Computación , Adsorción , Catálisis
3.
PLoS One ; 17(2): e0263296, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35113924

RESUMEN

G-protein-coupled receptors (GPCR) are a family of membrane receptors that play important roles in the regulation of various physiological phenomena. LPA receptors (LPA1-6) are members of the class A GPCRs, which transduce a lysophosphatidic acid (LPA) signal across the cell membrane and evoke various responses, including cellular survival, proliferation, differentiation, and migration. The crystal structure of LPA6 revealed a gap between its transmembrane helices (TMs), which is opened toward the membrane side. This led to the proposal of the "lateral access model," in which its lipophilic ligand directly enters the binding pocket through the gap structure at the membrane. In this study, we performed molecular dynamics (MD) simulations and Markov state model (MSM) analyses of LPA6 and LPA, to elucidate the long timescale dynamics of the ligand binding process. The results from the 71.4-µs MD simulation suggested that the flexibility of the TMs constituting the gap structure enables the lateral entrance of the ligand, and the key interactions between the receptor and ligand facilitate the transition state of the ligand binding process.


Asunto(s)
Receptores Acoplados a Proteínas G/metabolismo , Receptores del Ácido Lisofosfatídico/química , Membrana Celular/metabolismo , Simulación por Computador , Cristalografía por Rayos X , Células HEK293 , Humanos , Ligandos , Cadenas de Markov , Simulación de Dinámica Molecular , Unión Proteica , Transducción de Señal , Factor de Crecimiento Transformador alfa/química
4.
Elife ; 102021 03 23.
Artículo en Inglés | MEDLINE | ID: mdl-33752801

RESUMEN

Channelrhodopsins (ChRs) are microbial light-gated ion channels utilized in optogenetics to control neural activity with light . Light absorption causes retinal chromophore isomerization and subsequent protein conformational changes visualized as optically distinguished intermediates, coupled with channel opening and closing. However, the detailed molecular events underlying channel gating remain unknown. We performed time-resolved serial femtosecond crystallographic analyses of ChR by using an X-ray free electron laser, which revealed conformational changes following photoactivation. The isomerized retinal adopts a twisted conformation and shifts toward the putative internal proton donor residues, consequently inducing an outward shift of TM3, as well as a local deformation in TM7. These early conformational changes in the pore-forming helices should be the triggers that lead to opening of the ion conducting pore.


Asunto(s)
Proteínas Algáceas/genética , Channelrhodopsins/genética , Chlamydomonas reinhardtii/genética , Proteínas Algáceas/química , Proteínas Algáceas/metabolismo , Secuencia de Aminoácidos , Channelrhodopsins/química , Channelrhodopsins/metabolismo , Chlamydomonas reinhardtii/metabolismo , Cristalografía , Isomerismo , Conformación Proteica , Estructura Secundaria de Proteína , Alineación de Secuencia
5.
Commun Biol ; 3(1): 240, 2020 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-32415200

RESUMEN

Members of the leucine-rich repeat-containing 8 (LRRC8) protein family, composed of the five LRRC8A-E isoforms, are pore-forming components of the volume-regulated anion channel (VRAC). LRRC8A and at least one of the other LRRC8 isoforms assemble into heteromers to generate VRAC transport activities. Despite the availability of the LRRC8A structures, the structural basis of how LRRC8 isoforms other than LRRC8A contribute to the functional diversity of VRAC has remained elusive. Here, we present the structure of the human LRRC8D isoform, which enables the permeation of organic substrates through VRAC. The LRRC8D homo-hexamer structure displays a two-fold symmetric arrangement, and together with a structure-based electrophysiological analysis, revealed two key features. The pore constriction on the extracellular side is wider than that in the LRRC8A structures, which may explain the increased permeability of organic substrates. Furthermore, an N-terminal helix protrudes into the pore from the intracellular side and may be critical for gating.


Asunto(s)
Transporte Iónico/fisiología , Transducción de Señal , Microscopía por Crioelectrón , Dominios Proteicos , Isoformas de Proteínas/química , Isoformas de Proteínas/ultraestructura , Canales Aniónicos Dependientes del Voltaje/química , Canales Aniónicos Dependientes del Voltaje/ultraestructura
6.
J Med Chem ; 63(6): 3188-3204, 2020 03 26.
Artículo en Inglés | MEDLINE | ID: mdl-32134652

RESUMEN

Autotaxin (ATX, also known as ENPP2) is a predominant lysophosphatidic acid (LPA)-producing enzyme in the body, and LPA regulates various physiological functions, such as angiogenesis and wound healing, as well as pathological functions, including proliferation, metastasis, and fibrosis, via specific LPA receptors. Therefore, the ATX-LPA axis is a promising therapeutic target for dozens of diseases, including cancers, pulmonary and liver fibroses, and neuropathic pain. Previous structural studies revealed that the catalytic domain of ATX has a hydrophobic pocket and a hydrophobic channel; these serve to recognize the substrate, lysophosphatidylcholine (LPC), and deliver generated LPA to LPA receptors on the plasma membrane. Most reported ATX inhibitors bind to either the hydrophobic pocket or the hydrophobic channel. Herein, we present a unique ATX inhibitor that binds mainly to the hydrophobic pocket and also partly to the hydrophobic channel, inhibiting ATX activity with high potency and selectivity in vitro and in vivo. Notably, our inhibitor can rescue the cardia bifida (two hearts) phenotype in ATX-overexpressing zebrafish embryos.


Asunto(s)
Imidazoles/uso terapéutico , Inhibidores de Fosfodiesterasa/uso terapéutico , Hidrolasas Diéster Fosfóricas/metabolismo , Pirimidinas/uso terapéutico , Animales , Dominio Catalítico , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Cristalografía por Rayos X , Cardiopatías/prevención & control , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Imidazoles/síntesis química , Imidazoles/metabolismo , Masculino , Ratones Endogámicos C57BL , Estructura Molecular , Inhibidores de Fosfodiesterasa/síntesis química , Inhibidores de Fosfodiesterasa/metabolismo , Unión Proteica , Pirimidinas/síntesis química , Pirimidinas/metabolismo , Relación Estructura-Actividad , Pez Cebra
8.
Nat Commun ; 11(1): 858, 2020 02 12.
Artículo en Inglés | MEDLINE | ID: mdl-32051406

RESUMEN

PIWI-clade Argonaute proteins associate with PIWI-interacting RNAs (piRNAs), and silence transposons in animal gonads. Here, we report the crystal structure of the Drosophila PIWI-clade Argonaute Piwi in complex with endogenous piRNAs, at 2.9 Å resolution. A structural comparison of Piwi with other Argonautes highlights the PIWI-specific structural features, such as the overall domain arrangement and metal-dependent piRNA recognition. Our structural and biochemical data reveal that, unlike other Argonautes including silkworm Siwi, Piwi has a non-canonical DVDK tetrad and lacks the RNA-guided RNA cleaving slicer activity. Furthermore, we find that the Piwi mutant with the canonical DEDH catalytic tetrad exhibits the slicer activity and readily dissociates from less complementary RNA targets after the slicer-mediated cleavage, suggesting that the slicer activity could compromise the Piwi-mediated co-transcriptional silencing. We thus propose that Piwi lost the slicer activity during evolution to serve as an RNA-guided RNA-binding platform, thereby ensuring faithful co-transcriptional silencing of transposons.


Asunto(s)
Proteínas Argonautas/clasificación , Proteínas de Drosophila/química , Drosophila/metabolismo , Animales , Proteínas Argonautas/química , Proteínas Argonautas/genética , Bombyx/metabolismo , Línea Celular , Cristalografía por Rayos X , Elementos Transponibles de ADN/genética , Drosophila/genética , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Silenciador del Gen , Enlace de Hidrógeno , Modelos Moleculares , Conformación Proteica , Dominios Proteicos , ARN Guía de Kinetoplastida/metabolismo , ARN Interferente Pequeño/metabolismo , ARN no Traducido
9.
Biochim Biophys Acta Biomembr ; 1862(12): 183154, 2020 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-31866287

RESUMEN

Xenobiotic and metabolite extrusion is an important process for the proper functions of cells and their compartments, including acidic organelles. MATE (multidrug and toxic compound extrusion) is a large family of secondary active transporters involved in the transport of various compounds across cellular and organellar membranes, and is present in the three domains of life. The major substrates of the bacterial MATE transporters are cationic compounds, including clinically important antibiotics, and thereby MATE transporters confer multi-drug resistance to pathogenic bacteria. The plant MATE transporters are important for the accumulation of various metabolites in organelles, including vacuoles. The human MATE transporters are expressed in the brush-border membrane of the kidney, and are involved in the clearance of cationic drugs from the body. During the past decade, progress in structural biology has clarified the transport mechanism of these MATE transporters in atomic detail. The present review summarizes the reported structures of MATE family transporters, along with their structure-guided functional analyses. This integrated view of the structures of MATE transporters provides novel insights into their transport mechanism.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/metabolismo , Proteínas de Transporte de Catión Orgánico/metabolismo , Proteínas de Plantas/metabolismo , Transportadoras de Casetes de Unión a ATP/química , Transportadoras de Casetes de Unión a ATP/clasificación , Archaea/metabolismo , Proteínas Arqueales/química , Proteínas Arqueales/clasificación , Proteínas Arqueales/metabolismo , Humanos , Simulación de Dinámica Molecular , Proteínas de Transporte de Catión Orgánico/química , Proteínas de Transporte de Catión Orgánico/clasificación , Proteínas de Plantas/química , Proteínas de Plantas/clasificación , Plantas/metabolismo , Estructura Terciaria de Proteína , Especificidad por Sustrato
10.
Nat Struct Mol Biol ; 26(6): 510-517, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-31160781

RESUMEN

The L-type amino acid transporter 1 (LAT1 or SLC7A5) transports large neutral amino acids across the membrane and is crucial for brain drug delivery and tumor growth. LAT1 forms a disulfide-linked heterodimer with CD98 heavy chain (CD98hc, 4F2hc or SLC3A2), but the mechanism of assembly and amino acid transport are poorly understood. Here we report the cryo-EM structure of the human LAT1-CD98hc heterodimer at 3.3-Å resolution. LAT1 features a canonical Leu T-fold and exhibits an unusual loop structure on transmembrane helix 6, creating an extended cavity that might accommodate bulky amino acids and drugs. CD98hc engages with LAT1 through the extracellular, transmembrane and putative cholesterol-mediated interactions. We also show that two anti-CD98 antibodies recognize distinct, multiple epitopes on CD98hc but not its glycans, explaining their robust reactivities. These results reveal the principles of glycoprotein-solute carrier assembly and provide templates for improving preclinical drugs and antibodies targeting LAT1 or CD98hc.


Asunto(s)
Cadena Pesada de la Proteína-1 Reguladora de Fusión/química , Transportador de Aminoácidos Neutros Grandes 1/química , Microscopía por Crioelectrón , Cadena Pesada de la Proteína-1 Reguladora de Fusión/metabolismo , Cadena Pesada de la Proteína-1 Reguladora de Fusión/ultraestructura , Humanos , Transportador de Aminoácidos Neutros Grandes 1/metabolismo , Transportador de Aminoácidos Neutros Grandes 1/ultraestructura , Modelos Moleculares , Conformación Proteica , Pliegue de Proteína , Multimerización de Proteína
11.
Acta Crystallogr F Struct Biol Commun ; 75(Pt 5): 348-358, 2019 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-31045564

RESUMEN

Proton-dependent oligopeptide transporters (POTs) belong to the major facilitator superfamily (MFS) and transport dipeptides and tripeptides from the extracellular environment into the target cell. The human POTs PepT1 and PepT2 are also involved in the absorption of various orally ingested drugs. Previously reported structures revealed that the bacterial POTs possess 14 helices, of which H1-H6 and H7-H12 constitute the typical MFS fold and the residual two helices are involved in the cytoplasmic linker. PepTSo2 from Shewanella oneidensis is a unique POT which reportedly assembles as a 200 kDa tetramer. Although the previously reported structures suggested the importance of H12 for tetramer formation, the structural basis for the PepTSo2-specific oligomerization remains unclear owing to the lack of a high-resolution tetrameric structure. In this study, the expression and purification conditions for tetrameric PepTSo2 were optimized. A single-particle cryo-EM analysis revealed the tetrameric structure of PepTSo2 incorporated into Salipro nanoparticles at 4.1 Šresolution. Furthermore, a combination of lipidic cubic phase (LCP) crystallization and an automated data-processing system for multiple microcrystals enabled crystal structures of PepTSo2 to be determined at resolutions of 3.5 and 3.9 Å. The present structures in a lipid bilayer revealed the detailed mechanism for the tetrameric assembly of PepTSo2, in which a characteristic extracellular loop (ECL) interacts with two asparagine residues on H12 which were reported to be important for tetramerization and plays an essential role in oligomeric assembly. This study provides valuable insights into the oligomerization mechanism of this MFS-type transporter, which will further pave the way for understanding other oligomeric membrane proteins.


Asunto(s)
Proteínas Bacterianas/química , Proteínas Portadoras/química , Shewanella/química , Secuencia de Aminoácidos , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Sitios de Unión , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Clonación Molecular , Microscopía por Crioelectrón , Cristalografía por Rayos X , Escherichia coli/genética , Escherichia coli/metabolismo , Expresión Génica , Vectores Genéticos/química , Vectores Genéticos/metabolismo , Modelos Moleculares , Unión Proteica , Conformación Proteica en Hélice alfa , Conformación Proteica en Lámina beta , Dominios y Motivos de Interacción de Proteínas , Multimerización de Proteína , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Shewanella/metabolismo , Especificidad por Sustrato
12.
Nat Commun ; 10(1): 1968, 2019 04 29.
Artículo en Inglés | MEDLINE | ID: mdl-31036811

RESUMEN

The RNA-guided DNA endonuclease Cas9 cleaves double-stranded DNA targets bearing a protospacer adjacent motif (PAM) and complementarity to an RNA guide. Unlike other Cas9 orthologs, Corynebacterium diphtheriae Cas9 (CdCas9) recognizes the promiscuous NNRHHHY PAM. However, the CdCas9-mediated PAM recognition mechanism remains unknown. Here, we report the crystal structure of CdCas9 in complex with the guide RNA and its target DNA at 2.9 Å resolution. The structure reveals that CdCas9 recognizes the NNRHHHY PAM via a combination of van der Waals interactions and base-specific hydrogen bonds. Moreover, we find that CdCas9 exhibits robust DNA cleavage activity with the optimal 22-nucleotide length guide RNAs. Our findings highlight the mechanistic diversity of the PAM recognition by Cas9 orthologs, and provide a basis for the further engineering of the CRISPR-Cas9 genome-editor nucleases.


Asunto(s)
Proteína 9 Asociada a CRISPR/química , Proteína 9 Asociada a CRISPR/metabolismo , Enzimas de Restricción-Modificación del ADN/química , Enzimas de Restricción-Modificación del ADN/metabolismo , Sistemas CRISPR-Cas , Línea Celular , Corynebacterium diphtheriae/enzimología , Corynebacterium diphtheriae/metabolismo , Cristalografía por Rayos X , División del ADN , Células HEK293 , Humanos , Enlace de Hidrógeno
13.
Nat Plants ; 5(3): 308-315, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30742036

RESUMEN

The iron ion is an essential cofactor in several vital enzymatic reactions, such as DNA replication, oxygen transport, and respiratory and photosynthetic electron transfer chains, but its excess accumulation induces oxidative stress in cells. Vacuolar iron transporter 1 (VIT1) is important for iron homeostasis in plants, by transporting cytoplasmic ferrous ions into vacuoles. Modification of the VIT1 gene leads to increased iron content in crops, which could be used for the treatment of human iron deficiency diseases. Furthermore, a VIT1 from the malaria-causing parasite Plasmodium is considered as a potential drug target for malaria. Here we report the crystal structure of VIT1 from rose gum Eucalyptus grandis, which probably functions as a H+-dependent antiporter for Fe2+ and other transition metal ions. VIT1 adopts a novel protein fold forming a dimer of five membrane-spanning domains, with an ion-translocating pathway constituted by the conserved methionine and carboxylate residues at the dimer interface. The second transmembrane helix protrudes from the lipid membrane by about 40 Å and connects to a three-helical bundle, triangular cytoplasmic domain, which binds to the substrate metal ions and stabilizes their soluble form, thus playing an essential role in their transport. These mechanistic insights will provide useful information for the further design of genetically modified crops and the development of anti-malaria drugs.


Asunto(s)
Proteínas de Transporte de Catión/química , Aceite de Eucalipto/química , Proteínas de Transporte de Catión/metabolismo , Cristalografía por Rayos X , Citoplasma/metabolismo , Hierro/metabolismo , Modelos Moleculares , Conformación Proteica , Pliegue de Proteína , Multimerización de Proteína , Vacuolas/metabolismo
14.
Science ; 363(6423)2019 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-30467178

RESUMEN

N 6-methyladenosine (m6A), a major modification of messenger RNAs (mRNAs), plays critical roles in RNA metabolism and function. In addition to the internal m6A, N 6, 2'-O-dimethyladenosine (m6Am) is present at the transcription start nucleotide of capped mRNAs in vertebrates. However, its biogenesis and functional role remain elusive. Using a reverse genetics approach, we identified PCIF1, a factor that interacts with the serine-5-phosphorylated carboxyl-terminal domain of RNA polymerase II, as a cap-specific adenosine methyltransferase (CAPAM) responsible for N 6-methylation of m6Am. The crystal structure of CAPAM in complex with substrates revealed the molecular basis of cap-specific m6A formation. A transcriptome-wide analysis revealed that N 6-methylation of m6Am promotes the translation of capped mRNAs. Thus, a cap-specific m6A writer promotes translation of mRNAs starting from m6Am.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/química , Metiltransferasas/química , Proteínas Nucleares/química , Caperuzas de ARN/química , ARN Polimerasa II/química , Técnicas de Inactivación de Genes , Células HEK293 , Humanos , Espectrometría de Masas , Metilación , Biosíntesis de Proteínas , Dominios Proteicos , Sitio de Iniciación de la Transcripción
15.
Structure ; 27(2): 293-301.e3, 2019 02 05.
Artículo en Inglés | MEDLINE | ID: mdl-30449688

RESUMEN

Multidrug and toxic compound extrusion (MATE) transporters efflux toxic compounds using a Na+ or H+ gradient across the membrane. Although the structures of MATE transporters have been reported, the cation-coupled substrate transport mechanism remains controversial. Here we report crystal structures of VcmN, a Vibrio cholerae MATE transporter driven by the H+ gradient. High-resolution structures in two distinct conformations associated with different pHs revealed that the rearrangement of the hydrogen-bonding network around the conserved Asp35 induces the bending of transmembrane helix 1, as in the case of the H+-coupled Pyrococcus furiosus MATE transporter. We also determined the crystal structure of the D35N mutant, which captured a unique conformation of TM1 facilitated by an altered hydrogen-bonding network. Based on the present results, we propose a common step in the transport cycle shared among prokaryotic H+-coupled MATE transporters.


Asunto(s)
Mutación , Proteínas de Transporte de Catión Orgánico/química , Vibrio cholerae/metabolismo , Asparagina/metabolismo , Proteínas Bacterianas/metabolismo , Cristalografía por Rayos X , Enlace de Hidrógeno , Modelos Moleculares , Proteínas de Transporte de Catión Orgánico/genética , Conformación Proteica , Vibrio cholerae/genética
16.
Acta Crystallogr F Struct Biol Commun ; 74(Pt 12): 810-816, 2018 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-30511676

RESUMEN

The type VI secretion system (T6SS) comprises needle-shaped multisubunit complexes that play a role in the microbial defense systems of Gram-negative bacteria. Some Gram-negative bacteria harboring a T6SS deliver toxic effector proteins into the cytoplasm or periplasm of competing bacteria in order to lyse and kill them. To avoid self-cell disruption, these bacteria have cognate immunity proteins that inhibit their toxic effector proteins. T6SS amidase effector protein 4 (Tae4) and T6SS amidase immunity protein 4 (Tai4) are a representative of the toxic effector-immunity pairs of the T6SS. Here, the three-dimensional structures of Tai4 and the Tae4-Tai4 complex from Agrobacterium tumefaciens are reported at 1.55 and 1.9 Šresolution, respectively. A structural comparison with other Tae4-Tai4 homologs revealed similarities and differences in the catalytic and inhibitory mechanisms among the Tae4 and Tai4 family proteins.


Asunto(s)
Agrobacterium tumefaciens/química , Agrobacterium tumefaciens/inmunología , Proteínas Bacterianas/química , Proteínas Bacterianas/inmunología , Sistemas de Secreción Bacterianos/química , Sistemas de Secreción Bacterianos/inmunología , Agrobacterium tumefaciens/genética , Secuencia de Aminoácidos , Proteínas Bacterianas/genética , Sistemas de Secreción Bacterianos/genética , Cristalización , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína
17.
Nat Commun ; 9(1): 4424, 2018 10 24.
Artículo en Inglés | MEDLINE | ID: mdl-30356045

RESUMEN

ENPP1 (Ecto-nucleotide pyrophosphatase phosphodiesterase 1), a type II transmembrane glycoprotein, hydrolyzes ATP to produce AMP and diphosphate, thereby inhibiting bone mineralization. A recent study showed that ENPP1 also preferentially hydrolyzes 2'3'-cGAMP (cyclic GMP-AMP) but not its linkage isomer 3'3'-cGAMP, and negatively regulates the cGAS-STING pathway in the innate immune system. Here, we present the high-resolution crystal structures of ENPP1 in complex with 3'3'-cGAMP and the reaction intermediate pA(3',5')pG. The structures revealed that the adenine and guanine bases of the dinucleotides are recognized by nucleotide- and guanine-pockets, respectively. Furthermore, the structures indicate that 2'3'-cGAMP, but not 3'3'-cGAMP, binds to the active site in a conformation suitable for catalysis, thereby explaining the specific degradation of 2'3'-cGAMP by ENPP1. Our findings provide insights into how ENPP1 hydrolyzes both ATP and cGAMP to participate in the two distinct biological processes.


Asunto(s)
Nucleótidos Cíclicos/química , Nucleótidos Cíclicos/metabolismo , Hidrolasas Diéster Fosfóricas/metabolismo , Pirofosfatasas/metabolismo , Adenosina Trifosfato/metabolismo , Catálisis , Línea Celular , Línea Celular Tumoral , Células HEK293 , Humanos , Proteínas de la Membrana/metabolismo , Hidrolasas Diéster Fosfóricas/química , Estructura Secundaria de Proteína , Pirofosfatasas/química , Transducción de Señal/fisiología
18.
Science ; 361(6408): 1259-1262, 2018 09 21.
Artículo en Inglés | MEDLINE | ID: mdl-30166441

RESUMEN

The RNA-guided endonuclease Cas9 cleaves its target DNA and is a powerful genome-editing tool. However, the widely used Streptococcus pyogenes Cas9 enzyme (SpCas9) requires an NGG protospacer adjacent motif (PAM) for target recognition, thereby restricting the targetable genomic loci. Here, we report a rationally engineered SpCas9 variant (SpCas9-NG) that can recognize relaxed NG PAMs. The crystal structure revealed that the loss of the base-specific interaction with the third nucleobase is compensated by newly introduced non-base-specific interactions, thereby enabling the NG PAM recognition. We showed that SpCas9-NG induces indels at endogenous target sites bearing NG PAMs in human cells. Furthermore, we found that the fusion of SpCas9-NG and the activation-induced cytidine deaminase (AID) mediates the C-to-T conversion at target sites with NG PAMs in human cells.


Asunto(s)
Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Sistemas CRISPR-Cas , Endonucleasas/química , Endonucleasas/genética , Edición Génica , Proteína 9 Asociada a CRISPR , Cristalografía por Rayos X , Células HEK293 , Humanos , Ingeniería de Proteínas
19.
J Phys Chem B ; 122(42): 9681-9696, 2018 10 25.
Artículo en Inglés | MEDLINE | ID: mdl-30252477

RESUMEN

Magnesium ions (Mg2+) are crucial for various biological processes. A bacterial Mg2+ channel, MgtE, tightly regulates the intracellular Mg2+ concentration. Previous X-ray crystal structures showed that MgtE forms a dimeric structure composed of a total of 10 transmembrane α helices forming a central pore, and intracellular soluble domains constituting a Mg2+ sensor. The ion selectivity for Mg2+ over Ca2+ resides at a central cavity in the transmembrane pore of MgtE, involving a conserved aspartate residue (Asp432) from each monomer. Here, we applied ion-exchange-induced difference FTIR spectroscopy to analyze the interactions between MgtE and divalent cations, Mg2+ and Ca2+. Using site-directed mutagenesis, vibrational bands at 1421 (Mg2+), 1407 (Mg2+), ∼1440 (Ca2+), and 1390 (Ca2+) cm-1 were assigned to symmetric carboxylate stretching modes of Asp432, involved in the ion coordination. Conservative modifications of the central cavity by Asp432Glu or Ala417Leu mutations resulted in the disappearance of the Mg2+-sensitive carboxylate bands, suggesting a highly optimized geometry for accommodating a Mg2+ ion. The dependency of the vibrational changes on Mg2+ and Ca2+ concentrations revealed the presence of a two different classes of binding sites: a high affinity site for Mg2+ ( Kd ≈ 0.3 mM) with low Ca2+ affinity ( Kd ≈ 80 mM), and a medium affinity site for Mg2+ ( Kd ≈ 2 mM) and Ca2+ ( Kd ≈ 6 mM), tentatively assigned to the central cavity and the sensor domain, respectively. With the aid of molecular dynamics simulation and normal-mode analysis by quantum chemistry, we confirm that changes in carboxylate bands of the high affinity binding site originate from Asp432 in the central cavity.

20.
Biophys J ; 115(7): 1281-1291, 2018 10 02.
Artículo en Inglés | MEDLINE | ID: mdl-30236783

RESUMEN

Channelrhodopsins (ChRs) are microbial light-gated ion channels with a retinal chromophore and are widely utilized in optogenetics to precisely control neuronal activity with light. Despite increasing understanding of their structures and photoactivation kinetics, the atomistic mechanism of light gating and ion conduction remains elusive. Here, we present an atomic structural model of a chimeric ChR in a precursor state of the channel opening determined by an accurate hybrid molecular simulation technique and a statistical theory of internal water distribution. The photoactivated structure features extensive tilt of the chromophore accompanied by redistribution of water molecules in its binding pocket, which is absent in previously known photoactivated structures of analogous photoreceptors, and widely agrees with structural and spectroscopic experimental evidence of ChRs. The atomistic model manifests a photoactivated ion-conduction pathway that is markedly different from a previously proposed one and successfully explains experimentally observed mutagenic effects on key channel properties.


Asunto(s)
Channelrhodopsins/química , Channelrhodopsins/metabolismo , Activación del Canal Iónico/efectos de la radiación , Luz , Modelos Moleculares , Conformación Proteica/efectos de la radiación , Termodinámica
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...