Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Bioorg Med Chem Lett ; 71: 128807, 2022 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-35605837

RESUMEN

SST5 receptor activation potently inhibits insulin secretion from pancreatic ß-cells, and an orally available nonpeptide selective SST5 agonist may be used to effectively manage the blood glucose levels of congenital HI patients to avoid severe hypoglycemia. Our medicinal chemistry efforts have led to the discovery of 4-(3-aminopyrrolidinyl)-3-aryl-5-(benzimidazol-2-yl)-pyridine analogs as potent SST5 agonists. This class of molecules exhibits excellent human SST5 potency and selectivity against SST1, SST2, SST3 and SST4 receptors. Leading compound 3-{4-[(3S)-3-aminopyrrolidin-1-yl]-5-(4-methyl-1H-1,3-benzodiazol-2-yl)pyridin-3-yl-5-fluorobenzonitrile (28, CRN02481) showed limited off-target activity and good pharmacokinetic profiles in both male Sprague Dawley rats and Beagle dogs to advance into further preclinical evaluations.


Asunto(s)
Hiperinsulinismo Congénito , Somatostatina , Animales , Hiperinsulinismo Congénito/tratamiento farmacológico , Perros , Humanos , Masculino , Piridinas/farmacología , Ratas , Ratas Sprague-Dawley , Receptores de Somatostatina/agonistas , Somatostatina/farmacología , Somatostatina/fisiología
2.
J Am Chem Soc ; 141(36): 14210-14219, 2019 09 11.
Artículo en Inglés | MEDLINE | ID: mdl-31418572

RESUMEN

Peptide agonists of GPCRs and other receptors are powerful signaling molecules with high potential as biological tools and therapeutics, but they are typically plagued by instability and short half-lives in vivo. Nature uses protein glycosylation to increase the serum stability of secreted proteins. However, these extracellular modifications are complex and heterogeneous in structure, making them an impractical solution. In contrast, intracellular proteins are subjected to a simple version of glycosylation termed O-GlcNAc modification. In our studies of this modification, we found that O-GlcNAcylation inhibits proteolysis, and strikingly, this stabilization occurs despite large distances in primary sequence (10-15 amino acids) between the O-GlcNAc and the site of cleavage. We therefore hypothesized that this "remote stabilization" concept could be useful to engineer the stability and potentially additional properties of peptide or protein therapeutics. Here, we describe the application of O-GlcNAcylation to two clinically important peptides: glucagon-like peptide-1 (GLP-1) and the parathyroid hormone (PTH), which respectively help control glucose and calcium levels in the blood. For both peptides, we found O-GlcNAcylated analogs that are equipotent to unmodified peptide in cell-based activation assays, while several GLP-1 analogs were biased agonists relative to GLP-1. As we predicted, O-GlcNAcylation can improve the stability of both GLP-1 and PTH in serum despite the fact that the O-GlcNAc can be quite remote from characterized sites of peptide cleavage. The O-GlcNAcylated GLP-1 and PTH also displayed significantly improved in vivo activity. Finally, we employed structure-based molecular modeling and receptor mutagenesis to predict how O-GlcNAcylation can be accommodated by the receptors and the potential interactions that contribute to peptide activity. This approach demonstrates the potential of O-GlcNAcylation for generating analogs of therapeutic peptides with enhanced proteolytic stability.


Asunto(s)
Péptido 1 Similar al Glucagón/farmacología , Hormona Paratiroidea/farmacología , Ingeniería de Proteínas , Receptores Acoplados a Proteínas G/agonistas , Péptido 1 Similar al Glucagón/sangre , Péptido 1 Similar al Glucagón/química , Glicosilación , Humanos , Hormona Paratiroidea/sangre , Hormona Paratiroidea/química , Conformación Proteica , Receptores Acoplados a Proteínas G/metabolismo
3.
Proc Natl Acad Sci U S A ; 116(17): 8370-8379, 2019 04 23.
Artículo en Inglés | MEDLINE | ID: mdl-30971491

RESUMEN

Metabotropic GABAB receptors mediate a significant fraction of inhibitory neurotransmission in the brain. Native GABAB receptor complexes contain the principal subunits GABAB1 and GABAB2, which form an obligate heterodimer, and auxiliary subunits, known as potassium channel tetramerization domain-containing proteins (KCTDs). KCTDs interact with GABAB receptors and modify the kinetics of GABAB receptor signaling. Little is known about the molecular mechanism governing the direct association and functional coupling of GABAB receptors with these auxiliary proteins. Here, we describe the high-resolution structure of the KCTD16 oligomerization domain in complex with part of the GABAB2 receptor. A single GABAB2 C-terminal peptide is bound to the interior of an open pentamer formed by the oligomerization domain of five KCTD16 subunits. Mutation of specific amino acids identified in the structure of the GABAB2-KCTD16 interface disrupted both the biochemical association and functional modulation of GABAB receptors and G protein-activated inwardly rectifying K+ channel (GIRK) channels. These interfacial residues are conserved among KCTDs, suggesting a common mode of KCTD interaction with GABAB receptors. Defining the binding interface of GABAB receptor and KCTD reveals a potential regulatory site for modulating GABAB-receptor function in the brain.


Asunto(s)
Péptidos y Proteínas de Señalización Intracelular , Proteínas del Tejido Nervioso , Receptores de GABA-B , Sitios de Unión/genética , Cristalografía , Humanos , Péptidos y Proteínas de Señalización Intracelular/química , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Modelos Moleculares , Proteínas del Tejido Nervioso/química , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Unión Proteica/genética , Receptores de GABA-B/química , Receptores de GABA-B/genética , Receptores de GABA-B/metabolismo , Transducción de Señal/genética
4.
Assay Drug Dev Technol ; 16(5): 278-288, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-30019946

RESUMEN

GPR119 drug discovery efforts in the pharmaceutical industry for the treatment of type 2 diabetes mellitus (T2DM) and obesity, were initiated based on its restricted distribution in pancreas and GI tract, and its possible role in glucose homeostasis. While a number of lead series have emerged, the pharmacological endpoints they provide have not been clear. In particular, many lead series have demonstrated loss of efficacy and significant toxic side effects. Thus, we sought to identify novel, potent, positive modulators of GPR119. In this study, we have successfully developed and optimized a high-throughput screening strategy to identify GPR119 modulators using a live cell assay format that utilizes a cyclic nucleotide-gated channel as a biosensor for cAMP production. Our high-throughput screening (HTS) approach is unique to that of previous HTS approaches targeting this receptor, as changes in cAMP were measured both in the presence and absence of an EC10 of the endogenous ligand, oleoylethanolamide, enabling detection of both agonists and potential allosteric modulators in a single assay. From these efforts, we have identified positive modulators of GPR119 with similar as well as unique scaffolds compared to existing compounds and similar as well as unique signaling properties. Our compounds will not only serve as novel molecular probes to better understand GPR119 pleiotropic signaling and the underlying physiological consequences of receptor activation, but are also well-suited for translation as potential therapeutic agents.


Asunto(s)
Endocannabinoides/farmacología , Hipoglucemiantes/farmacología , Ácidos Oléicos/farmacología , Receptores Acoplados a Proteínas G/agonistas , Bibliotecas de Moléculas Pequeñas/química , Bibliotecas de Moléculas Pequeñas/farmacología , Células Cultivadas , Endocannabinoides/química , Células HEK293 , Ensayos Analíticos de Alto Rendimiento , Humanos , Hipoglucemiantes/química , Estructura Molecular , Ácidos Oléicos/química , Receptores Acoplados a Proteínas G/metabolismo
5.
Psychopharmacology (Berl) ; 234(9-10): 1633-1644, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28382542

RESUMEN

RATIONALE: GABAB receptors (GABABR) play a critical role in GABAergic neurotransmission in the brain and are thought to be one of the most promising targets for the treatment of drug addiction. GABABR positive allosteric modulators (PAMs) have shown promise as potential anti-addictive therapies, as they lack the sedative and muscle relaxant properties of full GABAB receptor agonists such as baclofen. OBJECTIVES: The present study was aimed at developing novel, selective, and potent GABABR PAMs with efficacy on abuse-related effects of nicotine. RESULTS: We synthetized ~100 analogs of BHF177, a GABABR PAM that has been shown to inhibit nicotine taking and seeking, and tested their activity in multiple cell-based functional assays. Among these compounds, KK-92A displayed superior PAM properties at the GABABR. Interestingly, our results revealed the existence of pathway-selective differential modulation of GABABR signaling by the structurally related GABABR allosteric modulators BHF177 and KK-92A. In vivo, similarly to BHF177, KK-92A inhibited intravenous nicotine self-administration under both fixed- and progressive-ratio schedules of reinforcement in rats. In contrast to BHF177, KK-92A had no effect on food self-administration. Furthermore, KK-92A decreased cue-induced nicotine-seeking behavior without affecting food seeking. CONCLUSIONS: These results indicate that KK-92A is a selective GABABR PAM with efficacy in inhibition of the primary reinforcing and incentive motivational effects of nicotine, and attenuation of nicotine seeking, further confirming that GABABR PAMs may be useful antismoking medications.


Asunto(s)
Conducta Adictiva/prevención & control , Señales (Psicología) , Moduladores del GABA/uso terapéutico , Nicotina/administración & dosificación , Receptores de GABA-B/fisiología , Regulación Alostérica/efectos de los fármacos , Regulación Alostérica/fisiología , Animales , Conducta Adictiva/psicología , Células CHO , Cricetinae , Cricetulus , Relación Dosis-Respuesta a Droga , Moduladores del GABA/farmacología , Agonistas de Receptores GABA-B/farmacología , Agonistas de Receptores GABA-B/uso terapéutico , Humanos , Masculino , Motivación/efectos de los fármacos , Motivación/fisiología , Nicotina/antagonistas & inhibidores , Ratas , Ratas Wistar , Refuerzo en Psicología , Autoadministración/métodos
6.
Pharmacol Res Perspect ; 5(2): e00288, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28357120

RESUMEN

Positive modulation of the GABAB receptor (GABABR) represents a potentially useful therapeutic approach for the treatment of nicotine addiction. The positive allosteric modulators (PAMs) of GABABR GS39783 and BHF177 enhance GABA-stimulated [35S]GTP γS-binding, and have shown efficacy in a rodent nicotine self-administration procedure reflecting aspects of nicotine dependence. Interestingly, the structural related analog, NVP998, had no effect on nicotine self-administration in rats despite demonstrating similar pharmacokinetic properties. Extensive in vitro characterization of GS39783, BHF177, and NVP998 activity on GABABR-regulated signaling events, including modulation of cAMP, intracellular calcium levels, and ERK activation, revealed that these structurally related molecules display distinct pathway-specific signaling activities that correlate with the dissimilarities observed in rodent models and may be predictive of in vivo efficacy. Furthermore, these GABABR allosteric modulators exhibit species-dependent activity. Collectively, these data will be useful in guiding the development of GABABR allosteric modulators that display optimal in vivo efficacy in a preclinical model of nicotine dependence, and will identify those that have the potential to lead to novel antismoking therapies.

7.
Elife ; 52016 07 19.
Artículo en Inglés | MEDLINE | ID: mdl-27434672

RESUMEN

Human calcium-sensing receptor (CaSR) is a G-protein-coupled receptor (GPCR) that maintains extracellular Ca(2+) homeostasis through the regulation of parathyroid hormone secretion. It functions as a disulfide-tethered homodimer composed of three main domains, the Venus Flytrap module, cysteine-rich domain, and seven-helix transmembrane region. Here, we present the crystal structures of the entire extracellular domain of CaSR in the resting and active conformations. We provide direct evidence that L-amino acids are agonists of the receptor. In the active structure, L-Trp occupies the orthosteric agonist-binding site at the interdomain cleft and is primarily responsible for inducing extracellular domain closure to initiate receptor activation. Our structures reveal multiple binding sites for Ca(2+) and PO4(3-) ions. Both ions are crucial for structural integrity of the receptor. While Ca(2+) ions stabilize the active state, PO4(3-) ions reinforce the inactive conformation. The activation mechanism of CaSR involves the formation of a novel dimer interface between subunits.


Asunto(s)
Calcio/metabolismo , Receptores Sensibles al Calcio/agonistas , Receptores Sensibles al Calcio/química , Triptófano/química , Triptófano/metabolismo , Sitios de Unión , Cristalografía por Rayos X , Humanos , Modelos Moleculares , Fosfatos/metabolismo , Unión Proteica , Conformación Proteica , Multimerización de Proteína
8.
Pharmacol Res Perspect ; 4(2): e00226, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-27069636

RESUMEN

The known angiotensin II (AngII) physiological effect of aldosterone synthesis and secretion induction, a steroid hormone that contributes to the pathology of postmyocardial infarction (MI) heart failure (HF), is mediated by both Gq/11 proteins and ß-arrestins, both of which couple to the AngII type 1 receptors (AT1Rs) of adrenocortical zona glomerulosa (AZG) cells. Over the past several years, AngII analogs with increased selectivity ("bias") toward ß-arrestin-dependent signaling at the AT1R have been designed and described, starting with SII, the gold-standard ß-arrestin-"biased" AngII analog. In this study, we examined the relative potencies of an extensive series of AngII peptide analogs at relative activation of G proteins versus ß-arrestins by the AT1R. The major structural difference of these peptides from SII was their varied substitutions at position 5, rather than position 4 of native AngII. Three of them were found biased for ß-arrestin activation and extremely potent at stimulating aldosterone secretion in AZG cells in vitro, much more potent than SII in that regard. Finally, the most potent of these three ([Sar(1), Cys(Et)(5), Leu(8)]-AngII, CORET) was further examined in post-MI rats progressing to HF and overexpressing adrenal ß-arrestin1 in vivo. Consistent with the in vitro studies, CORET was found to exacerbate the post-MI hyperaldosteronism, and, consequently, cardiac function of the post-MI animals in vivo. Finally, our data suggest that increasing the size of position 5 of the AngII peptide sequence results in directly proportional increases in AT1R-dependent ß-arrestin activation. These findings provide important insights for AT1R pharmacology and future AngII-targeted drug development.

9.
Nat Commun ; 6: 8918, 2015 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-26621478

RESUMEN

Glucagon-like peptide-1 (GLP-1) receptor (GLP-1R) agonists have emerged as treatment options for type 2 diabetes mellitus (T2DM). GLP-1R signals through G-protein-dependent, and G-protein-independent pathways by engaging the scaffold protein ß-arrestin; preferential signalling of ligands through one or the other of these branches is known as 'ligand bias'. Here we report the discovery of the potent and selective GLP-1R G-protein-biased agonist, P5. We identified P5 in a high-throughput autocrine-based screening of large combinatorial peptide libraries, and show that P5 promotes G-protein signalling comparable to GLP-1 and Exendin-4, but exhibited a significantly reduced ß-arrestin response. Preclinical studies using different mouse models of T2DM demonstrate that P5 is a weak insulin secretagogue. Nevertheless, chronic treatment of diabetic mice with P5 increased adipogenesis, reduced adipose tissue inflammation as well as hepatic steatosis and was more effective at correcting hyperglycaemia and lowering haemoglobin A1c levels than Exendin-4, suggesting that GLP-1R G-protein-biased agonists may provide a novel therapeutic approach to T2DM.


Asunto(s)
Diabetes Mellitus Tipo 2/tratamiento farmacológico , Receptor del Péptido 1 Similar al Glucagón/antagonistas & inhibidores , Hipoglucemiantes/administración & dosificación , Péptidos/administración & dosificación , Adipogénesis/efectos de los fármacos , Tejido Adiposo/efectos de los fármacos , Tejido Adiposo/metabolismo , Animales , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/fisiopatología , Evaluación Preclínica de Medicamentos , Receptor del Péptido 1 Similar al Glucagón/genética , Receptor del Péptido 1 Similar al Glucagón/metabolismo , Humanos , Insulina/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL
10.
Sci Rep ; 5: 8116, 2015 Jan 29.
Artículo en Inglés | MEDLINE | ID: mdl-25631300

RESUMEN

The known angiotensin II (AngII) physiological effect of aldosterone synthesis and secretion is mediated by either Gq/11 proteins or ßarrestin1 (ßarr1), both of which can couple to its type 1 receptors (AT1Rs), present in adrenocortical zona glomerulosa (AZG) cell membranes. In the present study, we examined the relative potencies of all the currently used in the clinic AT1R antagonist drugs (angiotensin receptor blockers, ARBs, or sartans) at preventing activation of these two signaling mediators (G proteins and ßarrs) at the AngII-bound AT1R and, consequently, at suppression of aldosterone in vitro. All ARBs were found to be potent inhibitors of G protein activation at the AT1R. However, candesartan and valsartan were the most potent at blocking AngII-induced ßarr activation at this receptor, among the tetrazolo-biphenyl-methyl derivatives, translating into excellent efficacies at aldosterone suppression in H295R cells. Conversely, irbesartan and losartan were largely G protein-selective inhibitors at the AT1R, with very low potency towards ßarr inhibition. As a result, they were very weak suppressors of ßarr1-dependent aldosterone production in H295R cells. These findings provide important pharmacological insights into the drug class of ARBs and medicinal chemistry insights for future drug development in the field of AngII antagonism.


Asunto(s)
Glándulas Suprarrenales/metabolismo , Aldosterona/biosíntesis , Antagonistas de Receptores de Angiotensina/farmacología , Arrestinas/metabolismo , Angiotensina II/farmacología , Antagonistas de Receptores de Angiotensina/química , Animales , Bencimidazoles/farmacología , Compuestos de Bifenilo , Células CHO , Cricetinae , Cricetulus , Proteínas de Unión al GTP/metabolismo , Humanos , Fosfoproteínas/metabolismo , Ratas , Receptor de Angiotensina Tipo 1/metabolismo , Tetrazoles/farmacología , Regulación hacia Arriba/efectos de los fármacos , Valina/análogos & derivados , Valina/farmacología , Valsartán , Zona Glomerular , beta-Arrestinas
12.
Assay Drug Dev Technol ; 12(4): 229-37, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24831789

RESUMEN

Genetic target validation studies have demonstrated that the apoptosis signal-regulating kinase 1 (ASK1) represents an important target for the treatment of rheumatoid arthritis, cardiac diseases, and several neurodegenerative disorders. To identify small-molecule inhibitors of ASK1, we have developed a high-throughput screening-compatible, homogenous, biochemical assay using AlphaScreen technology. This novel assay design utilizes purified stress-activated ASK1 signalosome complex, and it monitors phosphorylation of its full-length native substrate, MKK6. The assay has been optimized in a 384-well format and validated by screening the Sigma LOPAC library. The results presented here demonstrate that the assay is sensitive and robust with a Z' factor value of 0.88±0.04 and a signal-to-background ratio of 11, indicating that this assay can be used to screen large chemical libraries to discover novel inhibitors of ASK1.


Asunto(s)
Ensayos Analíticos de Alto Rendimiento/métodos , MAP Quinasa Quinasa Quinasa 5/antagonistas & inhibidores , Biotinilación , Inhibidores Enzimáticos , Células HEK293 , Humanos , MAP Quinasa Quinasa 6/metabolismo , MAP Quinasa Quinasa Quinasa 5/biosíntesis , Transducción de Señal/efectos de los fármacos , Bibliotecas de Moléculas Pequeñas , Estaurosporina , Estreptavidina/metabolismo
13.
Assay Drug Dev Technol ; 11(8): 468-77, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24116939

RESUMEN

The galanin 3 receptor (GalR3) belongs to the large G protein-coupled receptor (GPCR) family of proteins. GalR3 and two other closely related receptors, GalR1 and GalR2, together with their endogenous ligand galanin, are involved in a variety of physiological and pathophysiological processes. GalR3 in particular has been strongly implicated in addiction and mood-related disorders such as anxiety and depression. It has been the target of many drug discovery programs within the pharmaceutical industry, but despite the significant resources and effort devoted to discovery of galanin receptor subtype selective small molecule modulators, there have been very few reports for the discovery of such molecules. GalR3 has proven difficult to enable in cell-based functional assays due to its apparent poor cell surface expression in recombinant systems. Here, we describe the generation of a modified GalR3 that facilitates its cell surface expression while maintaining wild-type receptor pharmacology. The modified GalR3 has been used to develop a high-throughput screening-compatible, cell-based, cAMP biosensor assay to detect selective small molecule modulators of GalR3. The performance of the assay has been validated by challenging it against a test library of small molecules with known pharmacological activities (LOPAC; Sigma Aldrich). This approach will enable identification of GalR3 selective modulators (chemical probes) that will facilitate dissection of the biological role(s) that GalR3 plays in normal physiological processes as well as in disease states.


Asunto(s)
Receptor de Galanina Tipo 3/efectos de los fármacos , Técnicas Biosensibles , Membrana Celular/metabolismo , Clonación Molecular , AMP Cíclico/metabolismo , ADN Complementario/biosíntesis , ADN Complementario/genética , Técnica del Anticuerpo Fluorescente Indirecta , Galanina/metabolismo , Células HEK293 , Ensayos Analíticos de Alto Rendimiento , Humanos , Ligandos , Receptor de Galanina Tipo 3/genética , Bibliotecas de Moléculas Pequeñas
14.
Biochim Biophys Acta ; 1833(3): 583-92, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23123190

RESUMEN

The apoptosis signal-regulating kinase 1 (ASK1) is activated in response to a wide variety of extracellular stressors. Consequently, dysregulation of ASK1 is associated with multiple pathologies. Here, we show that ASK1 translocates from the cytoplasm to the nucleus in HEK293 cells and human cardiomyocytes in response to hydrogen peroxide (H(2)O(2)) or angiotensin respectively. Immunoprecipitation and mass spectrometry experiments reveal that ASK1 physically interacts with the karyopherin α2/ß1 heterodimer in response to stress and genetic knockdown experiments confirm that this association mediates H(2)O(2)-induced ASK1 nuclear translocation. In addition, we have identified a nuclear localization signal (NLS)-like motif within the primary amino acid sequence of ASK1 composed of two clusters of basic amino acids separated by an intervening 16 amino acid spacer, KR[ACANDLLVDEFLKVSS]KKKK. Mutation of the downstream lysine cluster markedly reduces the H(2)O(2)-induced ASK1-karyopherin α2/ß1 interaction and inhibits ASK1 nuclear translocation. Furthermore, we demonstrate that nuclear ASK1 is active and participates in H(2)O(2)-induced ASK1-mediated cell death. Collectively, our findings have identified a functional interaction between ASK1 and the karyopherin α2/ß1 heterodimer and have also revealed a novel mechanism by which nuclear trafficking regulates the apoptotic function of ASK1 in response to stress.


Asunto(s)
Apoptosis/efectos de los fármacos , Peróxido de Hidrógeno/farmacología , MAP Quinasa Quinasa Quinasa 5/metabolismo , Miocitos Cardíacos/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , alfa Carioferinas/metabolismo , beta Carioferinas/metabolismo , Secuencia de Aminoácidos , Angiotensinas/farmacología , Western Blotting , Núcleo Celular/metabolismo , Proliferación Celular , Células Cultivadas , Citoplasma/metabolismo , Técnica del Anticuerpo Fluorescente , Humanos , Inmunoprecipitación , Riñón/citología , Riñón/efectos de los fármacos , Riñón/metabolismo , MAP Quinasa Quinasa Quinasa 5/genética , Datos de Secuencia Molecular , Mutación/genética , Miocitos Cardíacos/citología , Miocitos Cardíacos/metabolismo , Señales de Localización Nuclear/efectos de los fármacos , Oxidantes/farmacología , Unión Proteica , Multimerización de Proteína , Homología de Secuencia de Aminoácido , Transducción de Señal/efectos de los fármacos , alfa Carioferinas/genética , beta Carioferinas/genética
15.
Cardiovasc Psychiatry Neurol ; 2010: 539581, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20672051

RESUMEN

Increasing evidence suggests that the small EF-hand calcium-binding protein S100B plays an important role in Alzheimer's disease. Among other evidences are the increased levels of both S100B and its receptor, the Receptor for Advanced Glycation Endproducts (RAGEs) in the AD diseased brain. The regulation of RAGE signaling by S100B is complex and probably involves other ligands including the amyloid beta peptide (Abeta), the Advanced Glycation Endproducts (AGEs), or transtheyretin. In this paper we discuss the current literature regarding the role of S100B/RAGE activation in Alzheimer's disease.

16.
Biochemistry ; 49(19): 4094-102, 2010 May 18.
Artículo en Inglés | MEDLINE | ID: mdl-20364819

RESUMEN

Apoptosis signal-regulating kinase 1 (ASK1) is a serine/threonine kinase that responds to a plethora of stress-inducing signals. In turn, activation of ASK1 is associated with a number of human pathological conditions, including neurodegenerative disease, inflammation, and heart failure. In response to oxidative stress, ASK1 activates the cell death-associated p38 MAPK pathway by phosphorylating MKK6. Here, we investigated the regulation of oxidative stress-induced ASK1-catalyzed phosphorylation of MKK6. MKK6 phosphorylation levels increased immediately after H(2)O(2) treatment in intact cells and decreased following treatment for 30 min. When expressed in HEK293T cells, ASK1 was reproducibly purified within a high-molecular mass complex ( approximately 1500 kDa) known as the ASK1 signalosome. Measurement of the in vitro kinetic parameters revealed that the catalytic efficiency (k(cat)/K(m)) of ASK1 was 4000-fold greater in cells treated with H(2)O(2) for 3 min than in untreated cells. Interestingly, although the K(m(ATP)) values were found to be unchanged, the K(m(MKK6)) was dramatically decreased ( approximately 1000-fold). The increased affinity was specific for MKK6 and short-lived, as the K(m(MKK6)) returned to basal levels 30 min after treatment. Consistently, endogenous MKK6 was found within the ASK1 signalosome in intact cells and in addition copurified with ASK1 following treatment for 3 min. In contrast, proteins modulating ASK1 activity and degradation were found to interact with the ASK1 signalosome once MKK6 activation was completed. Taken together, these data suggest that oxidative stress rapidly increases ASK1 catalytic efficiency for MKK6 phosphorylation by increasing MKK6 binding affinity within the ASK1 signalosome prior to induction of inactivation and degradation of the complex.


Asunto(s)
MAP Quinasa Quinasa 6/metabolismo , MAP Quinasa Quinasa Quinasa 5/metabolismo , Estrés Oxidativo/fisiología , Catálisis , Células Cultivadas , Humanos , Cinética , MAP Quinasa Quinasa 6/genética , MAP Quinasa Quinasa 6/aislamiento & purificación , MAP Quinasa Quinasa Quinasa 5/genética , MAP Quinasa Quinasa Quinasa 5/aislamiento & purificación , Fosforilación
17.
Rev Neurosci ; 20(2): 95-110, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19774788

RESUMEN

Hallmarks of Alzheimer's disease (AD) include the accumulation of amyloid beta peptide (Abeta), hyperphosphorylation of tau protein, and increased inflammatory activity in the hippocampus and cerebral cortex. The receptor for advanced glycation endproducts (RAGE) has been shown to interact with Abeta and to modulate Abeta transport across the blood-brain barrier. Furthermore, RAGE is upregulated at sites of inflammation and its activation results in distinct intracellular signaling cascades in respect to Abeta conformers. Besides Abeta, RAGE interacts with several members of the calcium binding S100 protein family, amphoterin and advanced glycation endproducts. Mounting evidence suggests that RAGE is a key player in the signaling pathways triggered by Abeta and S100 proteins in AD. In this review, we discuss recent discoveries about the crosstalk between RAGE, Abeta and S100 proteins in the pathophysiology of AD.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Calcio/metabolismo , Receptores Inmunológicos/metabolismo , Enfermedad de Alzheimer/fisiopatología , Animales , Humanos , Modelos Biológicos , Receptor para Productos Finales de Glicación Avanzada , Proteínas S100/metabolismo
18.
J Neurosci ; 28(20): 5149-58, 2008 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-18480271

RESUMEN

In the genesis of Alzheimer's disease (AD), converging lines of evidence suggest that amyloid-beta peptide (Abeta) triggers a pathogenic cascade leading to neuronal loss. It was long assumed that Abeta had to be assembled into extracellular amyloid fibrils or aggregates to exert its cytotoxic effects. Over the past decade, characterization of soluble oligomeric Abeta species in the brains of AD patients and in transgenic models has raised the possibility that different conformations of Abeta may contribute to AD pathology via different mechanisms. The receptor for advanced glycation end products (RAGE), a member of the Ig superfamily, is a cellular binding site for Abeta. Here, we investigate the role of RAGE in apoptosis induced by distinct well characterized Abeta conformations: Abeta oligomers (AbetaOs), Abeta fibrils (AbetaFs), and Abeta aggregates (AbetaAs). In our in vitro system, treatment with polyclonal anti-RAGE antibodies significantly improves SHSY-5Y cell and neuronal survival exposed to either AbetaOs or AbetaAs but does not affect AbetaF toxicity. Interestingly, using site-specific antibodies, we demonstrate that targeting of the V(d) domain of RAGE attenuates AbetaO-induced toxicity in both SHSY-5Y cells and rat cortical neurons, whereas inhibition of AbetaA-induced apoptosis requires the neutralization of the C(1d) domain of the receptor. Thus, our data indicate that distinct regions of RAGE are involved in Abeta-induced cellular and neuronal toxicity with respect to the Abeta aggregation state, and they suggest the blockage of particular sites of the receptor as a potential therapeutic strategy to attenuate neuronal death.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Encéfalo/metabolismo , Neuronas/metabolismo , Receptores Inmunológicos/metabolismo , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/fisiopatología , Péptidos beta-Amiloides/antagonistas & inhibidores , Animales , Anticuerpos/farmacología , Especificidad de Anticuerpos/inmunología , Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Sitios de Unión/efectos de los fármacos , Sitios de Unión/fisiología , Encéfalo/efectos de los fármacos , Encéfalo/fisiopatología , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/inmunología , Humanos , Neuronas/efectos de los fármacos , Neurotoxinas/antagonistas & inhibidores , Neurotoxinas/metabolismo , Placa Amiloide/efectos de los fármacos , Placa Amiloide/metabolismo , Conformación Proteica/efectos de los fármacos , Estructura Terciaria de Proteína/efectos de los fármacos , Estructura Terciaria de Proteína/fisiología , Ratas , Receptor para Productos Finales de Glicación Avanzada , Receptores Inmunológicos/química , Receptores Inmunológicos/efectos de los fármacos
19.
J Biol Chem ; 281(50): 38905-17, 2006 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-17030513

RESUMEN

S100A16 protein is a new and unique member of the EF-hand Ca(2+)-binding proteins. S100 proteins are cell- and tissue-specific and are involved in many intra- and extracellular processes through interacting with specific target proteins. In the central nervous system S100 proteins are implicated in cell proliferation, differentiation, migration, and apoptosis as well as in cognition. S100 proteins became of major interest because of their close association with brain pathologies, for example depression or Alzheimer's disease. Here we report for the first time the purification and biochemical characterization of human and mouse recombinant S100A16 proteins. Flow dialysis revealed that both homodimeric S100A16 proteins bind two Ca(2+) ions with the C-terminal EF-hand of each subunit, the human protein exhibiting a 2-fold higher affinity. Trp fluorescence variations indicate conformational changes in the orthologous proteins upon Ca(2+) binding, whereas formation of a hydrophobic patch, implicated in target protein recognition, only occurs in the human S100A16 protein. In situ hybridization analysis and immunohistochemistry revealed a widespread distribution in the mouse brain. Furthermore, S100A16 expression was found to be astrocyte-specific. Finally, we investigated S100A16 intracellular localization in human glioblastoma cells. The protein was found to accumulate within nucleoli and to translocate to the cytoplasm in response to Ca(2+) stimulation.


Asunto(s)
Proteínas S100/metabolismo , Animales , Secuencia de Bases , Western Blotting , Cationes , Células Cultivadas , Cartilla de ADN , ADN Complementario , Humanos , Inmunohistoquímica , Ratones , Reacción en Cadena de la Polimerasa , Unión Proteica , ARN Mensajero/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteínas S100/genética , Espectrometría de Fluorescencia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA