Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Artículo en Inglés | MEDLINE | ID: mdl-39356593

RESUMEN

In this article, we present a novel approach for designing a low-power, low-area impulse radio ultra-wideband (IR-UWB) noncoherent receiver capable of achieving a data rate of 100 Mbps. Our proposed receiver demonstrates the ability to demodulate ON-OFF keying pulse streams across the entire lower frequency band defined by the Federal Communication Commission for UWB applications. The key components of the proposed receiver include a reconfigurable differential two-stage low-noise amplifier, a fully differential squarer, narrow-band interface rejection filters, and variable gain baseband amplifiers. These circuits work cohesively to ensure efficient signal reception and processing. To validate the performance of the proposed receiver, we implemented the design using TSMC 40-nm CMOS process technology. A short-range communication including a 1.5 cm tissue layer is tested utilizing a typical upconversion UWB transmitter fabricated in the same technology. Remarkably, the proposed receiver achieves a data rate of 100 Mbps with an impressively low energy efficiency of 78.8 pJ/b and occupies an area of 0.705 mm2. The compact size, remarkable energy efficiency, and high data rate capabilities of the proposed receiver meet the stringent requirements of neural recording implants.

2.
IEEE Trans Biomed Circuits Syst ; 18(2): 369-382, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-37938944

RESUMEN

Brain-machine interfaces (BMI) are widely adopted in neuroscience investigations and neural prosthetics, with sensing channel counts constantly increasing. These Investigations place increasing demands for high data rates and low-power implantable devices despite high tissue losses. The Impulse radio ultra-wideband (IR-UWB), a revived wireless technology for short-range radios, has been widely used in various applications. Since the requirements and solutions are application-oriented, in this review paper we focus on neural recording implants with high-data rates and ultra-low power requirements. We examine in detail the working principle, design methodology, performance, and implementations of different architectures of IR-UWB transceivers in a quantitative manner to draw a deep comparison and extract the bottlenecks and possible solutions concerning the dedicated application. Our analysis shows that current solutions rely on enhanced or combined modulation techniques to improve link margin. An in-depth study of prior-art publications that achieved Gbps data rates concludes that edge-combination architecture and non-coherent detectors are remarkable for transmitter and receiver, respectively. Although the aim to minimize power and improve data rate - defined as energy efficiency (pJ/b) - extending communication distance despite high tissue losses and limited power budget, good narrow-band interference (NBI) tolerance coexisted in the same frequency band of UWB systems, and compatibility with energy harvesting designs are among the critical challenges remained unsolved. Furthermore, we expect that the combination of artificial intelligence (AI) and the inherent advantages of UWB radios will pave the way for future improvements in BMIs.


Asunto(s)
Interfaces Cerebro-Computador , Prótesis Neurales , Inteligencia Artificial , Prótesis e Implantes , Tecnología Inalámbrica
3.
Comput Biol Chem ; 80: 249-258, 2019 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-31029750

RESUMEN

An efficient, borax-catalyzed protocol for the synthesis of novel 4-aryl-substituted-4H-pyran derivatives fused to α-pyrone ring in a one-pot is described. By this achievement, some novel 4-aryl substituted 4H-pyrans fused to the α-pyrone ring as potential acetylcholinesterase inhibitors (AChEIs) with good to excellent yields are obtained from a one-pot three-component reaction between various aryl aldehydes, 4-hydroxy-6-methyl-2H-pyran-2-one and malononitrile. The method is a facile, inexpensive, practical and highly efficient one to obtain target compounds. The chemical structures of all compounds were characterized by FT-IR, FT-13CNMR and FT-1HNMR, MS spectroscopy and also elemental analyses data. Furthermore, the purity of all novel compounds was checked by HPLC. In addition, both molecular modelling studies and Absorption, Distribution, Metabolism, Excretion and Toxicity (ADMETox) prediction nominated all compounds as good acetylcholinesterase inhibitors to the potential treatment of Alzheimer, Parkinson and Autism diseases that among them compound 4f showed the best activity against acetylcholinesterase enzyme.


Asunto(s)
Acetilcolinesterasa/química , Inhibidores de la Colinesterasa/química , Pironas/química , Acetilcolinesterasa/metabolismo , Boratos/química , Catálisis , Inhibidores de la Colinesterasa/síntesis química , Inhibidores de la Colinesterasa/metabolismo , Inhibidores de la Colinesterasa/farmacocinética , Tecnología Química Verde/métodos , Humanos , Cinética , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Unión Proteica , Pironas/síntesis química , Pironas/metabolismo , Pironas/farmacocinética
4.
J Am Chem Soc ; 140(45): 15300-15308, 2018 11 14.
Artículo en Inglés | MEDLINE | ID: mdl-30296064

RESUMEN

The modification of proteins with O-linked N-acetylglucosamine ( O-GlcNAc) by the enzyme O-GlcNAc transferase (OGT) has emerged as an important regulator of cellular physiology. Metabolic labeling strategies to monitor O-GlcNAcylation in cells have proven of great value for uncovering the molecular roles of O-GlcNAc. These strategies rely on two-step labeling procedures, which limits the scope of experiments that can be performed. Here, we report on the creation of fluorescent uridine 5'-diphospho- N-acetylglucosamine (UDP-GlcNAc) analogues in which the N-acyl group of glucosamine is modified with a suitable linker and fluorophore. Using human OGT, we show these donor sugar substrates permit direct monitoring of OGT activity on protein substrates in vitro. We show that feeding cells with a corresponding fluorescent metabolic precursor for the last step of the hexosamine biosynthetic pathway (HBP) leads to its metabolic assimilation and labeling of O-GlcNAcylated proteins within live cells. This one-step metabolic feeding strategy permits labeling of O-GlcNAcylated proteins with a fluorescent glucosamine-nitrobenzoxadiazole (GlcN-NBD) conjugate that accumulates in a time- and dose-dependent manner. Because no genetic engineering of cells is required, we anticipate this strategy should be generally amenable to studying the roles of O-GlcNAc in cellular physiology as well as to gain an improved understanding of the regulation of OGT within cells. The further expansion of this one-step in-cell labeling strategy should enable performing a range of experiments including two-color pulse chase experiments and monitoring OGT activity on specific protein substrates in live cells.


Asunto(s)
Acetilglucosamina/química , Fluorescencia , N-Acetilglucosaminiltransferasas/química , Acetilglucosamina/metabolismo , Glicosilación , Células HeLa , Humanos , Estructura Molecular , N-Acetilglucosaminiltransferasas/metabolismo
5.
ACS Chem Biol ; 12(1): 206-213, 2017 01 20.
Artículo en Inglés | MEDLINE | ID: mdl-27935279

RESUMEN

O-GlcNAc transferase (OGT) catalyzes the installation of N-acetylglucosamine (GlcNAc) O-linked to nucleocytoplasmic proteins (O-GlcNAc) within multicellular eukaryotes. OGT shows surprising tolerance for structural changes in the sugar component of its nucleotide sugar donor substrate, uridine diphosphate N-acetylglucosamine (UDP-GlcNAc). Here, we find that OGT uses UDP-glucose to install O-linked glucose (O-Glc) onto proteins only 25-fold less efficiently than O-GlcNAc. Spurred by this observation, we show that OGT transfers 2-azido-2-deoxy-d-glucose (GlcAz) in vitro from UDP-GlcAz to proteins. Further, feeding cells with per-O-acetyl GlcAz (AcGlcAz), in combination with inhibition or inducible knockout of OGT, shows OGT-dependent modification of nuclear and cytoplasmic proteins with O-GlcAz as detected using microscopy, immunoblot, and proteomics. We find that O-GlcAz is reversible within cells, and an unidentified cellular enzyme exists to cleave O-Glc that can also process O-GlcAz. We anticipate that AcGlcAz will prove to be a useful tool to study the O-GlcNAc modification. We also speculate that, given the high concentration of UDP-Glc within certain mammalian tissues, O-Glc may exist within mammals and serve as a physiologically relevant modification.


Asunto(s)
Azidas/química , Desoxiglucosa/análogos & derivados , Glucosa/química , N-Acetilglucosaminiltransferasas/metabolismo , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Azidas/metabolismo , Células COS , Proteína Quinasa Tipo 4 Dependiente de Calcio Calmodulina/metabolismo , Chlorocebus aethiops , Desoxiglucosa/química , Glucosa/análogos & derivados , Glucosa/metabolismo , Glicosilación , Humanos , Glicoproteínas de Membrana/metabolismo , Ingeniería Metabólica , Ratones , N-Acetilglucosaminiltransferasas/genética , Proteínas de Complejo Poro Nuclear/metabolismo , Especificidad por Sustrato , Tritio , Uridina Difosfato Glucosa/análogos & derivados , Uridina Difosfato Glucosa/química , Uridina Difosfato Glucosa/metabolismo , beta-N-Acetilhexosaminidasas/química , Proteínas tau/metabolismo
6.
Nat Chem Biol ; 11(5): 319-25, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25774941

RESUMEN

Nucleocytoplasmic glycosylation of proteins with O-linked N-acetylglucosamine residues (O-GlcNAc) is recognized as a conserved post-translational modification found in all metazoans. O-GlcNAc has been proposed to regulate diverse cellular processes. Impaired cellular O-GlcNAcylation has been found to lead to decreases in the levels of various proteins, which is one mechanism by which O-GlcNAc seems to exert its varied physiological effects. Here we show that O-GlcNAcylation also occurs cotranslationally. This process protects nascent polypeptide chains from premature degradation by decreasing cotranslational ubiquitylation. Given that hundreds of proteins are O-GlcNAcylated within cells, our findings suggest that cotranslational O-GlcNAcylation may be a phenomenon regulating proteostasis of an array of nucleocytoplasmic proteins. These findings set the stage to assess whether O-GlcNAcylation has a role in protein quality control in a manner that bears similarity with the role played by N-glycosylation within the secretory pathway.


Asunto(s)
Acetilglucosamina/química , Péptidos/química , Animales , Glicosilación , Células HEK293 , Humanos , Ratones , Ratones Noqueados , N-Acetilglucosaminiltransferasas/metabolismo , Plásmidos , Procesamiento Proteico-Postraduccional , Receptores Citoplasmáticos y Nucleares/efectos de los fármacos , Factor de Transcripción Sp1/química , Ubiquitinación
7.
Acc Chem Res ; 47(1): 211-25, 2014 Jan 21.
Artículo en Inglés | MEDLINE | ID: mdl-23964564

RESUMEN

In humans, four different enzymes mediate the digestion of ingested carbohydrates. First salivary and pancreatic α-amylases, the two endoacting retaining glucosidases, break down the complex starch molecules into smaller linear maltose-oligomers (LM) and branched α-limit dextrins (αLDx). Then two retaining exoglucosidases, maltase-glucoamylase (MGAM) and sucrase-isomaltase (SI), convert those molecules into glucose in the small intestine. The small intestinal brush-border epithelial cells anchor MGAM and SI, and each contains a catalytic N- and C-terminal subunit, ntMGAM, ctMGAM, ntSI, and ctSI, respectively. All four catalytic domains have, to varying extents, α-1,4-exohydrolytic glucosidase activity and belong to the glycoside hydrolase family 31 (GH31). ntSI and ctSI show additional activity toward α-1,6 (isomaltose substrates) and α-1,2 (sucrose) glycosidic linkages, respectively. Because they mediate the final steps of starch digestion, both MGAM and SI are important target enzymes for the treatment of type-2 diabetes. Because of their potent inhibitory activities against the mammalian intestinal α-glucosidases, sulfonium-ion glucosidase inhibitors isolated from the antidiabetic herbal extracts of various Salacia species have received considerable attention recently. Thus far, researchers have isolated eight sulfonium-ion glucosidase inhibitors from Salacia species: salaprinol, salacinol, ponkoranol, kotalanol, and four of their corresponding de-O-sulfonated compounds, the structures of which comprise a 1,4-anhydro-4-thio-d-arabinitol and a polyhydroxylated acyclic side chain. Some of these compounds more strongly inhibit human intestinal α-glucosidases than the currently available antidiabetic drugs, acarbose and miglitol, and could serve as lead candidates in the treatment of type-2 diabetes. In this Account, we summarize progress in the field since 2010 with this class of inhibitors, with particular focus on their selective inhibitory activities against the intestinal glucosidases. Through structure-activity relationship (SAR) studies, we have modified the natural compounds to derive more potent, nanomolar inhibitors of human MGAM and SI. This structural optimization also yielded the most potent inhibitors known to date for each subunit. Furthermore, we observed that some of our synthetic inhibitors selectively blocked the activity of some mucosal α-glucosidases. Those results led to our current working hypothesis that selective inhibitors can dampen the action of a fast digesting subunit or subunits which places the burden of digestion on slower digesting subunits. That strategy can control the rate of starch digestion and glucose release to the body. Decreasing the initial glucose spike after a carbohydrate-rich meal and extending postprandial blood glucose delivery to the body can be desirable for diabetics and patients with other metabolic syndrome-associated diseases.


Asunto(s)
Productos Biológicos/farmacología , Inhibidores Enzimáticos/farmacología , Glucosidasas/antagonistas & inhibidores , Hipoglucemiantes/farmacología , Compuestos de Azufre/farmacología , Animales , Productos Biológicos/química , Inhibidores Enzimáticos/química , Humanos , Hipoglucemiantes/química , Relación Estructura-Actividad , Compuestos de Azufre/química
10.
J Biol Chem ; 287(38): 31929-38, 2012 Sep 14.
Artículo en Inglés | MEDLINE | ID: mdl-22851177

RESUMEN

Starch digestion involves the breakdown by α-amylase to small linear and branched malto-oligosaccharides, which are in turn hydrolyzed to glucose by the mucosal α-glucosidases, maltase-glucoamylase (MGAM) and sucrase-isomaltase (SI). MGAM and SI are anchored to the small intestinal brush-border epithelial cells, and each contains a catalytic N- and C-terminal subunit. All four subunits have α-1,4-exohydrolytic glucosidase activity, and the SI N-terminal subunit has an additional exo-debranching activity on the α-1,6-linkage. Inhibition of α-amylase and/or α-glucosidases is a strategy for treatment of type 2 diabetes. We illustrate here the concept of "toggling": differential inhibition of subunits to examine more refined control of glucogenesis of the α-amylolyzed starch malto-oligosaccharides with the aim of slow glucose delivery. Recombinant MGAM and SI subunits were individually assayed with α-amylolyzed waxy corn starch, consisting mainly of maltose, maltotriose, and branched α-limit dextrins, as substrate in the presence of four different inhibitors: acarbose and three sulfonium ion compounds. The IC(50) values show that the four α-glucosidase subunits could be differentially inhibited. The results support the prospect of controlling starch digestion rates to induce slow glucose release through the toggling of activities of the mucosal α-glucosidases by selective enzyme inhibition. This approach could also be used to probe associated metabolic diseases.


Asunto(s)
Glucosa/metabolismo , Membrana Mucosa/enzimología , alfa-Glucosidasas/metabolismo , Animales , Diabetes Mellitus/metabolismo , Drosophila melanogaster , Glicósido Hidrolasas/química , Glicosilación , Humanos , Hidrólisis , Concentración 50 Inhibidora , Mucosa Intestinal/metabolismo , Cinética , Ratones , Modelos Químicos , Obesidad/metabolismo , Estructura Terciaria de Proteína , Proteínas Recombinantes/química
11.
Int J Antimicrob Agents ; 40(3): 273-6, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22784856

RESUMEN

Enzymes involved in N-glycan processing are targets of interest in the inhibition of host processes for the blockade of dengue virus (DENV) morphogenesis. Of the ten proteins encoded by DENV, three have N-glycosylation sites, namely pre-membrane/membrane protein (prM/M), envelope protein (E) and non-structural protein-1 (NS1). It is known that aberrations in the oligosaccharide portions at these N-glycan sites affect proper folding of these proteins during the translation process that, in turn, affects the morphogenesis of the budding DENV. Here we report on the testing for antiviral activity of four known sulfonium-ion α-glucosidase inhibitors and two 5-thiomannosylamine disaccharide derivatives against DENV. Two of the sulfonium ions tested, namely, kotalanol and its de-O-sulfonated derivative, naturally occurring potent intestinal α-glucosidase inhibitors, had comparable inhibitory activity [50% inhibitory concentration (IC(50))=25.1±13.1 µM and 50.4±8.6 µM, respectively] with that of ribavirin (IC(50)=25.2±8.3 µM), a commercially available antiviral agent. The 5-thiomannosylamines did not show any activity at the concentrations tested.


Asunto(s)
Amino Azúcares/farmacología , Antivirales/farmacología , Virus del Dengue/efectos de los fármacos , Disacáridos/farmacología , Inhibidores Enzimáticos/farmacología , Compuestos de Sulfonio/farmacología , Animales , Chlorocebus aethiops , Inhibidores de Glicósido Hidrolasas , Concentración 50 Inhibidora , Pruebas de Sensibilidad Microbiana , Células Vero
12.
Chemistry ; 17(52): 14817-25, 2011 Dec 23.
Artículo en Inglés | MEDLINE | ID: mdl-22127878

RESUMEN

The synthesis and glucosidase inhibitory activities of two C-3'- and C-5'-ß-maltose-extended analogues of the naturally occurring sulfonium-ion inhibitor, de-O-sulfonated ponkoranol, are described. The compounds are designed to test the specificity towards four intestinal glycoside hydrolase family 31 (GH31) enzyme activities, responsible for the hydrolysis of terminal starch products and sugars into glucose, in humans. The target sulfonium-ion compounds were synthesized by means of nucleophilic attack of benzyl protected 1,4-anhydro-4-thio-D-arabinitol at the C-6 position of 6-O-trifluoromethanesulfonyl trisaccharides as alkylating agents. The alkylating agents were synthesized from D-glucose by glycosylation at C-4 or C-2 with maltosyl trichloroacetimidate. Deprotection of the coupled products by using a two-step sequence, followed by reduction afforded the final compounds. Evaluation of the target compounds for inhibition of the four glucosidase activities indicated that selective inhibition of one enzyme over the others is possible.


Asunto(s)
Mucosa Intestinal/metabolismo , Intestinos/enzimología , Maltosa/antagonistas & inhibidores , Maltosa/química , Maltosa/síntesis química , Almidón/química , Complejo Sacarasa-Isomaltasa/antagonistas & inhibidores , Complejo Sacarasa-Isomaltasa/metabolismo , Compuestos de Sulfonio/antagonistas & inhibidores , Compuestos de Sulfonio/química , Tiofenos/antagonistas & inhibidores , Tiofenos/síntesis química , alfa-Glucosidasas/química , alfa-Glucosidasas/síntesis química , Catálisis , Inhibidores Enzimáticos/farmacología , Humanos , Estructura Molecular , Estereoisomerismo , Tiofenos/química
13.
Bioorg Med Chem Lett ; 21(21): 6491-4, 2011 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-21924903

RESUMEN

Human maltase glucoamylase (MGAM) and sucrase isomaltase (SI) are two human intestinal glucosidases responsible for the final step of starch hydrolysis. MGAM and SI are anchored to the small intestinal brush border epithelial cells and contain two catalytic N-terminal and C-terminal subunits. In this study, we report the inhibition profile of 3'-O-methylponkoranol for the individual recombinant N and C terminal enzymes and compare the inhibitory activities of this compound with de-O-sulfonated ponkoranol. We show that 3'-O-methylponkoranol inhibits the different subunits to different extents, with extraordinary selectivity for C-terminal SI (K(i)=7±2nM). The enzymes themselves could serve as therapeutic targets for the treatment of digestive disorders or their sequelae.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Enfermedades Gastrointestinales/tratamiento farmacológico , Inhibidores de Glicósido Hidrolasas , Complejo Sacarasa-Isomaltasa/antagonistas & inhibidores , Tiofenos/farmacología , Amilopectina/química , Amilosa/química , Secuencia de Carbohidratos , Inhibidores Enzimáticos/uso terapéutico , Humanos , Datos de Secuencia Molecular , Tiofenos/uso terapéutico
14.
Chem Commun (Camb) ; 47(32): 9134-6, 2011 Aug 28.
Artículo en Inglés | MEDLINE | ID: mdl-21750824

RESUMEN

The synthesis of selenium analogues of de-O-sulfonated ponkoranol, a naturally occurring sulfonium-ion glucosidase inhibitor isolated from Salacia reticulata, and their evaluation as glucosidase inhibitors against two recombinant intestinal enzymes maltase glucoamylase (MGAM) and sucrase isomaltase (SI) are described.


Asunto(s)
Inhibidores Enzimáticos/química , Inhibidores de Glicósido Hidrolasas , Intestinos/enzimología , Salacia/química , Selenio/química , Tiofenos/química , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Inhibidores Enzimáticos/farmacología , Humanos , Modelos Moleculares , Proteínas Recombinantes/antagonistas & inhibidores , Proteínas Recombinantes/metabolismo , Selenio/farmacología , Azufre/química , Tiofenos/farmacología , alfa-Glucosidasas/metabolismo
15.
Bioorg Med Chem Lett ; 20(19): 5686-9, 2010 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-20801033

RESUMEN

Ponkoranol is a naturally occurring glucosidase inhibitor isolated from the plant Salacia reticulata. The compound comprises a sulfonium ion with an internal sulfate counter ion. We report here an efficient synthetic route to 3'-O-methyl ponkoranol to test the hypothesis that occupation of a hydrophobic pocket by a methyl group instead of the polar sulfate ion within the active site of human N-terminal maltase glucoamylase would be beneficial. The synthetic strategy relies on the nucleophilic attack of 2,3,5-tri-O-benzyl-1,4-anhydro-4-thio-D-arabinitol at the C-6 position of benzyl 6-O-p-toluenesulfonyl ß-D-glucopyranoside, followed by deprotection using boron trichloride and reduction with sodium borohydride. The target compound inhibited the N-terminal catalytic domain of intestinal human maltase glucoamylase (ntMGAM) with a K(i) value of 0.50 ± 0.04 µM, higher than those of de-O-sulfonated ponkoranol (K(i)=43 ± 3 nM), or its 5'-stereoisomer (K(i)=15 ± 1 nM). We conclude that the interaction of the methyl group with hydrophobic residues in the active site is not as beneficial to inhibition of ntMGAM as the other interactions of the polyhydroxylated chain with active-site residues.


Asunto(s)
Inhibidores de Glicósido Hidrolasas , Intestinos/enzimología , Éteres Metílicos/química , Sulfatos/química , Tiofenos/química , Sitios de Unión , Dominio Catalítico , Cristalografía por Rayos X , Humanos , Estructura Terciaria de Proteína , Tiofenos/farmacología , alfa-Glucosidasas/metabolismo
16.
Bioorg Med Chem ; 18(8): 2829-35, 2010 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-20363144

RESUMEN

The syntheses of an isomer of kotalanol, a naturally occurring glucosidase inhibitor, and of kotalanol itself are described. The target compounds were synthesized by nucleophilic attack of PMB-protected 1,4-anhydro-4-thio-d-arabinitol at the least hindered carbon atom of two 1,3-cyclic sulfates, which were synthesized from d-mannose. Methoxymethyl ether and isopropylidene were chosen as protecting groups. The latter group was critical to ensure the facile deprotection of the coupled products in a one-step sequence to yield kotalanol and its isomer. The stereoisomer of kotalanol, with the opposite stereochemistry at the C-6' stereogenic centre, inhibited the N-terminal catalytic domain of intestinal human maltase glucoamylase (ntMGAM) with a K(i) value of 0.20+/-0.02microM; this compares to a K(i) value for kotalanol of 0.19+/-0.03microM. The results indicate that the configuration at C-6' is inconsequential for inhibitory activity against this enzyme.


Asunto(s)
Inhibidores Enzimáticos/síntesis química , Inhibidores de Glicósido Hidrolasas , Monosacáridos/síntesis química , Sulfatos/síntesis química , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Humanos , Isomerismo , Manosa/química , Monosacáridos/química , Monosacáridos/farmacología , Sulfatos/química , Sulfatos/farmacología , alfa-Glucosidasas/metabolismo
17.
Org Lett ; 12(7): 1632-5, 2010 Apr 02.
Artículo en Inglés | MEDLINE | ID: mdl-20218632

RESUMEN

Ponkoranol, a glucosidase inhibitor isolated from the plant Salacia reticulata, comprises a sulfonium ion with an internal sulfate counterion. An efficient synthetic route to de-O-sulfonated ponkoranol and its 5'-stereoisomer is reported, and it is shown that these compounds are potent glucosidase inhibitors that inhibit a key intestinal human glucosidase, the N-terminal catalytic domain of maltase glucoamylase, with K(i) values of 43 +/- 3 and 15 +/- 1 nM, respectively.


Asunto(s)
Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/farmacología , Inhibidores de Glicósido Hidrolasas , Tiofenos/síntesis química , Tiofenos/farmacología , Inhibidores Enzimáticos/química , Humanos , Conformación Molecular , Estereoisomerismo , Tiofenos/química , alfa-Glucosidasas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA