Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 73
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Med Chem ; 62(23): 10816-10832, 2019 12 12.
Artículo en Inglés | MEDLINE | ID: mdl-31729873

RESUMEN

Retinoic acid receptor-related orphan receptor gamma-t (RORγt) is considered to be the master transcription factor for the development of Th17 cells that produce proinflammatory cytokines such as IL-17A. Overproportionate Th17 cell abundance is associated with the pathogenesis of many inflammatory conditions including psoriasis. In a high-throughput fluorescence resonance energy transfer (FRET) screen, we identified compound 1 as a hit with promising lipophilic efficiency (LipE). Using structure-based drug design based on a number of X-ray cocrystal structures, we morphed this hit class into potent imidazoles, exemplified by compound 3. To improve the poor absorption, distribution, metabolism, and excretion (ADME) properties of neutral imidazoles, we extended our ligands with carboxylic acid substituents toward a polar, water-rich area of the protein. This highly lipophilicity-efficient modification ultimately led to the discovery of compound 14, a potent and selective inhibitor of RORγt with good ADME properties and excellent in vivo pharmacokinetics. This compound showed good efficacy in an in vivo delayed-type hypersensitivity pharmacology model in rats.


Asunto(s)
Hipersensibilidad Tardía/tratamiento farmacológico , Imidazoles/farmacología , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/antagonistas & inhibidores , Administración Oral , Animales , Relación Dosis-Respuesta a Droga , Diseño de Fármacos , Femenino , Transferencia Resonante de Energía de Fluorescencia , Semivida , Imidazoles/química , Imidazoles/farmacocinética , Masculino , Modelos Moleculares , Estructura Molecular , Ratas
2.
FEBS J ; 285(16): 3097-3113, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29971953

RESUMEN

A large body of data demonstrates that interferon regulatory factor 5 (IRF5) and nuclear factor kappa B (NF-κB) are the two major transcription factors in classically activated macrophages responsible for the transcriptional control of proinflammatory genes. Although recent evidence suggests that IRF5 interacts with certain members of the nuclear factor kappa B pathway, the extent of cooperation and its implications in disease are ambiguous. Since both pathways are known for their strong contributions in TLR8 signaling we used the human monocytic cell line THP-1.Dual, featuring gene reporters for NF-κB and IRFs, to simultaneously study the roles of IRF5 and the NF-κB subunit p65 in TLR8-mediated gene reporter activities. Furthermore, we profiled from these cells the proinflammatory cytokines involved in the differentiation of TH1 and TH17 cells. After ablation of IRF5 and/or p65 we activated the resultant cells with the TLR8 agonists R848 or the psoriasis-associated antimicrobial peptide LL-37 complexed with ssRNA and demonstrate that IRF5 deficiency drastically impairs the secretion of IL-1ß, IL-6, IL-12, IL-23 and TNFα. In contrast, the lack of p65 impaired only IL-6, IL-12, and IL-23 secretion. Furthermore, we discovered that upon TLR8 stimulation, IRF5 but not NF-κB signaling is essential to provide a cytokine milieu supporting TH1 responses. Additionally, we demonstrate that IRF5 and NF-κB cooperate to provide a cytokine milieu supporting TH17 responses. Therefore, the distinct role of IRF5 in the intricate signaling network downstream of TLR8 may open new treatment options interfering with but not disrupting NF-κB signaling in human diseases.


Asunto(s)
Inflamación/metabolismo , Factores Reguladores del Interferón/metabolismo , FN-kappa B/metabolismo , Células TH1/inmunología , Células Th17/inmunología , Péptidos Catiónicos Antimicrobianos/farmacología , Sistemas CRISPR-Cas , Línea Celular , Citocinas/metabolismo , Técnicas de Inactivación de Genes , Genes Reporteros , Humanos , Imidazoles/farmacología , Factores Reguladores del Interferón/genética , FN-kappa B/genética , Elementos de Respuesta , Transducción de Señal , Células TH1/efectos de los fármacos , Células TH1/metabolismo , Células Th17/efectos de los fármacos , Células Th17/metabolismo , Receptor Toll-Like 8/agonistas , Receptor Toll-Like 8/inmunología , Receptor Toll-Like 8/metabolismo , Catelicidinas
3.
J Med Chem ; 61(15): 6724-6735, 2018 08 09.
Artículo en Inglés | MEDLINE | ID: mdl-29990434

RESUMEN

The transcription factor RORγt is an attractive drug-target due to its role in the differentiation of IL-17 producing Th17 cells that play a critical role in the etiopathology of several autoimmune diseases. Identification of starting points for RORγt inverse agonists with good properties has been a challenge. We report the identification of a fragment hit and its conversion into a potent inverse agonist through fragment optimization, growing and merging efforts. Further analysis of the binding mode revealed that inverse agonism was achieved by an unusual mechanism. In contrast to other reported inverse agonists, there is no direct interaction or displacement of helix 12 observed in the crystal structure. Nevertheless, compound 9 proved to be efficacious in a delayed-type hypersensitivity (DTH) inflammation model in rats.


Asunto(s)
Descubrimiento de Drogas , Agonismo Inverso de Drogas , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/agonistas , Animales , Dominio Catalítico , Modelos Animales de Enfermedad , Femenino , Inflamación/metabolismo , Modelos Moleculares , Ratas
4.
PLoS One ; 12(11): e0188391, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29155882

RESUMEN

Retinoic acid receptor-related-orphan-receptor-C (RORγt) is the key transcription factor that is driving the differentiation of IL-17 producing T-helper 17 (Th17) cells that are implicated in the pathology of various autoimmune and inflammatory diseases. Based on the importance of RORγt in promoting Th17-driven pathology, there is considerable interest to develop low-molecular-weight compounds with the aim of inhibiting the transcriptional activity of this nuclear hormone receptor. In this article, we describe the in vitro and in vivo pharmacology of a potent and selective small-molecular-weight RORγt inverse agonist. The compound binds to the ligand binding domain (LBD) of RORγt leading to displacement of a co-activator peptide. We show for the first time that a RORγt inverse agonist down-regulates permissive histone H3 acetylation and methylation at the IL17A and IL23R promoter regions, thereby providing insight into the transcriptional inhibition of RORγt-dependent genes. Consistent with this, the compound effectively reduced IL-17A production by polarized human T-cells and γδT-cells and attenuated transcription of RORγt target genes. The inhibitor showed good in vivo efficacy in an antigen-induced arthritis model in rats and reduced the frequencies of IL-17A producing cells in ex vivo recall assays. In summary, we demonstrate that inhibiting RORγt by a low-molecular-weight inhibitor results in efficient and selective blockade of the pro-inflammatory Th17/IL-17A pathway making it an attractive target for Th17-mediated disorders.


Asunto(s)
Artritis Experimental/tratamiento farmacológico , Imidazoles/farmacología , Interleucina-17/antagonistas & inhibidores , Linfocitos Intraepiteliales/efectos de los fármacos , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/antagonistas & inhibidores , Piridinas/farmacología , Pirimidinas/farmacología , Células Th17/efectos de los fármacos , Animales , Artritis Experimental/genética , Artritis Experimental/inmunología , Artritis Experimental/patología , Línea Celular Tumoral , Femenino , Regulación de la Expresión Génica , Células HEK293 , Humanos , Imidazoles/síntesis química , Interleucina-17/genética , Interleucina-17/inmunología , Linfocitos Intraepiteliales/inmunología , Linfocitos Intraepiteliales/patología , Cinética , Masculino , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/genética , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/inmunología , Regiones Promotoras Genéticas , Unión Proteica , Piridinas/síntesis química , Pirimidinas/síntesis química , Ratas , Ratas Endogámicas Lew , Receptores de Antígenos de Linfocitos T gamma-delta/genética , Receptores de Antígenos de Linfocitos T gamma-delta/inmunología , Receptores de Interleucina/genética , Receptores de Interleucina/inmunología , Transducción de Señal , Células Th17/inmunología , Células Th17/patología
5.
Sci Rep ; 7(1): 13591, 2017 10 19.
Artículo en Inglés | MEDLINE | ID: mdl-29051536

RESUMEN

Leukotriene A4 Hydrolase (LTA4H) is a bifunctional zinc metalloenzyme that comprises both epoxide hydrolase and aminopeptidase activity, exerted by two overlapping catalytic sites. The epoxide hydrolase function of the enzyme catalyzes the biosynthesis of the pro-inflammatory lipid mediator leukotriene (LT) B4. Recent literature suggests that the aminopeptidase function of LTA4H is responsible for degradation of the tripeptide Pro-Gly-Pro (PGP) for which neutrophil chemotactic activity has been postulated. It has been speculated that the design of epoxide hydrolase selective LTA4H inhibitors that spare the aminopeptidase pocket may therefore lead to more efficacious anti-inflammatory drugs. In this study, we conducted a high throughput screen (HTS) for LTA4H inhibitors and attempted to rationally design compounds that would spare the PGP degrading function. While we were able to identify compounds with preference for the epoxide hydrolase function, absolute selectivity was not achievable for highly potent compounds. In order to assess the relevance of designing such aminopeptidase-sparing LTA4H inhibitors, we studied the role of PGP in inducing inflammation in different settings in wild type and LTA4H deficient (LTA4H KO) animals but could not confirm its chemotactic potential.  Attempting to design highly potent epoxide hydrolase selective LTA4H inhibitors, therefore seems to be neither feasible nor relevant.


Asunto(s)
Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Epóxido Hidrolasas/antagonistas & inhibidores , Epóxido Hidrolasas/química , Oligopéptidos/metabolismo , Prolina/análogos & derivados , Aminopeptidasas/metabolismo , Animales , Antiinflamatorios no Esteroideos/química , Antiinflamatorios no Esteroideos/farmacología , Dominio Catalítico , Cristalografía por Rayos X , Diseño de Fármacos , Epóxido Hidrolasas/metabolismo , Ensayos Analíticos de Alto Rendimiento/métodos , Humanos , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Neutrófilos/metabolismo , Neutrófilos/patología , Neumonía/metabolismo , Neumonía/patología , Prolina/metabolismo , Relación Estructura-Actividad
6.
JCI Insight ; 2(5): e91127, 2017 03 09.
Artículo en Inglés | MEDLINE | ID: mdl-28289717

RESUMEN

Retinoic-acid-orphan-receptor-C (RORC) is a master regulator of Th17 cells, which are pathogenic in several autoimmune diseases. Genetic Rorc deficiency in mice, while preventing autoimmunity, causes early lethality due to metastatic thymic T cell lymphomas. We sought to determine whether pharmacological RORC inhibition could be an effective and safe therapy for autoimmune diseases by evaluating its effects on Th17 cell functions and intrathymic T cell development. RORC inhibitors effectively inhibited Th17 differentiation and IL-17A production, and delayed-type hypersensitivity reactions. In vitro, RORC inhibitors induced apoptosis, as well as Bcl2l1 and BCL2L1 mRNA downregulation, in mouse and nonhuman primate thymocytes, respectively. Chronic, 13-week RORC inhibitor treatment in rats caused progressive thymic alterations in all analyzed rats similar to those in Rorc-deficient mice prior to T cell lymphoma development. One rat developed thymic cortical hyperplasia with preneoplastic features, including increased mitosis and reduced IKAROS expression, albeit without skewed T cell clonality. In summary, pharmacological inhibition of RORC not only blocks Th17 cell development and related cytokine production, but also recapitulates thymic aberrations seen in Rorc-deficient mice. While RORC inhibition may offer an effective therapeutic principle for Th17-mediated diseases, T cell lymphoma with chronic therapy remains an apparent risk.


Asunto(s)
Receptores de Ácido Retinoico/antagonistas & inhibidores , Células Th17/citología , Timo/patología , Animales , Regulación hacia Abajo , Femenino , Expresión Génica , Humanos , Células Jurkat , Masculino , Ratones , Ratones Endogámicos C57BL , Ratas , Ratas Endogámicas Lew , Ratas Sprague-Dawley , Receptores de Ácido Retinoico/genética , Células Th17/metabolismo
7.
ChemMedChem ; 11(24): 2640-2648, 2016 12 16.
Artículo en Inglés | MEDLINE | ID: mdl-27902884

RESUMEN

Retinoic-acid-related orphan receptor γt (RORγt) is a key transcription factor implicated in the production of pro-inflammatory Th17 cytokines, which drive a number of autoimmune diseases. Despite diverse chemical series having been reported, combining high potency with a good physicochemical profile has been a very challenging task in the RORγt inhibitor field. Based on available chemical structures and incorporating in-house knowledge, a new series of triazolo- and imidazopyridine RORγt inverse agonists was designed. In addition, replacement of the terminal cyclopentylamide metabolic soft spot by five-membered heterocycles was investigated. From our efforts, we identified an optimal 6,7,8-substituted imidazo[1,2-a]pyridine core system and a 5-tert-butyl-1,2,4-oxadiazole as cyclopentylamide replacement leading to compounds 10 ((S)-N-(8-((4-(cyclopentanecarbonyl)-3-methylpiperazin-1-yl)methyl)-7-methylimidazo[1,2-a]pyridin-6-yl)-2-methylpyrimidine-5-carboxamide) and 33 ((S)-N-(8-((4-(5-(tert-butyl)-1,2,4-oxadiazol-3-yl)-3-methylpiperazin-1-yl)methyl)-7-methylimidazo[1,2-a]pyridin-6-yl)-2-methylpyrimidine-5-carboxamide). Both derivatives showed good pharmacological potencies in biochemical and cell-based assays combined with excellent physicochemical properties, including low to medium plasma protein binding across species. Finally, 10 and 33 were shown to be active in a rodent pharmacokinetic/pharmacodynamic (PK/PD) model after oral gavage at 15 mg kg-1 , lowering IL-17 cytokine production in ex vivo antigen recall assays.


Asunto(s)
Agonismo Inverso de Drogas , Imidazoles , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/agonistas , Piridinas/síntesis química , Receptores de Ácido Retinoico/agonistas , Triazoles , Animales , Sitios de Unión , Células Cultivadas , Cristalografía por Rayos X , Humanos , Imidazoles/síntesis química , Imidazoles/química , Imidazoles/farmacología , Concentración 50 Inhibidora , Interleucina-17/sangre , Estructura Molecular , Unión Proteica/efectos de los fármacos , Piridinas/química , Piridinas/farmacología , Ratas , Triazoles/síntesis química , Triazoles/química , Triazoles/farmacología
8.
Circ Res ; 117(4): 376-87, 2015 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-26129975

RESUMEN

RATIONALE: Platelets are known to play a crucial role in hemostasis. Sphingosine kinases (Sphk) 1 and 2 catalyze the conversion of sphingosine to the bioactive metabolite sphingosine 1-phosphate (S1P). Although platelets are able to secrete S1P on activation, little is known about a potential intrinsic effect of S1P on platelet function. OBJECTIVE: To investigate the role of Sphk1- and Sphk2-derived S1P in the regulation of platelet function. METHODS AND RESULTS: We found a 100-fold reduction in intracellular S1P levels in platelets derived from Sphk2(-/-) mutants compared with Sphk1(-/-) or wild-type mice, as analyzed by mass spectrometry. Sphk2(-/-) platelets also failed to secrete S1P on stimulation. Blood from Sphk2-deficient mice showed decreased aggregation after protease-activated receptor 4-peptide and adenosine diphosphate stimulation in vitro, as assessed by whole blood impedance aggregometry. We revealed that S1P controls platelet aggregation via the sphingosine 1-phosphate receptor 1 through modulation of protease-activated receptor 4-peptide and adenosine diphosphate-induced platelet activation. Finally, we show by intravital microscopy that defective platelet aggregation in Sphk2-deficient mice translates into reduced arterial thrombus stability in vivo. CONCLUSIONS: We demonstrate that Sphk2 is the major Sphk isoform responsible for the generation of S1P in platelets and plays a pivotal intrinsic role in the control of platelet activation. Correspondingly, Sphk2-deficient mice are protected from arterial thrombosis after vascular injury, but have normal bleeding times. Targeting this pathway could therefore present a new therapeutic strategy to prevent thrombosis.


Asunto(s)
Plaquetas/enzimología , Lisofosfolípidos/sangre , Fosfotransferasas (Aceptor de Grupo Alcohol)/sangre , Agregación Plaquetaria , Esfingosina/análogos & derivados , Animales , Ácido Araquidónico/sangre , Coagulación Sanguínea , Pruebas de Coagulación Sanguínea , Traumatismos de las Arterias Carótidas/sangre , Traumatismos de las Arterias Carótidas/enzimología , Modelos Animales de Enfermedad , Eritrocitos/enzimología , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Fosfotransferasas (Aceptor de Grupo Alcohol)/deficiencia , Fosfotransferasas (Aceptor de Grupo Alcohol)/genética , Adhesividad Plaquetaria , Pruebas de Función Plaquetaria , Receptores de Lisoesfingolípidos/sangre , Transducción de Señal , Esfingosina/sangre , Receptores de Esfingosina-1-Fosfato , Trombosis/sangre , Trombosis/enzimología , Trombosis/prevención & control , Tromboxano A2/sangre , Lesiones del Sistema Vascular/sangre , Lesiones del Sistema Vascular/enzimología
9.
Toxicol Pathol ; 43(5): 694-703, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25630683

RESUMEN

Sphingosine-1-phosphate (S1P) lyase is considered as a drug target in autoimmune diseases based on the protective effect of reducing activity of the enzyme in animal models of inflammation. Since S1P lyase deficiency in mice causes a severe, lethal phenotype, it was of interest to investigate any pathological alterations associated with only partially reduced activity of S1P lyase as may be encountered upon pharmacological inhibition. Both genetic reduction of S1P lyase activity in mice and inhibition of S1P lyase with a low-molecular-weight compound in rats consistently resulted in podocyte-based kidney toxicity, which is the most severe finding. In addition, skin irritation and platelet activation were observed in both instances. The similarity of the findings in both the genetic model and the pharmacological study supports the value of analyzing inducible partially target-deficient mice for safety assessment. If the findings described in rodents translate to humans, target-related toxicity, particularly podocyte dysfunction, may limit chronic systemic treatment of autoimmune diseases with S1P lyase inhibitors. Furthermore, partial deficiency or inhibition of S1P lyase appears to provide an in vivo rodent model to enable studies on the mechanism of podocyte dysfunction.


Asunto(s)
Aldehído-Liasas/antagonistas & inhibidores , Aldehído-Liasas/metabolismo , Activación Plaquetaria/fisiología , Podocitos/enzimología , Proteinuria/enzimología , Aldehído-Liasas/genética , Animales , Femenino , Riñón/enzimología , Riñón/patología , Masculino , Ratones , Proteinuria/sangre , Ratas , Piel/enzimología , Piel/patología , Tamoxifeno/farmacología
10.
J Med Chem ; 57(12): 5074-84, 2014 Jun 26.
Artículo en Inglés | MEDLINE | ID: mdl-24809814

RESUMEN

Sphingosine 1-phosphate (S1P) lyase has recently been implicated as a therapeutic target for the treatment of multiple sclerosis (MS), based on studies in a genetic mouse model. Potent active site directed inhibitors of the enzyme are not known so far. Here we describe the discovery of (4-benzylphthalazin-1-yl)-2-methylpiperazin-1-yl]nicotinonitrile 5 in a high-throughput screen using a biochemical assay, and its further optimization. This class of compounds was found to inhibit catalytic activity of S1PL by binding to the active site of the enzyme, as seen in the cocrystal structure of derivative 31 with the homodimeric human S1P lyase. 31 induces profound reduction of peripheral T cell numbers after oral dosage and confers pronounced protection in a rat model of multiple sclerosis. In conclusion, this novel class of direct S1P lyase inhibitors provides excellent tools to further explore the therapeutic potential of T cell-targeted therapies in multiple sclerosis and other autoimmune and inflammatory diseases.


Asunto(s)
Aldehído-Liasas/antagonistas & inhibidores , Esclerosis Múltiple/tratamiento farmacológico , Ftalazinas/química , Piridinas/química , Administración Oral , Aldehído-Liasas/química , Aldehído-Liasas/genética , Animales , Dominio Catalítico , Células Cultivadas , Cristalografía por Rayos X , Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Encefalomielitis Autoinmune Experimental/enzimología , Femenino , Humanos , Masculino , Modelos Moleculares , Esclerosis Múltiple/enzimología , Mutación , Ftalazinas/farmacocinética , Ftalazinas/farmacología , Conformación Proteica , Piridinas/farmacocinética , Piridinas/farmacología , Ratas Sprague-Dawley , Estereoisomerismo , Relación Estructura-Actividad
11.
Biochim Biophys Acta ; 1841(5): 745-58, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24239768

RESUMEN

Multiple Sclerosis (MS) is a chronic autoimmune disorder affecting the central nervous system (CNS) through demyelination and neurodegeneration. Until recently, major therapeutic treatments have relied on agents requiring injection delivery. In September 2010, fingolimod/FTY720 (Gilenya, Novartis) was approved as the first oral treatment for relapsing forms of MS. Fingolimod causes down-modulation of S1P1 receptors on lymphocytes which prevents the invasion of autoaggressive T cells into the CNS. In astrocytes, down-modulation of S1P1 by the drug reduces astrogliosis, a hallmark of MS, thereby allowing restoration of productive astrocyte communication with other neural cells and the blood brain barrier. Animal data further suggest that the drug directly supports the recovery of nerve conduction and remyelination. In human MS, such mechanisms may explain the significant decrease in the number of inflammatory markers on brain magnetic resonance imaging in recent clinical trials, and the reduction of brain atrophy by the drug. Fingolimod binds to 4 of the 5 known S1P receptor subtypes, and significant efforts were made over the past 5 years to develop next generation S1P receptor modulators and determine the minimal receptor selectivity needed for maximal therapeutic efficacy in MS patients. Other approaches considered were competitive antagonists of the S1P1 receptor, inhibitors of the S1P lyase to prevent S1P degradation, and anti-S1P antibodies. Below we discuss the current status of the field, and the functional properties of the most advanced compounds. This article is part of a Special Issue entitled New Frontiers in Sphingolipid Biology.


Asunto(s)
Medicina Clínica , Inmunosupresores/farmacología , Lisofosfolípidos/metabolismo , Glicoles de Propileno/farmacología , Proyectos de Investigación , Transducción de Señal/efectos de los fármacos , Esfingosina/análogos & derivados , Investigación Biomédica , Clorhidrato de Fingolimod , Humanos , Receptores de Lisoesfingolípidos/metabolismo , Esfingosina/metabolismo , Esfingosina/farmacología
12.
Chem Biol ; 20(7): 912-21, 2013 Jul 25.
Artículo en Inglés | MEDLINE | ID: mdl-23890009

RESUMEN

Toll-like receptor (TLR) signaling is a key component of innate immunity. Aberrant TLR activation leads to immune disorders via dysregulation of cytokine production, such as IL-12/IL-23. Herein, we identify and characterize PIKfyve, a lipid kinase, as a critical player in TLR signaling using apilimod as an affinity tool. Apilimod is a potent small molecular inhibitor of IL-12/IL-23 with an unknown target and has been evaluated in clinical trials for patients with Crohn's disease or rheumatoid arthritis. Using a chemical genetic approach, we show that it binds to PIKfyve and blocks its phosphotransferase activity, leading to selective inhibition of IL-12/IL-23p40. Pharmacological or genetic inactivation of PIKfyve is necessary and sufficient for suppression of IL-12/IL-23p40 expression. Thus, we have uncovered a phosphoinositide-mediated regulatory mechanism that controls TLR signaling.


Asunto(s)
Interleucina-12/antagonistas & inhibidores , Interleucina-23/antagonistas & inhibidores , Morfolinas/farmacología , Fosfatidilinositol 3-Quinasas/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3 , Transducción de Señal/efectos de los fármacos , Receptores Toll-Like/metabolismo , Triazinas/farmacología , Animales , Línea Celular , Citocinas/metabolismo , Inhibidores Enzimáticos/metabolismo , Inhibidores Enzimáticos/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Hidrazonas , Ratones , Morfolinas/metabolismo , Unión Proteica , Pirimidinas , Especificidad por Sustrato , Triazinas/metabolismo
13.
PLoS One ; 8(3): e59630, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23544080

RESUMEN

BACKGROUND: Sphingosine-1-phosphate (S1P) regulates the egress of T cells from lymphoid organs; levels of S1P in the tissues are controlled by S1P lyase (Sgpl1). Hence, Sgpl1 offers a target to block T cell-dependent inflammatory processes. However, the involvement of Sgpl1 in models of disease has not been fully elucidated yet, since Sgpl1 KO mice have a short life-span. METHODOLOGY: We generated inducible Sgpl1 KO mice featuring partial reduction of Sgpl1 activity and analyzed them with respect to sphingolipid levels, T-cell distribution, and response in models of inflammation. PRINCIPAL FINDINGS: The partially Sgpl1 deficient mice are viable but feature profound reduction of peripheral T cells, similar to the constitutive KO mice. While thymic T cell development in these mice appears normal, mature T cells are retained in thymus and lymph nodes, leading to reduced T cell numbers in spleen and blood, with a skewing towards increased proportions of memory T cells and T regulatory cells. The therapeutic relevance of Sgpl1 is demonstrated by the fact that the inducible KO mice are protected in experimental autoimmune encephalomyelitis (EAE). T cell immigration into the CNS was found to be profoundly reduced. Since S1P levels in the brain of the animals are unchanged, we conclude that protection in EAE is due to the peripheral effect on T cells, leading to reduced CNS immigration, rather than on local effects in the CNS. SIGNIFICANCE: The data suggest Sgpl1 as a novel therapeutic target for the treatment of multiple sclerosis.


Asunto(s)
Aldehído-Liasas/deficiencia , Encefalomielitis Autoinmune Experimental/enzimología , Encefalomielitis Autoinmune Experimental/prevención & control , Aldehído-Liasas/metabolismo , Animales , Encéfalo/metabolismo , Linfocitos T CD4-Positivos/inmunología , Encefalomielitis Autoinmune Experimental/sangre , Encefalomielitis Autoinmune Experimental/complicaciones , Factores de Transcripción Forkhead/metabolismo , Hipersensibilidad Tardía/sangre , Hipersensibilidad Tardía/complicaciones , Hipersensibilidad Tardía/inmunología , Hipersensibilidad Tardía/patología , Memoria Inmunológica/inmunología , Integrasas/metabolismo , Ganglios Linfáticos/inmunología , Ganglios Linfáticos/patología , Recuento de Linfocitos , Ratones , Ratones Noqueados , Ovinos , Esfingolípidos/metabolismo , Bazo/inmunología , Bazo/patología , Análisis de Supervivencia , Timo/inmunología , Timo/patología
15.
Biochem Biophys Res Commun ; 433(3): 345-8, 2013 Apr 12.
Artículo en Inglés | MEDLINE | ID: mdl-23499842

RESUMEN

Inhibitors of the sphingosine-1-phosphate (S1P) degrading enzyme S1P lyase (SPL) may be useful in the therapy of inflammatory diseases by preventing lymphocyte recruitment to diseased tissues. Here we describe a cellular assay for such inhibitors, which takes advantage of the observation that a fraction of the intracellular S1P accumulated in the presence of SPL inhibitors is secreted into the medium of cultured cells. The secreted S1P is then quantified using an S1P-sensitive reporter cell line. In the routine assay protocol, human HEK293T cells are treated with SPL inhibitors in the presence of phosphatase inhibitors and sphingosine; while the phosphatase inhibitors are included to prevent the degradation of S1P secreted from the cells, sphingosine is added as source for intracellular S1P that is prone to SPL degradation. The secreted S1P in the supernatant of the cell cultures is then quantified by measuring calcium flux induced in CHO-K1 cells expressing the human S1P3 receptor. Using this method SPL inhibitors were shown to induce a concentration-dependent increase of extracellular S1P under the conditions used; thus, the assay allows for the ranking of SPL inhibitors according to their potency on living cells.


Asunto(s)
Aldehído-Liasas/antagonistas & inhibidores , Bioensayo , Calcio/metabolismo , Inhibidores Enzimáticos/análisis , Lisofosfolípidos/metabolismo , Esfingosina/análogos & derivados , Aldehído-Liasas/metabolismo , Animales , Células CHO , Calcio/análisis , Cricetinae , Medios de Cultivo/química , Citoplasma/química , Inhibidores Enzimáticos/farmacología , Células HEK293 , Humanos , Cinética , Lisofosfolípidos/análisis , Fosfoproteínas Fosfatasas/antagonistas & inhibidores , Fosfoproteínas Fosfatasas/metabolismo , Receptores de Lisoesfingolípidos/metabolismo , Transducción de Señal/efectos de los fármacos , Esfingosina/análisis , Esfingosina/metabolismo , Esfingosina/farmacología
16.
Anal Biochem ; 434(2): 247-53, 2013 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-23246729

RESUMEN

Sphingosine-1-phosphate (S1P) lyase represents a target for therapeutic intervention in immune regulation. Inhibitors of the lyase can be identified by established biochemical assays, but a cellular test system for such inhibitors has not been described so far. We found that silencing or inhibition of S1P lyase with short interfering RNA (siRNA) or active site-directed inhibitors in cultured mammalian cells does not cause a relevant increase of S1P in the cells as measured by liquid chromatography-tandem mass spectrometry (LC-MS/MS). However, the addition of sphingosine to cultures of cell lines or primary cells provides a source of intracellular S1P that is susceptible to degradation by the lyase and, hence, increases on inhibition or silencing of the enzyme. The assay was optimized with respect to sphingosine concentration, incubation time, and cell density and was established for routine use with HEK293 cells. The assay was found to be suitable for the testing of novel active site-directed S1P lyase inhibitors, providing important information on their relative potency in intact cells.


Asunto(s)
Bioensayo , Inhibidores Enzimáticos/análisis , Lisofosfolípidos/antagonistas & inhibidores , Esfingosina/análogos & derivados , Animales , Dominio Catalítico , Línea Celular Tumoral , Activación Enzimática/efectos de los fármacos , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Células HEK293 , Humanos , Lisofosfolípidos/genética , Ratones , Ratones Noqueados , Estructura Molecular , Esfingosina/antagonistas & inhibidores , Esfingosina/genética
17.
J Exp Med ; 209(12): 2165-81, 2012 Nov 19.
Artículo en Inglés | MEDLINE | ID: mdl-23148237

RESUMEN

Millions of platelets are produced each hour by bone marrow (BM) megakaryocytes (MKs). MKs extend transendothelial proplatelet (PP) extensions into BM sinusoids and shed new platelets into the blood. The mechanisms that control platelet generation remain incompletely understood. Using conditional mutants and intravital multiphoton microscopy, we show here that the lipid mediator sphingosine 1-phosphate (S1P) serves as a critical directional cue guiding the elongation of megakaryocytic PP extensions from the interstitium into BM sinusoids and triggering the subsequent shedding of PPs into the blood. Correspondingly, mice lacking the S1P receptor S1pr1 develop severe thrombocytopenia caused by both formation of aberrant extravascular PPs and defective intravascular PP shedding. In contrast, activation of S1pr1 signaling leads to the prompt release of new platelets into the circulating blood. Collectively, our findings uncover a novel function of the S1P-S1pr1 axis as master regulator of efficient thrombopoiesis and might raise new therapeutic options for patients with thrombocytopenia.


Asunto(s)
Lisofosfolípidos/metabolismo , Megacariocitos/fisiología , Receptores de Lisoesfingolípidos/fisiología , Transducción de Señal/fisiología , Esfingosina/análogos & derivados , Trombocitopenia/metabolismo , Trombopoyesis/fisiología , Animales , Plaquetas/fisiología , Western Blotting , Extensiones de la Superficie Celular/fisiología , Células Cultivadas , Citometría de Flujo , Ratones , Ratones Endogámicos C57BL , Microscopía Electrónica de Transmisión , Receptores de Lisoesfingolípidos/deficiencia , Receptores de Lisoesfingolípidos/metabolismo , Esfingosina/metabolismo , Receptores de Esfingosina-1-Fosfato , Estadísticas no Paramétricas , Trombopoyesis/genética
18.
ChemMedChem ; 6(4): 667-77, 2011 Apr 04.
Artículo en Inglés | MEDLINE | ID: mdl-21280229

RESUMEN

FTY720 (fingolimod, Gilenya®) is a sphingosine 1-phosphate (S1P) receptor modulator that shows significant therapeutic efficacy after oral administration to patients of multiple sclerosis. Because FTY720 does not contain any atom whose PET or SPECT radioisotope would have a half-life compatible with its pharmacokinetic properties, it cannot be used directly for imaging. Instead, we propose BZM055 as a surrogate tracer to study its pharmacokinetics and organ distribution in patients and, given that FTY720 accumulates in myelin sheaths, for myelin imaging. BZM055 (2 a, 2-iodo-FTY720) can be easily radiolabeled with ¹²³I (for SPECT) or ¹²4I (for PET). Not only does it closely mimic the pharmacokinetics and organ distribution of FTY720, but also its affinity, selectivity for S1P receptors, phosphorylation kinetics, and overall physicochemical properties. [¹²³I]BZM055 is currently under development for clinical imaging.


Asunto(s)
Encéfalo/metabolismo , Radioisótopos de Yodo , Vaina de Mielina/metabolismo , Tomografía de Emisión de Positrones/métodos , Tomografía Computarizada de Emisión de Fotón Único/métodos , Encéfalo/diagnóstico por imagen , Encéfalo/patología , Humanos , Radioisótopos de Yodo/química , Cinética , Esclerosis Múltiple/metabolismo , Esclerosis Múltiple/patología , Vaina de Mielina/diagnóstico por imagen , Vaina de Mielina/patología , Fosforilación , Radiografía , Receptores de Lisoesfingolípidos/metabolismo
19.
Nat Rev Drug Discov ; 9(11): 883-97, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21031003

RESUMEN

The discovery of fingolimod (FTY720/Gilenya; Novartis), an orally active immunomodulatory drug, has opened up new approaches to the treatment of multiple sclerosis, the most common inflammatory disorder of the central nervous system. Elucidation of the effects of fingolimod--mediated by the modulation of sphingosine 1-phosphate (S1P) receptors--has indicated that its therapeutic activity could be due to regulation of the migration of selected lymphocyte subsets into the central nervous system and direct effects on neural cells, particularly astrocytes. An improved understanding of the biology of S1P receptors has also been gained. This article describes the discovery and development of fingolimod, which was approved by the US Food and Drug Administration in September 2010 as a first-line treatment for relapsing forms of multiple sclerosis, thereby becoming the first oral disease-modifying therapy to be approved for multiple sclerosis in the United States.


Asunto(s)
Descubrimiento de Drogas/tendencias , Esclerosis Múltiple/tratamiento farmacológico , Glicoles de Propileno/administración & dosificación , Investigación/tendencias , Esfingosina/análogos & derivados , Administración Oral , Animales , Clorhidrato de Fingolimod , Humanos , Mediadores de Inflamación/administración & dosificación , Esclerosis Múltiple/patología , Esclerosis Múltiple Recurrente-Remitente/tratamiento farmacológico , Esclerosis Múltiple Recurrente-Remitente/patología , Esfingosina/administración & dosificación
20.
Lipids Health Dis ; 9: 1, 2010 Jan 06.
Artículo en Inglés | MEDLINE | ID: mdl-20053284

RESUMEN

Ceramide kinase (CERK) produces the bioactive lipid ceramide-1-phosphate (C1P) and is a key regulator of ceramide and dihydroceramide levels. It is likely that CERK and C1P play a role in inflammatory processes but the cells involved and the mechanisms used remain to be clarified. In particular, the impact of CERK on T-cell biology has not been studied so far. Here, we used Cerk-/- mice backcrossed with DO11.10/RAG1-/- mice to probe the effect of CERK ablation on T-cell activation. Levels of interleukin (IL)-2, IL-4, IL-5, IL-13, of tumor necrosis factor (TNF)-alpha, and of interferon (INF)-gamma were recorded following ovalbumin challenge in vivo and using ovalbumin-treated splenocytes ex- vivo. Absence of CERK led to a significant decrease in the production of IL-4, thus suggesting that CERK may polarize T cells towards the TH2 cell subtype. However, the importance of CERK to TH2 cell biology will have to be investigated further because in a model of asthma, which is TH2-cell driven, Cerk-/- mice responded like wild-type animals.


Asunto(s)
Interleucina-4/sangre , Ovalbúmina/metabolismo , Fosfotransferasas (Aceptor de Grupo Alcohol)/genética , Animales , Asma/genética , Asma/metabolismo , Líquido del Lavado Bronquioalveolar , Regulación Enzimológica de la Expresión Génica , Proteínas de Homeodominio/genética , Interferón gamma/metabolismo , Ratones , Ratones Transgénicos , Bazo/citología , Linfocitos T/citología , Células Th2/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA