Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
Nature ; 535(7613): 517-522, 2016 07 28.
Artículo en Inglés | MEDLINE | ID: mdl-27437577

RESUMEN

Developmental signals of the Hedgehog (Hh) and Wnt families are transduced across the membrane by Frizzledclass G-protein-coupled receptors (GPCRs) composed of both a heptahelical transmembrane domain (TMD) and an extracellular cysteine-rich domain (CRD). How the large extracellular domains of GPCRs regulate signalling by the TMD is unknown. We present crystal structures of the Hh signal transducer and oncoprotein Smoothened, a GPCR that contains two distinct ligand-binding sites: one in its TMD and one in the CRD. The CRD is stacked a top the TMD, separated by an intervening wedge-like linker domain. Structure-guided mutations show that the interface between the CRD, linker domain and TMD stabilizes the inactive state of Smoothened. Unexpectedly, we find a cholesterol molecule bound to Smoothened in the CRD binding site. Mutations predicted to prevent cholesterol binding impair the ability of Smoothened to transmit native Hh signals. Binding of a clinically used antagonist, vismodegib, to the TMD induces a conformational change that is propagated to the CRD, resulting in loss of cholesterol from the CRD-linker domain-TMD interface. Our results clarify the structural mechanism by which the activity of a GPCR is controlled by ligand-regulated interactions between its extracellular and transmembrane domains.


Asunto(s)
Espacio Extracelular/metabolismo , Receptores Acoplados a Proteínas G/química , Receptores Acoplados a Proteínas G/metabolismo , Anilidas/química , Anilidas/metabolismo , Anilidas/farmacología , Antineoplásicos/metabolismo , Antineoplásicos/farmacología , Sitios de Unión/genética , Colesterol/metabolismo , Colesterol/farmacología , Cristalografía por Rayos X , Cisteína/química , Cisteína/genética , Cisteína/metabolismo , Proteínas Hedgehog/metabolismo , Humanos , Ligandos , Modelos Moleculares , Unión Proteica/genética , Estabilidad Proteica/efectos de los fármacos , Estructura Terciaria de Proteína/efectos de los fármacos , Estructura Terciaria de Proteína/genética , Piridinas/química , Piridinas/metabolismo , Piridinas/farmacología , Receptores Acoplados a Proteínas G/antagonistas & inhibidores , Receptores Acoplados a Proteínas G/genética , Transducción de Señal/efectos de los fármacos , Receptor Smoothened
2.
Proc Natl Acad Sci U S A ; 115(12): E2819-E2828, 2018 03 20.
Artículo en Inglés | MEDLINE | ID: mdl-29507247

RESUMEN

Treatment of bacterial infections is becoming a serious clinical challenge due to the global dissemination of multidrug antibiotic resistance, necessitating the search for alternative treatments to disarm the virulence mechanisms underlying these infections. Uropathogenic Escherichia coli (UPEC) employs multiple chaperone-usher pathway pili tipped with adhesins with diverse receptor specificities to colonize various host tissues and habitats. For example, UPEC F9 pili specifically bind galactose or N-acetylgalactosamine epitopes on the kidney and inflamed bladder. Using X-ray structure-guided methods, virtual screening, and multiplex ELISA arrays, we rationally designed aryl galactosides and N-acetylgalactosaminosides that inhibit the F9 pilus adhesin FmlH. The lead compound, 29ß-NAc, is a biphenyl N-acetyl-ß-galactosaminoside with a Ki of ∼90 nM, representing a major advancement in potency relative to the characteristically weak nature of most carbohydrate-lectin interactions. 29ß-NAc binds tightly to FmlH by engaging the residues Y46 through edge-to-face π-stacking with its A-phenyl ring, R142 in a salt-bridge interaction with its carboxylate group, and K132 through water-mediated hydrogen bonding with its N-acetyl group. Administration of 29ß-NAc in a mouse urinary tract infection (UTI) model significantly reduced bladder and kidney bacterial burdens, and coadministration of 29ß-NAc and mannoside 4Z269, which targets the type 1 pilus adhesin FimH, resulted in greater elimination of bacteria from the urinary tract than either compound alone. Moreover, FmlH specifically binds healthy human kidney tissue in a 29ß-NAc-inhibitable manner, suggesting a key role for F9 pili in human kidney colonization. Thus, these glycoside antagonists of FmlH represent a rational antivirulence strategy for UPEC-mediated UTI treatment.


Asunto(s)
Adhesinas de Escherichia coli/química , Antibacterianos/química , Antibacterianos/farmacología , Adhesión Bacteriana/efectos de los fármacos , Infecciones Urinarias/microbiología , Adhesinas de Escherichia coli/metabolismo , Animales , Cristalografía por Rayos X , Evaluación Preclínica de Medicamentos/métodos , Infecciones por Escherichia coli/tratamiento farmacológico , Infecciones por Escherichia coli/microbiología , Femenino , Galactósidos/síntesis química , Galactósidos/química , Humanos , Riñón/efectos de los fármacos , Riñón/metabolismo , Riñón/microbiología , Ligandos , Ratones Endogámicos C3H , Simulación del Acoplamiento Molecular , Imitación Molecular , Infecciones Urinarias/tratamiento farmacológico , Escherichia coli Uropatógena/efectos de los fármacos , Escherichia coli Uropatógena/patogenicidad
3.
J Lipid Res ; 60(3): 707-716, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30617147

RESUMEN

Cholesterol is an essential structural component of cellular membranes and precursor molecule for oxysterol, bile acid, and hormone synthesis. The study of intracellular cholesterol trafficking pathways has been limited in part due to a lack of suitable cholesterol analogues. Herein, we developed three novel diazirine alkyne cholesterol probes: LKM38, KK174, and KK175. We evaluated these probes as well as a previously described diazirine alkyne cholesterol analogue, trans-sterol, for their fidelity as cholesterol mimics and for study of cholesterol trafficking. LKM38 emerged as a promising cholesterol mimic because it both sustained the growth of cholesterol-auxotrophic cells and appropriately regulated key cholesterol homeostatic pathways. When presented as an ester in lipoprotein particles, LKM38 initially localized to the lysosome and subsequently trafficked to the plasma membrane and endoplasmic reticulum. LKM38 bound to diverse, established cholesterol binding proteins. Through a detailed characterization of the cellular behavior of a panel of diazirine alkyne probes using cell biological, biochemical trafficking assays and immunofluorescence approaches, we conclude that LKM38 can serve as a powerful tool for the study of cholesterol protein interactions and trafficking.


Asunto(s)
Alquinos/química , Colesterol/metabolismo , Diazometano/síntesis química , Diazometano/metabolismo , Espacio Intracelular/metabolismo , Sondas Moleculares/síntesis química , Sondas Moleculares/metabolismo , Transporte Biológico , Línea Celular Tumoral , Técnicas de Química Sintética , Diazometano/química , Homeostasis , Humanos , Lipoproteínas/metabolismo , Lisosomas/metabolismo , Sondas Moleculares/química
4.
J Biol Chem ; 292(22): 9294-9304, 2017 06 02.
Artículo en Inglés | MEDLINE | ID: mdl-28396346

RESUMEN

Voltage-dependent anion channel-1 (VDAC1) is a highly regulated ß-barrel membrane protein that mediates transport of ions and metabolites between the mitochondria and cytosol of the cell. VDAC1 co-purifies with cholesterol and is functionally regulated by cholesterol, among other endogenous lipids. Molecular modeling studies based on NMR observations have suggested five cholesterol-binding sites in VDAC1, but direct experimental evidence for these sites is lacking. Here, to determine the sites of cholesterol binding, we photolabeled purified mouse VDAC1 (mVDAC1) with photoactivatable cholesterol analogues and analyzed the photolabeled sites with both top-down mass spectrometry (MS), and bottom-up MS paired with a clickable, stable isotope-labeled tag, FLI-tag. Using cholesterol analogues with a diazirine in either the 7 position of the steroid ring (LKM38) or the aliphatic tail (KK174), we mapped a binding pocket in mVDAC1 localized to Thr83 and Glu73, respectively. When Glu73 was mutated to a glutamine, KK174 no longer photolabeled this residue, but instead labeled the nearby Tyr62 within this same binding pocket. The combination of analytical strategies employed in this work permits detailed molecular mapping of a cholesterol-binding site in a protein, including an orientation of the sterol within the site. Our work raises the interesting possibility that cholesterol-mediated regulation of VDAC1 may be facilitated through a specific binding site at the functionally important Glu73 residue.


Asunto(s)
Colesterol/química , Canal Aniónico 1 Dependiente del Voltaje/química , Marcadores de Afinidad , Animales , Sitios de Unión , Ratones , Resonancia Magnética Nuclear Biomolecular , Canal Aniónico 1 Dependiente del Voltaje/genética
5.
J Biol Chem ; 289(16): 11095-11110, 2014 Apr 18.
Artículo en Inglés | MEDLINE | ID: mdl-24596093

RESUMEN

Oxysterols, oxidized metabolites of cholesterol, are endogenous small molecules that regulate lipid metabolism, immune function, and developmental signaling. Although the cell biology of cholesterol has been intensively studied, fundamental questions about oxysterols, such as their subcellular distribution and trafficking pathways, remain unanswered. We have therefore developed a useful method to image intracellular 20(S)-hydroxycholesterol with both high sensitivity and spatial resolution using click chemistry and fluorescence microscopy. The metabolic labeling of cells with an alkynyl derivative of 20(S)-hydroxycholesterol has allowed us to directly visualize this oxysterol by attaching an azide fluorophore through cyclo-addition. Unexpectedly, we found that this oxysterol selectively accumulates in the Golgi membrane using a pathway that is sensitive to ATP levels, temperature, and lysosome function. Although previous models have proposed nonvesicular pathways for the rapid equilibration of oxysterols between membranes, direct imaging of oxysterols suggests that a vesicular pathway is responsible for differential accumulation of oxysterols in organelle membranes. More broadly, clickable alkynyl sterols may represent useful tools for sterol cell biology, both to investigate the functions of these important lipids and to decipher the pathways that determine their cellular itineraries.


Asunto(s)
Química Clic , Colorantes Fluorescentes , Aparato de Golgi/metabolismo , Hidroxicolesteroles , Membranas Intracelulares/metabolismo , Animales , Transporte Biológico Activo/fisiología , Células CHO , Cricetinae , Cricetulus , Colorantes Fluorescentes/química , Colorantes Fluorescentes/metabolismo , Hidroxicolesteroles/síntesis química , Hidroxicolesteroles/química , Hidroxicolesteroles/metabolismo , Ratones , Microscopía Fluorescente , Células 3T3 NIH
6.
Biochemistry ; 53(18): 3042-51, 2014 May 13.
Artículo en Inglés | MEDLINE | ID: mdl-24758724

RESUMEN

Side-chain oxysterols, such as 25-hydroxycholesterol (25-HC), are key regulators of cholesterol homeostasis. New evidence suggests that the alteration of membrane structure by 25-HC contributes to its regulatory effects. We have examined the role of oxysterol membrane effects on cholesterol accessibility within the membrane using perfringolysin O (PFO), a cholesterol-dependent cytolysin that selectively binds accessible cholesterol, as a sensor of membrane cholesterol accessibility. We show that 25-HC increases cholesterol accessibility in a manner dependent on the membrane lipid composition. Structural analysis of molecular dynamics simulations reveals that increased cholesterol accessibility is associated with membrane thinning, and that the effects of 25-HC on cholesterol accessibility are driven by these changes in membrane thickness. Further, we find that the 25-HC antagonist LY295427 (agisterol) abrogates the membrane effects of 25-HC in a nonenantioselective manner, suggesting that agisterol antagonizes the cholesterol-homeostatic effects of 25-HC indirectly through its membrane interactions. These studies demonstrate that oxysterols regulate cholesterol accessibility, and thus the availability of cholesterol to be sensed and transported throughout the cell, by modulating the membrane environment. This work provides new insights into how alterations in membrane structure can be used to relay cholesterol regulatory signals.


Asunto(s)
Membrana Celular/efectos de los fármacos , Colesterol/química , Toxinas Bacterianas/farmacología , Colestanoles/farmacología , Colesterol/metabolismo , Proteínas Hemolisinas/farmacología , Homeostasis/efectos de los fármacos , Hidroxicolesteroles/farmacología , Liposomas/metabolismo , Lípidos de la Membrana/química , Simulación de Dinámica Molecular , Relación Estructura-Actividad
7.
Langmuir ; 30(41): 12114-8, 2014 Oct 21.
Artículo en Inglés | MEDLINE | ID: mdl-25290635

RESUMEN

One of the long-standing issues surrounding cholesterol (Chol) relates to its two-faced character. In particular, the consequences of its having a rough ß-face and a smooth α-face on its structural influence in cell membranes has remained elusive. In this study, direct comparisons have been made between cholesterol and a "smoothened" analog, DChol (i.e., 18,19-dinorcholesterol) using model membranes and a combination of nearest-neighbor recognition, differential scanning calorimetry, fluorescence, and monolayer measurements. Taken together, these results indicate that subtle differences exist between the interaction of these two sterols with the different states of 1,2-dipalmitoyl-sn-glycero-3-phosphocholine (DPPC). Chol has a greater condensing power than DChol, but only slightly so, i.e., on the order of a few tens of calories per mole.


Asunto(s)
Colesterol/química , 1,2-Dipalmitoilfosfatidilcolina/análogos & derivados , 1,2-Dipalmitoilfosfatidilcolina/química , Conformación Molecular
8.
J Org Chem ; 79(12): 5636-43, 2014 Jun 20.
Artículo en Inglés | MEDLINE | ID: mdl-24823889

RESUMEN

Herein, we report the first synthesis of a demethylated form of cholesterol (18,19-di-nor-cholesterol), in which the C18 and C19 methyl groups of the ß-face were eliminated. Recent molecular simulations modeling 18,19-di-nor-cholesterol have suggested that cholesterol's opposing rough ß-face and smooth α-face play necessary roles in cholesterol's membrane condensing abilities and, additionally, that specific facial preferences are displayed as cholesterol interacts with different neighboring lipids and transmembrane proteins. Inspired by these poorly characterized biochemical interactions, an extensive 18-step synthesis was completed as part of a collaborative effort, wherein synthesizing a "smoothened" cholesterol analogue would provide a direct way to experimentally measure the significance of the ß-face methyl groups. Starting from known perhydrochrysenone A, the synthesis of 18,19-di-nor-cholesterol was accomplished with an excellent overall yield of 3.5%. The use of the highly stereoselective Dieckmann condensation and the employment of Evans' chiral auxiliary were both key to ensuring the success of this synthesis.


Asunto(s)
Colestenonas/síntesis química , Colesterol/síntesis química , Proteínas Ligadas a Lípidos/química , Colestenonas/química , Colesterol/química , Proteínas Ligadas a Lípidos/metabolismo , Espectroscopía de Resonancia Magnética , Estereoisomerismo , Relación Estructura-Actividad
9.
Sci Adv ; 8(22): eabm5563, 2022 06 03.
Artículo en Inglés | MEDLINE | ID: mdl-35658032

RESUMEN

Smoothened (SMO) transduces the Hedgehog (Hh) signal across the plasma membrane in response to accessible cholesterol. Cholesterol binds SMO at two sites: one in the extracellular cysteine-rich domain (CRD) and a second in the transmembrane domain (TMD). How these two sterol-binding sites mediate SMO activation in response to the ligand Sonic Hedgehog (SHH) remains unknown. We find that mutations in the CRD (but not the TMD) reduce the fold increase in SMO activity triggered by SHH. SHH also promotes the photocrosslinking of a sterol analog to the CRD in intact cells. In contrast, sterol binding to the TMD site boosts SMO activity regardless of SHH exposure. Mutational and computational analyses show that these sites are in allosteric communication despite being 45 angstroms apart. Hence, sterols function as both SHH-regulated orthosteric ligands at the CRD and allosteric ligands at the TMD to regulate SMO activity and Hh signaling.


Asunto(s)
Cisteína , Proteínas Hedgehog , Colesterol/metabolismo , Proteínas Hedgehog/química , Ligandos , Esteroles/química
11.
Biochim Biophys Acta Mol Cell Biol Lipids ; 1864(2): 128-136, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30471426

RESUMEN

Cholesterol is an essential component of cell membranes, and is required for mammalian pentameric ligand-gated ion channel (pLGIC) function. Computational studies suggest direct interactions between cholesterol and pLGICs but experimental evidence identifying specific binding sites is limited. In this study, we mapped cholesterol binding to Gloeobacter ligand-gated ion channel (GLIC), a model pLGIC chosen for its high level of expression, existing crystal structure, and previous use as a prototypic pLGIC. Using two cholesterol analogue photolabeling reagents with the photoreactive moiety on opposite ends of the sterol, we identified two cholesterol binding sites: an intersubunit site between TM3 and TM1 of adjacent subunits and an intrasubunit site between TM1 and TM4. In both the inter- and intrasubunit sites, cholesterol is oriented such that the 3­OH group points toward the center of the transmembrane domains rather than toward either the cytosolic or extracellular surfaces. We then compared this binding to that of the cholesterol metabolite, allopregnanolone, a neurosteroid that allosterically modulates pLGICs. The same binding pockets were identified for allopregnanolone and cholesterol, but the binding orientation of the two ligands was markedly different, with the 3­OH group of allopregnanolone pointing to the intra- and extracellular termini of the transmembrane domains rather than to their centers. We also found that cholesterol increases, whereas allopregnanolone decreases the thermal stability of GLIC. These data indicate that cholesterol and neurosteroids bind to common hydrophobic pockets in the model pLGIC, GLIC, but that their effects depend on the orientation and specific molecular interactions unique to each sterol.


Asunto(s)
Colesterol/metabolismo , Canales Iónicos Activados por Ligandos/fisiología , Neurotransmisores/metabolismo , Sitios de Unión/fisiología , Membrana Celular/metabolismo , Colesterol/fisiología , Cianobacterias/metabolismo , Canales Iónicos Activados por Ligandos/metabolismo , Ligandos , Modelos Moleculares , Neurotransmisores/fisiología , Etiquetas de Fotoafinidad/metabolismo , Pregnanolona/metabolismo , Unión Proteica/fisiología , Dominios Proteicos/fisiología
12.
Expert Opin Drug Discov ; 12(7): 711-731, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28506090

RESUMEN

INTRODUCTION: The bacterial adhesin FimH is a virulence factor and an attractive therapeutic target for urinary tract infection (UTI) and Crohn's Disease (CD). Located on type 1 pili of uropathogenic E. coli (UPEC), the FimH adhesin plays an integral role in the pathogenesis of UPEC. Recent efforts have culminated in the development of small-molecule mannoside FimH antagonists that target the mannose-binding lectin domain of FimH, inhibiting its function and preventing UPEC from binding mannosylated host cells in the bladder, thereby circumventing infection. Areas covered: The authors describe the structure-guided design of mannoside ligands, and review the structural biology of the FimH lectin domain. Additionally, they discuss the lead optimization of mannosides for therapeutic application in UTI and CD, and describe various assays used to measure mannoside potency in vitro and mouse models used to determine efficacy in vivo. Expert opinion: To date, mannoside optimization has led to a diverse set of small-molecule FimH antagonists with oral bioavailability. With clinical trials already initiated in CD and on the horizon for UTI, it is the authors, opinion that mannosides will be a 'first-in-class' treatment strategy for UTI and CD, and will pave the way for treatment of other Gram-negative bacterial infections.


Asunto(s)
Enfermedad de Crohn/tratamiento farmacológico , Proteínas Fimbrias/antagonistas & inhibidores , Infecciones Urinarias/tratamiento farmacológico , Adhesinas de Escherichia coli , Animales , Antibacterianos/administración & dosificación , Antibacterianos/farmacocinética , Antibacterianos/farmacología , Disponibilidad Biológica , Enfermedad de Crohn/microbiología , Modelos Animales de Enfermedad , Diseño de Fármacos , Infecciones por Escherichia coli/tratamiento farmacológico , Infecciones por Escherichia coli/microbiología , Humanos , Manósidos/administración & dosificación , Manósidos/farmacocinética , Manósidos/farmacología , Ratones , Relación Estructura-Actividad , Infecciones Urinarias/microbiología , Escherichia coli Uropatógena/aislamiento & purificación
13.
Science ; 355(6331): 1306-1311, 2017 03 24.
Artículo en Inglés | MEDLINE | ID: mdl-28336668

RESUMEN

The mechanistic target of rapamycin complex 1 (mTORC1) protein kinase is a master growth regulator that becomes activated at the lysosome in response to nutrient cues. Here, we identify cholesterol, an essential building block for cellular growth, as a nutrient input that drives mTORC1 recruitment and activation at the lysosomal surface. The lysosomal transmembrane protein, SLC38A9, is required for mTORC1 activation by cholesterol through conserved cholesterol-responsive motifs. Moreover, SLC38A9 enables mTORC1 activation by cholesterol independently from its arginine-sensing function. Conversely, the Niemann-Pick C1 (NPC1) protein, which regulates cholesterol export from the lysosome, binds to SLC38A9 and inhibits mTORC1 signaling through its sterol transport function. Thus, lysosomal cholesterol drives mTORC1 activation and growth signaling through the SLC38A9-NPC1 complex.


Asunto(s)
Sistemas de Transporte de Aminoácidos/metabolismo , Proteínas Portadoras/metabolismo , Colesterol/metabolismo , Lisosomas/metabolismo , Complejos Multiproteicos/metabolismo , Proteínas Nucleares/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Secuencias de Aminoácidos , Sistemas de Transporte de Aminoácidos/genética , Animales , Transporte Biológico , Células CHO , HDL-Colesterol/metabolismo , Cricetulus , Activación Enzimática , Fibroblastos , Células HEK293 , Humanos , Diana Mecanicista del Complejo 1 de la Rapamicina , Ratones , Complejos Multiproteicos/antagonistas & inhibidores , Mutación , Transducción de Señal , Serina-Treonina Quinasas TOR/antagonistas & inhibidores
14.
Expert Opin Ther Pat ; 26(2): 175-97, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26651364

RESUMEN

INTRODUCTION: Type 1 pili are utilized by Gram-negative bacteria to adhere to host tissue and thus are a key virulence factor in urinary tract infections (UTIs) and Crohn's disease (CD). This adhesion is mediated through specific binding of the terminal adhesin, FimH, to mannosylated host glycoproteins. FimH is essential for UTI pathogenesis and thus is a promising therapeutic target. AREAS COVERED: Herein, we review the structural frameworks of FimH antagonists disclosed in the patent literature. X-ray crystallographic binding studies of D-mannose and early FimH antagonists have uncovered key molecular interactions. Exploiting this knowledge, mannosides with extraordinarily high binding affinities have been designed. Structure-activity relationships (SAR) and structure-property relationship (SPR) studies have resulted in the rapid development of orally bioavailable FimH antagonists with promising therapeutic potential for UTI and CD. EXPERT OPINION: It is our opinion that biaryl or 'two-ring' mannosides, which represent the largest and most thoroughly tested class of FimH antagonists, also hold the most promise as a novel treatment for UTIs. These antagonists have also been shown to have efficacy in treating CD. Judging from the strong preclinical data, we predict that one or more FimH antagonists will be entering the clinic within the next 1-2 years.


Asunto(s)
Enfermedad de Crohn/tratamiento farmacológico , Proteínas Fimbrias/antagonistas & inhibidores , Infecciones Urinarias/tratamiento farmacológico , Adhesinas de Escherichia coli , Animales , Antibacterianos/administración & dosificación , Antibacterianos/química , Antibacterianos/farmacología , Enfermedad de Crohn/microbiología , Diseño de Fármacos , Bacterias Gramnegativas/efectos de los fármacos , Bacterias Gramnegativas/patogenicidad , Infecciones por Bacterias Gramnegativas/tratamiento farmacológico , Infecciones por Bacterias Gramnegativas/microbiología , Humanos , Manosa/química , Patentes como Asunto , Relación Estructura-Actividad , Infecciones Urinarias/microbiología
15.
J Med Chem ; 59(20): 9390-9408, 2016 Oct 27.
Artículo en Inglés | MEDLINE | ID: mdl-27689912

RESUMEN

Gram-negative uropathogenic Escherichia coli (UPEC) bacteria are a causative pathogen of urinary tract infections (UTIs). Previously developed antivirulence inhibitors of the type 1 pilus adhesin, FimH, demonstrated oral activity in animal models of UTI but were found to have limited compound exposure due to the metabolic instability of the O-glycosidic bond (O-mannosides). Herein, we disclose that compounds having the O-glycosidic bond replaced with carbon linkages had improved stability and inhibitory activity against FimH. We report on the design, synthesis, and in vivo evaluation of this promising new class of carbon-linked C-mannosides that show improved pharmacokinetic (PK) properties relative to O-mannosides. Interestingly, we found that FimH binding is stereospecifically modulated by hydroxyl substitution on the methylene linker, where the R-hydroxy isomer has a 60-fold increase in potency. This new class of C-mannoside antagonists have significantly increased compound exposure and, as a result, enhanced efficacy in mouse models of acute and chronic UTI.


Asunto(s)
Antibacterianos/farmacología , Escherichia coli/efectos de los fármacos , Escherichia coli/patogenicidad , Manósidos/administración & dosificación , Manósidos/farmacología , Infecciones Urinarias/tratamiento farmacológico , Infecciones Urinarias/microbiología , Administración Oral , Animales , Antibacterianos/administración & dosificación , Antibacterianos/química , Relación Dosis-Respuesta a Droga , Femenino , Manósidos/química , Ratones , Ratones Endogámicos C3H , Pruebas de Sensibilidad Microbiana , Modelos Moleculares , Estructura Molecular , Relación Estructura-Actividad , Virulencia/efectos de los fármacos
16.
ChemMedChem ; 11(4): 367-73, 2016 Feb 17.
Artículo en Inglés | MEDLINE | ID: mdl-26812660

RESUMEN

Uropathogenic E. coli (UPEC) employ the mannose-binding adhesin FimH to colonize the bladder epithelium during urinary tract infection (UTI). Previously reported FimH antagonists exhibit good potency and efficacy, but low bioavailability and a short half-life in vivo. In a rational design strategy, we obtained an X-ray structure of lead mannosides and then designed mannosides with improved drug-like properties. We show that cyclizing the carboxamide onto the biphenyl B-ring aglycone of biphenyl mannosides into a fused heterocyclic ring, generates new biaryl mannosides such as isoquinolone 22 (2-methyl-4-(1-oxo-1,2-dihydroisoquinolin-7-yl)phenyl α-d-mannopyranoside) with enhanced potency and in vivo efficacy resulting from increased oral bioavailability. N-Substitution of the isoquinolone aglycone with various functionalities produced a new potent subseries of FimH antagonists. All analogues of the subseries have higher FimH binding affinity than unsubstituted lead 22, as determined by thermal shift differential scanning fluorimetry assay. Mannosides with pyridyl substitution on the isoquinolone group inhibit bacteria-mediated hemagglutination and prevent biofilm formation by UPEC with single-digit nanomolar potency, which is unprecedented for any FimH antagonists or any other antivirulence compounds reported to date.


Asunto(s)
Adhesinas de Escherichia coli/metabolismo , Antibacterianos/farmacología , Infecciones por Escherichia coli/tratamiento farmacológico , Proteínas Fimbrias/metabolismo , Manósidos/farmacología , Infecciones Urinarias/tratamiento farmacológico , Escherichia coli Uropatógena/efectos de los fármacos , Antibacterianos/química , Enfermedad Crónica , Infecciones por Escherichia coli/microbiología , Humanos , Isoquinolinas/química , Isoquinolinas/farmacología , Manósidos/química , Simulación del Acoplamiento Molecular , Vejiga Urinaria/microbiología , Infecciones Urinarias/microbiología , Escherichia coli Uropatógena/metabolismo
17.
Sci Transl Med ; 8(337): 337ra63, 2016 05 04.
Artículo en Inglés | MEDLINE | ID: mdl-27147587

RESUMEN

Niemann-Pick disease type C (NPC) is a fatal, neurodegenerative, cholesterol storage disorder. With new therapeutics in clinical trials, it is imperative to improve diagnostics and facilitate early intervention. We used metabolomic profiling to identify potential markers and discovered three unknown bile acids that were increased in plasma from NPC but not control subjects. The bile acids most elevated in the NPC subjects were identified as 3ß,5α,6ß-trihydroxycholanic acid and its glycine conjugate, which were shown to be metabolites of cholestane-3ß,5α,6ß-triol, an oxysterol elevated in NPC. A high-throughput mass spectrometry-based method was developed and validated to measure the glycine-conjugated bile acid in dried blood spots. Analysis of dried blood spots from 4992 controls, 134 NPC carriers, and 44 NPC subjects provided 100% sensitivity and specificity in the study samples. Quantification of the bile acid in dried blood spots, therefore, provides the basis for a newborn screen for NPC that is ready for piloting in newborn screening programs.


Asunto(s)
Ácidos y Sales Biliares/sangre , Biomarcadores/sangre , Enfermedad de Niemann-Pick Tipo C/sangre , Enfermedad de Niemann-Pick Tipo C/diagnóstico , Pruebas con Sangre Seca , Humanos , Recién Nacido , Sensibilidad y Especificidad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA