Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 780
Filtrar
1.
Prep Biochem Biotechnol ; 53(6): 672-682, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36244016

RESUMEN

Lipopolysaccharide (LPS), as a stubborn contamination, should be monitored and kept in an acceptable level during the pharmaceutical production process. Recombinant hepatitis B surface antigen (r-HBsAg) is one of the recombinant biological products, which is probable to suffer from extrinsic endotoxin due to its long and complex production process. This research aims to assess the potential interaction between LPS and r-HBsAg by recruiting immunoaffinity chromatography (IAC) as a novel tool to quantify the interaction. Molecular modeling was performed on the HBsAg molecule to theoretically predict its potential binding and interaction sites. Then dynamic light scattering (DLS) analysis was implemented on HBsAg, LPS, and mixtures of them to reveal the interaction. The virus-like particle (VLP) structure of HBsAg and the ribbon-like structure of LPS were visualized by transmission electron microscopy (TEM). Finally, the interaction was quantified by applying various LPS/HBsAg ratios ranging from 1.67 to 120 EU/dose in the IAC. Consequently, the LPS/HBsAg ratios in the eluate were measured from 1.67 to a maximum of 92.5 EU/dose. The results indicated that 77 to 100% of total LPS interacted with HBsAg by an inverse relationship to the incubated LPS concentration. The findings implied that the introduced procedure is remarkably practical in the quantification of LPS interaction with a target recombinant protein.


Asunto(s)
Cromatografía de Afinidad , Antígenos de Superficie de la Hepatitis B , Lipopolisacáridos , Proteínas Recombinantes , Lipopolisacáridos/análisis , Proteínas Recombinantes/aislamiento & purificación , Proteínas Recombinantes/ultraestructura , Antígenos de Superficie de la Hepatitis B/química , Antígenos de Superficie de la Hepatitis B/genética , Antígenos de Superficie de la Hepatitis B/aislamiento & purificación , Antígenos de Superficie de la Hepatitis B/ultraestructura , Microscopía Electrónica de Transmisión , Vacunas contra Hepatitis B/química , Vacunas contra Hepatitis B/genética , Vacunas contra Hepatitis B/aislamiento & purificación , Modelos Químicos , Secuencia de Aminoácidos , Dispersión Dinámica de Luz , Cromatografía de Afinidad/métodos
2.
Nat Commun ; 13(1): 734, 2022 02 08.
Artículo en Inglés | MEDLINE | ID: mdl-35136046

RESUMEN

AMPA-type glutamate receptors (AMPARs) mediate rapid signal transmission at excitatory synapses in the brain. Glutamate binding to the receptor's ligand-binding domains (LBDs) leads to ion channel activation and desensitization. Gating kinetics shape synaptic transmission and are strongly modulated by transmembrane AMPAR regulatory proteins (TARPs) through currently incompletely resolved mechanisms. Here, electron cryo-microscopy structures of the GluA1/2 TARP-γ8 complex, in both open and desensitized states (at 3.5 Å), reveal state-selective engagement of the LBDs by the large TARP-γ8 loop ('ß1'), elucidating how this TARP stabilizes specific gating states. We further show how TARPs alter channel rectification, by interacting with the pore helix of the selectivity filter. Lastly, we reveal that the Q/R-editing site couples the channel constriction at the filter entrance to the gate, and forms the major cation binding site in the conduction path. Our results provide a mechanistic framework of how TARPs modulate AMPAR gating and conductance.


Asunto(s)
Canales de Calcio/metabolismo , Receptores AMPA/metabolismo , Animales , Canales de Calcio/genética , Canales de Calcio/aislamiento & purificación , Canales de Calcio/ultraestructura , Microscopía por Crioelectrón , Ácido Glutámico/metabolismo , Células HEK293 , Humanos , Mutación , Técnicas de Placa-Clamp , Dominios Proteicos/genética , Ratas , Receptores AMPA/genética , Receptores AMPA/aislamiento & purificación , Receptores AMPA/ultraestructura , Proteínas Recombinantes/genética , Proteínas Recombinantes/aislamiento & purificación , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/ultraestructura , Transmisión Sináptica , Transfección
3.
Nat Commun ; 13(1): 731, 2022 02 08.
Artículo en Inglés | MEDLINE | ID: mdl-35136060

RESUMEN

Lysophospholipids are bioactive lipids and can signal through G-protein-coupled receptors (GPCRs). The best studied lysophospholipids are lysophosphatidic acid (LPA) and sphingosine 1-phosphate (S1P). The mechanisms of lysophospholipid recognition by an active GPCR, and the activations of lysophospholipid GPCR-G-protein complexes remain unclear. Here we report single-particle cryo-EM structures of human S1P receptor 1 (S1P1) and heterotrimeric Gi complexes formed with bound S1P or the multiple sclerosis (MS) treatment drug Siponimod, as well as human LPA receptor 1 (LPA1) and Gi complexes in the presence of LPA. Our structural and functional data provide insights into how LPA and S1P adopt different conformations to interact with their cognate GPCRs, the selectivity of the homologous lipid GPCRs for S1P versus LPA, and the different activation mechanisms of these GPCRs by LPA and S1P. Our studies also reveal specific optimization strategies to improve the MS-treating S1P1-targeting drugs.


Asunto(s)
Subunidades alfa de la Proteína de Unión al GTP Gi-Go/metabolismo , Receptores del Ácido Lisofosfatídico/metabolismo , Receptores de Esfingosina-1-Fosfato/metabolismo , Animales , Azetidinas/farmacología , Azetidinas/uso terapéutico , Compuestos de Bencilo/farmacología , Compuestos de Bencilo/uso terapéutico , Microscopía por Crioelectrón , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/genética , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/aislamiento & purificación , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/ultraestructura , Humanos , Lisofosfolípidos/metabolismo , Conformación Molecular/efectos de los fármacos , Simulación del Acoplamiento Molecular , Esclerosis Múltiple/tratamiento farmacológico , Esclerosis Múltiple/patología , Receptores del Ácido Lisofosfatídico/genética , Receptores del Ácido Lisofosfatídico/aislamiento & purificación , Receptores del Ácido Lisofosfatídico/ultraestructura , Proteínas Recombinantes/genética , Proteínas Recombinantes/aislamiento & purificación , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/ultraestructura , Células Sf9 , Imagen Individual de Molécula , Esfingosina/análogos & derivados , Esfingosina/metabolismo , Receptores de Esfingosina-1-Fosfato/genética , Receptores de Esfingosina-1-Fosfato/aislamiento & purificación , Receptores de Esfingosina-1-Fosfato/ultraestructura , Spodoptera
4.
Nat Commun ; 13(1): 749, 2022 02 08.
Artículo en Inglés | MEDLINE | ID: mdl-35136069

RESUMEN

Tousled-like kinases (TLKs) are nuclear serine-threonine kinases essential for genome maintenance and proper cell division in animals and plants. A major function of TLKs is to phosphorylate the histone chaperone proteins ASF1a and ASF1b to facilitate DNA replication-coupled nucleosome assembly, but how TLKs selectively target these critical substrates is unknown. Here, we show that TLK2 selectivity towards ASF1 substrates is achieved in two ways. First, the TLK2 catalytic domain recognizes consensus phosphorylation site motifs in the ASF1 C-terminal tail. Second, a short sequence at the TLK2 N-terminus docks onto the ASF1a globular N-terminal domain in a manner that mimics its histone H3 client. Disrupting either catalytic or non-catalytic interactions through mutagenesis hampers ASF1 phosphorylation by TLK2 and cell growth. Our results suggest that the stringent selectivity of TLKs for ASF1 is enforced by an unusual interaction mode involving mutual recognition of a short sequence motifs by both kinase and substrate.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Chaperonas Moleculares/metabolismo , Imitación Molecular , Proteínas Quinasas/metabolismo , Secuencias de Aminoácidos/genética , Secuencia de Aminoácidos , Dominio Catalítico/genética , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/aislamiento & purificación , Proteínas de Ciclo Celular/ultraestructura , Secuencia Conservada , Cristalografía por Rayos X , Histonas/metabolismo , Humanos , Chaperonas Moleculares/genética , Chaperonas Moleculares/aislamiento & purificación , Chaperonas Moleculares/ultraestructura , Simulación del Acoplamiento Molecular , Mutagénesis , Biblioteca de Péptidos , Fosforilación , Proteínas Quinasas/genética , Proteínas Quinasas/aislamiento & purificación , Proteínas Quinasas/ultraestructura , Proteínas Recombinantes/genética , Proteínas Recombinantes/aislamiento & purificación , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/ultraestructura , Especificidad por Sustrato
5.
Nat Commun ; 13(1): 923, 2022 02 17.
Artículo en Inglés | MEDLINE | ID: mdl-35177668

RESUMEN

N-methyl-D-aspartate receptors (NMDARs) are critically involved in basic brain functions and neurodegeneration as well as tumor invasiveness. Targeting specific subtypes of NMDARs with distinct activities has been considered an effective therapeutic strategy for neurological disorders and diseases. However, complete elimination of off-target effects of small chemical compounds has been challenging and thus, there is a need to explore alternative strategies for targeting NMDAR subtypes. Here we report identification of a functional antibody that specifically targets the GluN1-GluN2B NMDAR subtype and allosterically down-regulates ion channel activity as assessed by electrophysiology. Through biochemical analysis, x-ray crystallography, single-particle electron cryomicroscopy, and molecular dynamics simulations, we show that this inhibitory antibody recognizes the amino terminal domain of the GluN2B subunit and increases the population of the non-active conformational state. The current study demonstrates that antibodies may serve as specific reagents to regulate NMDAR functions for basic research and therapeutic objectives.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Receptores de N-Metil-D-Aspartato/antagonistas & inhibidores , Animales , Anticuerpos Monoclonales/genética , Anticuerpos Monoclonales/aislamiento & purificación , Anticuerpos Monoclonales/ultraestructura , Clonación Molecular , Microscopía por Crioelectrón , Cristalografía por Rayos X , Fragmentos Fab de Inmunoglobulinas/genética , Fragmentos Fab de Inmunoglobulinas/aislamiento & purificación , Fragmentos Fab de Inmunoglobulinas/farmacología , Fragmentos Fab de Inmunoglobulinas/ultraestructura , Región Variable de Inmunoglobulina/genética , Región Variable de Inmunoglobulina/aislamiento & purificación , Región Variable de Inmunoglobulina/farmacología , Región Variable de Inmunoglobulina/ultraestructura , Simulación de Dinámica Molecular , Oocitos , Ratas , Receptores de N-Metil-D-Aspartato/genética , Receptores de N-Metil-D-Aspartato/aislamiento & purificación , Proteínas Recombinantes/genética , Proteínas Recombinantes/aislamiento & purificación , Proteínas Recombinantes/farmacología , Proteínas Recombinantes/ultraestructura , Células Sf9 , Spodoptera , Xenopus laevis
6.
Nat Commun ; 12(1): 7057, 2021 12 03.
Artículo en Inglés | MEDLINE | ID: mdl-34862374

RESUMEN

In response to genotoxic stress, the tumor suppressor p53 acts as a transcription factor by regulating the expression of genes critical for cancer prevention. Mutations in the gene encoding p53 are associated with cancer development. PRIMA-1 and eprenetapopt (APR-246/PRIMA-1MET) are small molecules that are converted into the biologically active compound, methylene quinuclidinone (MQ), shown to reactivate mutant p53 by binding covalently to cysteine residues. Here, we investigate the structural basis of mutant p53 reactivation by MQ based on a series of high-resolution crystal structures of cancer-related and wild-type p53 core domains bound to MQ in their free state and in complexes with their DNA response elements. Our data demonstrate that MQ binds to several cysteine residues located at the surface of the core domain. The structures reveal a large diversity in MQ interaction modes that stabilize p53 and its complexes with DNA, leading to a common global effect that is pertinent to the restoration of non-functional p53 proteins.


Asunto(s)
Antineoplásicos/farmacología , Compuestos Aza/farmacología , Compuestos Bicíclicos Heterocíclicos con Puentes/farmacología , Neoplasias/tratamiento farmacológico , Quinuclidinas/farmacología , Proteína p53 Supresora de Tumor/agonistas , Antineoplásicos/uso terapéutico , Compuestos Aza/química , Compuestos Aza/uso terapéutico , Compuestos Bicíclicos Heterocíclicos con Puentes/química , Compuestos Bicíclicos Heterocíclicos con Puentes/uso terapéutico , Cristalografía por Rayos X , Humanos , Mutación con Pérdida de Función/efectos de los fármacos , Neoplasias/genética , Dominios Proteicos/efectos de los fármacos , Quinuclidinas/química , Quinuclidinas/uso terapéutico , Proteínas Recombinantes/genética , Proteínas Recombinantes/aislamiento & purificación , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/ultraestructura , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/aislamiento & purificación , Proteína p53 Supresora de Tumor/ultraestructura
7.
Biochemistry ; 60(47): 3644-3658, 2021 11 30.
Artículo en Inglés | MEDLINE | ID: mdl-34730940

RESUMEN

The aggregation of α-synuclein (αSN) and increased oxidative stress leading to lipid peroxidation are pathological characteristics of Parkinson's disease (PD). Here, we report that aggregation of αSN in the presence of lipid peroxidation products 4-hydroxy-2-nonenal (HNE) and 4-oxo-2-nonenal (ONE) increases the stability and the yield of αSN oligomers (αSO). Further, we show that ONE is more efficient than HNE at inducing αSO. In addition, we demonstrate that the two αSO differ in both size and shape. ONE-αSO are smaller in size than HNE-αSO, except when they are formed at a high molar excess of aldehyde. In both monomeric and oligomeric αSN, His50 is the main target of HNE modification, and HNE-induced oligomerization is severely retarded in the mutant His50Ala αSN. In contrast, ONE-induced aggregation of His50Ala αSN occurs readily, demonstrating the different pathways for inducing αSN aggregation by HNE and ONE. Our results show different morphologies of the HNE-treated and ONE-treated αSO and different roles of His50 in their modification of αSN, but we also observe structural similarities between these αSO and the non-treated αSO, e.g., flexible C-terminus, a folded core composed of the N-terminal and NAC region. Furthermore, HNE-αSO show a similar deuterium uptake as a previously characterized oligomer formed by non-treated αSO, suggesting that the backbone conformational dynamics of their folded cores resemble one another.


Asunto(s)
Aldehídos/metabolismo , Enfermedad de Parkinson/patología , alfa-Sinucleína/metabolismo , Aldehídos/química , Línea Celular Tumoral , Humanos , Peroxidación de Lípido , Resonancia Magnética Nuclear Biomolecular , Agregado de Proteínas , Proteínas Recombinantes/química , Proteínas Recombinantes/aislamiento & purificación , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/ultraestructura , Dispersión del Ángulo Pequeño , Difracción de Rayos X , alfa-Sinucleína/química , alfa-Sinucleína/aislamiento & purificación , alfa-Sinucleína/ultraestructura
8.
Nat Commun ; 12(1): 6805, 2021 11 23.
Artículo en Inglés | MEDLINE | ID: mdl-34815401

RESUMEN

GPR158, a class C orphan GPCR, functions in cognition, stress-induced mood control, and synaptic development. Among class C GPCRs, GPR158 is unique as it lacks a Venus flytrap-fold ligand-binding domain and terminates Gαi/o protein signaling through the RGS7-Gß5 heterodimer. Here, we report the cryo-EM structures of GPR158 alone and in complex with one or two RGS7-Gß5 heterodimers. GPR158 dimerizes through Per-Arnt-Sim-fold extracellular and transmembrane (TM) domains connected by an epidermal growth factor-like linker. The TM domain (TMD) reflects both inactive and active states of other class C GPCRs: a compact intracellular TMD, conformations of the two intracellular loops (ICLs) and the TMD interface formed by TM4/5. The ICL2, ICL3, TM3, and first helix of the cytoplasmic coiled-coil provide a platform for the DHEX domain of one RGS7 and the second helix recruits another RGS7. The unique features of the RGS7-binding site underlie the selectivity of GPR158 for RGS7.


Asunto(s)
Subunidades beta de la Proteína de Unión al GTP/ultraestructura , Proteínas RGS/ultraestructura , Receptores Acoplados a Proteínas G/ultraestructura , Microscopía por Crioelectrón , Subunidades beta de la Proteína de Unión al GTP/genética , Subunidades beta de la Proteína de Unión al GTP/aislamiento & purificación , Subunidades beta de la Proteína de Unión al GTP/metabolismo , Células HEK293 , Humanos , Proteínas RGS/genética , Proteínas RGS/aislamiento & purificación , Proteínas RGS/metabolismo , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/aislamiento & purificación , Receptores Acoplados a Proteínas G/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/aislamiento & purificación , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/ultraestructura
9.
Nat Commun ; 12(1): 6103, 2021 10 20.
Artículo en Inglés | MEDLINE | ID: mdl-34671049

RESUMEN

Multiple SARS-CoV-2 variants of concern (VOCs) have been emerging and some have been linked to an increase in case numbers globally. However, there is yet a lack of understanding of the molecular basis for the interactions between the human ACE2 (hACE2) receptor and these VOCs. Here we examined several VOCs including Alpha, Beta, and Gamma, and demonstrate that five variants receptor-binding domain (RBD) increased binding affinity for hACE2, and four variants pseudoviruses increased entry into susceptible cells. Crystal structures of hACE2-RBD complexes help identify the key residues facilitating changes in hACE2 binding affinity. Additionally, soluble hACE2 protein efficiently prevent most of the variants pseudoviruses. Our findings provide important molecular information and may help the development of novel therapeutic and prophylactic agents targeting these emerging mutants.


Asunto(s)
Enzima Convertidora de Angiotensina 2/metabolismo , COVID-19/virología , Dominios y Motivos de Interacción de Proteínas/genética , SARS-CoV-2/metabolismo , Glicoproteína de la Espiga del Coronavirus/metabolismo , Secuencia de Aminoácidos , Enzima Convertidora de Angiotensina 2/genética , Enzima Convertidora de Angiotensina 2/aislamiento & purificación , Enzima Convertidora de Angiotensina 2/ultraestructura , Animales , Línea Celular Tumoral , Cristalografía por Rayos X , Células HEK293 , Humanos , Simulación de Dinámica Molecular , Mutación , Proteínas Recombinantes/genética , Proteínas Recombinantes/aislamiento & purificación , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/ultraestructura , SARS-CoV-2/genética , Células Sf9 , Glicoproteína de la Espiga del Coronavirus/genética , Glicoproteína de la Espiga del Coronavirus/aislamiento & purificación , Glicoproteína de la Espiga del Coronavirus/ultraestructura , Spodoptera , Resonancia por Plasmón de Superficie , Acoplamiento Viral , Internalización del Virus
10.
Molecules ; 26(20)2021 Oct 14.
Artículo en Inglés | MEDLINE | ID: mdl-34684779

RESUMEN

Caveolin-1 (CAV1), a membrane protein that is necessary for the formation and maintenance of caveolae, is a promising drug target for the therapy of various diseases, such as cancer, diabetes, and liver fibrosis. The biology and pathology of caveolae have been widely investigated; however, very little information about the structural features of full-length CAV1 is available, as well as its biophysical role in reshaping the cellular membrane. Here, we established a method, with high reliability and reproducibility, for the expression and purification of CAV1. Amyloid-like properties of CAV1 and its C-terminal peptide CAV1(168-178) suggest a structural basis for the short linear CAV1 assemblies that have been recently observed in caveolin polyhedral cages in Escherichia coli (E. coli). Reconstitution of CAV1 into artificial lipid membranes induces a caveolae-like membrane curvature. Structural characterization of CAV1 in the membrane by solid-state nuclear magnetic resonance (ssNMR) indicate that it is largely α-helical, with very little ß-sheet content. Its scaffolding domain adopts a α-helical structure as identified by chemical shift analysis of threonine (Thr). Taken together, an in vitro model was developed for the CAV1 structural study, which will further provide meaningful evidences for the design and screening of bioactive compounds targeting CAV1.


Asunto(s)
Caveolina 1/química , Secuencia de Aminoácidos , Caveolina 1/genética , Caveolina 1/ultraestructura , Humanos , Espectroscopía de Resonancia Magnética , Lípidos de la Membrana/química , Membranas Artificiales , Microscopía Electrónica de Transmisión , Conformación Proteica en Hélice alfa , Conformación Proteica en Lámina beta , Estructura Cuaternaria de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/ultraestructura
11.
Nat Commun ; 12(1): 5690, 2021 09 28.
Artículo en Inglés | MEDLINE | ID: mdl-34584093

RESUMEN

SLC4 transporters play significant roles in pH regulation and cellular sodium transport. The previously solved structures of the outward facing (OF) conformation for AE1 (SLC4A1) and NBCe1 (SLC4A4) transporters revealed an identical overall fold despite their different transport modes (chloride/bicarbonate exchange versus sodium-carbonate cotransport). However, the exact mechanism determining the different transport modes in the SLC4 family remains unknown. In this work, we report the cryo-EM 3.4 Å structure of the OF conformation of NDCBE (SLC4A8), which shares transport properties with both AE1 and NBCe1 by mediating the electroneutral exchange of sodium-carbonate with chloride. This structure features a fully resolved extracellular loop 3 and well-defined densities corresponding to sodium and carbonate ions in the tentative substrate binding pocket. Further, we combine computational modeling with functional studies to unravel the molecular determinants involved in NDCBE and SLC4 transport.


Asunto(s)
Simportadores de Sodio-Bicarbonato/ultraestructura , Células HEK293 , Humanos , Proteínas Recombinantes/genética , Proteínas Recombinantes/aislamiento & purificación , Proteínas Recombinantes/ultraestructura , Simportadores de Sodio-Bicarbonato/genética , Simportadores de Sodio-Bicarbonato/aislamiento & purificación
12.
Biol Pharm Bull ; 44(9): 1196-1201, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34471047

RESUMEN

Peroxisome proliferator-activated receptor gamma (PPARγ) is a nuclear receptor and the molecular target of thiazolidinedione-class antidiabetic drugs. It has been reported that the loss of function R288H mutation in the human PPARγ ligand-binding domain (LBD) may be associated with the onset of colon cancer. A previous in vitro study showed that this mutation dampens 15-deoxy-Δ12,14-prostaglandin J2 (15d-PGJ2, a natural PPARγ agonist)-dependent transcriptional activation; however, it is poorly understood why the function of the R288H mutant is impaired and what role this arginine (Arg) residue plays. In this study, we found that the apo-form of R288H PPARγ mutant displays several altered conformational arrangements of the amino acid side chains in LBD: 1) the loss of a salt bridge between Arg288 and Glu295 leads to increased helix 3 movement; 2) closer proximity of Gln286 and His449 via a hydrogen bond, and closer proximity of Cys285 and Phe363 via hydrophobic interaction, stabilize the helix 3-helix 11 interaction; and 3) there is steric hindrance between Cys285/Gln286/Ser289/His449 and the flexible ligands 15d-PGJ2, 6-oxotetracosahexaenoic acid (6-oxoTHA), and 17-oxodocosahexaenoic acid (17-oxoDHA). These results suggest why Arg288 plays an important role in ligand binding and why the R288H mutation is disadvantageous for flexible ligand binding.


Asunto(s)
PPAR gamma/genética , Sustitución de Aminoácidos , Animales , Arginina/genética , Células COS , Chlorocebus aethiops , Cristalografía por Rayos X , Histidina/genética , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Ligandos , Mutación con Pérdida de Función , PPAR gamma/aislamiento & purificación , PPAR gamma/metabolismo , PPAR gamma/ultraestructura , Dominios Proteicos/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/aislamiento & purificación , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/ultraestructura , Relación Estructura-Actividad
13.
Biol Pharm Bull ; 44(9): 1202-1209, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34471048

RESUMEN

Peroxisome proliferator-activated receptor (PPAR)α, a member of the nuclear receptor family, is a transcription factor that regulates the expression of genes related to lipid metabolism in a ligand-dependent manner, and has attracted attention as a target for hypolipidemic drugs. We have been developing phenylpropaonic acid derivatives as PPARα-targeted drug candidates for the treatment of metabolic diseases. Recently, we have developed the "ligand-exchange soaking method," which crystallizes the recombinant PPARα ligand-binding domain (LBD) as a complex with intrinsic fatty acids derived from an expression host Escherichia (E.) coli and thereafter replaces them with other higher-affinity ligands by soaking. Here we applied this method for preparation of cocrystals of PPARα LBD with its ligands that have not been obtained with the conventional cocrystallization method. We revealed the high-resolution structures of the cocrystals of PPARα LBD and the three synthetic phenylpropaonic acid derivatives: TIPP-703, APHM19, and YN4pai, the latter two of which are the first observations. The overall structures of cocrystals obtained from the two methods are identical and illustrate the close interaction between these ligands and the surrounding amino acid residues of PPARα LBD. This ligand-exchange soaking method could be applicable to high throughput preparations of co-crystals with another subtype PPARδ LBD for high resolution X-ray crystallography, because it also crystallizes in complex with intrinsic fatty acid(s) while not in the apo-form.


Asunto(s)
PPAR alfa/ultraestructura , Fenilpropionatos/metabolismo , Dominios Proteicos , Humanos , Ligandos , PPAR alfa/aislamiento & purificación , PPAR alfa/metabolismo , Fenilpropionatos/química , Proteínas Recombinantes/aislamiento & purificación , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/ultraestructura , Difracción de Rayos X
14.
Biol Pharm Bull ; 44(9): 1210-1219, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34471049

RESUMEN

Peroxisome proliferator-activated receptors (PPARs) are nuclear receptor-type transcription factors that consist of three subtypes (α, γ, and ß/δ) with distinct functions and PPAR dual/pan agonists are expected to be the next generation of drugs for metabolic diseases. Saroglitazar is the first clinically approved PPARα/γ dual agonist for treatment of diabetic dyslipidemia and is currently in clinical trials to treat non-alcoholic fatty liver disease (NAFLD); however, the structural information of its interaction with PPARα/γ remains unknown. We recently revealed the high-resolution co-crystal structure of saroglitazar and the PPARα-ligand binding domain (LBD) through X-ray crystallography, and in this study, we report the structure of saroglitazar and the PPARγ-LBD. Saroglitazar was located at the center of "Y"-shaped PPARγ-ligand-binding pocket (LBP), just as it was in the respective region of PPARα-LBP. Its carboxylic acid was attached to four amino acids (Ser289/His323/His449/Thr473), which contributes to the stabilization of Activating Function-2 helix 12, and its phenylpyrrole moiety was rotated 121.8 degrees in PPARγ-LBD from that in PPARα-LBD to interact with Phe264. PPARδ-LBD has the consensus four amino acids (Thr253/His287/His413/Tyr437) towards the carboxylic acids of its ligands, but it seems to lack sufficient space to accept saroglitazar because of the steric hindrance between the Trp228 or Arg248 residue of PPARδ-LBD and its methylthiophenyl moiety. Accordingly, in a coactivator recruitment assay, saroglitazar activated PPARα-LBD and PPARγ-LBD but not PPARδ-LBD, whereas glycine substitution of either Trp228, Arg248, or both of PPARδ-LBD conferred saroglitazar concentration-dependent activation. Our findings may be valuable in the molecular design of PPARα/γ dual or PPARα/γ/δ pan agonists.


Asunto(s)
Hipolipemiantes/farmacología , PPAR alfa/ultraestructura , PPAR gamma/ultraestructura , Fenilpropionatos/farmacología , Pirroles/farmacología , Sitios de Unión , Cristalografía por Rayos X , Diabetes Mellitus Tipo 2/complicaciones , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Dislipidemias/tratamiento farmacológico , Dislipidemias/etiología , Humanos , Hipolipemiantes/química , Hipolipemiantes/uso terapéutico , Metabolismo de los Lípidos , Enfermedad del Hígado Graso no Alcohólico/tratamiento farmacológico , PPAR alfa/agonistas , PPAR alfa/aislamiento & purificación , PPAR alfa/metabolismo , PPAR gamma/agonistas , PPAR gamma/aislamiento & purificación , PPAR gamma/metabolismo , Fenilpropionatos/química , Fenilpropionatos/uso terapéutico , Dominios Proteicos , Pirroles/química , Pirroles/uso terapéutico , Proteínas Recombinantes/aislamiento & purificación , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/ultraestructura
15.
Nat Commun ; 12(1): 5708, 2021 09 29.
Artículo en Inglés | MEDLINE | ID: mdl-34588452

RESUMEN

Ufmylation is a post-translational modification essential for regulating key cellular processes. A three-enzyme cascade involving E1, E2 and E3 is required for UFM1 attachment to target proteins. How UBA5 (E1) and UFC1 (E2) cooperatively activate and transfer UFM1 is still unclear. Here, we present the crystal structure of UFC1 bound to the C-terminus of UBA5, revealing how UBA5 interacts with UFC1 via a short linear sequence, not observed in other E1-E2 complexes. We find that UBA5 has a region outside the adenylation domain that is dispensable for UFC1 binding but critical for UFM1 transfer. This region moves next to UFC1's active site Cys and compensates for a missing loop in UFC1, which exists in other E2s and is needed for the transfer. Overall, our findings advance the understanding of UFM1's conjugation machinery and may serve as a basis for the development of ufmylation inhibitors.


Asunto(s)
Procesamiento Proteico-Postraduccional , Proteínas/metabolismo , Enzimas Activadoras de Ubiquitina/metabolismo , Enzimas Ubiquitina-Conjugadoras/metabolismo , Dominio Catalítico/genética , Humanos , Simulación del Acoplamiento Molecular , Resonancia Magnética Nuclear Biomolecular , Unión Proteica/genética , Proteínas/genética , Proteínas/aislamiento & purificación , Proteínas/ultraestructura , Proteínas Recombinantes/genética , Proteínas Recombinantes/aislamiento & purificación , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/ultraestructura , Enzimas Activadoras de Ubiquitina/genética , Enzimas Activadoras de Ubiquitina/aislamiento & purificación , Enzimas Activadoras de Ubiquitina/ultraestructura , Enzimas Ubiquitina-Conjugadoras/genética , Enzimas Ubiquitina-Conjugadoras/aislamiento & purificación , Enzimas Ubiquitina-Conjugadoras/ultraestructura , Difracción de Rayos X
16.
Nat Commun ; 12(1): 5709, 2021 09 29.
Artículo en Inglés | MEDLINE | ID: mdl-34588453

RESUMEN

The gastric H+,K+-ATPase mediates electroneutral exchange of 1H+/1K+ per ATP hydrolysed across the membrane. Previous structural analysis of the K+-occluded E2-P transition state of H+,K+-ATPase showed a single bound K+ at cation-binding site II, in marked contrast to the two K+ ions occluded at sites I and II of the closely-related Na+,K+-ATPase which mediates electrogenic 3Na+/2K+ translocation across the membrane. The molecular basis of the different K+ stoichiometry between these K+-counter-transporting pumps is elusive. We show a series of crystal structures and a cryo-EM structure of H+,K+-ATPase mutants with changes in the vicinity of site I, based on the structure of the sodium pump. Our step-wise and tailored construction of the mutants finally gave a two-K+ bound H+,K+-ATPase, achieved by five mutations, including amino acids directly coordinating K+ (Lys791Ser, Glu820Asp), indirectly contributing to cation-binding site formation (Tyr340Asn, Glu936Val), and allosterically stabilizing K+-occluded conformation (Tyr799Trp). This quintuple mutant in the K+-occluded E2-P state unambiguously shows two separate densities at the cation-binding site in its 2.6 Å resolution cryo-EM structure. These results offer new insights into how two closely-related cation pumps specify the number of K+ accommodated at their cation-binding site.


Asunto(s)
Mucosa Gástrica/enzimología , ATPasa Intercambiadora de Hidrógeno-Potásio/metabolismo , Potasio/metabolismo , Sitios de Unión/genética , Cationes Monovalentes/metabolismo , Membrana Celular/enzimología , Microscopía por Crioelectrón , Cristalización , Pruebas de Enzimas , Mucosa Gástrica/citología , ATPasa Intercambiadora de Hidrógeno-Potásio/genética , ATPasa Intercambiadora de Hidrógeno-Potásio/aislamiento & purificación , ATPasa Intercambiadora de Hidrógeno-Potásio/ultraestructura , Células HEK293 , Humanos , Modelos Moleculares , Mutación , Ingeniería de Proteínas , Proteínas Recombinantes/genética , Proteínas Recombinantes/aislamiento & purificación , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/ultraestructura , Especificidad por Sustrato/genética
17.
Nat Commun ; 12(1): 5707, 2021 09 29.
Artículo en Inglés | MEDLINE | ID: mdl-34588455

RESUMEN

Bacillus subtilis can form structurally complex biofilms on solid or liquid surfaces, which requires expression of genes for matrix production. The transcription of these genes is activated by regulatory protein RemA, which binds to poorly conserved, repetitive DNA regions but lacks obvious DNA-binding motifs or domains. Here, we present the structure of the RemA homologue from Geobacillus thermodenitrificans, showing a unique octameric ring with the potential to form a 16-meric superstructure. These results, together with further biochemical and in vivo characterization of B. subtilis RemA, suggests that the protein can wrap DNA around its ring-like structure through a LytTR-related domain.


Asunto(s)
Proteínas Bacterianas/metabolismo , Biopelículas/crecimiento & desarrollo , ADN Bacteriano/metabolismo , Geobacillus/fisiología , Factores de Transcripción/metabolismo , Bacillus subtilis/fisiología , Proteínas Bacterianas/genética , Proteínas Bacterianas/aislamiento & purificación , Proteínas Bacterianas/ultraestructura , Cristalografía por Rayos X , Regulación Bacteriana de la Expresión Génica , Modelos Genéticos , Mutagénesis Sitio-Dirigida , Dominios y Motivos de Interacción de Proteínas/genética , Multimerización de Proteína/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/aislamiento & purificación , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/ultraestructura , Secuencias Reguladoras de Ácidos Nucleicos , Factores de Transcripción/genética , Factores de Transcripción/aislamiento & purificación , Factores de Transcripción/ultraestructura
18.
Sci Rep ; 11(1): 18885, 2021 09 23.
Artículo en Inglés | MEDLINE | ID: mdl-34556749

RESUMEN

Since 1993, when the structure of Escherichia coli type II L-asparaginase (EcAII) in complex with L-aspartate was firstly reported, many structures of the wild type and mutated enzyme have been deposited in the Protein Data Bank. None of them report the full structure of the monomer in its ligand-free, open conformation, mainly because of the high dynamic and flexibility of the active site flexible loop. Here we report for the first time the structure of EcAII wild type in its open conformation comprising, for at least one protomer, clear electron density for the active site flexible loop (PDB ID: 6YZI). The structural element is highly mobile and it is transposed onto the rigid part of the active site upon substrate binding to allow completion of the enzyme catalytic center, thanks to key residues that serve as hinges and anchoring points. In the substrate binding pocket, several highly conserved water molecules are coordinated by residues involved in substrate binding, comprising two water molecules very likely involved in the enzyme catalytic process. We also describe, by molecular dynamics simulations, how the transposition of the loop, besides providing the proximity of residues needed for catalysis, causes a general stabilization of the protein.


Asunto(s)
Asparaginasa/ultraestructura , Proteínas de Escherichia coli/ultraestructura , Proteínas Recombinantes/ultraestructura , Asparaginasa/aislamiento & purificación , Dominio Catalítico , Escherichia coli/enzimología , Proteínas de Escherichia coli/aislamiento & purificación , Simulación de Dinámica Molecular , Estabilidad Proteica , Proteínas Recombinantes/aislamiento & purificación , Difracción de Rayos X
19.
Nat Commun ; 12(1): 4417, 2021 07 20.
Artículo en Inglés | MEDLINE | ID: mdl-34285212

RESUMEN

Endoperoxide-containing natural products are a group of compounds with structurally unique cyclized peroxide moieties. Although numerous endoperoxide-containing compounds have been isolated, the biosynthesis of the endoperoxides remains unclear. NvfI from Aspergillus novofumigatus IBT 16806 is an endoperoxidase that catalyzes the formation of fumigatonoid A in the biosynthesis of novofumigatonin. Here, we describe our structural and functional analyses of NvfI. The structural elucidation and mutagenesis studies indicate that NvfI does not utilize a tyrosyl radical in the reaction, in contrast to other characterized endoperoxidases. Further, the crystallographic analysis reveals significant conformational changes of two loops upon substrate binding, which suggests a dynamic movement of active site during the catalytic cycle. As a result, NvfI installs three oxygen atoms onto a substrate in a single enzyme turnover. Based on these results, we propose a mechanism for the NvfI-catalyzed, unique endoperoxide formation reaction to produce fumigatonoid A.


Asunto(s)
Aspergillus/enzimología , Biocatálisis , Proteínas Fúngicas/metabolismo , Oxigenasas/metabolismo , Peróxidos/metabolismo , Aspergillus/genética , Dominio Catalítico , Cristalografía por Rayos X , Compuestos Ferrosos/metabolismo , Proteínas Fúngicas/genética , Proteínas Fúngicas/aislamiento & purificación , Proteínas Fúngicas/ultraestructura , Ácidos Cetoglutáricos/metabolismo , Mutagénesis Sitio-Dirigida , Oxidación-Reducción , Oxígeno/metabolismo , Oxigenasas/genética , Oxigenasas/aislamiento & purificación , Oxigenasas/ultraestructura , Conformación Proteica en Lámina beta , Proteínas Recombinantes/genética , Proteínas Recombinantes/aislamiento & purificación , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/ultraestructura , Terpenos/metabolismo
20.
Nat Commun ; 12(1): 4621, 2021 07 30.
Artículo en Inglés | MEDLINE | ID: mdl-34330928

RESUMEN

Cytochromes bd are ubiquitous amongst prokaryotes including many human-pathogenic bacteria. Such complexes are targets for the development of antimicrobial drugs. However, an understanding of the relationship between the structure and functional mechanisms of these oxidases is incomplete. Here, we have determined the 2.8 Å structure of Mycobacterium smegmatis cytochrome bd by single-particle cryo-electron microscopy. This bd oxidase consists of two subunits CydA and CydB, that adopt a pseudo two-fold symmetrical arrangement. The structural topology of its Q-loop domain, whose function is to bind the substrate, quinol, is significantly different compared to the C-terminal region reported for cytochromes bd from Geobacillus thermodenitrificans (G. th) and Escherichia coli (E. coli). In addition, we have identified two potential oxygen access channels in the structure and shown that similar tunnels also exist in G. th and E. coli cytochromes bd. This study provides insights to develop a framework for the rational design of antituberculosis compounds that block the oxygen access channels of this oxidase.


Asunto(s)
Proteínas Bacterianas/ultraestructura , Microscopía por Crioelectrón/métodos , Grupo Citocromo b/ultraestructura , Proteínas del Complejo de Cadena de Transporte de Electrón/ultraestructura , Mycobacterium smegmatis/enzimología , Oxidorreductasas/ultraestructura , Proteínas Bacterianas/química , Proteínas Bacterianas/metabolismo , Grupo Citocromo b/química , Grupo Citocromo b/metabolismo , Transporte de Electrón , Proteínas del Complejo de Cadena de Transporte de Electrón/química , Proteínas del Complejo de Cadena de Transporte de Electrón/metabolismo , Hemo/química , Hemo/metabolismo , Modelos Moleculares , Mycobacterium smegmatis/genética , Oxidorreductasas/química , Oxidorreductasas/metabolismo , Oxígeno/metabolismo , Conformación Proteica , Subunidades de Proteína/química , Subunidades de Proteína/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/ultraestructura , Especificidad por Sustrato
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA