Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Biochem Pharmacol ; 168: 330-338, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31348898

RESUMEN

The signaling mechanisms of the angiotensin II type 2 receptor (AT2R), a heptahelical receptor, have not yet been clearly and completely defined. In the present contribution, we set out to identify the molecular determinants involved in AT2R activation. Although AT2R has not been shown to engage Gq/11, G12, Gi2, and ß-arrestin (ßarr) pathways as does the AT1R upon angiotensin II (AngII) stimulation, the atypical positioning of helix VIII in the recently published AT2R structure may play a role in the receptor's capacity to couple to downstream effectors. In the AT2R structure, helix VIII points inwards and towards intracellular loop 3 (ICL3) to form tertiary interactions with transmembrane domain 6 (TM6), possibly impeding access to signaling effectors. On the other hand, in most class A GPCRs, helix VIII is found to be engaged in tertiary interactions with ICL1 and away from the effector binding site. Upon closer examination of the AT2R structure, we found that the residues contained within intracellular loop 1 (ICL1) may be involved in driving this unusual conformation of helix VIII. To explore this hypothesis, we designed a series of AT1R/AT2R receptor chimeras to validate the roles of ICL1 and helix VIII in AT2R signaling. Substituting the AT1R ICL1 into AT2R led to a mutant receptor that coupled to Gi2. The substitution of the helix VIII and C-terminal domains of AT2R into the AT1R backbone led to a mutant receptor that retained AT1R-like signaling properties. These results suggest that the C-terminal portion of AT2R is compatible with canonical GPCR signaling and that ICL1 of AT2R is involved in repositioning helix VIII, which impedes engagement of classical GPCR effectors such as G proteins or ßarrs.


Asunto(s)
Secuencias Hélice-Asa-Hélice/fisiología , Membranas Intracelulares/química , Membranas Intracelulares/metabolismo , Receptor de Angiotensina Tipo 2/química , Receptor de Angiotensina Tipo 2/metabolismo , Angiotensina II/farmacología , Sitios de Unión/efectos de los fármacos , Sitios de Unión/fisiología , Relación Dosis-Respuesta a Droga , Células HEK293 , Secuencias Hélice-Asa-Hélice/efectos de los fármacos , Humanos , Membranas Intracelulares/efectos de los fármacos , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Receptor de Angiotensina Tipo 2/agonistas
2.
Pharmacol Res ; 136: 108-120, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-29959993

RESUMEN

Angiotensin II (AngII) type 1 receptor (AT1R) is a G protein-coupled receptor known for its role in numerous physiological processes and its implication in many vascular diseases. Its functions are mediated through G protein dependent and independent signaling pathways. AT1R has several endogenous peptidic agonists, all derived from angiotensinogen, as well as several synthetic ligands known to elicit biased signaling responses. Here, surface plasmon resonance (SPR) was used as a cell-based and label-free technique to quantify, in real time, the response of HEK293 cells stably expressing the human AT1R. The goal was to take advantage of the integrative nature of this assay to identify specific signaling pathways in the features of the response profiles generated by numerous endogenous and synthetic ligands of AT1R. First, we assessed the contributions of Gq, G12/13, Gi, Gßγ, ERK1/2 and ß-arrestins pathways in the cellular responses measured by SPR where Gq, G12/Rho/ROCK together with ß-arrestins and ERK1/2 were found to play significant roles. More specifically, we established a major role for G12 in the early events of the AT1R-dependent response, which was followed by a robust ERK1/2 component associated to the later phase of the signal. Interestingly, endogenous AT1R ligands (AngII, AngIII and AngIV) exhibited distinct responses signatures with a significant increase of the ERK1/2-like components for both AngIII and AngIV, which points toward possibly distinct physiological roles for the later. We also tested AT1R biased ligands, all of which affected both the early and later events. Our results support SPR-based integrative cellular assays as a powerful approach to delineate the contribution of specific signaling pathways for a given cell response and reveal response differences associated with ligands with distinct pharmacological properties.


Asunto(s)
Angiotensina II/análogos & derivados , Angiotensina II/farmacología , Proteínas de Unión al GTP/fisiología , Receptor de Angiotensina Tipo 1/fisiología , Células HEK293 , Humanos , ARN Interferente Pequeño/genética , Transducción de Señal , Resonancia por Plasmón de Superficie
3.
Pharmacol Res ; 104: 176-85, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26707030

RESUMEN

Advanced glycation end-products (AGE) and the receptor for AGE (RAGE) have been linked to numerous diabetic vascular complications. RAGE activation promotes a self-sustaining state of chronic inflammation and has been shown to induce apoptosis in various cell types. Although previous studies in vascular smooth muscle cells (VSMC) showed that RAGE activation increases vascular calcification and interferes with their contractile phenotype, little is known on the potential of RAGE to induce apoptosis in VSMC. Using a combination of apoptotic assays, we showed that RAGE stimulation with its ligand CML-HSA promotes apoptosis of VSMC. The formation of stress granules and the increase in the level of the associated protein HuR point toward RAGE-dependent endoplasmic reticulum (ER) stress, which is proposed as a key contributor of RAGE-induced apoptosis in VSMC as it has been shown to promote cell death via numerous mechanisms, including up-regulation of caspase-9. Chronic NF-κB activation and modulation of Bcl-2 homologs are also suspected to contribute to RAGE-dependent apoptosis in VSMC. With the goal of reducing RAGE signaling and its detrimental impact on VSMC, we designed a RAGE antagonist (iRAGE) derived from the primary amino acid sequence of HSA. The resulting CML peptide was selected for the high glycation frequency of the primary sequence in the native protein in vivo. Pretreatment with iRAGE blocked 69.6% of the increase in NF-κB signaling caused by RAGE activation with CML-HSA after 48h. Preincubation with iRAGE was successful in reducing RAGE-induced apoptosis, as seen through enhanced cell survival by SPR and reduced PARP cleavage. Activation of executioner caspases was 63.5% lower in cells treated with iRAGE before stimulation with CML-HSA. To our knowledge, iRAGE is the first antagonist shown to block AGE-RAGE interaction and we propose the molecule as an initial candidate for drug discovery.


Asunto(s)
Lisina/análogos & derivados , Péptidos/farmacología , Receptor para Productos Finales de Glicación Avanzada/antagonistas & inhibidores , Albúmina Sérica/farmacología , Animales , Apoptosis/efectos de los fármacos , Caspasa 3/metabolismo , Caspasa 7/metabolismo , Línea Celular , Estrés del Retículo Endoplásmico/efectos de los fármacos , Productos Finales de Glicación Avanzada/metabolismo , Lisina/química , Lisina/farmacología , Músculo Liso Vascular/citología , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/metabolismo , FN-kappa B/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Ratas , Receptor para Productos Finales de Glicación Avanzada/metabolismo , Albúmina Sérica/química
4.
PLoS One ; 10(8): e0128881, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26248341

RESUMEN

Increased blood glucose concentrations promote reactions between glucose and proteins to form advanced glycation end-products (AGE). Circulating AGE in the blood plasma can activate the receptor for advanced end-products (RAGE), which is present on both endothelial and vascular smooth muscle cells (VSMC). RAGE exhibits a complex signaling that involves small G-proteins and mitogen activated protein kinases (MAPK), which lead to increased nuclear factor kappa B (NF-κB) activity. While RAGE signaling has been previously addressed in endothelial cells, little is known regarding its impact on the function of VSMC. Therefore, we hypothesized that RAGE signaling leads to alterations in the mechanical and functional properties of VSMC, which could contribute to complications associated with diabetes. We demonstrated that RAGE is expressed and functional in the A7r5 VSMC model, and its activation by AGE significantly increased NF-κB activity, which is known to interfere with the contractile phenotype of VSMC. The protein levels of the contraction-related transcription factor myocardin were also decreased by RAGE activation with a concomitant decrease in the mRNA and protein levels of transgelin (SM-22α), a regulator of VSMC contraction. Interestingly, we demonstrated that RAGE activation increased the overall cell rigidity, an effect that can be related to an increase in myosin activity. Finally, although RAGE stimulation amplified calcium signaling and slightly myosin activity in VSMC challenged with vasopressin, their contractile capacity was negatively affected. Overall, RAGE activation in VSMC could represent a keystone in the development of vascular diseases associated with diabetes by interfering with the contractile phenotype of VSMC through the modification of their mechanical and functional properties.


Asunto(s)
Músculo Liso Vascular/citología , Receptor para Productos Finales de Glicación Avanzada/metabolismo , Transducción de Señal , Animales , Fenómenos Biomecánicos , Calcio/metabolismo , Línea Celular , Complicaciones de la Diabetes/etiología , Complicaciones de la Diabetes/metabolismo , Productos Finales de Glicación Avanzada/metabolismo , Humanos , Contracción Muscular , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/fisiología , FN-kappa B/metabolismo , Ratas , Albúmina Sérica/metabolismo , Enfermedades Vasculares/etiología , Enfermedades Vasculares/metabolismo , Albúmina Sérica Glicada
5.
Anal Biochem ; 478: 73-81, 2015 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-25800564

RESUMEN

Protein glycation is a ubiquitous process involved in vascular complications observed in diabetes. Glyoxal (GO), an intracellular reactive oxoaldehyde that is one of the most potent glycation agents, readily reacts with amines present on proteins to produce the lysine-derived adduct carboxymethyllysine, which is a prevalent advanced glycation end-product (AGE). Our group previously showed that cell exposure to GO leads to an alteration in the cell contractile activity that could occur as a result of the glycation of various proteins regulating the cell contractile machinery. Here, we measured the extent of glycation on three functionally distinct proteins known to participate in cell contraction and cytoskeletal organization-Rho-kinase (ROCK), actin, and gelsolin (GSN)-using an assay based on the reaction of the cell membrane-permeable fluorescent probe carboxyfluorescein diacetate succinimidyl ester (CFDA-SE), which reacts with primary amine groups of proteins. By combining CFDA-SE fluorescence and Western blot detection, we observed (following GO incubation) increased glycation of actin and ROCK as well as an increased interaction between actin and GSN as observed by co-immunoprecipitation. Thus, we conclude that the use of the fluorescent probe CFDA-SE offers an interesting alternative to perform a comparative analysis of the extent of intracellular protein glycation in live cells.


Asunto(s)
Fluoresceínas/metabolismo , Colorantes Fluorescentes/metabolismo , Glicosilación , Succinimidas/metabolismo , Actinas/análisis , Actinas/metabolismo , Western Blotting , Línea Celular , Fluoresceínas/análisis , Colorantes Fluorescentes/análisis , Gelsolina/análisis , Gelsolina/metabolismo , Glioxal/metabolismo , Humanos , Microscopía Fluorescente , Succinimidas/análisis , Quinasas Asociadas a rho/análisis , Quinasas Asociadas a rho/metabolismo
6.
Integr Biol (Camb) ; 6(4): 411-21, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24503653

RESUMEN

Glyoxal (GO), a highly reactive metabolite of glucose, is associated with diabetic vascular complications via the formation of advanced glycation end-products. Considering its ability to react with proteins' amino acids and its crosslinking potential, we suggest that GO affects cellular mechanical functions such as contractility. Therefore, we tested the effects of GO on cellular contractile response following AngII stimulation of human embryonic kidney cells over-expressing the AT1 receptor (HEK 293 AT1aR). Prior to cell stimulation with AngII, cells exposed to GO exhibited carboxymethyllysine-adduct formation and an increase in cellular stiffness, which could be prevented by pre-treatment with aminoguanidine. The time-dependent cellular contractile response to AngII was measured by monitoring cell membrane displacement by atomic force atomic force microscopy (AFM) and by quantifying myosin light chain phosphorylation (p-MLC) via immunoblotting. Interestingly, short-term GO exposure increased by 2.6 times the amplitude of cell contraction induced by AngII and this was also associated with a sustained rise in p-MLC. This increased response to AngII induced by GO appears to be linked to its glycation potential, as aminoguanidine pre-treatment prevented this increased cellular mechanical response. Our results also suggest that GO could have an impact on ROCK activity, as ROCK inhibition with Y-27632 blocked the enhanced contractile response (p = 0.011) measured under GO conditions. Together, these results indicate that GO enhances the cellular response to AngII and modifies cellular mechanical properties via a mechanism that relies on its glycation potential and on the activation of the ROCK-dependent pathway.


Asunto(s)
Citoesqueleto de Actina/fisiología , Actomiosina/metabolismo , Amidas/farmacología , Angiotensina II/metabolismo , Productos Finales de Glicación Avanzada/metabolismo , Glioxal/metabolismo , Piridinas/farmacología , Citoesqueleto de Actina/ultraestructura , Células HEK293 , Humanos , Microscopía de Fuerza Atómica , Microscopía Fluorescente , Cadenas Ligeras de Miosina/metabolismo , Fosforilación/fisiología
7.
PLoS One ; 8(11): e80532, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24255721

RESUMEN

Over the last decade, it has been established that G-protein-coupled receptors (GPCRs) signal not only through canonical G-protein-mediated mechanisms, but also through the ubiquitous cellular scaffolds ß-arrestin-1 and ß-arrestin-2. Previous studies have implicated ß-arrestins as regulators of actin reorganization in response to GPCR stimulation while also being required for membrane protrusion events that accompany cellular motility. One of the most critical events in the active movement of cells is the cyclic phosphorylation and activation of myosin light chain (MLC), which is required for cellular contraction and movement. We have identified the myosin light chain phosphatase Targeting Subunit (MYPT-1) as a binding partner of the ß-arrestins and found that ß-arrestins play a role in regulating the turnover of phosphorylated myosin light chain. In response to stimulation of the angiotensin Type 1a Receptor (AT1aR), MLC phosphorylation is induced quickly and potently. We have found that ß-arrestin-2 facilitates dephosphorylation of MLC, while, in a reciprocal fashion, ß-arrestin 1 limits dephosphorylation of MLC. Intriguingly, loss of either ß-arrestin-1 or 2 blocks phospho-MLC turnover and causes a decrease in the contraction of cells as monitored by atomic force microscopy (AFM). Furthermore, by employing the ß-arrestin biased ligand [Sar(1),Ile(4),Ile(8)]-Ang, we demonstrate that AT1aR-mediated cellular motility involves a ß-arrestin dependent component. This suggests that the reciprocal regulation of MLC phosphorylation status by ß-arrestins-1 and 2 causes turnover in the phosphorylation status of MLC that is required for cell contractility and subsequent chemotaxic motility.


Asunto(s)
Arrestinas/metabolismo , Cadenas Ligeras de Miosina/metabolismo , Receptor de Angiotensina Tipo 1/metabolismo , Secuencia de Aminoácidos , Angiotensina II/farmacología , Arrestinas/química , Arrestinas/genética , Línea Celular , Movimiento Celular/efectos de los fármacos , Movimiento Celular/genética , Humanos , Datos de Secuencia Molecular , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/metabolismo , Fosfatasa de Miosina de Cadena Ligera/metabolismo , Fosforilación , Unión Proteica , Mapeo de Interacción de Proteínas , Receptor de Angiotensina Tipo 1/genética , Técnicas del Sistema de Dos Híbridos , beta-Arrestina 1 , Arrestina beta 2 , beta-Arrestinas
8.
Biochim Biophys Acta ; 1833(6): 1294-303, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23458835

RESUMEN

STIM1 plays a crucial role in Ca(2+) homeostasis, particularly in replenishing the intracellular Ca(2+) store following its depletion. In cardiomyocytes, the Ca(2+) content of the sarcoplasmic reticulum must be tightly controlled to sustain contractile activity. The presence of STIM1 in cardiomyocytes suggests that it may play a role in regulating the contraction of cardiomyocytes. The aim of the present study was to determine how STIM1 participates in the regulation of cardiac contractility. Atomic force microscopy revealed that knocking down STIM1 disrupts the contractility of cardiomyocyte-derived HL-1 cells. Ca(2+) imaging also revealed that knocking down STIM1 causes irregular spontaneous Ca(2+) oscillations in HL-1 cells. Action potential recordings further showed that knocking down STIM1 induces early and delayed afterdepolarizations. Knocking down STIM1 increased the peak amplitude and current density of T-type voltage-dependent Ca(2+) channels (T-VDCC) and shifted the activation curve toward more negative membrane potentials in HL-1 cells. Biotinylation assays revealed that knocking down STIM1 increased T-VDCC surface expression and co-immunoprecipitation assays suggested that STIM1 directly regulates T-VDCC activity. Thus, STIM1 is a negative regulator of T-VDCC activity and maintains a constant cardiac rhythm by preventing a Ca(2+) overload that elicits arrhythmogenic events.


Asunto(s)
Canales de Calcio Tipo T/metabolismo , Señalización del Calcio/fisiología , Calcio/metabolismo , Glicoproteínas de Membrana/metabolismo , Contracción Muscular , Miocitos Cardíacos/metabolismo , Taquicardia , Animales , Western Blotting , Canales de Calcio , Células Cultivadas , Electrofisiología , Inmunoprecipitación , Glicoproteínas de Membrana/antagonistas & inhibidores , Glicoproteínas de Membrana/genética , Ratones , Miocitos Cardíacos/citología , Molécula de Interacción Estromal 1
9.
J Pharmacol Exp Ther ; 328(2): 540-8, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18987301

RESUMEN

The aim of this study was to identify the role of chymase in the conversion of exogenously administered Big endothelin-1 in the mouse in vivo. Real-time polymerase chain reaction analysis detected mRNA of mucosal mast cell chymases 4 and 5, endothelin-converting enzyme 1a, and neutral endopeptidase 24.11 in pulmonary, cardiac, and aorta homogenates derived from C57BL/6J mice, with the latter tissue expressing the highest levels of both chymase isoforms. Furthermore, hydrolysis of a fluorogenic peptide substrate, Suc-Leu-Leu-Val-Tyr-7-amino-4-methylcoumarin, was sensitive to the chymase inhibitors Suc-Val-Pro-Phe(P)(OPh)(2) (200 microM) and chymostatin [(S)-1-carboxy-2-phenylethyl]-carbamoyl-alpha-[2-iminohexahydro-4(S)-pyrimidyl]-(S)-Gly-X-Phe-al, where X can be the amino acid Leu, Val, or Ile) (100 microM) in supernatants extracted from the same tissue homogenates. In anesthetized mice, Big endothelin-1, endothelin-1 (1-31), and endothelin-1 triggered pressor responses (ED(50)s, 0.67, 0.89, and 0.16 nmol/kg) that were all reduced or potentiated by selective endothelin ET(A) or ET(B) receptor antagonists, respectively, BQ-123 (cyclo[D-Asp-Pro-D-Val-Leu-D-Trp]) or BQ-788 (N-[N-[N-[(2,6-dimethyl-1-piperidinyl)carbonyl]-4-methyl-l-leucyl]-1-(methoxycarbonyl)-D-tryptophyl]-d-norleucine sodium salt), each at 1 mg/kg. The pressor responses to big endothelin-1 were significantly reduced by the neutral endopeptidase inhibitor thiorphan (dl-3-mercapto-2-benzylpropanoylglycine) (1 mg/kg) or the endothelin-converting enzyme inhibitor CGS 35066 [alpha-[(S)-(phosphonomethyl)amino]-3-dibenzofuranopropanoic acid] (0.1 mg/kg). In contrast, the responses to endothelin-1 (1-31) were abolished by thiorphan but unaffected by CGS 35066. In addition, Suc-Val-Pro-Phe(P)(OPh)(2) (20-40 mg/kg) reduced, by more than 60%, the hemodynamic response to big endothelin-1 but not to endothelin-1 (1-31) and endothelin-1. Finally, intravenous administration of big endothelin-1 induced Suc-Val-Pro-Phe(P)-(OPh)(2)-sensitive increases in plasma-immunoreactive levels of endothelin-1 (1-31) and endothelin-1. The present study suggests that chymase plays a pivotal role in the conversion and cardiovascular properties of big endothelin-1 in vivo.


Asunto(s)
Quimasas/metabolismo , Endotelina-1/metabolismo , Anestesia , Animales , Masculino , Ratones , Ratones Endogámicos C57BL , ARN Mensajero/metabolismo , Receptores de Endotelina/metabolismo , Serina Endopeptidasas/genética , Serina Endopeptidasas/metabolismo
10.
Can J Physiol Pharmacol ; 86(8): 516-25, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18758499

RESUMEN

We hypothesized that constitutive endothelial NO synthase (eNOS) and inducible NO synthase (iNOS) have opposite effects on the regulation of endothelin and its receptors. We therefore sought to determine whether deletions of iNOS or eNOS genes in mice modulate pressor responses to endothelin and the expression of ETA and ETB receptors in a similar fashion. Despite unchanged baseline hemodynamic parameters, anesthetized iNOS-/- mice displayed reduced pressor responses to endothelin-1, but not to that of IRL-1620, a selective ETB agonist. Protein content of cardiac ETA receptors was reduced in iNOS-/- mice compared with wild-type mice, but that of ETB receptors was unchanged. Anesthetized eNOS-/- mice presented a hypertensive state, accompanied by an enhanced pressor response to intravenous endothelin-1, whereas the pressor response to IRL-1620 was reduced. Protein levels were also found to be increased for ETA receptors, but reduced for ETB receptors, in cardiac tissues of eNOS-/- mice. In conscious animals, both strains responded equally to the hypotensive effect of an ETA antagonist, ABT-627, whereas orally administered A-192621, an ETB antagonist, increased MAP to a greater extent in eNOS-/- than in wild-type mice. Furthermore, significant levels of immunoreactive endothelin were found in mesenteric arteries in eNOS-/- but not in iNOS-/- or wild-type congeners. Our study shows that repression of iNOS or eNOS has differential effects on endothelin-1 and its receptors. We have also shown that the heart is the main organ in which iNOS or eNOS repression induces important alterations in protein content of endothelin receptors in adult mice.


Asunto(s)
Endotelina-1/fisiología , Óxido Nítrico Sintasa de Tipo III/genética , Óxido Nítrico Sintasa de Tipo II/genética , Transducción de Señal/fisiología , Animales , Cartilla de ADN , ADN Complementario/biosíntesis , ADN Complementario/genética , Antagonistas de los Receptores de la Endotelina A , Antagonistas de los Receptores de la Endotelina B , Endotelina-1/genética , Endotelina-1/farmacología , Endotelinas/farmacología , Ratones , Ratones Noqueados , Nitratos/metabolismo , Óxido Nítrico Sintasa de Tipo II/fisiología , Óxido Nítrico Sintasa de Tipo III/fisiología , Nitritos/metabolismo , Fragmentos de Péptidos/farmacología , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Receptor de Endotelina A/agonistas , Receptor de Endotelina B/agonistas , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/genética , Superóxidos/metabolismo , Telemetría
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA