Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 71
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
J Physiol ; 602(8): 1623-1636, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38598430

RESUMEN

Two-pore channels and TRP mucolipins are ubiquitous endo-lysosomal cation channels of pathophysiological relevance. Both are Ca2+-permeable and regulated by phosphoinositides, principally PI(3,5)P2. Accumulating evidence has uncovered synergistic channel activation by PI(3,5)P2 and endogenous metabolites such as the Ca2+ mobilizing messenger NAADP, synthetic agonists including approved drugs and physical cues such as voltage and osmotic pressure. Here, we provide an overview of this coordination.


Asunto(s)
Canales de Calcio , Canales de Potencial de Receptor Transitorio , Canales de Calcio/metabolismo , Canales de Dos Poros , Calcio/metabolismo , Lisosomas/metabolismo , NADP/metabolismo , Presión Osmótica , Canales de Potencial de Receptor Transitorio/metabolismo
2.
J Cell Physiol ; 238(6): 1354-1367, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-37042220

RESUMEN

The voltage-gated sodium channel NaV 1.7 is involved in various pain phenotypes and is physiologically regulated by the NaV -ß3-subunit. Venom toxins ProTx-II and OD1 modulate NaV 1.7 channel function and may be useful as therapeutic agents and/or research tools. Here, we use patch-clamp recordings to investigate how the ß3-subunit can influence and modulate the toxin-mediated effects on NaV 1.7 function, and we propose a putative binding mode of OD1 on NaV 1.7 to rationalise its activating effects. The inhibitor ProTx-II slowed the rate of NaV 1.7 activation, whilst the activator OD1 reduced the rate of fast inactivation and accelerated recovery from inactivation. The ß3-subunit partially abrogated these effects. OD1 induced a hyperpolarising shift in the V1/2 of steady-state activation, which was not observed in the presence of ß3. Consequently, OD1-treated NaV 1.7 exhibited an enhanced window current compared with OD1-treated NaV 1.7-ß3 complex. We identify candidate OD1 residues that are likely to prevent the upward movement of the DIV S4 helix and thus impede fast inactivation. The binding sites for each of the toxins and the predicted location of the ß3-subunit on the NaV 1.7 channel are distinct. Therefore, we infer that the ß3-subunit influences the interaction of toxins with NaV 1.7 via indirect allosteric mechanisms. The enhanced window current shown by OD1-treated NaV 1.7 compared with OD1-treated NaV 1.7-ß3 is discussed in the context of differing cellular expressions of NaV 1.7 and the ß3-subunit in dorsal root ganglion (DRG) neurons. We propose that ß3, as the native binding partner for NaV 1.7 in DRG neurons, should be included during screening of molecules against NaV 1.7 in relevant analgesic discovery campaigns.


Asunto(s)
Ponzoñas , Canales de Sodio Activados por Voltaje , Humanos , Ponzoñas/uso terapéutico , Péptidos/farmacología , Péptidos/uso terapéutico , Analgésicos/uso terapéutico , Dolor/tratamiento farmacológico
3.
J Chem Inf Model ; 63(21): 6912-6924, 2023 11 13.
Artículo en Inglés | MEDLINE | ID: mdl-37883148

RESUMEN

Polo-like kinase 1 (PLK1) and p38γ mitogen-activated protein kinase (p38γ) play important roles in cancer pathogenesis by controlling cell cycle progression and are therefore attractive cancer targets. The design of multitarget inhibitors may offer synergistic inhibition of distinct targets and reduce the risk of drug-drug interactions to improve the balance between therapeutic efficacy and safety. We combined deep-learning-based quantitative structure-activity relationship (QSAR) modeling and hybrid-based consensus scoring to screen for inhibitors with potential activity against the targeted proteins. Using this combination strategy, we identified a potent PLK1 inhibitor (compound 4) that inhibited PLK1 activity and liver cancer cell growth in the nanomolar range. Next, we deployed both our QSAR models for PLK1 and p38γ on the Enamine compound library to identify dual-targeting inhibitors against PLK1 and p38γ. Likewise, the identified hits were subsequently subjected to hybrid-based consensus scoring. Using this method, we identified a promising compound (compound 14) that could inhibit both PLK1 and p38γ activities. At nanomolar concentrations, compound 14 inhibited the growth of human hepatocellular carcinoma and hepatoblastoma cells in vitro. This study demonstrates the combined screening strategy to identify novel potential inhibitors for existing targets.


Asunto(s)
Inhibidores de Proteínas Quinasas , Proteínas Serina-Treonina Quinasas , Relación Estructura-Actividad Cuantitativa , Humanos , Proteínas de Ciclo Celular/metabolismo , Consenso , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Serina-Treonina Quinasas/metabolismo , Quinasa Tipo Polo 1
4.
Int J Mol Sci ; 24(8)2023 Apr 17.
Artículo en Inglés | MEDLINE | ID: mdl-37108523

RESUMEN

Protein kinase p38γ is an attractive target against cancer because it plays a pivotal role in cancer cell proliferation by phosphorylating the retinoblastoma tumour suppressor protein. Therefore, inhibition of p38γ with active small molecules represents an attractive alternative for developing anti-cancer drugs. In this work, we present a rigorous and systematic virtual screening framework to identify potential p38γ inhibitors against cancer. We combined the use of machine learning-based quantitative structure activity relationship modelling with conventional computer-aided drug discovery techniques, namely molecular docking and ligand-based methods, to identify potential p38γ inhibitors. The hit compounds were filtered using negative design techniques and then assessed for their binding stability with p38γ through molecular dynamics simulations. To this end, we identified a promising compound that inhibits p38γ activity at nanomolar concentrations and hepatocellular carcinoma cell growth in vitro in the low micromolar range. This hit compound could serve as a potential scaffold for further development of a potent p38γ inhibitor against cancer.


Asunto(s)
Antineoplásicos , Simulación de Dinámica Molecular , Antineoplásicos/farmacología , Bioensayo , Descubrimiento de Drogas , Ligandos , Simulación del Acoplamiento Molecular , Relación Estructura-Actividad Cuantitativa , Proteína Quinasa 12 Activada por Mitógenos/metabolismo
5.
Biochem Biophys Res Commun ; 610: 56-60, 2022 06 25.
Artículo en Inglés | MEDLINE | ID: mdl-35436631

RESUMEN

The store-operated Ca2+ entry (SOCE) represents an important route for generating cellular Ca2+ signals that are implicated in physiological and various pathological scenarios that include diabetic cardiomyopathy (DM-CMP) which is well known to have Ca2+ dysregulation among other salient features. In this study, we investigated the role of SOCE in Ca2+ handling of cardiomyocytes obtained from adult male Wistar rats that were made diabetic by intraperitoneal administration of streptozotocin (STZ 50 mg/kg). We also included another group of rats with diabetes induced by STZ administration but received an angiotensin II receptor blocker - losartan. In whole cell recordings with isolated cardiomyocytes, the SOCE-representative whole-cell current ICRAC was found to be significantly reduced for the diabetic group compared to the control group and chronic losartan treatment could restore ICRAC to a level comparable to the control group. However, in contrast to the observed reduction in ICRAC, Orai1 and Orai3 proteins were found to be significantly upregulated in diabetic condition whereas no significant change in the expression levels of Stim1, Stim2 and Orai2 was observed. Also, losartan treatment did not affect the expression pattern of these key proteins for SOCE in diabetic group. The observed imbalance between the functional read out of SOCE (peak ICRAC size) and expression levels of the underlying proteins was puzzling but could be, among other possibilities, due to impairment of interaction between Stim and Orai proteins. We argue that the observed changes in SOCE with diabetes could be a contributing factor for the Ca2+ dyshomeostasis associated with diabetic cardiomyopathies and blockade of angiotensin II receptor may potentially restore normal SOCE in diabetic cardiomyocytes.


Asunto(s)
Antagonistas de Receptores de Angiotensina , Canales de Calcio , Diabetes Mellitus , Miocitos Cardíacos , Antagonistas de Receptores de Angiotensina/farmacología , Animales , Calcio/metabolismo , Canales de Calcio/metabolismo , Señalización del Calcio , Diabetes Mellitus/metabolismo , Losartán/farmacología , Masculino , Proteínas de la Membrana/metabolismo , Miocitos Cardíacos/metabolismo , Ratas , Ratas Wistar , Receptores de Angiotensina/metabolismo , Molécula de Interacción Estromal 1/metabolismo
6.
J Chem Inf Model ; 62(10): 2586-2599, 2022 05 23.
Artículo en Inglés | MEDLINE | ID: mdl-35533315

RESUMEN

Lipoteichoic acid synthase (LtaS) is a key enzyme for the cell wall biosynthesis of Gram-positive bacteria. Gram-positive bacteria that lack lipoteichoic acid (LTA) exhibit impaired cell division and growth defects. Thus, LtaS appears to be an attractive antimicrobial target. The pharmacology around LtaS remains largely unexplored with only two small-molecule LtaS inhibitors reported, namely "compound 1771" and the Congo red dye. Structure-based drug discovery efforts against LtaS remain unattempted due to the lack of an inhibitor-bound structure of LtaS. To address this, we combined the use of a molecular docking technique with molecular dynamics (MD) simulations to model a plausible binding mode of compound 1771 to the extracellular catalytic domain of LtaS (eLtaS). The model was validated using alanine mutagenesis studies combined with isothermal titration calorimetry. Additionally, lead optimization driven by our computational model resulted in an improved version of compound 1771, namely, compound 4 which showed greater affinity for binding to eLtaS than compound 1771 in biophysical assays. Compound 4 reduced LTA production in S. aureus dose-dependently, induced aberrant morphology as seen for LTA-deficient bacteria, and significantly reduced bacteria titers in the lung of mice infected with S. aureus. Analysis of our MD simulation trajectories revealed the possible formation of a transient cryptic pocket in eLtaS. Virtual screening (VS) against the cryptic pocket led to the identification of a new class of inhibitors that could potentiate ß-lactams against methicillin-resistant S. aureus. Our overall workflow and data should encourage further drug design campaign against LtaS. Finally, our work reinforces the importance of considering protein conformational flexibility to a successful VS endeavor.


Asunto(s)
Staphylococcus aureus Resistente a Meticilina , Staphylococcus aureus , Animales , Lipopolisacáridos/metabolismo , Lipopolisacáridos/farmacología , Staphylococcus aureus Resistente a Meticilina/metabolismo , Ratones , Simulación del Acoplamiento Molecular , Staphylococcus aureus/metabolismo , Ácidos Teicoicos/metabolismo
7.
Platelets ; 33(7): 1090-1095, 2022 Oct 03.
Artículo en Inglés | MEDLINE | ID: mdl-35417662

RESUMEN

Thrombin is a potent platelet activator, acting through proteinase-activated receptors -1 and -4 (PAR1 and PAR4). Of these, PAR-1 is activated more rapidly and by lower thrombin concentrations. Consequently, PAR-1 has been extensively investigated as a target for anti-platelet drugs to prevent myocardial infarction. Q94 has been reported to act as an allosteric modulator of PAR1, potently and selectively inhibiting PAR1-Gαq coupling in multiple cell lines, but its effects on human platelet activation have not been previously studied. Platelet Ca2+ signaling, integrin αIIbß3 activation and α-granule secretion were monitored following stimulation by a PAR1-activating peptide (PAR1-AP). Although Q94 inhibited these responses, its potency was low compared to other PAR1 antagonists. In addition, αIIbß3 activation and α-granule secretion in response to other platelet activators were also inhibited with similar potency. Finally, in endothelial cells, Q94 did not inhibit PAR1-dependent Ca2+ signaling. Our data suggest that Q94 may have PAR1-independent off-target effects in platelets, precluding its use as a selective PAR1 allosteric modulator.


Asunto(s)
Receptor PAR-1 , Trombina , Plaquetas/metabolismo , Células Endoteliales/metabolismo , Humanos , Activación Plaquetaria , Agregación Plaquetaria , Complejo GPIIb-IIIa de Glicoproteína Plaquetaria/metabolismo , Receptor PAR-1/metabolismo , Receptores de Trombina/metabolismo , Trombina/metabolismo , Trombina/farmacología
8.
J Biol Chem ; 295(48): 16411-16426, 2020 11 27.
Artículo en Inglés | MEDLINE | ID: mdl-32943550

RESUMEN

Clinical isolates of the opportunistic pathogen Pseudomonas aeruginosa from patients with cystic fibrosis (CF) frequently contain mutations in the gene encoding an elongation factor, FusA1. Recent work has shown that fusA1 mutants often display elevated aminoglycoside resistance due to increased expression of the efflux pump, MexXY. However, we wondered whether these mutants might also be affected in other virulence-associated phenotypes. Here, we isolated a spontaneous gentamicin-resistant fusA1 mutant (FusA1P443L) in which mexXY expression was increased. Proteomic and transcriptomic analyses revealed that the fusA1 mutant also exhibited discrete changes in the expression of key pathogenicity-associated genes. Most notably, the fusA1 mutant displayed greatly increased expression of the Type III secretion system (T3SS), widely considered to be the most potent virulence factor in the P. aeruginosa arsenal, and also elevated expression of the Type VI (T6) secretion machinery. This was unexpected because expression of the T3SS is usually reciprocally coordinated with T6 secretion system expression. The fusA1 mutant also displayed elevated exopolysaccharide production, dysregulated siderophore production, elevated ribosome synthesis, and transcriptomic signatures indicative of translational stress. Each of these phenotypes (and almost all of the transcriptomic and proteomic changes associated with the fusA1 mutation) were restored to levels comparable with that in the progenitor strain by expression of the WT fusA1 gene in trans, indicating that the mutant gene is recessive. Our data show that in addition to elevating antibiotic resistance through mexXY expression (and also additional contributory resistance mechanisms), mutations in fusA1 can lead to highly selective dysregulation of virulence gene expression.


Asunto(s)
Proteínas Bacterianas , Farmacorresistencia Bacteriana/genética , Regulación Bacteriana de la Expresión Génica , Factor G de Elongación Peptídica , Polimorfismo de Nucleótido Simple , Pseudomonas aeruginosa , Factores de Virulencia , Proteínas Bacterianas/biosíntesis , Proteínas Bacterianas/genética , Mutación , Factor G de Elongación Peptídica/genética , Factor G de Elongación Peptídica/metabolismo , Pseudomonas aeruginosa/genética , Pseudomonas aeruginosa/metabolismo , Pseudomonas aeruginosa/patogenicidad , Sistemas de Secreción Tipo III/genética , Sistemas de Secreción Tipo III/metabolismo , Sistemas de Secreción Tipo VI/genética , Sistemas de Secreción Tipo VI/metabolismo , Factores de Virulencia/biosíntesis , Factores de Virulencia/genética
9.
Molecules ; 26(4)2021 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-33672721

RESUMEN

The ongoing coronavirus pandemic has been a burden on the worldwide population, with mass fatalities and devastating socioeconomic consequences. It has particularly drawn attention to the lack of approved small-molecule drugs to inhibit SARS coronaviruses. Importantly, lessons learned from the SARS outbreak of 2002-2004, caused by severe acute respiratory syndrome coronavirus 1 (SARS-CoV-1), can be applied to current drug discovery ventures. SARS-CoV-1 and SARS-CoV-2 both possess two cysteine proteases, the main protease (Mpro) and the papain-like protease (PLpro), which play a significant role in facilitating viral replication, and are important drug targets. The non-covalent inhibitor, GRL-0617, which was found to inhibit replication of SARS-CoV-1, and more recently SARS-CoV-2, is the only PLpro inhibitor co-crystallised with the recently solved SARS-CoV-2 PLpro crystal structure. Therefore, the GRL-0617 structural template and pharmacophore features are instrumental in the design and development of more potent PLpro inhibitors. In this work, we conducted scaffold hopping using GRL-0617 as a reference to screen over 339,000 ligands in the chemical space using the ChemDiv, MayBridge, and Enamine screening libraries. Twenty-four distinct scaffolds with structural and electrostatic similarity to GRL-0617 were obtained. These proceeded to molecular docking against PLpro using the AutoDock tools. Of two compounds that showed the most favourable predicted binding affinities to the target site, as well as comparable protein-ligand interactions to GRL-0617, one was chosen for further analogue-based work. Twenty-seven analogues of this compound were further docked against the PLpro, which resulted in two additional hits with promising docking profiles. Our in silico pipeline consisted of an integrative four-step approach: (1) ligand-based virtual screening (scaffold-hopping), (2) molecular docking, (3) an analogue search, and, (4) evaluation of scaffold drug-likeness, to identify promising scaffolds and eliminate those with undesirable properties. Overall, we present four novel, and lipophilic, scaffolds obtained from an exhaustive search of diverse and uncharted regions of chemical space, which may be further explored in vitro through structure-activity relationship (SAR) studies in the search for more potent inhibitors. Furthermore, these scaffolds were predicted to have fewer off-target interactions than GRL-0617. Lastly, to our knowledge, this work contains the largest ligand-based virtual screen performed against GRL-0617.


Asunto(s)
Antivirales/química , COVID-19/enzimología , Proteasas 3C de Coronavirus , Inhibidores de Cisteína Proteinasa/química , Simulación del Acoplamiento Molecular , SARS-CoV-2/enzimología , Antivirales/uso terapéutico , Proteasas 3C de Coronavirus/antagonistas & inhibidores , Proteasas 3C de Coronavirus/química , Cristalografía por Rayos X , Inhibidores de Cisteína Proteinasa/uso terapéutico , Evaluación Preclínica de Medicamentos , Humanos , Tratamiento Farmacológico de COVID-19
10.
Trends Biochem Sci ; 41(6): 475-477, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-27156118

RESUMEN

Two-pore channels (TPCs) are intracellular Ca(2+)-permeable ion channels that are expressed on acidic Ca(2+) stores. They are co-regulated by voltage and Ca(2+) in plant vacuoles and by the second messenger NAADP in animal endo-lysosomes. Two new studies of plant TPC structures reveal essential features of their architecture and provide mechanistic insight into their workings.


Asunto(s)
Proteínas de Arabidopsis/química , Arabidopsis/metabolismo , Canales de Calcio/química , Calcio/metabolismo , NADP/análogos & derivados , Vacuolas/metabolismo , Arabidopsis/genética , Proteínas de Arabidopsis/genética , Proteínas de Arabidopsis/metabolismo , Sitios de Unión , Canales de Calcio/genética , Canales de Calcio/metabolismo , Señalización del Calcio , Cristalografía por Rayos X , Endosomas/metabolismo , Expresión Génica , Activación del Canal Iónico , Lisosomas/metabolismo , NADP/química , NADP/metabolismo , Unión Proteica , Conformación Proteica en Hélice alfa , Dominios y Motivos de Interacción de Proteínas , Vacuolas/química
11.
J Biol Chem ; 294(42): 15505-15516, 2019 10 18.
Artículo en Inglés | MEDLINE | ID: mdl-31484721

RESUMEN

Unlike many other well-characterized bacteria, the opportunistic human pathogen Pseudomonas aeruginosa relies exclusively on the Entner-Doudoroff pathway (EDP) for glycolysis. Pyruvate kinase (PK) is the main "pacemaker" of the EDP, and its activity is also relevant for P. aeruginosa virulence. Two distinct isozymes of bacterial PK have been recognized, PykA and PykF. Here, using growth and expression analyses of relevant PK mutants, we show that PykA is the dominant isoform in P. aeruginosa Enzyme kinetics assays revealed that PykA displays potent K-type allosteric activation by glucose 6-phosphate and by intermediates from the pentose phosphate pathway. Unexpectedly, the X-ray structure of PykA at 2.4 Å resolution revealed that glucose 6-phosphate binds in a pocket that is distinct from the binding site reported for this metabolite in the PK from Mycobacterium tuberculosis (the only other available bacterial PK structure containing bound glucose 6-phosphate). We propose a mechanism by which glucose 6-phosphate binding at the allosteric site communicates with the PykA active site. Taken together, our findings indicate remarkable evolutionary plasticity in the mechanism(s) by which PK senses and responds to allosteric signals.


Asunto(s)
Proteínas Bacterianas/química , Pseudomonas aeruginosa/enzimología , Piruvato Quinasa/química , Regulación Alostérica , Sitio Alostérico , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Dominio Catalítico , Glucosa-6-Fosfato/metabolismo , Isoenzimas/química , Isoenzimas/genética , Isoenzimas/metabolismo , Modelos Moleculares , Vía de Pentosa Fosfato , Pseudomonas aeruginosa/química , Pseudomonas aeruginosa/genética , Pseudomonas aeruginosa/crecimiento & desarrollo , Piruvato Quinasa/genética , Piruvato Quinasa/metabolismo
12.
Molecules ; 25(23)2020 Nov 24.
Artículo en Inglés | MEDLINE | ID: mdl-33255326

RESUMEN

The ongoing pandemic caused by the novel coronavirus has been the greatest global health crisis since the Spanish flu pandemic of 1918. Thus far, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has resulted in over 1 million deaths, and there is no cure or vaccine to date. The recently solved crystal structure of the SARS-CoV-2 main protease has been a major focus for drug-discovery efforts. Here, we present a fragment-guided approach using ZINCPharmer, where 17 active fragments known to bind to the catalytic centre of the SARS-CoV-2 main protease (SARS-CoV-2 Mpro) were used as pharmacophore queries to search the ZINC databases of natural compounds and natural derivatives. This search yielded 134 hits that were then subjected to multiple rounds of in silico analyses, including blind and focused docking against the 3D structure of the main protease. We scrutinised the poses, scores, and protein-ligand interactions of 15 hits and selected 7. The scaffolds of the seven hits were structurally distinct from known inhibitor scaffolds, thus indicating scaffold novelty. Our work presents several novel scaffolds as potential candidates for experimental validation against SARS-CoV-2 Mpro.


Asunto(s)
Tratamiento Farmacológico de COVID-19 , Proteasas 3C de Coronavirus/antagonistas & inhibidores , Pandemias , SARS-CoV-2/química , COVID-19/virología , Proteasas 3C de Coronavirus/química , Humanos , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , SARS-CoV-2/efectos de los fármacos , SARS-CoV-2/patogenicidad
13.
Biochemistry ; 56(41): 5539-5549, 2017 10 17.
Artículo en Inglés | MEDLINE | ID: mdl-28985053

RESUMEN

Pseudomonas aeruginosa is an opportunistic human pathogen recognized as a critical threat by the World Health Organization because of the dwindling number of effective therapies available to treat infections. Over the past decade, it has become apparent that the glyoxylate shunt plays a vital role in sustaining P. aeruginosa during infection scenarios. The glyoxylate shunt comprises two enzymes: isocitrate lyase and malate synthase isoform G. Inactivation of these enzymes has been reported to abolish the ability of P. aeruginosa to establish infection in a mammalian model system, yet we still lack the structural information to support drug design efforts. In this work, we describe the first X-ray crystal structure of P. aeruginosa malate synthase G in the apo form at 1.62 Å resolution. The enzyme is a monomer composed of four domains and is highly conserved with homologues found in other clinically relevant microorganisms. It is also dependent on Mg2+ for catalysis. Metal ion binding led to a change in the intrinsic fluorescence of the protein, allowing us to quantitate its affinity for Mg2+. We also identified putative drug binding sites in malate synthase G using computational analysis and, because of the high resolution of the experimental data, were further able to characterize its hydration properties. Our data reveal two promising binding pockets in malate synthase G that may be exploited for drug design.


Asunto(s)
Proteínas Bacterianas/metabolismo , Malato Sintasa/metabolismo , Modelos Moleculares , Pseudomonas aeruginosa/enzimología , Acetilcoenzima A/química , Acetilcoenzima A/metabolismo , Secuencia de Aminoácidos , Apoenzimas/química , Apoenzimas/genética , Apoenzimas/metabolismo , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Sitios de Unión , Dominio Catalítico , Biología Computacional , Secuencia Conservada , Cristalografía por Rayos X , Sistemas Especialistas , Glioxilatos/química , Glioxilatos/metabolismo , Indoles/química , Indoles/metabolismo , Ligandos , Magnesio/química , Magnesio/metabolismo , Malato Sintasa/química , Malato Sintasa/genética , Simulación del Acoplamiento Molecular , Estructura Molecular , Conformación Proteica , Estructura Secundaria de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Alineación de Secuencia , Homología Estructural de Proteína
14.
Bioorg Med Chem Lett ; 27(4): 733-739, 2017 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-28129976

RESUMEN

Drug efflux pumps confer multidrug resistance to dangerous pathogens which makes these pumps important drug targets. We have synthesised a novel series of compounds based on a 2-naphthamide pharmacore aimed at inhibiting the efflux pumps from Gram-negative bacteria. The archeatypical transporter AcrB from Escherichia coli was used as model efflux pump as AcrB is widely conserved throughout Gram-negative organisms. The compounds were tested for their antibacterial action, ability to potentiate the action of antibiotics and for their ability to inhibit Nile Red efflux by AcrB. None of the compounds were antimicrobial against E. coli wild type cells. Most of the compounds were able to inhibit Nile Red efflux indicating that they are substrates of the AcrB efflux pump. Three compounds were able to synergise with antibiotics and reverse resistance in the resistant phenotype. Compound A3, 4-(isopentyloxy)-2-naphthamide, reduced the MICs of erythromycin and chloramphenicol to the MIC levels of the drug sensitive strain that lacks an efflux pump. A3 had no effect on the MIC of the non-substrate rifampicin indicating that this compound acts specifically through the AcrB efflux pump. A3 also does not act through non-specific mechanisms such as outer membrane or inner membrane permeabilisation and is not cytotoxic against mammalian cell lines. Therefore, we have designed and synthesised a novel chemical compound with great potential to further optimisation as inhibitor of drug efflux pumps.


Asunto(s)
Amidas/química , Antiinfecciosos/química , Proteínas de Escherichia coli/antagonistas & inhibidores , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/antagonistas & inhibidores , Amidas/farmacología , Amidas/toxicidad , Antiinfecciosos/farmacología , Antiinfecciosos/toxicidad , Sitios de Unión , Supervivencia Celular/efectos de los fármacos , Cloranfenicol/farmacología , Farmacorresistencia Bacteriana/efectos de los fármacos , Eritromicina/farmacología , Escherichia coli/efectos de los fármacos , Escherichia coli/metabolismo , Proteínas de Escherichia coli/metabolismo , Células HEK293 , Células Hep G2 , Humanos , Enlace de Hidrógeno , Pruebas de Sensibilidad Microbiana , Simulación del Acoplamiento Molecular , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/metabolismo , Naftoles/química , Estructura Terciaria de Proteína
15.
Proc Natl Acad Sci U S A ; 110(21): 8507-12, 2013 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-23650371

RESUMEN

Calcium-binding protein 1 (CaBP1) is a neuron-specific member of the calmodulin superfamily that regulates several Ca(2+) channels, including inositol 1,4,5-trisphosphate receptors (InsP3Rs). CaBP1 alone does not affect InsP3R activity, but it inhibits InsP3-evoked Ca(2+) release by slowing the rate of InsP3R opening. The inhibition is enhanced by Ca(2+) binding to both the InsP3R and CaBP1. CaBP1 binds via its C lobe to the cytosolic N-terminal region (NT; residues 1-604) of InsP3R1. NMR paramagnetic relaxation enhancement analysis demonstrates that a cluster of hydrophobic residues (V101, L104, and V162) within the C lobe of CaBP1 that are exposed after Ca(2+) binding interact with a complementary cluster of hydrophobic residues (L302, I364, and L393) in the ß-domain of the InsP3-binding core. These residues are essential for CaBP1 binding to the NT and for inhibition of InsP3R activity by CaBP1. Docking analyses and paramagnetic relaxation enhancement structural restraints suggest that CaBP1 forms an extended tetrameric turret attached by the tetrameric NT to the cytosolic vestibule of the InsP3R pore. InsP3 activates InsP3Rs by initiating conformational changes that lead to disruption of an intersubunit interaction between a "hot-spot" loop in the suppressor domain (residues 1-223) and the InsP3-binding core ß-domain. Targeted cross-linking of residues that contribute to this interface show that InsP3 attenuates cross-linking, whereas CaBP1 promotes it. We conclude that CaBP1 inhibits InsP3R activity by restricting the intersubunit movements that initiate gating.


Asunto(s)
Proteínas de Unión al Calcio/metabolismo , Receptores de Inositol 1,4,5-Trifosfato/metabolismo , Activación del Canal Iónico/fisiología , Proteínas del Tejido Nervioso/metabolismo , Animales , Proteínas de Unión al Calcio/química , Proteínas de Unión al Calcio/genética , Línea Celular , Interacciones Hidrofóbicas e Hidrofílicas , Receptores de Inositol 1,4,5-Trifosfato/química , Receptores de Inositol 1,4,5-Trifosfato/genética , Simulación del Acoplamiento Molecular , Proteínas del Tejido Nervioso/química , Proteínas del Tejido Nervioso/genética , Resonancia Magnética Nuclear Biomolecular , Unión Proteica , Estructura Cuaternaria de Proteína , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Ratas
16.
Biochem Biophys Res Commun ; 450(1): 384-9, 2014 Jul 18.
Artículo en Inglés | MEDLINE | ID: mdl-24942880

RESUMEN

Acid sensing ion channels (ASICs) are proton-gated cation channels that are expressed throughout the nervous system and have been implicated in mediating sensory perception of noxious stimuli. Amongst the six ASIC isoforms, ASIC1a, 1b, 2a and 3 form proton-gated homomers, which differ in their activation and inactivation kinetics, expression profiles and pharmacological modulation; protons do not gate ASIC2b and ASIC4. As with many other ion channels, structure-function studies of ASICs have been greatly aided by the discovery of some toxins that act in isoform-specific ways. ASIC3 is predominantly expressed by sensory neurons of the peripheral nervous system where it acts to detect acid as a noxious stimulus and thus plays an important role in nociception. ASIC3 is the only ASIC subunit that is inhibited by the sea anemone (Anthopleura elegantissima)-derived toxin APETx2. However, the molecular mechanism by which APETx2 interacts with ASIC3 remains largely unknown. In this study, we made a homology model of ASIC3 and used extensive protein-protein docking to predict for the first time, the probable sites of APETx2 interaction on ASIC3. Additionally, using computational alanine scanning, we also suggest the 'hot-spots' that are likely to be critical for ASIC3-APETx2 interaction.


Asunto(s)
Canales Iónicos Sensibles al Ácido/química , Canales Iónicos Sensibles al Ácido/ultraestructura , Venenos de Cnidarios/química , Membrana Dobles de Lípidos/química , Modelos Químicos , Modelos Moleculares , Animales , Sitios de Unión , Pollos , Simulación por Computador , Unión Proteica , Conformación Proteica , Ratas
17.
Biochem Soc Trans ; 42(1): 63-70, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24450629

RESUMEN

In the 30 years since IP3 (inositol 1,4,5-trisphosphate) was first shown to release Ca2+ from intracellular stores, the importance of spatially organized interactions within IP3-regulated signalling pathways has been universally recognized. Recent evidence that addresses three different levels of the structural determinants of IP3-evoked Ca2+ signalling is described in the present review. High-resolution structures of the N-terminal region of the IP3R (IP3 receptor) have established that the two essential phosphate groups of IP3 bind to opposite sides of the IP3-binding site, pulling its two domains together. This conformational change is proposed to disrupt an interaction between adjacent subunits within the tetrameric IP3R that normally holds the channel in a closed state. Similar structural changes are thought to allow gating of ryanodine receptors. cAMP increases the sensitivity of IP3Rs and thereby potentiates the Ca2+ signals evoked by receptors that stimulate IP3 formation. We speculate that both IP3 and cAMP are delivered to IP3Rs within signalling junctions, wherein the associated IP3Rs are exposed to a saturating concentration of either messenger. The concentration-dependent effects of extracellular stimuli come from recruitment of junctions rather than from a graded increase in the activity of individual junctions. IP3Rs within 'IP3 junctions' respond directly to receptors that stimulate phospholipase C, whereas extra-junctional IP3Rs are exposed to suboptimal concentrations of IP3 and open only when they are sensitized by cAMP. These results highlight the importance of selective delivery of diffusible messengers to IP3Rs. The spatial organization of IP3Rs also allows them to direct Ca2+ to specific intracellular targets that include other IP3Rs, mitochondria and Ca2+-regulated channels and enzymes. IP3Rs also interact functionally with lysosomes because Ca2+ released by IP3Rs, but not that entering cells via store-operated Ca2+ entry pathways, is selectively accumulated by lysosomes. This Ca2+ uptake shapes the Ca2+ signals evoked by IP3 and it may regulate lysosomal behaviour.


Asunto(s)
Señalización del Calcio , Receptores de Inositol 1,4,5-Trifosfato/fisiología , Calcio/metabolismo , Retículo Endoplásmico/metabolismo , Humanos , Receptores de Inositol 1,4,5-Trifosfato/química , Lisosomas/metabolismo , Estructura Cuaternaria de Proteína , Estructura Terciaria de Proteína
18.
Biochem J ; 449(1): 39-49, 2013 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-23009366

RESUMEN

Binding of IP3 (inositol 1,4,5-trisphosphate) to the IP3-binding core (residues 224-604) of IP3Rs (IP3 receptors) initiates opening of these ubiquitous intracellular Ca2+ channels. The mechanisms are unresolved, but require conformational changes to pass through the suppressor domain (residues 1-223). A calmodulin-binding peptide derived from myosin light chain kinase uncouples these events. We identified a similar conserved 1-8-14 calmodulin-binding motif within the suppressor domain of IP3R1 and, using peptides and mutagenesis, we demonstrate that it is essential for IP3R activation, whether assessed by IP3-evoked Ca2+ release or patch-clamp recoding of nuclear IP3R. Mimetic peptides specifically inhibit activation of IP3R by uncoupling the IP3-binding core from the suppressor domain. Mutations of key hydrophobic residues within the endogenous 1-8-14 motif mimic the peptides. Our results show that an endogenous 1-8-14 motif mediates conformational changes that are essential for IP3R activation. The inhibitory effects of calmodulin and related proteins may result from disruption of this essential interaction.


Asunto(s)
Calmodulina/metabolismo , Receptores de Inositol 1,4,5-Trifosfato/metabolismo , Secuencias de Aminoácidos/genética , Secuencia de Aminoácidos , Animales , Sitios de Unión/fisiología , Calmodulina/química , Calmodulina/genética , Bovinos , Línea Celular , Receptores de Inositol 1,4,5-Trifosfato/química , Receptores de Inositol 1,4,5-Trifosfato/genética , Datos de Secuencia Molecular , Unión Proteica/fisiología , Conformación Proteica , Ratas
19.
Biochem J ; 453(1): 147-51, 2013 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-23634879

RESUMEN

TPCs (two-pore channels) are NAADP (nicotinic acid-adenine dinucleotide phosphate)-sensitive Ca2+-permeable ion channels expressed on acidic organelles. In the present study we show that deletion of the N-terminal region redirects TPC1 to the ER (endoplasmic reticulum). The introduction of fluorophores at the N-terminus of TPC1 does not affect its subcellular location, but does reversibly abolish NAADP sensitivity. Our results reveal a dual role for the N-terminus in localization and function of TPC1.


Asunto(s)
Canales de Calcio/fisiología , Señalización del Calcio/fisiología , Retículo Endoplásmico/metabolismo , NADP/análogos & derivados , Humanos , NADP/fisiología , Fragmentos de Péptidos/farmacología
20.
ACS Omega ; 9(2): 2250-2262, 2024 Jan 16.
Artículo en Inglés | MEDLINE | ID: mdl-38250404

RESUMEN

The protein c-Myc is a transcription factor that remains largely intrinsically disordered and is known to be involved in various biological processes and is overexpressed in various cancers, making it an attractive drug target. However, intrinsically disordered proteins such as c-Myc do not show funnel-like basins in their free-energy landscapes; this makes their druggability a challenge. For the first time, we propose a heterodimer model of c-Myc/Max in full length in this work. We used Gaussian-accelerated molecular dynamics (GaMD) simulations to explore the behavior of c-Myc and its various regions, including the transactivation domain (TAD) and the basic helix-loop-helix-leucine-zipper (bHLH-Zipper) motif in three different conformational states: (a) monomeric c-Myc, (b) c-Myc when bound to its partner protein, Max, and (c) when Max was removed after binding. We analyzed the GaMD trajectories using root-mean-square deviation (RMSD), radius of gyration, root-mean-square fluctuation, and free-energy landscape (FEL) calculations to elaborate the behaviors of these regions. The results showed that the monomeric c-Myc structure showed a higher RMSD fluctuation as compared with the c-Myc/Max heterodimer in the bHLH-Zipper motif. This indicated that the bHLH-Zipper motif of c-Myc is more stable when it is bound to Max. The TAD region in both monomeric and Max-bound states showed similar plasticity in terms of RMSD. We also conducted residue decomposition calculations and showed that the c-Myc and Max interaction could be driven mainly by electrostatic interactions and the residues Arg299, Ile403, and Leu420 seemed to play important roles in the interaction. Our work provides insights into the behavior of c-Myc and its regions that could support the development of drugs that target c-Myc and other intrinsically disordered proteins.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA