Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
J Pediatr Neurosci ; 16(2): 149-152, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-35018184

RESUMEN

ß-Mannosidosis is a rare lysosomal storage disorder that is caused by a deficiency of ß-mannosidase activity, which is due to mutations of the MANBA gene. Two Indian siblings born out of a third-degree consanguineous marriage presented during late infancy with global developmental delay. On examination, both the siblings had hypotonia; hepatosplenomegaly was present in the first sibling whereas it was absent in the second sibling. Fundus evaluation, hearing assessment, and skeletal survey were normal in both siblings. Enzyme assay showed the absence of the ß-mannosidase enzyme. Next-generation sequencing showed a homozygous variation of c.1317 + 1G>A in intron 10 of the MANBA (-) gene in the elder sibling. Sanger sequencing confirmed the same mutation in the homozygous state in both siblings and in the heterozygous state in both parents.

3.
Neurosci Lett ; 467(1): 1-5, 2009 Dec 18.
Artículo en Inglés | MEDLINE | ID: mdl-19788911

RESUMEN

Membranous adenylyl cyclase (AC) subtypes play differential roles in the regulation of cell functions. The C1- and C2-subunits of AC form a heterodimer that efficiently catalyzes cAMP formation and constitutes a very useful model system for AC analysis at a molecular level. Intriguingly, C1 and C2 homodimers exist, too. The C2 homodimer is catalytically inactive and possesses two forskolin binding sites. However, little is known about the C1 homodimer. Therefore, in this study, we examined the C1 homodimer. C1 exhibited exceedingly low catalytic activity but high substrate-affinity. Fluorescence studies with the AC inhibitor 2',3'-O-(2,4,6-trinitrophenyl)-ATP suggested that 2 mol of C1 binds 1 mol of nucleotide, pointing to homodimerization. C1 also bound the AC inhibitor 2',3'-O-(N-methylanthraniloyl)-GTP as assessed by direct fluorescence and fluorescence resonance energy transfer studies. Molecular modelling revealed that in the C1 homodimer, the catalytic base arginine is exchanged against histidine. The lower basicity and shorter side chain of histidine probably account for the low catalytic activity. In conclusion, the C1 homodimer of AC binds nucleotides with high affinity, but exhibits only exceedingly low catalytic activity. The low catalytic activity of the C1 homodimer may constitute a mechanism by which in intact cells dimeric AC molecules exhibit a high signal-to-noise ratio upon stimulation by receptor agonists.


Asunto(s)
Adenilil Ciclasas/química , Adenilil Ciclasas/metabolismo , Nucleótidos/metabolismo , Adenosina Trifosfato/análogos & derivados , Adenosina Trifosfato/química , Adenosina Trifosfato/metabolismo , Inhibidores de Adenilato Ciclasa , Arginina/química , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/metabolismo , Fluorescencia , Transferencia Resonante de Energía de Fluorescencia , Guanosina Trifosfato/análogos & derivados , Guanosina Trifosfato/química , Guanosina Trifosfato/metabolismo , Histidina/química , Manganeso/química , Modelos Químicos , Modelos Moleculares , Nucleótidos/química , Multimerización de Proteína
4.
Biochem Pharmacol ; 78(3): 224-30, 2009 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-19492438

RESUMEN

Anthrax disease is caused by the spore-forming bacterium, Bacillus anthracis. B. anthracis produces a calmodulin-activated adenylyl cyclase (AC) toxin, edema factor (EF). Through excessive cAMP accumulation EF disrupts host defence. In a recent study [Taha HM, Schmidt J, Göttle M, Suryanarayana S, Shen Y, Tang WJ, et al. Molecular analysis of the interaction of anthrax adenylyl cyclase toxin, edema factor, with 2'(3')-O-(N-(methyl)anthraniloyl)-substituted purine and pyrimidine nucleotides. Mol Pharmacol 2009;75:693-703] we showed that various 2'(3')-O-N-(methyl)anthraniloyl (MANT)-substituted nucleoside 5'-triphosphates are potent inhibitors (K(i) values in the 0.1-5 microM range) of purified EF. Upon interaction with calmodulin we observed efficient fluorescence resonance energy transfer (FRET) between tryptophan and tyrosine residues of EF and the MANT-group of MANT-ATP. Molecular modelling suggested that both the 2'- and 3'-MANT-isomers can bind to EF. The aim of the present study was to examine the effects of defined 2'- and 3'-MANT-isomers of ATP and GTP on EF. 3'-MANT-2'-deoxy-ATP inhibited EF more potently than 2'-MANT-3'-deoxy-ATP, whereas the opposite was the case for the corresponding GTP analogs. Calmodulin-dependent direct MANT fluorescence and FRET was much larger with 2'-MANT-3'-deoxy-ATP and 2'-MANT-3'-deoxy-GTP compared to the corresponding 3'-MANT-2'-deoxy-isomers and the 2'(3')-racemates. K(i) values of MANT-nucleotides for inhibition of catalysis correlated with K(d) values of MANT-nucleotides in FRET studies. Molecular modelling indicated different positioning of the MANT-group in 2'-MANT-3'-deoxy-ATP/GTP and 3'-MANT-2'-deoxy-ATP/GTP bound to EF. Collectively, EF interacts differentially with 2'- and 3'-MANT-isomers of ATP and GTP, indicative for conformational flexibility of the catalytic site and offering a novel approach for the development of potent and selective EF inhibitors. Moreover, our present study may serve as a general model of how to use MANT-nucleotide isomers for the analysis of the molecular mechanisms of nucleotide/protein interactions.


Asunto(s)
Adenosina Trifosfato/análogos & derivados , Adenilil Ciclasas/química , Antígenos Bacterianos/química , Bacillus anthracis/enzimología , Toxinas Bacterianas/química , Guanosina Trifosfato/química , ortoaminobenzoatos/química , Adenosina Trifosfato/química , Transferencia Resonante de Energía de Fluorescencia , Isomerismo , Modelos Moleculares
5.
J Pharmacol Exp Ther ; 330(3): 687-95, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19494187

RESUMEN

Adenylyl cyclases (ACs) catalyze the conversion of ATP into the second messenger cAMP and play a key role in signal transduction. In a recent study (Mol Pharmacol 70:878-886, 2006), we reported that 2',3'-O-(2,4,6-trinitrophenyl)-substituted nucleoside 5'-triphosphates (TNP-NTPs) are potent inhibitors (K(i) values in the 10 nM range) of the purified catalytic subunits VC1 and IIC2 of membranous AC (mAC). The crystal structure of VC1:IIC2 in complex with TNP-ATP revealed that the nucleotide binds to the catalytic site with the TNP-group projecting into a hydrophobic pocket. The aims of this study were to analyze the interaction of TNP-nucleotides with VC1:IIC2 by fluorescence spectroscopy and to analyze inhibition of mAC isoforms, soluble AC (sAC), soluble guanylyl cyclase (sGC), and G-proteins by TNP-nucleotides. Interaction of VC1:IIC2 with TNP-NDPs and TNP-NTPs resulted in large fluorescence increases that were differentially reduced by a water-soluble forskolin analog. TNP-ATP turned out to be the most potent inhibitor for ACV (K(i), 3.7 nM) and sGC (K(i), 7.3 nM). TNP-UTP was identified as the most potent inhibitor for ACI (K(i), 7.1 nM) and ACII (K(i), 24 nM). TNP-NTPs inhibited sAC and GTP hydrolysis by G(s)- and G(i)-proteins only with low potencies. Molecular modeling revealed that TNP-GTP and TNP-ATP interact very similarly, but not identically, with VC1:IIC2. Collectively, our data show that TNP-nucleotides are useful fluorescent probes to monitor conformational changes in VC1:IIC2 and that TNP-NTPs are a promising starting point to develop isoform-selective AC and sGC inhibitors. TNP-ATP is the most potent sGC inhibitor known so far.


Asunto(s)
Inhibidores de Adenilato Ciclasa , Inhibidores Enzimáticos/farmacología , Guanilato Ciclasa/antagonistas & inhibidores , Nucleótidos/síntesis química , Nucleótidos/farmacología , Adenilil Ciclasas/genética , Membrana Celular/efectos de los fármacos , Membrana Celular/enzimología , Células Cultivadas , Colorantes Fluorescentes , GTP Fosfohidrolasas/antagonistas & inhibidores , GTP Fosfohidrolasas/metabolismo , Proteínas de Unión al GTP/metabolismo , Guanilato Ciclasa/genética , Humanos , Técnicas In Vitro , Isoenzimas/antagonistas & inhibidores , Modelos Moleculares , Unión Proteica , Transducción de Señal/efectos de los fármacos , Espectrometría de Fluorescencia
6.
Mol Pharmacol ; 75(3): 693-703, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19056899

RESUMEN

Bacillus anthracis causes anthrax disease and exerts its deleterious effects by the release of three exotoxins: lethal factor, protective antigen, and edema factor (EF), a highly active calmodulin-dependent adenylyl cyclase (AC). However, conventional antibiotic treatment is ineffective against either toxemia or antibiotic-resistant strains. Thus, more effective drugs for anthrax treatment are needed. Previous studies from our laboratory showed that mammalian membranous AC (mAC) exhibits broad specificity for purine and pyrimidine nucleotides ( Mol Pharmacol 70: 878-886, 2006 ). Here, we investigated structural requirements for EF inhibition by natural purine and pyrimidine nucleotides and nucleotides modified with N-methylanthraniloyl (MANT)- or anthraniloyl groups at the 2'(3')-O-ribosyl position. MANT-CTP was the most potent EF inhibitor (K(i), 100 nM) among 16 compounds studied. MANT-nucleotides inhibited EF competitively. Activation of EF by calmodulin resulted in effective fluorescence resonance energy transfer (FRET) from tryptophan and tyrosine residues located in the vicinity of the catalytic site to MANT-ATP, but FRET to MANT-CTP was only small. Mutagenesis studies revealed that Phe586 is crucial for FRET to MANT-ATP and MANT-CTP and that the mutations N583Q, K353A, and K353R differentially alter the inhibitory potencies of MANT-ATP and MANT-CTP. Docking approaches relying on crystal structures of EF indicate similar binding modes of the MANT nucleotides with subtle differences in the region of the nucleobases. In conclusion, like mAC, EF accommodates both purine and pyrimidine nucleotides. The unique preference of EF for the base cytosine offers an excellent starting point for the development of potent and selective EF inhibitors.


Asunto(s)
Adenilil Ciclasas/metabolismo , Vacunas contra el Carbunco/metabolismo , Antígenos Bacterianos/metabolismo , Toxinas Bacterianas/metabolismo , Nucleótidos de Purina/metabolismo , Nucleótidos de Pirimidina/metabolismo , Adenosina Difosfato/análogos & derivados , Adenosina Difosfato/química , Adenosina Difosfato/metabolismo , Adenilil Ciclasas/química , Adenilil Imidodifosfato/análogos & derivados , Adenilil Imidodifosfato/química , Adenilil Imidodifosfato/metabolismo , Animales , Vacunas contra el Carbunco/química , Vacunas contra el Carbunco/genética , Antígenos Bacterianos/química , Antígenos Bacterianos/genética , Toxinas Bacterianas/química , Toxinas Bacterianas/genética , Catálisis , Bovinos , Cristalografía por Rayos X , Transferencia Resonante de Energía de Fluorescencia , Mutagénesis Sitio-Dirigida , Unión Proteica/genética , Nucleótidos de Purina/química , Nucleótidos de Pirimidina/química , ortoaminobenzoatos/química , ortoaminobenzoatos/metabolismo
7.
Mol Pharmacol ; 70(3): 878-86, 2006 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16766715

RESUMEN

Membrane adenylyl cyclases (mACs) play an important role in signal transduction and are therefore potential drug targets. Earlier, we identified 2',3'-O-(N-methylanthraniloyl) (MANT)-substituted purine nucleotides as a novel class of highly potent competitive mAC inhibitors (Ki values in the 10 nM range). MANT nucleotides discriminate among various mAC isoforms through differential interactions with a binding pocket localized at the interface between the C1 and C2 domains of mAC. In this study, we examine the structure/activity relationships for 2',3'-substituted nucleotides and compare the crystal structures of mAC catalytic domains (VC1:IIC2) bound to MANT-GTP, MANT-ATP, and 2',3'-(2,4,6-trinitrophenyl) (TNP)-ATP. TNP-substituted purine and pyrimidine nucleotides inhibited VC1:IIC2 with moderately high potency (Ki values in the 100 nM range). Elongation of the linker between the ribosyl group and the MANT group and substitution of N-adenine atoms with MANT reduces inhibitory potency. Crystal structures show that MANT-GTP, MANT-ATP, and TNP-ATP reside in the same binding pocket in the VC1:IIC2 protein complex, but there are substantial differences in interactions of base, fluorophore, and polyphosphate chain of the inhibitors with mAC. Fluorescence emission and resonance transfer spectra also reflect differences in the interaction between MANT-ATP and VC1:IIC2 relative to MANT-GTP. Our data are indicative of a three-site mAC pharmacophore; the 2',3'-O-ribosyl substituent and the polyphosphate chain have the largest impact on inhibitor affinity and the nucleotide base has the least. The mAC binding site exhibits broad specificity, accommodating various bases and fluorescent groups at the 2',3'-O-ribosyl position. These data should greatly facilitate the rational design of potent, isoform-selective mAC inhibitors.


Asunto(s)
Adenosina Trifosfato/análogos & derivados , Adenilil Ciclasas/química , Adenilil Ciclasas/metabolismo , ortoaminobenzoatos/metabolismo , Adenosina Trifosfato/metabolismo , Adenosina Trifosfato/farmacología , Animales , Dominio Catalítico/efectos de los fármacos , Cristalografía por Rayos X , Electrones , Fluorescencia , Modelos Moleculares , Especificidad por Sustrato/efectos de los fármacos , ortoaminobenzoatos/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...