Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Cell Chem Biol ; 23(8): 992-1001, 2016 08 18.
Artículo en Inglés | MEDLINE | ID: mdl-27499529

RESUMEN

Heat shock protein 70 (Hsp70) is a chaperone that normally scans the proteome and initiates the turnover of some proteins (termed clients) by linking them to the degradation pathways. This activity is critical to normal protein homeostasis, yet it appears to fail in diseases associated with abnormal protein accumulation. It is not clear why Hsp70 promotes client degradation under some conditions, while sparing that protein under others. Here, we used a combination of chemical biology and genetic strategies to systematically perturb the affinity of Hsp70 for the model client, tau. This approach revealed that tight complexes between Hsp70 and tau were associated with enhanced turnover while transient interactions favored tau retention. These results suggest that client affinity is one important parameter governing Hsp70-mediated quality control.


Asunto(s)
Benzotiazoles/farmacología , Proteínas HSP70 de Choque Térmico/metabolismo , Modelos Biológicos , Tauopatías/tratamiento farmacológico , Tauopatías/metabolismo , Tiazolidinas/farmacología , Proteínas tau/metabolismo , Benzotiazoles/química , Relación Dosis-Respuesta a Droga , Proteínas HSP70 de Choque Térmico/química , Células HeLa , Humanos , Estructura Molecular , Estabilidad Proteica/efectos de los fármacos , Relación Estructura-Actividad , Tiazolidinas/química , Células Tumorales Cultivadas , Proteínas tau/química
2.
Cell Rep ; 16(6): 1518-1526, 2016 08 09.
Artículo en Inglés | MEDLINE | ID: mdl-27452469

RESUMEN

G-protein-coupled receptors (GPCRs) initiate a variety of signaling cascades, depending on effector coupling. ß-arrestins, which were initially characterized by their ability to "arrest" GPCR signaling by uncoupling receptor and G protein, have recently emerged as important signaling effectors for GPCRs. ß-arrestins engage signaling pathways that are distinct from those mediated by G protein. As such, arrestin-dependent signaling can play a unique role in regulating cell function, but whether neuromodulatory GPCRs utilize ß-arrestin-dependent signaling to regulate neuronal excitability remains unclear. Here, we find that D3 dopamine receptors (D3R) regulate axon initial segment (AIS) excitability through ß-arrestin-dependent signaling, modifying CaV3 voltage dependence to suppress high-frequency action potential generation. This non-canonical D3R signaling thereby gates AIS excitability via pathways distinct from classical GPCR signaling pathways.


Asunto(s)
Segmento Inicial del Axón/metabolismo , Canales de Calcio/metabolismo , Dopamina/metabolismo , beta-Arrestinas/metabolismo , Animales , Calcio/metabolismo , Células HEK293 , Humanos , Fosforilación , Receptores Acoplados a Proteínas G/metabolismo
3.
J Neuropathol Exp Neurol ; 75(3): 256-62, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26851378

RESUMEN

Drug discovery for neurodegenerative diseases is particularly challenging because of the discrepancies in drug effects between in vitro and in vivo studies. These discrepancies occur in part because current cell culture systems used for drug screening have many limitations. First, few cell culture systems accurately model human aging or neurodegenerative diseases. Second, drug efficacy may differ between dividing and stationary cells, the latter resembling nondividing neurons in the CNS. Brain aggregates (BrnAggs) derived from embryonic day 15 gestation mouse embryos may represent neuropathogenic processes in prion disease and reflect in vivo drug efficacy. Here, we report a new method for the production of BrnAggs suitable for drug screening and suggest that BrnAggs can model additional neurological diseases such as tauopathies. We also report a functional assay with BrnAggs by measuring electrophysiological activities. Our data suggest that BrnAggs could serve as an effective in vitro cell culture system for drug discovery for neurodegenerative diseases.


Asunto(s)
Encéfalo/citología , Técnicas de Cultivo de Célula/métodos , Modelos Biológicos , Red Nerviosa/fisiología , Enfermedades Neurodegenerativas , Potenciales de Acción/efectos de los fármacos , Potenciales de Acción/fisiología , Factores de Edad , Animales , Células Cultivadas , Embrión de Mamíferos , Femenino , Humanos , Masculino , Ratones , Ratones Transgénicos , Red Nerviosa/efectos de los fármacos , Enfermedades Neurodegenerativas/genética , Enfermedades Neurodegenerativas/patología , Enfermedades Neurodegenerativas/fisiopatología , Neuroglía/fisiología , Neuronas/fisiología , Embarazo , Bloqueadores de los Canales de Sodio/farmacología , Proteínas tau/genética , Proteínas tau/metabolismo
4.
PLoS One ; 9(5): e98496, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24866748

RESUMEN

Prion disease is caused by a single pathogenic protein (PrPSc), an abnormal conformer of the normal cellular prion protein PrPC. Depletion of PrPC in prion knockout mice makes them resistant to prion disease. Thus, gene silencing of the Prnp gene is a promising effective therapeutic approach. Here, we examined adeno-associated virus vector type 2 encoding a short hairpin RNA targeting Prnp mRNA (AAV2-PrP-shRNA) to suppress PrPC expression both in vitro and in vivo. AAV2-PrP-shRNA treatment suppressed PrP levels and prevented dendritic degeneration in RML-infected brain aggregate cultures. Infusion of AAV2-PrP-shRNA-eGFP into the thalamus of CD-1 mice showed that eGFP was transported to the cerebral cortex via anterograde transport and the overall PrPC levels were reduced by ∼ 70% within 4 weeks. For therapeutic purposes, we treated RML-infected CD-1 mice with AAV2-PrP-shRNA beginning at 50 days post inoculation. Although AAV2-PrP-shRNA focally suppressed PrPSc formation in the thalamic infusion site by ∼ 75%, it did not suppress PrPSc formation efficiently in other regions of the brain. Survival of mice was not extended compared to the untreated controls. Global suppression of PrPC in the brain is required for successful therapy of prion diseases.


Asunto(s)
Dependovirus/genética , Enfermedades por Prión/terapia , Priones/antagonistas & inhibidores , ARN Interferente Pequeño/metabolismo , Tálamo/metabolismo , Animales , Femenino , Técnicas de Inactivación de Genes , Terapia Genética , Vectores Genéticos/administración & dosificación , Ratones , Proteínas PrPC/metabolismo , Enfermedades por Prión/metabolismo , Enfermedades por Prión/patología , Proteínas Priónicas , ARN Interferente Pequeño/genética , Tálamo/patología
5.
PLoS One ; 8(1): e55575, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23383230

RESUMEN

Prion diseases are rare but invariably fatal neurodegenerative disorders. They are associated with spongiform encephalopathy, a histopathology characterized by the presence of large, membrane-bound vacuolar structures in the neuropil of the brain. While the primary cause is recognized as conversion of the normal form of prion protein (PrP(C)) to a conformationally distinct, pathogenic form (PrP(Sc)), the cellular pathways and mechanisms that lead to spongiform change, neuronal dysfunction and death are not known. Mice lacking the Mahogunin Ring Finger 1 (MGRN1) E3 ubiquitin ligase develop spongiform encephalopathy by 9 months of age but do not become ill. In cell culture, PrP aberrantly present in the cytosol was reported to interact with and sequester MGRN1. This caused endo-lysosomal trafficking defects similar to those observed when Mgrn1 expression is knocked down, implicating disrupted MGRN1-dependent trafficking in the pathogenesis of prion disease. As these defects were rescued by over-expression of MGRN1, we investigated whether reduced or elevated Mgrn1 expression influences the onset, progression or pathology of disease in mice inoculated with PrP(Sc). No differences were observed, indicating that disruption of MGRN1-dependent pathways does not play a significant role in the pathogenesis of transmissible spongiform encephalopathy.


Asunto(s)
Enfermedades por Prión/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Animales , Encéfalo/metabolismo , Encéfalo/patología , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Femenino , Expresión Génica , Humanos , Ratones , Ratones Transgénicos , Enfermedades por Prión/genética , Enfermedades por Prión/mortalidad , Priones/metabolismo , Ubiquitina-Proteína Ligasas/genética
6.
PLoS One ; 7(7): e39112, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22768295

RESUMEN

The lipophilic cationic compound quinacrine has been used as an antimalarial drug for over 75 years but its pharmacokinetic profile is limited. Here, we report on the pharmacokinetic properties of quinacrine in mice. Following an oral dose of 40 mg/kg/day for 30 days, quinacrine concentration in the brain of wild-type mice was maintained at a concentration of ∼1 µM. As a substrate of the P-glycoprotein (P-gp) efflux transporter, quinacrine is actively exported from the brain, preventing its accumulation to levels that may show efficacy in some disease models. In the brains of P-gp-deficient Mdr1(0/0) mice, we found quinacrine reached concentrations of ∼80 µM without any signs of acute toxicity. Additionally, we examined the distribution and metabolism of quinacrine in the wild-type and Mdr1(0/0) brains. In wild-type mice, the co-administration of cyclosporin A, a known P-gp inhibitor, resulted in a 6-fold increase in the accumulation of quinacrine in the brain. Our findings argue that the inhibition of the P-gp efflux transporter should improve the poor pharmacokinetic properties of quinacrine in the CNS.


Asunto(s)
Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/metabolismo , Antineoplásicos/farmacocinética , Encéfalo/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Quinacrina/farmacocinética , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/antagonistas & inhibidores , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/genética , Animales , Ciclosporina/farmacología , Inhibidores Enzimáticos/farmacología , Femenino , Masculino , Ratones , Ratones Mutantes , Proteínas del Tejido Nervioso/antagonistas & inhibidores , Proteínas del Tejido Nervioso/genética
7.
J Neuropathol Exp Neurol ; 71(5): 449-66, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22507918

RESUMEN

Brain aggregates (BrnAggs) derived from fetal mouse brains contain mature neurons and glial cells. We determined that BrnAggs are consistently infected with Rocky Mountain Laboratory scrapie strain prions and produce increasing levels of the pathogenic form of the prion protein (PrP). Their abundant dendrites undergo degeneration shortly after prion infection. Treatment of prion-infected BrnAggs with drugs, such as a γ-secretase inhibitors and quinacrine (Qa), which stop PrP formation and dendritic degeneration, mirrors the results from rodent studies. Because PrP is trafficked into lysosomes by endocytosis and autophagosomes by phagocytosis in neurons of prion strain-infected BrnAggs, we studied the effects of drugs that modulate subcellular trafficking. Rapamycin (Rap), which activates autophagy, markedly increased light-chain 3-II (LC3-II)-positive autophagosomes and cathepsin D-positive lysosomes in BrnAggs but could not eliminate the intracellular PrP within them. Adding Qa to Rap markedly reduced the number of LC3-II-positive autolysosomes. Rap + Qa created a competition between Rap increasing and Qa decreasing LC3-II. Rapamycin + Qa decreased total PrP by 56% compared with that of Qa alone, which reduced PrP by 37% relative to Rap alone. We conclude that the decrease was dominated by the ability of Qa to decrease the formation of PrP. Therefore, BrnAggs provide an efficient in vitro tool for screening drug therapies and studying the complex biology of prions.


Asunto(s)
Encéfalo/metabolismo , Encéfalo/patología , Inhibidores Enzimáticos/uso terapéutico , Proteínas PrPSc/metabolismo , Enfermedades por Prión , Priones/metabolismo , Alanina/análogos & derivados , Alanina/farmacología , Animales , Azepinas/farmacología , Encéfalo/efectos de los fármacos , Encéfalo/embriología , Catepsina D/metabolismo , Dendritas/efectos de los fármacos , Dendritas/metabolismo , Dendritas/patología , Modelos Animales de Enfermedad , Embrión de Mamíferos , Inhibidores Enzimáticos/farmacología , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Inmunosupresores/farmacología , Técnicas In Vitro , Lisosomas/fisiología , Masculino , Ratones , Ratones Noqueados , Microglía/efectos de los fármacos , Microglía/metabolismo , Microglía/patología , Microscopía Confocal , Proteínas Asociadas a Microtúbulos/metabolismo , Degeneración Nerviosa/etiología , Degeneración Nerviosa/patología , Degeneración Nerviosa/prevención & control , Proteínas del Tejido Nervioso/metabolismo , Proteínas PrPSc/antagonistas & inhibidores , Embarazo , Enfermedades por Prión/tratamiento farmacológico , Enfermedades por Prión/metabolismo , Enfermedades por Prión/patología , Proteínas Priónicas , Priones/genética , Transporte de Proteínas/efectos de los fármacos , Quinacrina/farmacología , Sirolimus/farmacología , Fracciones Subcelulares/efectos de los fármacos , Fracciones Subcelulares/metabolismo , Fracciones Subcelulares/patología , Factores de Tiempo
8.
J Biol Chem ; 285(14): 10415-23, 2010 Apr 02.
Artículo en Inglés | MEDLINE | ID: mdl-19955177

RESUMEN

Prion-infected cells accumulate a heterogeneous population of aberrantly folded PrP conformers, including the disease-causing isoform (PrP(Sc)). We found that specific chemicals can modulate the levels of various PrP conformers in cultured cells. Positively charged polyamidoamines (dendrimers) eliminated protease-resistant (r) PrP(Sc) from prion-infected cells and induced the formation of insoluble, protease-sensitive PrP aggregates (designated PrP(A)). Larger, positively charged polyamidoamines more efficaciously induced the formation of PrP(A) and cleared rPrP(Sc), whereas negatively charged polyamidoamines neither induced PrP(A) nor cleared rPrP(Sc). Although the biochemical properties of PrP(A) were shown to be similar to protease-sensitive (s) PrP(Sc), bioassays of PrP(A) indicated that it is not infectious. Our studies argue that PrP(A) represents an aggregated PrP species that is off-pathway relative to the formation of rPrP(Sc). It remains to be established whether the formation of PrP(A) inhibits the formation of rPrP(Sc) by sequestering PrP(C) in the form of benign, insoluble aggregates.


Asunto(s)
Dendrímeros/farmacología , Nylons/farmacología , Proteínas PrPC/química , Proteínas PrPSc/química , Pliegue de Proteína/efectos de los fármacos , Multimerización de Proteína/efectos de los fármacos , Animales , Western Blotting , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Cromatografía en Gel , Dimerización , Endopeptidasas/metabolismo , Ratones , Ratones Transgénicos , Neuroblastoma/tratamiento farmacológico , Neuroblastoma/metabolismo , Neuroblastoma/patología , Proteínas PrPC/metabolismo , Proteínas PrPSc/metabolismo , Conformación Proteica , Isoformas de Proteínas , ARN Mensajero/genética , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células Tumorales Cultivadas
9.
PLoS Pathog ; 5(11): e1000673, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19956709

RESUMEN

Quinacrine is a potent antiprion compound in cell culture models of prion disease but has failed to show efficacy in animal bioassays and human clinical trials. Previous studies demonstrated that quinacrine inefficiently penetrates the blood-brain barrier (BBB), which could contribute to its lack of efficacy in vivo. As quinacrine is known to be a substrate for P-glycoprotein multi-drug resistance (MDR) transporters, we circumvented its poor BBB permeability by utilizing MDR(0/0) mice that are deficient in mdr1a and mdr1b genes. Mice treated with 40 mg/kg/day of quinacrine accumulated up to 100 microM of quinacrine in their brains without acute toxicity. PrP(Sc) levels in the brains of prion-inoculated MDR(0/0) mice diminished upon the initiation of quinacrine treatment. However, this reduction was transient and PrP(Sc) levels recovered despite the continuous administration of quinacrine. Treatment with quinacrine did not prolong the survival times of prion-inoculated, wild-type or MDR(0/0) mice compared to untreated mice. A similar phenomenon was observed in cultured differentiated prion-infected neuroblastoma cells: PrP(Sc) levels initially decreased after quinacrine treatment then rapidly recovered after 3 d of continuous treatment. Biochemical characterization of PrP(Sc) that persisted in the brains of quinacrine-treated mice had a lower conformational stability and different immunoaffinities compared to that found in the brains of untreated controls. These physical properties were not maintained upon passage in MDR(0/0) mice. From these data, we propose that quinacrine eliminates a specific subset of PrP(Sc) conformers, resulting in the survival of drug-resistant prion conformations. Transient accumulation of this drug-resistant prion population provides a possible explanation for the lack of in vivo efficacy of quinacrine and other antiprion drugs.


Asunto(s)
Resistencia a Medicamentos , Enfermedades por Prión/tratamiento farmacológico , Priones/efectos de los fármacos , Subfamilia B de Transportador de Casetes de Unión a ATP/deficiencia , Animales , Química Encefálica , Células Cultivadas , Ratones , Ratones Noqueados , Neuroblastoma/patología , Proteínas PrPSc/análisis , Proteínas PrPSc/química , Priones/química , Conformación Proteica , Quinacrina/administración & dosificación , Quinacrina/farmacología , Quinacrina/uso terapéutico , Tasa de Supervivencia , Miembro 4 de la Subfamilia B de Casete de Unión a ATP
10.
J Neurosci ; 27(43): 11533-42, 2007 Oct 24.
Artículo en Inglés | MEDLINE | ID: mdl-17959796

RESUMEN

Voltage-gated sodium channels are responsible for action potential initiation and propagation in neurons, and modulation of their function has an important impact on neuronal excitability. Sodium channels are regulated by a Src-family tyrosine kinase pathway, and this modulation can be reversed by specifically bound receptor phosphoprotein tyrosine phosphatase-beta. However, the specific tyrosine kinase and signaling pathway are unknown. We found that the sodium channels in rat brain interact with Fyn, one of four Src-family tyrosine kinases expressed in the brain. Na(V)1.2 channels and Fyn are localized together in the axons of cultured hippocampal neurons, the mossy fibers of the hippocampus, and cell bodies, dendrites, and axons of neurons in many other brain areas, and they coimmunoprecipitate with Fyn from cotransfected tsA-201 cells. Coexpression of Fyn with Na(V)1.2 channels decreases sodium currents by increasing the rate of inactivation and causing a negative shift in the voltage dependence of inactivation. Reconstitution of a signaling pathway from brain-derived neurotrophic factor (BDNF) to sodium channels via the tyrosine receptor kinase B (TrkB)/p75 neurotrophin receptor and Fyn kinase in transfected cells resulted in an increased rate of inactivation of sodium channels and a negative shift in the voltage dependence of inactivation after treatment with BDNF. These results indicate that Fyn kinase is associated with sodium channels in brain neurons and can modulate Na(V)1.2 channels by tyrosine phosphorylation after activation of TrkB/p75 signaling by BDNF.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo/fisiología , Proteínas del Tejido Nervioso/metabolismo , Proteínas Proto-Oncogénicas c-fyn/fisiología , Receptor trkB/fisiología , Canales de Sodio/metabolismo , Animales , Células Cultivadas , Hipocampo/metabolismo , Hipocampo/fisiología , Canal de Sodio Activado por Voltaje NAV1.2 , Proteínas del Tejido Nervioso/antagonistas & inhibidores , Técnicas de Placa-Clamp , Fosforilación , Proteínas Proto-Oncogénicas c-fyn/genética , Proteínas Proto-Oncogénicas c-fyn/metabolismo , Ratas , Receptor trkB/genética , Transducción de Señal/genética , Transducción de Señal/fisiología , Tirosina/metabolismo
11.
J Neurosci ; 27(43): 11543-51, 2007 Oct 24.
Artículo en Inglés | MEDLINE | ID: mdl-17959797

RESUMEN

Voltage-gated sodium channels are important targets for modulation of electrical excitability by neurotransmitters and neurotrophins acting through protein phosphorylation. Fast inactivation of Na(V)1.2 channels is regulated via tyrosine phosphorylation by Fyn kinase and dephosphorylation by receptor phosphoprotein tyrosine phosphatase-beta, which are associated in a signaling complex. Here we have identified the amino acid residues on Na(V)1.2 channels that coordinate binding of Fyn kinase and mediate inhibition of sodium currents by enhancing fast inactivation. Fyn kinase binds to a Src homology 3 (SH3)-binding motif in the second half of the intracellular loop connecting domains I and II (L(I-II)) of Na(V)1.2, and mutation of that SH3-binding motif prevents Fyn binding and Fyn enhancement of fast inactivation of sodium currents. Analysis of tyrosine phosphorylation sites by mutagenesis and functional expression revealed a multisite regulatory mechanism. Y66 and Y1893, which are in consensus sequences appropriate for binding to the Fyn SH2 domain after phosphorylation, are both required for optimal binding and regulation by Fyn. Y730, which is located near the SH3-binding motif in L(I-II), and Y1497 and Y1498 in the inactivation gate in L(III-IV), are also required for optimal regulation. Phosphorylation of these sites likely promotes fast inactivation. Fast inactivation of the closely related Na(V)1.1 channels is not modulated by Fyn, and these channels do not contain an SH3-binding motif in L(I-II). Subtype-selective modulation by tyrosine phosphorylation/dephosphorylation provides a mechanism for differential regulation of sodium channels by neurotrophins and tyrosine phosphorylation in unmyelinated axons and dendrites, where Na(V)1.2 channels are expressed in brain neurons.


Asunto(s)
Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Proteínas Proto-Oncogénicas c-fyn/genética , Proteínas Proto-Oncogénicas c-fyn/metabolismo , Canales de Sodio/genética , Canales de Sodio/metabolismo , Animales , Sitios de Unión/genética , Sitios de Unión/fisiología , Mutagénesis Sitio-Dirigida , Canal de Sodio Activado por Voltaje NAV1.2 , Proteínas del Tejido Nervioso/biosíntesis , Ratas , Canales de Sodio/biosíntesis
12.
J Am Chem Soc ; 124(44): 12991-8, 2002 Nov 06.
Artículo en Inglés | MEDLINE | ID: mdl-12405825

RESUMEN

The structure-based design of multivalent ligands offers an attractive strategy toward high affinity protein inhibitors. The spatial arrangement of the receptor-binding sites of cholera toxin, the causative agent of the severe diarrheal disease cholera and a member of the AB(5) bacterial toxin family, provides the opportunity of designing branched multivalent ligands with 5-fold symmetry. Our modular synthesis enabled the construction of a family of complex ligands with five flexible arms each ending with a bivalent ligand. The largest of these ligands has a molecular weight of 10.6 kDa. These ligands are capable of simultaneously binding to two toxin B pentamer molecules with high affinity, thus blocking the receptor-binding process of cholera toxin. A more than million-fold improvement over the monovalent ligand in inhibitory power was achieved with the best branched decavalent ligand. This is better than the improvement observed earlier for the corresponding nonbranched pentavalent ligand. Dynamic light scattering studies demonstrate the formation of concentration-dependent unique 1:1 and 1:2 ligand/toxin complexes in solution with no sign of nonspecific aggregation. This is in complete agreement with a crystal structure of the branched multivalent ligand/toxin B pentamer complex solved at 1.45 A resolution that shows the specific 1:2 ligand/toxin complex formation in the solid state. These results reiterate the power of the structure-based design of multivalent protein ligands as a general strategy for achieving high affinity and potent inhibition.


Asunto(s)
Toxina del Cólera/antagonistas & inhibidores , Gangliósido G(M1)/antagonistas & inhibidores , Galactosa/análogos & derivados , Receptores de Superficie Celular/antagonistas & inhibidores , Sitios de Unión , Toxina del Cólera/metabolismo , Cristalografía por Rayos X , Gangliósido G(M1)/metabolismo , Galactosa/química , Galactosa/farmacología , Ligandos , Luz , Modelos Moleculares , Conformación Proteica , Receptores de Superficie Celular/metabolismo , Dispersión de Radiación , Soluciones
13.
J Am Chem Soc ; 124(30): 8818-24, 2002 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-12137534

RESUMEN

Multivalent ligand design constitutes an attractive avenue to the inhibition of receptor recognition and other biological events mediated by oligomeric proteins with multiple binding sites. One example is the design of multivalent receptor blockers targeting members of the AB(5) bacterial toxin family. We report here the synthesis and characterization of a pentavalent inhibitor for cholera toxin and Escherichia coli heat-labile enterotoxin. This inhibitor is an advance over the symmetric pentacyclen-derived inhibitor described in our earlier work in that it presents five copies of m-nitrophenyl-alpha-D-galactoside (MNPG) rather than five copies of beta-D-galactose. The approximately 100-fold higher single-site affinity of MNPG for the toxin receptor binding site relative to galactose is found to yield a proportionate increase in the affinity and IC50 measured for the respective pentavalent constructs. We show by dynamic light scattering that inhibition of receptor binding by the pentavalent inhibitor is due to 1:1 inhibitor:toxin association rather than to inhibitor-mediated aggregation. This 1:1 association is in complete agreement with a 1.46 A resolution crystal structure of the pentavalent inhibitor:toxin complex, which shows that the favorable single-site binding interactions of MNPG are retained by the five arms of the 5256 Da pentavalent MNPG-based inhibitor and that the initial segment of the linking groups interacts with the surface of the toxin B pentamer.


Asunto(s)
Toxinas Bacterianas/antagonistas & inhibidores , Toxina del Cólera/antagonistas & inhibidores , Enterotoxinas/antagonistas & inhibidores , Proteínas de Escherichia coli , Gangliósido G(M1)/antagonistas & inhibidores , Nitrofenilgalactósidos/química , Receptores de Superficie Celular/antagonistas & inhibidores , Receptores Inmunológicos/antagonistas & inhibidores , Toxinas Bacterianas/química , Toxinas Bacterianas/metabolismo , Toxina del Cólera/química , Toxina del Cólera/metabolismo , Cristalografía por Rayos X , Enterotoxinas/química , Enterotoxinas/metabolismo , Escherichia coli/metabolismo , Gangliósido G(M1)/química , Gangliósido G(M1)/metabolismo , Cinética , Ligandos , Luz , Modelos Moleculares , Nitrofenilgalactósidos/síntesis química , Nitrofenilgalactósidos/metabolismo , Receptores de Superficie Celular/química , Receptores de Superficie Celular/metabolismo , Receptores Inmunológicos/química , Receptores Inmunológicos/metabolismo , Dispersión de Radiación
14.
Chem Biol ; 9(2): 215-24, 2002 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-11880036

RESUMEN

The action of cholera toxin and E. coli heat-labile enterotoxin can be inhibited by blocking their binding to the cell-surface receptor GM1. We have used anchor-based design to create 15 receptor binding inhibitors that contain the previously characterized inhibitor MNPG as a substructure. In ELISA assays, all 15 compounds exhibited increased potency relative to MNPG. Binding affinities for two compounds, each containing a morpholine ring linked to MNPG via a hydrophobic tail, were characterized by pulsed ultrafiltration (PUF) and isothermal titration calorimetry (ITC). Crystal structures for these compounds bound to toxin B pentamer revealed a conserved binding mode for the MNPG moiety, with multiple binding modes adopted by the attached morpholine derivatives. The observed binding interactions can be exploited in the design of improved toxin binding inhibitors.


Asunto(s)
Toxinas Bacterianas/química , Toxina del Cólera/química , Enterotoxinas/química , Proteínas de Escherichia coli , Galactosa/química , Receptores de Superficie Celular/antagonistas & inhibidores , Receptores de Superficie Celular/química , Toxinas Bacterianas/metabolismo , Unión Competitiva , Toxina del Cólera/metabolismo , Cristalografía , Diseño de Fármacos , Enterotoxinas/metabolismo , Ensayo de Inmunoadsorción Enzimática , Escherichia coli/metabolismo , Galactosa/análogos & derivados , Galactosa/metabolismo , Modelos Moleculares , Conformación Proteica , Receptores de Superficie Celular/metabolismo , Relación Estructura-Actividad
15.
J Biol Chem ; 277(6): 4079-87, 2002 Feb 08.
Artículo en Inglés | MEDLINE | ID: mdl-11733497

RESUMEN

In skeletal muscle, voltage-dependent potentiation of L-type Ca(2+) channel (Ca(V)1.1) activity requires phosphorylation by cyclic AMP-dependent protein kinase (PKA) anchored via an A kinase-anchoring protein (AKAP15). However, the mechanism by which AKAP15 targets PKA to L-type Ca(2+) channels has not been elucidated. Here we report that AKAP15 directly interacts with the C-terminal domain of the alpha(1) subunit of Ca(V)1.1 via a leucine zipper (LZ) motif. Disruption of the LZ interaction effectively inhibits voltage-dependent potentiation of L-type Ca(2+) channels in skeletal muscle cells. Our results reveal a novel mechanism whereby anchoring of PKA to Ca(2+) channels via LZ interactions ensures rapid and efficient phosphorylation of Ca(2+) channels in response to local signals such as cAMP and depolarization.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales , Canales de Calcio Tipo L/metabolismo , Canales de Calcio Tipo L/fisiología , Proteínas Portadoras/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Leucina Zippers , Proteínas de la Membrana/metabolismo , Músculo Esquelético/metabolismo , Proteínas de Anclaje a la Quinasa A , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Animales , Sitios de Unión , Canales de Calcio Tipo L/química , Células Cultivadas , Ratones , Ratones Endogámicos BALB C , Datos de Secuencia Molecular , Unión Proteica , Homología de Secuencia de Aminoácido
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...