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










Base de datos
Intervalo de año de publicación
1.
Mol Pharmacol ; 89(1): 176-86, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26494861

RESUMEN

The molecular pharmacology of the G protein-coupled receptors for sphingosine 1-phosphate (S1P) provides important insight into established and new therapeutic targets. A new, potent bitopic S1P3 antagonist, SPM-354, with in vivo activity, has been used, together with S1P3-knockin and S1P3-knockout mice to define the spatial and functional properties of S1P3 in regulating cardiac conduction. We show that S1P3 is a key direct regulator of cardiac rhythm both in vivo and in isolated perfused hearts. 2-Amino-2-[2-(4-octylphenyl)ethyl]propane-1,3-diol in vivo and S1P in isolated hearts induced a spectrum of cardiac effects, ranging from sinus bradycardia to complete heart block, as measured by a surface electrocardiogram in anesthetized mice and in volume-conducted Langendorff preparations. The agonist effects on complete heart block are absent in S1P3-knockout mice and are reversed in wild-type mice with SPM-354, as characterized and described here. Homologous knockin of S1P3-mCherry is fully functional pharmacologically and is strongly expressed by immunohistochemistry confocal microscopy in Hyperpolarization Activated Cyclic Nucleotide Gated Potassium Channel 4 (HCN4)-positive atrioventricular node and His-Purkinje fibers, with relative less expression in the HCN4-positive sinoatrial node. In Langendorff studies, at constant pressure, SPM-354 restored sinus rhythm in S1P-induced complete heart block and fully reversed S1P-mediated bradycardia. S1P3 distribution and function in the mouse ventricular cardiac conduction system suggest a direct mechanism for heart block risk that should be further studied in humans. A richer understanding of receptor and ligand usage in the pacemaker cells of the cardiac system is likely to be useful in understanding ventricular conduction in health, disease, and pharmacology.


Asunto(s)
Bloqueo Cardíaco/tratamiento farmacológico , Bloqueo Cardíaco/genética , Frecuencia Cardíaca/efectos de los fármacos , Frecuencia Cardíaca/fisiología , Receptores de Lisoesfingolípidos/antagonistas & inhibidores , Receptores de Lisoesfingolípidos/genética , Animales , Cardiotónicos/farmacología , Cardiotónicos/uso terapéutico , Bloqueo Cardíaco/fisiopatología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores de Esfingosina-1-Fosfato
2.
Curr Top Microbiol Immunol ; 378: 1-21, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24728591

RESUMEN

The understanding of the role of the sphingosine 1-phosphate signaling system in immunology and host defense has deepened exponentially over the past 12 years since the discovery that lymphocyte egress was reversibly modulated by sphingosine 1-phosphate receptors, and with the development of fingolimod, a prodrug of a nonselective S1P receptor agonist, for therapeutic use in the treatment of relapsing, remitting multiple sclerosis. Innovative genetic and chemical approaches, together with structural biology, now provide a more detailed molecular understanding of a regulated lysophospholipid ligand with a variety of autocrine, paracrine, and systemic effects in physiology and pathology, based upon selective interactions with a high affinity and selective evolutionary cluster of G-protein-coupled receptors.


Asunto(s)
Lisofosfolípidos/metabolismo , Transducción de Señal , Esfingosina/análogos & derivados , Animales , Humanos , Esclerosis Múltiple/tratamiento farmacológico , Esclerosis Múltiple/genética , Esclerosis Múltiple/metabolismo , Receptores de Lisoesfingolípidos/agonistas , Receptores de Lisoesfingolípidos/genética , Receptores de Lisoesfingolípidos/metabolismo , Esfingosina/metabolismo
3.
F1000Prime Rep ; 6: 109, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25580263

RESUMEN

Development of sphingosine-1-phosphate receptor 1 (S1P1) modulators to dampen inflammation and its sequelae is becoming increasingly promising for treating medical conditions characterized by significant immunopathology. As shown by the non-selective S1P receptor modulator FTY720 (fingolimod [Gilenya(®)]) in the treatment of relapsing-remitting multiple sclerosis (MS), the ability to use S1P1 modulation to precisely block immune cell traffic-immunomodulation-while maintaining immunosurveillance, has opened therapeutic opportunities in various other immune-derived chronic pathologies, including inflammatory bowel disease (IBD), lupus, psoriasis, as well as, potentially, in early acute viral respiratory infection. Proof-of-concept studies across validated animal models with S1P receptor modulators highly selective for S1P1, such as BAF-312 (Siponimod), KRP-203, ONO-4641 (Ceralifimod), ponesimod and RPC-1063, and emerging clinical trials for safety and efficacy in humans, particularly in MS, ulcerative colitis (UC) and psoriasis, have set the stage for us to consider additional testing in various other autoimmune diseases.

4.
Mol Pharmacol ; 83(2): 316-21, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23204443

RESUMEN

Sphingosine 1-phosphate receptor 1 (S1P(1)) is a G protein-coupled receptor that is critical for proper lymphocyte development and recirculation. Agonists to S1P(1) are currently in use clinically for the treatment of multiple sclerosis, and these drugs may act on both S1P(1) expressed on lymphocytes and S1P(1) expressed within the central nervous system. Agonists to S1P(1) and deficiency in S1P(1) both cause lymphocyte sequestration in the lymph nodes. In the present study, we show that S1P(1) antagonism induces lymphocyte sequestration in the lymph nodes similar to that observed with S1P(1) agonists while upregulating S1P(1) on lymphocytes and endothelial cells. Additionally, we show that S1P(1) antagonism reverses experimental autoimmune encephalomyelitis in mice without acting on S1P(1) expressed within the central nervous system, demonstrating that lymphocyte sequestration via S1P(1) antagonism is sufficient to alleviate autoimmune pathology.


Asunto(s)
Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Encefalomielitis Autoinmune Experimental/genética , Inmunosupresores/farmacología , Receptores de Lisoesfingolípidos/antagonistas & inhibidores , Receptores de Lisoesfingolípidos/genética , Animales , Células CHO , Línea Celular , Sistema Nervioso Central/efectos de los fármacos , Sistema Nervioso Central/metabolismo , Cricetinae , Encefalomielitis Autoinmune Experimental/metabolismo , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Femenino , Células HEK293 , Humanos , Ganglios Linfáticos/efectos de los fármacos , Ganglios Linfáticos/metabolismo , Linfocitos/efectos de los fármacos , Linfocitos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Receptores de Lisoesfingolípidos/metabolismo , Regulación hacia Arriba/efectos de los fármacos
5.
Am J Physiol Cell Physiol ; 302(10): C1460-8, 2012 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-22357735

RESUMEN

Lymph nodes are highly organized structures specialized for efficient regulation of adaptive immunity. The blood and lymphatic systems within a lymph node play essential roles by providing functionally distinct environments for lymphocyte entry and egress, respectively. Direct imaging and measurement of vascular microenvironments by intravital multiphoton microscopy provide anatomical and mechanistic insights into the essential events of lymphocyte trafficking. Lymphocytes, blood endothelial cells, and lymphatic endothelial cells express sphingosine 1-phosphate receptor 1, a key G protein-coupled receptor regulating cellular egress and a modulator of endothelial permeability. Here we report the development of a differential vascular labeling (DVL) technique in which a single intravenous injection of a fluorescent dextran, in combination with fluorescent semiconductor quantum dot particles, differentially labels multiple blood and lymphatic compartments in a manner dependent on the size of the fluorescent particle used. Thus DVL allows measurement of endothelial integrity in multiple vascular compartments and the affects or pharmacological manipulation in vascular integrity. In addition, this technique allows for real-time observation of lymphocyte trafficking across physiological barriers differentiated by DVL. Last, single-field fluid movement dynamics can be derived, allowing for the simultaneous determination of fluid flow rates in diverse blood and lymphatic compartments.


Asunto(s)
Sistemas de Computación , Células Endoteliales/fisiología , Líquido Extracelular/química , Vasos Linfáticos/química , Flujo Sanguíneo Regional , Coloración y Etiquetado/métodos , Animales , Células Endoteliales/química , Líquido Extracelular/fisiología , Ganglios Linfáticos/irrigación sanguínea , Ganglios Linfáticos/química , Ganglios Linfáticos/fisiología , Vasos Linfáticos/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Receptores de Lisoesfingolípidos/biosíntesis , Receptores de Lisoesfingolípidos/sangre , Flujo Sanguíneo Regional/fisiología
6.
Mol Pharmacol ; 81(2): 166-74, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22031473

RESUMEN

Multiple sclerosis (MS) therapies modulate T-cell autoimmunity in the central nervous system (CNS) but may exacerbate latent infections. Fingolimod, a nonselective sphingosine-1-phosphate (S1P) receptor agonist that induces sustained lymphopenia and accumulates in the CNS, represents a new treatment modality for MS. We hypothesized that sustained lymphopenia would not be required for efficacy and that a selective, CNS-penetrant, peripherally short-acting, S1P(1) agonist would show full efficacy in a mouse MS model. Using daily treatment with 10 mg/kg 2-(4-(5-(3,4-diethoxyphenyl)-1,2,4-oxadiazol-3-yl)-2,3-dihydro-1H-inden-1-yl amino)ethanol (CYM-5442) at the onset of clinical signs in myelin oligodendrocyte glycoprotein MOG(35-55)- induced experimental allergic encephalomyelitis (EAE), we assessed clinical scores, CNS cellular infiltration, demyelination, and gliosis for 12 days with CYM-5442, vehicle, or fingolimod. CYM-5442 levels in CNS and plasma were determined at experiment termination, and blood lymphopenia was measured 3 and 24 h after the last injection. Plasma levels of cytokines were assayed at the end of the protocol. Changes in S1P(1)-enhanced green fluorescent protein expression on neurons and astrocytes during active EAE and upon CYM-5442 treatment were quantified with flow cytometry and Western blotting by using native-locus enhanced green fluorescent protein-tagged S1P(1) mice. S1P(1) agonism alone reduced pathological features as did fingolimod (maximally lymphopenic throughout), despite full reversal of lymphopenia within each dosing interval. CYM-5442 levels in CNS but not in plasma were sustained. Neuronal and astrocytic S1P(1) expression in EAE was suppressed by CYM-5442 treatment, relative to vehicle, and levels of key cytokines, such as interleukin 17A, were also significantly reduced in drug-treated mice. S1P(1)-selective agonists that induce reversible lymphopenia while persisting in the CNS may be effective MS treatments.


Asunto(s)
Linfopenia/tratamiento farmacológico , Esclerosis Múltiple/tratamiento farmacológico , Receptores de Lisoesfingolípidos/agonistas , Animales , Sistema Nervioso Central/metabolismo , Citocinas/efectos de los fármacos , Modelos Animales de Enfermedad , Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Clorhidrato de Fingolimod , Inmunosupresores , Indanos , Ratones , Oxadiazoles , Glicoles de Propileno/uso terapéutico , Receptores de Lisoesfingolípidos/metabolismo , Esfingosina/análogos & derivados , Esfingosina/uso terapéutico
7.
Nat Chem Biol ; 7(5): 254-6, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21445057

RESUMEN

Sphingosine 1-phosphate receptor 1 (S1P(1)) is critical for lymphocyte recirculation and is a clinical target for treatment of multiple sclerosis. By generating a short-duration S1P(1) agonist and mice in which fluorescently tagged S1P(1) replaces wild-type receptor, we elucidate physiological and agonist-perturbed changes in expression of S1P(1) at a subcellular level in vivo. We demonstrate differential downregulation of S1P(1) on lymphocytes and endothelia after agonist treatment.


Asunto(s)
Técnicas de Sustitución del Gen , Proteínas Fluorescentes Verdes/química , Esclerosis Múltiple/tratamiento farmacológico , Receptores de Lisoesfingolípidos/agonistas , Receptores de Lisoesfingolípidos/uso terapéutico , Animales , Regulación hacia Abajo/efectos de los fármacos , Endotelio/efectos de los fármacos , Endotelio/metabolismo , Citometría de Flujo , Colorantes Fluorescentes/química , Colorantes Fluorescentes/metabolismo , Proteínas Fluorescentes Verdes/metabolismo , Linfocitos/efectos de los fármacos , Linfocitos/metabolismo , Ratones , Esclerosis Múltiple/metabolismo , Esclerosis Múltiple/patología , Receptores de Lisoesfingolípidos/metabolismo , Factores de Tiempo
8.
Annu Rev Biochem ; 78: 743-68, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19231986

RESUMEN

The sphingosine 1-phosphate (S1P) receptor signaling system is a productive model system. A hydrophobic zwitterionic lysophospholipid ligand with difficult physical properties interacts with five high-affinity G protein-coupled receptors to generate multiple downstream signals. These signals modulate homeostasis and pathology on a steep agonist concentration-response curve. Ligand presence is essential for vascular development and endothelial integrity, while acute increases in ligand concentrations result in cardiac death. Understanding this integrated biochemical system has exemplified the impact of both genetics and chemistry. Developing specific tools with defined biochemical properties for the reversible modulation of signals in real time has been essential to complement insights gained from genetic approaches that may be irreversible and compensated. Despite its knife-edge between life and death, this system, based in part on receptor subtype-selectivity and in part on differential attenuation of deleterious signals, now appears to be on the cusp of meaningful therapy for multiple sclerosis.


Asunto(s)
Receptores de Lisoesfingolípidos/metabolismo , Transducción de Señal , Animales , Sistema Cardiovascular/embriología , Sistema Cardiovascular/metabolismo , Humanos , Tejido Linfoide/embriología , Tejido Linfoide/metabolismo , Esclerosis Múltiple/metabolismo , Receptores de Lisoesfingolípidos/química , Receptores de Lisoesfingolípidos/genética
9.
Mol Pharmacol ; 74(5): 1308-18, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18708635

RESUMEN

Strong evidence exists for interactions of zwitterionic phosphate and amine groups in sphingosine-1 phosphate (S1P) to conserved Arg and Glu residues present at the extracellular face of the third transmembrane domain of S1P receptors. The contribution of Arg(120) and Glu(121) for high-affinity ligand-receptor interactions is essential, because single-point R(120)A or E(121)A S1P(1) mutants neither bind S1P nor transduce S1P function. Because S1P receptors are therapeutically interesting, identifying potent selective agonists with different binding modes and in vivo efficacy is of pharmacological importance. Here we describe a modestly water-soluble highly selective S1P(1) agonist [2-(4-(5-(3,4-diethoxyphenyl)-1,2,4-oxadiazol-3-yl)-2,3-dihydro-1H-inden-1-yl amino) ethanol (CYM-5442)] that does not require Arg(120) or Glu(121) residues for activating S1P(1)-dependent p42/p44 mitogen-activated protein kinase phosphorylation, which defines a new hydrophobic pocket in S1P(1). CYM-5442 is a full agonist in vitro for S1P(1) internalization, phosphorylation, and ubiquitination. It is noteworthy that CYM-5442 was a full agonist for induction and maintenance of S1P(1)-dependent blood lymphopenia, decreasing B lymphocytes by 65% and T lymphocytes by 85% of vehicle. Induction of CYM-5442 lymphopenia was dose- and time-dependent, requiring serum concentrations in the 50 nM range. In vitro measures of S1P(1) activation by CYM-5442 were noncompetitively inhibited by a specific S1P(1) antagonist [(R)-3-amino-(3-hexylphenylamino)-4-oxobutylphosphonic acid (W146)], competitive for S1P, 2-amino-2-(4-octylphenethyl)propane-1,3-diol (FTY720-P), and 5-[4-phenyl-5-(trifluoromethyl)-2-thienyl]-3-[3-(trifluoromethyl)phenyl]-1,2, 4-oxadiazole (SEW2871). In addition, lymphopenia induced by CYM-5442 was reversed by W146 administration or upon pharmacokinetic agonist clearance. Pharmacokinetics in mice also indicated that CYM-5442 partitions significantly in central nervous tissue. These data show that CYM-5442 activates S1P(1)-dependent pathways in vitro and to levels of full efficacy in vivo through a hydrophobic pocket separate from the orthosteric site of S1P binding that is headgroup-dependent.


Asunto(s)
Indanos/farmacología , Oxadiazoles/farmacología , Receptores de Lisoesfingolípidos/agonistas , Animales , Células CHO , Cricetinae , Cricetulus , Ensayo de Inmunoadsorción Enzimática , Ligandos , Espectroscopía de Resonancia Magnética , Masculino , Ratones , Ratones Endogámicos C57BL , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Estructura Molecular , Unión Proteica , Ratas , Ratas Sprague-Dawley , Receptores de Lisoesfingolípidos/metabolismo
10.
ACS Chem Biol ; 3(8): 486-98, 2008 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-18590333

RESUMEN

We have studied the sphingosine 1-phosphate (S1P) receptor system to better understand why certain molecular targets within a closely related family are much more tractable when identifying compelling chemical leads. Five medically important G-protein-coupled receptors for S1P regulate heart rate, coronary artery caliber, endothelial barrier integrity, and lymphocyte trafficking. Selective S1P receptor agonist probes would be of great utility to study receptor subtype-specific function. Through systematic screening of the same libraries, we identified novel selective agonist chemotypes for each of the S1P1 and S1P3 receptors. Ultrahigh-throughput screening (uHTS) for S1P1 was more effective than that for S1P3, with many selective, low nanomolar hits of proven mechanism emerging. Receptor structure modeling and ligand docking reveal differences between the receptor binding pockets, which are the basis for subtype selectivity. Novel selective agonists interact primarily in the hydrophobic pocket of the receptor in the absence of headgroup interactions. Chemistry-space and shape-based analysis of the screening libraries in combination with the binding models explain the observed differential hit rates and enhanced efficiency for lead discovery for S1P1 versus S1P3 in this closely related receptor family.


Asunto(s)
Técnicas Biosensibles/métodos , Sondas Moleculares/química , Oxadiazoles/química , Receptores de Lisoesfingolípidos/agonistas , Animales , Sitios de Unión , Células CHO , Calcio/metabolismo , Análisis por Conglomerados , Cricetinae , Cricetulus , Ligandos , Modelos Moleculares , Sondas Moleculares/farmacología , Oxadiazoles/farmacología , Fosforilación , Unión Proteica , Conformación Proteica , Receptores de Lisoesfingolípidos/genética , Relación Estructura-Actividad
11.
Trends Immunol ; 28(3): 102-7, 2007 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17276731

RESUMEN

The lysophospholipid sphingosine 1-phosphate (S1P) is a pleiotropic signaling lipid present constitutively in plasma, and secreted locally at elevated concentrations at sites of inflammation. S1P maintains essential variable homeostatic functions in addition to inducing pathophysiology through the activation of five specific high-affinity G-protein-coupled receptors. Therefore, S1P can function as an extracellular rheostat regulating tonic and acutely evoked functions. Although S1P receptors can regulate lymphoid development and lymphocyte trafficking, and different opinions exist on the roles of receptor agonism and functional antagonism in regulating lymphocyte recirculation, this personal perspective highlights the pivotal control points regulated by constitutive and induced S1P receptor tone at vascular endothelial and lymphatic endothelial barriers, through which S1P agonism impacts on both innate and adaptive immunity. We also emphasize how specific, proof-of-concept chemical tools complement genetic approaches by enabling reversible perturbation of the S1P-S1P(1) receptor axis and, thus, clarifying in vivo mechanisms in the absence of developmental compensations.


Asunto(s)
Endotelio Vascular/metabolismo , Lisofosfolípidos/fisiología , Esfingosina/análogos & derivados , Animales , Antígenos CD/fisiología , Antígenos de Diferenciación de Linfocitos T/fisiología , Movimiento Celular , Clorhidrato de Fingolimod , Humanos , Lectinas Tipo C , Linfocitos/fisiología , Glicoles de Propileno/farmacología , Receptores de Lisoesfingolípidos/fisiología , Transducción de Señal , Esfingosina/farmacología , Esfingosina/fisiología
12.
J Biol Chem ; 282(10): 7254-64, 2007 Mar 09.
Artículo en Inglés | MEDLINE | ID: mdl-17218309

RESUMEN

Sphingosine 1-phosphate subtype 1 (S1P(1)) receptor agonists alter lymphocyte trafficking and endothelial barrier integrity in vivo. Among these is the potent, non-selective agonist, FTY720-P, whose mechanism of action has been suggested to correlate with S1P(1) down-regulation. Discovery of the in vivo active S1P(1)-selective agonist, SEW2871, has broadened our understanding of minimal requirements for S1P(1) function while highlighting differences regarding agonist effect on S1P(1) fate, because SEW2871 does not degrade S1P(1). To further understand the mechanism of agonist-induced S1P(1) down-regulation, we compared signaling and fate of human S1P(1)-green fluorescent protein (GFP) in stable 293 cells, using AFD-R, a chiral analog of FTY720-P, SEW2871, and S1P. Although all agonists acutely internalized S1P(1) to late endosomal vesicles and activated GTPgammaS(35) binding and pERK to similar maxima, only AFD-R led to significant S1P(1) down-regulation, as shown by GFP immunoprecipitation studies. Down-regulation was time- and concentration-dependent, was partially blocked by proteasomal inhibition and reversed by chloroquine and an antagonist to S1P(1). All agonists induced a receptor-associated increase in ubiquitination, with AFD-R inducing 3-fold more accumulation than S1P and being 3-4 logs more potent than SEW2871. The formation of AFD-R-receptor ubiquitin complex was inhibited by antagonist and chloroquine and was enhanced by proteasomal inhibition. Identification of proteins by PAGE liquid chromatography-tandem mass spectrometry in cells treated with AFD-R confirmed the co-migration of ubiquitin peptides with those of S1P(1) and GFP, relative to vehicle alone. These data suggest that the hierarchy of ubiquitin recruitment to S1P(1) (AFD-R > S1P > SEW2871) correlates with the efficiency of lysosomal receptor degradation and reflects intrinsic differences between agonists.


Asunto(s)
Lisofosfolípidos/metabolismo , Proteómica , Esfingosina/análogos & derivados , Células Cultivadas , Cloroquina/farmacología , Clorhidrato de Fingolimod , Guanosina 5'-O-(3-Tiotrifosfato)/metabolismo , Humanos , Ligandos , Oxadiazoles/farmacología , Glicoles de Propileno/farmacología , Transporte de Proteínas , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Lisoesfingolípidos/agonistas , Receptores de Lisoesfingolípidos/antagonistas & inhibidores , Receptores de Lisoesfingolípidos/metabolismo , Esfingosina/metabolismo , Esfingosina/farmacología , Tiofenos/farmacología , Ubiquitina/metabolismo
13.
Nat Chem Biol ; 2(8): 434-41, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16829954

RESUMEN

Sphingosine 1-phosphate (S1P, 1) regulates vascular barrier and lymphoid development, as well as lymphocyte egress from lymphoid organs, by activating high-affinity S1P1 receptors. We used reversible chemical probes (i) to gain mechanistic insights into S1P systems organization not accessible through genetic manipulations and (ii) to investigate their potential for therapeutic modulation. Vascular (but not airway) administration of the preferred R enantiomer of an in vivo-active chiral S1P1 receptor antagonist induced loss of capillary integrity in mouse skin and lung. In contrast, the antagonist did not affect the number of constitutive blood lymphocytes. Instead, alteration of lymphocyte trafficking and phenotype required supraphysiological elevation of S1P1 tone and was reversed by the antagonist. In vivo two-photon imaging of lymph nodes confirmed requirements for obligate agonism, and the data were consistent with the presence of a stromal barrier mechanism for gating lymphocyte egress. Thus, chemical modulation reveals differences in S1P-S1P1 'set points' among tissues and highlights both mechanistic advantages (lymphocyte sequestration) and risks (pulmonary edema) of therapeutic intervention.


Asunto(s)
Anilidas/farmacología , Linfocitos/efectos de los fármacos , Organofosfonatos/farmacología , Receptores de Lisoesfingolípidos/antagonistas & inhibidores , Anilidas/administración & dosificación , Anilidas/síntesis química , Animales , Células CHO , Permeabilidad Capilar/efectos de los fármacos , Permeabilidad Capilar/fisiología , Células Cultivadas , Cricetinae , Modelos Animales de Enfermedad , Azul de Evans/química , Humanos , Ganglios Linfáticos/efectos de los fármacos , Linfocitos/metabolismo , Lisofosfolípidos/química , Lisofosfolípidos/farmacología , Lisofosfolípidos/fisiología , Ratones , Ratones Endogámicos C57BL , Modelos Biológicos , Organofosfonatos/administración & dosificación , Organofosfonatos/síntesis química , Fenotipo , Edema Pulmonar/inducido químicamente , Edema Pulmonar/diagnóstico , Receptores de Lisoesfingolípidos/agonistas , Receptores de Lisoesfingolípidos/metabolismo , Esfingosina/análogos & derivados , Esfingosina/química , Esfingosina/farmacología , Esfingosina/fisiología , Estereoisomerismo
14.
Chem Biol ; 12(6): 703-15, 2005 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15975516

RESUMEN

The essential role of the sphingosine 1-phosphate (S1P) receptor S1P(1) in regulating lymphocyte trafficking was demonstrated with the S1P(1)-selective nanomolar agonist, SEW2871. Despite its lack of charged headgroup, the tetraaromatic compound SEW2871 binds and activates S1P(1) through a combination of hydrophobic and ion-dipole interactions. Both S1P and SEW2871 activated ERK, Akt, and Rac signaling pathways and induced S1P(1) internalization and recycling, unlike FTY720-phosphate, which induces receptor degradation. Agonism with receptor recycling is sufficient for alteration of lymphocyte trafficking by S1P and SEW2871. S1P(1) modeling and mutagenesis studies revealed that residues binding the S1P headgroup are required for kinase activation by both S1P and SEW2871. Therefore, SEW2871 recapitulates the action of S1P in all the signaling pathways examined and overlaps in interactions with key headgroup binding receptor residues, presumably replacing salt-bridge interactions with ion-dipole interactions.


Asunto(s)
Evaluación Preclínica de Medicamentos , Sondas Moleculares/metabolismo , Oxadiazoles/farmacología , Receptores de Lisoesfingolípidos/agonistas , Receptores de Lisoesfingolípidos/metabolismo , Transducción de Señal/efectos de los fármacos , Tiofenos/farmacología , Animales , Sitios de Unión , Línea Celular , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Cricetinae , Activación Enzimática/efectos de los fármacos , Humanos , Ligandos , Lisofosfolípidos/química , Lisofosfolípidos/farmacología , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Modelos Moleculares , Sondas Moleculares/química , Sondas Moleculares/farmacología , Mutación/genética , Oxadiazoles/química , Fosforilación/efectos de los fármacos , Proteínas Serina-Treonina Quinasas/metabolismo , Estructura Terciaria de Proteína , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-akt , Receptores de Lisoesfingolípidos/química , Receptores de Lisoesfingolípidos/genética , Esfingosina/análogos & derivados , Esfingosina/química , Esfingosina/farmacología , Tiofenos/química , Proteínas de Unión al GTP rac/metabolismo
15.
Endocrinology ; 145(11): 5157-67, 2004 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-15297446

RESUMEN

Alpha(1)-Adrenergic receptors have been implicated in growth-promoting pathways. A microarray study of individual alpha(1)-adrenergic receptor subtypes (alpha(1A), alpha(1B), and alpha(1D)) expressed in Rat-1 fibroblasts revealed that epinephrine altered the transcription of several cell cycle regulatory genes in a direction consistent with the alpha(1A)- and alpha(1D)-adrenergic receptors mediating G(1)-S cell cycle arrest and the alpha(1B-)mediating cell-cycle progression. A time course indicated that in alpha(1A) cells, epinephrine stimulated a G(1)-S arrest, which began after 8 h of stimulation and maximized at 16 h, at which point was completely blocked with cycloheximide. The alpha(1B)-adrenergic receptor profile also showed unchecked cell cycle progression, even under low serum conditions and induced foci formation. The G(1)-S arrest induced by alpha(1A)- and alpha(1D)-adrenergic receptors was associated with decreased cyclin-dependent kinase-6 and cyclin E-associated kinase activities and increased expression of the cyclin-dependent kinase inhibitor p27(Kip1), all of which were blocked by prazosin. There were no differences in kinase activities and/or expression of p27(Kip1) in epinephrine alpha(1B)-AR fibroblasts, although the microarray did indicate differences in p27(Kip1) RNA levels. Cell counts proved the antimitotic effect of epinephrine in alpha(1A) and alpha(1D) cells and indicated that alpha(1B)-adrenergic receptor subtype expression was sufficient to cause proliferation of Rat-1 fibroblasts independent of agonist stimulation. Analysis in transfected PC12 cells also confirmed the alpha(1A)- and alpha(1B)-adrenergic receptor effect. The alpha(1B)-subtype native to DDT1-MF2 cells, a smooth muscle cell line, caused progression of the cell cycle. These results indicate that the alpha(1A)- and alpha(1D)-adrenergic receptors mediate G(1)-S cell-cycle arrest, whereas alpha(1B)-adrenergic receptor expression causes a cell cycle progression and may induce transformation in sensitive cell lines.


Asunto(s)
Fibroblastos/citología , Fase G1/fisiología , Receptores Adrenérgicos alfa 1/metabolismo , Fase S/fisiología , Agonistas Adrenérgicos/farmacología , Animales , Recuento de Células , Proteínas de Ciclo Celular/metabolismo , División Celular/fisiología , Inhibidor p27 de las Quinasas Dependientes de la Ciclina , Quinasas Ciclina-Dependientes/metabolismo , Epinefrina/farmacología , Fibroblastos/fisiología , Citometría de Flujo , Fase G1/efectos de los fármacos , Humanos , Miocitos del Músculo Liso/citología , Miocitos del Músculo Liso/fisiología , Células PC12 , Ratas , Receptores Adrenérgicos alfa 1/genética , Fase S/efectos de los fármacos , Transcripción Genética/efectos de los fármacos , Proteínas Supresoras de Tumor/metabolismo
16.
Cardiovasc Res ; 60(3): 598-607, 2003 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-14659805

RESUMEN

OBJECTIVE: alpha(1)-Adrenergic receptors (ARs) are known mediators of a positive inotropy in the heart, which may play even more important roles in heart disease. Due to a lack of sufficiently selective ligands, the contribution of each of the three alpha(1)-AR subtypes (alpha(1A), alpha(1B) and alpha(1D)) to cardiac function is not clearly defined. In this study, we used a systemically expressing mouse model that overexpresses the alpha(1B)-AR to define the role of this subtype in cardiac function. METHODS: We used the mouse Langendorff heart model to assess changes in contractility under basal and phenylephrine-induced conditions. RESULTS: We find that a 50% increase of the alpha(1B)-AR in the heart does not change basal cardiac parameters compared to age-matched normals (heart rate, +/-dP/dT and coronary flow). However, the inotropic response to phenylephrine is blunted. The same results were obtained in isolated adult myocytes. The difference in inotropy could be blocked by the selective alpha(1A)-AR antagonist, 5-methylurapidil, which correlated with decreases in alpha(1A)-AR density, suggesting that the alpha(1B)-AR had caused a compensatory downregulation of the alpha(1A)-AR. CONCLUSIONS: These results suggest that the alpha(1B)-AR does not have a major role in the positive inotropic response in the mouse myocardium but may negatively modulate the response of the alpha(1A)-AR.


Asunto(s)
Corazón/fisiología , Contracción Miocárdica/efectos de los fármacos , Fenilefrina/farmacología , Receptores Adrenérgicos alfa 1/fisiología , Antagonistas Adrenérgicos alfa/farmacología , Animales , Femenino , Expresión Génica , Masculino , Ratones , Ratones Transgénicos , Miocitos Cardíacos/metabolismo , Perfusión , Piperazinas/farmacología , Receptores Adrenérgicos alfa 1/genética , Estimulación Química
17.
Brain ; 126(Pt 12): 2667-81, 2003 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-12937073

RESUMEN

The alpha1-adrenergic receptors (alpha1ARs) play an important role in mediating sympathetic neurotransmission in peripheral organ systems; however, central alpha1ARs are not well characterized. Additionally, due to the lack of sufficiently subtype-selective drugs or high avidity antibodies, the contribution of each alpha1AR subtype to various central functions is currently unclear. Transcription regulation through alpha1AR subtypes in the CNS is also unknown. Of interest, transgenic mice that systemically overexpress the alpha1BAR show central symptoms that include age-progressive impaired mobility, neurodegeneration and susceptibility to epileptic seizure. To investigate the molecular basis of this phenotype, oligonucleotide microarray studies of whole brains of various ages were carried out to compare gene expression profiles between transgenic and normal brains. The results indicated changes in expression of apoptotic, calcium regulatory, neurodegenerative and genes involved in neurotransmission. Defects in regulation of intracellular calcium are known to play a role in cell death; thus, these genes may provide clues as to the molecular basis of alpha1BAR-induced neurodegeneration. Epilepsy is a disorder that can be caused by an imbalance between excitatory (e.g. glutamate) and inhibitory (e.g. GABA) signals. Microarray analysis of transgenic brains showed increased N-methyl-d-aspartate (NMDA) receptors and decreased GABAA, which were confirmed with immunohistochemistry, western blot and radioligand binding studies. The alpha1BAR also co-localized with the glutamatergic distribution, suggesting a glutamate imbalance as a molecular rationale for the epileptic seizures.


Asunto(s)
Apoptosis/genética , Enfermedades Neurodegenerativas/genética , Receptores Adrenérgicos alfa 1/fisiología , Animales , Corteza Cerebral/metabolismo , Femenino , Expresión Génica , Perfilación de la Expresión Génica , Hipocampo/metabolismo , Masculino , Ratones , Ratones Transgénicos , Enfermedades Neurodegenerativas/metabolismo , Enfermedades Neurodegenerativas/patología , Análisis de Secuencia por Matrices de Oligonucleótidos , Receptores Adrenérgicos alfa 1/genética , Receptores de GABA-A/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo
18.
Mol Pharmacol ; 63(5): 1104-16, 2003 May.
Artículo en Inglés | MEDLINE | ID: mdl-12695539

RESUMEN

Alpha(1)-adrenoceptor subtypes (alpha(1A)-, alpha(1B)-, alpha(1D)-) are known to couple to similar signaling pathways, although differences among the subtypes do exist. As a more sensitive assay, we used oligonucleotide microarrays to identify gene expression changes in Rat-1 fibroblasts stably expressing each individual subtype. We report the gene expressions that change by at least a factor of 2 or more. Gene expression profiles significantly changed equally among all three subtypes, despite the unequal efficacy of the inositol phosphate response. Gene expressions were clustered into cytokines/growth factors, transcription factors, enzymes, and extracellular matrix proteins. There were also a number of individual subtype-specific changes in gene expression, suggesting a link to independent pathways. In addition, all three alpha(1)-AR subtypes robustly stimulated the transcription of the prohypertrophic cytokine interleukin (IL)-6, but differentially altered members of the IL-6 signaling pathway (gp-130 and STAT3). This was confirmed by measurement of secreted IL-6, activated STAT3, and gp-130 levels. Activation of STAT3 Tyr705 phosphorylation by the alpha(1)-ARs was not through IL-6 activation but was synergistic with IL-6, suggesting direct effects. Interestingly, alpha(1B)-AR stimulation caused the dimerization-dependent phosphorylation of Tyr705 on STAT3 but did not activate the transcriptional-dependent phosphorylation of Ser727. The alpha(1B)-AR also constitutively down-regulated the protein levels of gp-130. These results suggest that the alpha(1B)-AR has differential effects on the phosphorylation status of the STAT3 pathway and may not be as prohypertrophic as the other two subtypes.


Asunto(s)
Antígenos CD/metabolismo , Proteínas de Unión al ADN/metabolismo , Interleucina-6/metabolismo , Glicoproteínas de Membrana/metabolismo , Receptores Adrenérgicos alfa 1/genética , Transactivadores/metabolismo , Animales , Unión Competitiva , Northern Blotting , Células Cultivadas , Receptor gp130 de Citocinas , Epinefrina/farmacología , Fibroblastos/metabolismo , Expresión Génica , Perfilación de la Expresión Génica , Humanos , Fosfatos de Inositol/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Fosforilación , Ratas , Receptores Adrenérgicos alfa 1/clasificación , Factor de Transcripción STAT3 , Serina/metabolismo , Transducción de Señal/fisiología , Tritio
19.
J Pharmacol Exp Ther ; 303(1): 247-56, 2002 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-12235258

RESUMEN

alpha(1a)-Adrenergic receptors (ARs) couple to phosphoinositide hydrolysis, adenylyl cyclase, and mitogen-activated protein kinase (MAPK) pathways. However, the interaction among these signaling pathways in activating extracellular signal-regulated kinase 1/2 (ERK1/2) is not well understood. We investigated the coupling of alpha(1a)-ARs to ERK1/2 in Chinese hamster ovary (CHO)-K1 cells stably transfected with mouse alpha(1a)-ARs, as well as the interaction between ERK1/2 and norepinephrine-induced cAMP accumulation. alpha(1a)-AR activation by norepinephrine increased the cytosolic Ca(2+) concentration and phosphorylated ERK1/2 in a time- and concentration-dependent manner. ERK1/2 phosphorylation was blocked by the MAPK kinase 1/2 inhibitor 2'-amino-3'-methoxyflavone (PD 98059) and the alpha(1)-AR antagonist prazosin. A transient elevation in intracellular Ca(2+) was required for the phosphorylation of ERK1/2; however, activation of protein kinase C did not seem to be required for ERK1/2 phosphorylation. Norepinephrine also stimulated cAMP accumulation in transfected CHO-K1 cells in a concentration-dependent manner via alpha(1a)-ARs, which was blocked by the Ca(2+) chelator 1,2-bis(2-aminophenoxy)ethane-N,N,N',N'-tetraacetic acid. Norepinephrine-induced ERK1/2 phosphorylation was inhibited by the adenylyl cyclase activator forskolin and was enhanced by the adenylyl cyclase inhibitor 9-(tetrahydro-2-furanyl)-9H-purine-6-amine (SQ 22536) and the protein kinase A inhibitor 4-cyano-3-methylisoquinoline. In conclusion, in transfected CHO-K1 cells, alpha(1a)-AR activation activates both phospholipase C and adenylyl cyclase-mediated signaling pathways. alpha(1a)-AR-mediated ERK1/2 phosphorylation was dependent on a rise in intracellular Ca(2+), and this pathway was reciprocally regulated by the concomitant activation of adenylyl cyclase, which inhibits ERK1/2 phosphorylation. Thus, alpha(1a)-AR stimulation of cAMP production may play an important role in regulating ERK1/2 phosphorylation in cell lines and native tissues.


Asunto(s)
Calcio/metabolismo , AMP Cíclico/fisiología , Sistema de Señalización de MAP Quinasas/fisiología , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Norepinefrina/farmacología , Receptores Adrenérgicos alfa 1/fisiología , Animales , Células CHO , Clonación Molecular , Cricetinae , Cinética , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Ratones , Proteína Quinasa 3 Activada por Mitógenos , Fosforilación , Receptores Adrenérgicos alfa 1/efectos de los fármacos , Receptores Adrenérgicos alfa 1/genética , Proteínas Recombinantes/metabolismo , Acetato de Tetradecanoilforbol/farmacología , Transfección
20.
J Pharmacol Exp Ther ; 303(1): 403-11, 2002 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-12235277

RESUMEN

The endogenous adrenergic agonists norepinephrine (NE) and epinephrine regulate vascular tone by stimulating alpha(1)-adrenergic receptors (ARs) on smooth muscle cells to cause contraction. In addition, alpha(1)-ARs also couple to growth factor pathways, through stimulation of mitogen-activated protein kinases (MAPKs). MAPKs are a family of serine-threonine kinases that include extracellular signal-regulated kinase (ERK) and a variety of other kinases that are able to activate transcription factors when stimulated. We examined alpha(1)-AR stimulation of contraction and ERK activation in the bovine inferior alveolar artery (BIAA), using in vitro contraction studies and Western blotting. Using antagonists selective for individual adrenergic receptor types, we found that only alpha(1)-ARs were coupled to ERK activation and contraction. NE stimulated contraction (EC(50) = 11 microM) and ERK activation (EC(50) = 21 microM) with similar potency. Using alpha(1)-AR subtype-selective antagonists, we identified the alpha(1)-AR subtypes coupled to each response. Affinity values for alpha(1)-AR subtype-selective antagonists were consistent with alpha(1A)-AR-mediated contraction. In contrast, simultaneous treatment with concentrations of these antagonists selective for each alpha(1)-AR subtype (alpha(1A)-, alpha(1B)-, and alpha(1D)-AR) was required to inhibit ERK activation, suggesting that all three alpha(1)-ARs activate ERK in BIAA. Transmural electrical stimulation of BIAA segments resulted in activation of ERK, which was inhibited by the alpha(1)-AR-selective antagonist BE 2254 (2-[[beta-(4-hydroxyphenyl)ethyl]aminomethyl]-1-tetralone). These data suggest that in an intact artery, NE released from sympathetic nerves stimulates alpha(1)-ARs to cause contraction and ERK activation, and that redundancy among subtypes exists for alpha(1)-AR activation of ERK.


Asunto(s)
Agonistas alfa-Adrenérgicos/farmacología , Antagonistas Adrenérgicos alfa/farmacología , Sistema de Señalización de MAP Quinasas/fisiología , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Contracción Muscular/fisiología , Alveolos Pulmonares/irrigación sanguínea , Arteria Pulmonar/fisiología , Receptores Adrenérgicos alfa 1/fisiología , Animales , Bovinos , Desipramina/farmacología , Activación Enzimática , Hidrocortisona/farmacología , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos , Contracción Muscular/efectos de los fármacos , Norepinefrina/farmacología , Receptores Adrenérgicos alfa 1/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...