Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
PLoS One ; 19(5): e0303839, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38758765

RESUMEN

The interaction between SARS-CoV-2 non-structural protein Nsp9 and the nanobody 2NSP90 was investigated by NMR spectroscopy using the paramagnetic perturbation methodology PENELOP (Paramagnetic Equilibrium vs Nonequilibrium magnetization Enhancement or LOss Perturbation). The Nsp9 monomer is an essential component of the replication and transcription complex (RTC) that reproduces the viral gRNA for subsequent propagation. Therefore preventing Nsp9 recruitment in RTC would represent an efficient antiviral strategy that could be applied to different coronaviruses, given the Nsp9 relative invariance. The NMR results were consistent with a previous characterization suggesting a 4:4 Nsp9-to-nanobody stoichiometry with the occurrence of two epitope pairs on each of the Nsp9 units that establish the inter-dimer contacts of Nsp9 tetramer. The oligomerization state of Nsp9 was also analyzed by molecular dynamics simulations and both dimers and tetramers resulted plausible. A different distribution of the mapped epitopes on the tetramer surface with respect to the former 4:4 complex could also be possible, as well as different stoichiometries of the Nsp9-nanobody assemblies such as the 2:2 stoichiometry suggested by the recent crystal structure of the Nsp9 complex with 2NSP23 (PDB ID: 8dqu), a nanobody exhibiting essentially the same affinity as 2NSP90. The experimental NMR evidence, however, ruled out the occurrence in liquid state of the relevant Nsp9 conformational change observed in the same crystal structure.


Asunto(s)
Epítopos , Simulación de Dinámica Molecular , SARS-CoV-2 , Anticuerpos de Dominio Único , Proteínas no Estructurales Virales , Proteínas no Estructurales Virales/inmunología , Proteínas no Estructurales Virales/química , Proteínas no Estructurales Virales/metabolismo , Anticuerpos de Dominio Único/química , Anticuerpos de Dominio Único/inmunología , Anticuerpos de Dominio Único/metabolismo , SARS-CoV-2/inmunología , Epítopos/inmunología , Epítopos/química , Humanos , Espectroscopía de Resonancia Magnética , Unión Proteica , Multimerización de Proteína , COVID-19/inmunología , COVID-19/virología , Proteínas de Unión al ARN
2.
Vaccines (Basel) ; 9(11)2021 Nov 12.
Artículo en Inglés | MEDLINE | ID: mdl-34835247

RESUMEN

Type 2 Diabetes Mellitus (T2DM) is a chronic progressive disease, defined by insulin resistance and insufficient insulin secretion to maintain normoglycemia. Amyloidogenic aggregates are a hallmark of T2DM patients; they are cytotoxic for the insulin producing ß-cells, and cause inflammasome-dependent secretion of IL-1ß. To avoid the associated ß-cell loss and inflammation in advanced stage T2DM, we developed a novel monoclonal therapy targeting the major component of aggregates, islet amyloid polypeptide (IAPP). The here described monoclonal antibody (mAb) m81, specific for oligomeric and fibrils, but not for soluble free IAPP, is able to prevent oligomer growth and aggregate formation in vitro, and blocks islet inflammation and disease progression in vivo. Collectively, our data show that blocking fibril formation and prevention of new amyloidogenic aggregates by monoclonal antibody therapy may be a potential therapy for T2DM.

3.
Antibiotics (Basel) ; 10(2)2021 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-33562778

RESUMEN

Rapid spread of antibiotic resistance throughout the kingdom bacteria is inevitably bringing humanity towards the "post-antibiotic" era. The emergence of so-called "superbugs"-pathogen strains that develop resistance to multiple conventional antibiotics-is urging researchers around the globe to work on the development or perfecting of alternative means of tackling the pathogenic bacteria infections. Although various conceptually different approaches are being considered, each comes with its advantages and drawbacks. While drug-resistant pathogens are undoubtedly represented by both Gram(+) and Gram(-) bacteria, possible target spectrum across the proposed alternative approaches of tackling them is variable. Numerous anti-virulence strategies aimed at reducing the pathogenicity of target bacteria rather than eliminating them are being considered among such alternative approaches. Sortase A (SrtA) is a membrane-associated cysteine protease that catalyzes a cell wall sorting reaction by which surface proteins, including virulence factors, are anchored to the bacterial cell wall of Gram(+) bacteria. Although SrtA inhibition seems perspective among the Gram-positive pathogen-targeted antivirulence strategies, it still remains less popular than other alternatives. A decrease in virulence due to inactivation of SrtA activity has been extensively studied in Staphylococcus aureus, but it has also been demonstrated in other Gram(+) species. In this manuscript, results of past studies on the discovery of novel SrtA inhibitory compounds and evaluation of their potency were summarized and commented on. Here, we discussed the rationale behind the inhibition of SrtA, raised some concerns on the comparability of the results from different studies, and touched upon the possible resistance mechanisms as a response to implementation of such therapy in practice. The goal of this article is to encourage further studies of SrtA inhibitory compounds.

4.
ACS Infect Dis ; 6(2): 186-194, 2020 02 14.
Artículo en Inglés | MEDLINE | ID: mdl-31724850

RESUMEN

Because of its essential role as a bacterial virulence factor, enzyme sortase A (SrtA) has become an attractive target for the development of new antivirulence drugs against Gram-positive infections. Here we describe 27 compounds identified as covalent inhibitors of Staphylococcus aureus SrtA by screening a library of approximately 50 000 compounds using a FRET assay followed by NMR-based validation and binding reversibility analysis. Nineteen of these compounds displayed only moderate to weak cytotoxicity, with CC50 against NIH 3T3 mice fibroblast cells ranging from 12 to 740 µM. Analysis using covalent docking suggests that the inhibitors initially associate via hydrophobic interactions, followed by covalent bond formation between the SrtA active site cysteine and an electrophilic center of the inhibitor. The compounds represent good starting points that have the potential to be developed into broad spectrum antivirulence agents as exemplified by hit-to-lead optimization of one of the compounds.


Asunto(s)
Aminoaciltransferasas/antagonistas & inhibidores , Antibacterianos/química , Antibacterianos/farmacología , Proteínas Bacterianas/antagonistas & inhibidores , Staphylococcus aureus/efectos de los fármacos , Animales , Sitios de Unión , Dominio Catalítico , Cisteína Endopeptidasas , Descubrimiento de Drogas , Fibroblastos/efectos de los fármacos , Espectroscopía de Resonancia Magnética , Ratones , Simulación del Acoplamiento Molecular , Células 3T3 NIH , Bibliotecas de Moléculas Pequeñas , Staphylococcus aureus/enzimología , Factores de Virulencia/antagonistas & inhibidores
5.
Bioorg Med Chem ; 27(5): 721-728, 2019 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-30711310

RESUMEN

New compounds able to counteract staphylococcal biofilm formation are needed. In this study we investigate the mechanism of action of pyrrolomycins, whose potential as antimicrobial agents has been demonstrated. We performed a new efficient and easy method to use microwave organic synthesis suitable for obtaining pyrrolomycins in good yields and in suitable amount for their in vitro in-depth investigation. We evaluate the inhibitory activity towards Sortase A (SrtA), a transpeptidase responsible for covalent anchoring in Gram-positive peptidoglycan of many surface proteins involved in adhesion and in biofilm formation. All compounds show a good inhibitory activity toward SrtA, having IC50 values ranging from 130 to 300 µM comparable to berberine hydrochloride. Of note compound 1d shows a good affinity in docking experiment to SrtA and exhibits the highest capability to interfere with biofilm formation of S. aureus showing an IC50 of 3.4 nM. This compound is also effective in altering S. aureus murein hydrolase activity that is known to be responsible for degradation, turnover, and maturation of bacterial peptidoglycan and involved in the initial stages of S. aureus biofilm formation.


Asunto(s)
Antibacterianos/farmacología , Pirroles/farmacología , Aminoaciltransferasas/química , Aminoaciltransferasas/metabolismo , Antibacterianos/síntesis química , Antibacterianos/farmacocinética , Proteínas Bacterianas/química , Proteínas Bacterianas/metabolismo , Biopelículas/efectos de los fármacos , Cisteína Endopeptidasas/química , Cisteína Endopeptidasas/metabolismo , Pruebas de Enzimas , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/farmacocinética , Inhibidores Enzimáticos/farmacología , Pruebas de Sensibilidad Microbiana , Microondas , Simulación del Acoplamiento Molecular , N-Acetil Muramoil-L-Alanina Amidasa/antagonistas & inhibidores , Pseudomonas aeruginosa/efectos de los fármacos , Pirroles/síntesis química , Pirroles/farmacocinética , Staphylococcus aureus/efectos de los fármacos
6.
Molecules ; 21(2): 241, 2016 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-26907235

RESUMEN

A FRET-based random screening assay was used to generate hit compounds as sortase A inhibitors that allowed us to identify ethyl 3-oxo-2-(2-phenylhydrazinylidene)butanoate as an example of a new class of sortase A inhibitors. Other analogues were generated by changing the ethoxycarbonyl function for a carboxy, cyano or amide group, or introducing substituents in the phenyl ring of the ester and acid derivatives. The most active derivative found was 3-oxo-2-(2-(3,4dichlorophenyl)hydrazinylidene)butanoic acid (2b), showing an IC50 value of 50 µM. For a preliminary assessment of their antivirulence properties the new derivatives were tested for their antibiofilm activity. The most active compound resulted 2a, which showed inhibition of about 60% against S. aureus ATCC 29213, S. aureus ATCC 25923, S. aureus ATCC 6538 and S. epidermidis RP62A at a screening concentration of 100 µM.


Asunto(s)
Aminoaciltransferasas/antagonistas & inhibidores , Proteínas Bacterianas/antagonistas & inhibidores , Inhibidores Enzimáticos/química , Fenilhidrazinas/química , Aminoaciltransferasas/química , Antiinfecciosos/química , Antiinfecciosos/farmacología , Proteínas Bacterianas/química , Biopelículas/efectos de los fármacos , Cisteína Endopeptidasas/química , Inhibidores Enzimáticos/farmacología , Concentración 50 Inhibidora , Fenilhidrazinas/farmacología , Staphylococcus aureus/efectos de los fármacos , Staphylococcus epidermidis/efectos de los fármacos
7.
Bioorg Med Chem ; 22(21): 5988-6003, 2014 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-25282649

RESUMEN

Gram-positive bacteria, in general, and staphylococci, in particular, are the widespread cause of nosocomial and community-acquired infections. The rapid evolvement of strains resistant to antibiotics currently in use is a serious challenge. Novel antimicrobial compounds have to be developed to fight these resistant bacteria, and sortase A, a bacterial cell wall enzyme, is a promising target for novel therapies. As a transpeptidase that covalently attaches various virulence factors to the cell surface, this enzyme plays a crucial role in the ability of bacteria to invade the host's tissues and to escape the immune response. In this study we have screened a small molecule library against recombinant Staphylococcus aureus sortase A using an in vitro FRET-based assay. The selected hits were validated by NMR methods in order to exclude false positives and to analyze the reversibility of inhibition. Further structural and functional analysis of the best hit allowed the identification of a novel class of benzisothiazolinone-based compounds as potent and promising sortase inhibitors.


Asunto(s)
Aminoaciltransferasas/antagonistas & inhibidores , Proteínas Bacterianas/antagonistas & inhibidores , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Staphylococcus aureus/enzimología , Tiazoles/química , Tiazoles/farmacología , Aminoaciltransferasas/química , Aminoaciltransferasas/metabolismo , Proteínas Bacterianas/química , Proteínas Bacterianas/metabolismo , Cisteína Endopeptidasas/química , Cisteína Endopeptidasas/metabolismo , Transferencia Resonante de Energía de Fluorescencia , Ensayos Analíticos de Alto Rendimiento , Humanos , Simulación del Acoplamiento Molecular , Infecciones Estafilocócicas/tratamiento farmacológico , Infecciones Estafilocócicas/microbiología , Staphylococcus aureus/química , Staphylococcus aureus/efectos de los fármacos , Relación Estructura-Actividad
8.
BMC Struct Biol ; 13: 19, 2013 Oct 07.
Artículo en Inglés | MEDLINE | ID: mdl-24099525

RESUMEN

BACKGROUND: Increasing rates of antimicrobial resistance among uropathogens led, among other efforts, to the application of subtractive reverse vaccinology for the identification of antigens present in extraintestinal pathogenic E. coli (ExPEC) strains but absent or variable in non-pathogenic strains, in a quest for a broadly protective Escherichia coli vaccine. The protein coded by locus c5321 from CFT073 E. coli was identified as one of nine potential vaccine candidates against ExPEC and was able to confer protection with an efficacy of 33% in a mouse model of sepsis. c5321 (known also as EsiB) lacks functional annotation and structurally belongs to the Sel1-like repeat (SLR) family. Herein, as part of the general characterization of this potential antigen, we have focused on its structural properties. RESULTS: We report the 1.74 Å-resolution crystal structure of c5321 from CFT073 E. coli determined by Se-Met SAD phasing. The structure is composed of 11 SLR units in a topological organisation that highly resembles that found in HcpC from Helicobacter pylori, with the main difference residing in how the super-helical fold is stabilised. The stabilising effect of disulfide bridges in HcpC is replaced in c5321 by a strengthening of the inter-repeat hydrophobic core. A metal-ion binding site, uncharacteristic of SLR proteins, is detected between SLR units 3 and 4 in the region of the inter-repeat hydrophobic core. Crystal contacts are observed between the C-terminal tail of one molecule and the C-terminal amphipathic groove of a neighbouring one, resembling interactions between ligand and proteins containing tetratricopeptide-like repeats. CONCLUSIONS: The structure of antigen c5321 presents a mode of stabilization of the SLR fold different from that observed in close homologs of known structure. The location of the metal-ion binding site and the observed crystal contacts suggest a potential role in regulation of conformational flexibility and interaction with yet unidentified target proteins, respectively. These findings open new perspectives in both antigen design and for the identification of a functional role for this protective antigen.


Asunto(s)
Antígenos Bacterianos/química , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/metabolismo , Escherichia coli Uropatógena/química , Secuencia de Aminoácidos , Animales , Antígenos Bacterianos/inmunología , Antígenos Bacterianos/metabolismo , Antígenos CD1/inmunología , Sitios de Unión , Secuencia de Consenso , Cristalografía por Rayos X , Mapeo Epitopo , Proteínas de Escherichia coli/inmunología , Vacunas contra Escherichia coli/inmunología , Vacunas contra Escherichia coli/metabolismo , Helicobacter pylori/química , Helicobacter pylori/inmunología , Helicobacter pylori/metabolismo , Magnesio/metabolismo , Ratones , Modelos Moleculares , Conformación Proteica , Pliegue de Proteína , Estabilidad Proteica , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Homología Estructural de Proteína , Escherichia coli Uropatógena/inmunología
9.
J Biomol NMR ; 29(3): 351-61, 2004 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-15213433

RESUMEN

A single free Cys sidechain in the N-terminal domain of the E. coli arginine repressor was covalently derivatized with S-cysteaminyl-EDTA for site-specific attachment of paramagnetic metal ions. The effects of chelated metal ions were monitored with (15)N-HSQC spectra. Complexation of Co(2+), which has a fast relaxing electron spin, resulted in significant pseudocontact shifts, but also in peak doubling which was attributed to the possibility of forming two different stereoisomers of the EDTA-Co(2+) complex. In contrast, complexation of Cu(2+) or Mn(2+), which have slowly relaxing electron spins, did not produce chemical shift changes and yielded self-consistent sets of paramagnetic relaxation enhancements of the amide protons. T (1) relaxation enhancements with Cu(2+) combined with T (2) relaxation enhancements with Mn(2+) are shown to provide accurate distance restraints ranging from 9 to 25 A. These long-range distance restraints can be used for structural studies inaccessible to NOEs. As an example, the structure of a solvent-exposed loop in the N-terminal domain of the E. coli arginine repressor was refined by paramagnetic restraints. Electronic correlation times of Cu(2+) and Mn(2+) were determined from a comparison of T (1) and T (2) relaxation enhancements.


Asunto(s)
Quelantes/farmacología , Cisteína/química , Espectroscopía de Resonancia Magnética/métodos , Proteínas/química , Proteínas Bacterianas/química , Quelantes/química , Cobalto/química , Cobre/química , ADN/química , Ácido Edético/química , Espectroscopía de Resonancia por Spin del Electrón , Electrones , Escherichia coli/metabolismo , Gadolinio/química , Iones , Manganeso/química , Modelos Químicos , Modelos Moleculares , Estructura Terciaria de Proteína , Protones , Proteínas Represoras/química , Factores de Tiempo
10.
J Mol Biol ; 326(1): 217-23, 2003 Feb 07.
Artículo en Inglés | MEDLINE | ID: mdl-12547203

RESUMEN

The R3H domain is a conserved sequence motif, identified in over 100 proteins, that is thought to be involved in polynucleotide-binding, including DNA, RNA and single-stranded DNA. In this work the 3D structure of the R3H domain from human Smubp-2 was determined by NMR spectroscopy. It is the first 3D structure determination of an R3H domain. The fold presents a small motif, consisting of a three-stranded antiparallel beta-sheet and two alpha-helices, which is related to the structures of the YhhP protein and the C-terminal domain of the translational initiation factor IF3. The similarities are non-trivial, as the amino acid identities are below 10%. Three conserved basic residues cluster on the same face of the R3H domain and could play a role in nucleic acid recognition. An extended hydrophobic area at a different site of the molecular surface could act as a protein-binding site. A strong correlation between conservation of hydrophobic amino acids and side-chain solvent protection indicates that the structure of the Smubp-2 R3H domain is representative of R3H domains in general.


Asunto(s)
Proteínas de Unión al ADN/química , Proteínas de Escherichia coli , Resonancia Magnética Nuclear Biomolecular , Factores de Transcripción/química , Secuencia de Aminoácidos , Proteínas Bacterianas/química , Proteínas de Unión al ADN/metabolismo , Humanos , Modelos Moleculares , Datos de Secuencia Molecular , Factor 3 Procariótico de Iniciación/química , Estructura Terciaria de Proteína , Alineación de Secuencia , Soluciones , Relación Estructura-Actividad , Factores de Transcripción/metabolismo
11.
FEBS Lett ; 522(1-3): 93-8, 2002 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-12095625

RESUMEN

The glycine-alanine repeat (GAr) of the Epstein-Barr virus nuclear antigen-1 is a cis-acting transferable element that inhibits ubiquitin/proteasome-dependent proteolysis in vitro and in vivo. We have here examined the effect of a synthetic 20-mer GAr oligopeptide on the degradation of iodinated or biotin labeled lysozyme in a rabbit reticulocyte lysates in vitro assay. Micromolar concentrations of the GA-20 peptide inhibited the hydrolysis of lysozyme without significant effect on ubiquitination. Addition of the peptide did not inhibit the hydrolysis of fluorogenic substrate by purified proteasomes and did not affect the ubiquitination of lysozyme. An excess of the peptide failed to compete for binding of a synthetic tetra-ubiquitin complex to the S5a ubiquitin-binding subunit of the 19S regulator, confirming that the GAr does not block the access of ubiquitinated substrates to the proteasome. Our data suggest that the GAr may act by destabilizing the interaction of ubiquitinated substrates with the proteasome and promote the premature release of the substrate.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Antígenos Nucleares del Virus de Epstein-Barr/farmacología , Herpesvirus Humano 4 , Complejos Multienzimáticos/antagonistas & inhibidores , Muramidasa/antagonistas & inhibidores , Oligopéptidos/farmacología , Péptidos/farmacología , Ubiquitinas/metabolismo , Alanina , Secuencia de Aminoácidos , Animales , Biotina , Proteínas Portadoras/antagonistas & inhibidores , Cisteína Endopeptidasas , Glicina , Radioisótopos de Yodo , Marcaje Isotópico , Imitación Molecular , Datos de Secuencia Molecular , Polímeros , Complejo de la Endopetidasa Proteasomal , Conejos
12.
Proc Natl Acad Sci U S A ; 99(3): 1532-7, 2002 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-11805282

RESUMEN

Functional inactivation of the tumor suppressor protein p53 by accelerated ubiquitin/proteasome-dependent proteolysis is a common event in tumor progression. Proteasomal degradation is inhibited by the Gly-Ala repeat (GAr) of the Epstein-Barr virus nuclear antigen-1, which acts as a transferable element on a variety of proteasomal substrates. We demonstrate that p53 chimeras containing GAr domains of different lengths and positions within the protein are protected from proteolysis induced by the ubiquitin ligases murine double minute 2 and E6-associated protein but are still ubiquitinated and retain the capacity to interact with the S5a ubiquitin-binding subunit of the proteasome. The GAr chimeras transactivate p53 target genes, induce cell cycle arrest and apoptosis, and exhibit improved growth inhibitory activity in tumor cells with impaired endogenous p53 activity.


Asunto(s)
Proteínas de Unión al ADN , Dipéptidos/química , Genes p53 , Herpesvirus Humano 4/genética , Proteínas de Neoplasias/metabolismo , Proteínas Nucleares , Proteínas Oncogénicas Virales/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Proteína p53 Supresora de Tumor/genética , Apoptosis/fisiología , Secuencia de Bases , Ciclo Celular/fisiología , División Celular , Cisteína Endopeptidasas/metabolismo , Cartilla de ADN , Humanos , Datos de Secuencia Molecular , Complejos Multienzimáticos/metabolismo , Oligodesoxirribonucleótidos Antisentido , Proteínas Oncogénicas Virales/genética , Sistemas de Lectura Abierta , Osteosarcoma , Complejo de la Endopetidasa Proteasomal , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas c-mdm2 , Proteínas Recombinantes de Fusión , Secuencias Repetitivas de Aminoácido , Células Tumorales Cultivadas , Proteína p53 Supresora de Tumor/metabolismo , Ubiquitina/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA