Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Más filtros

Métodos Terapéuticos y Terapias MTCI
Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
J Bone Miner Res ; 34(12): 2264-2276, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31373726

RESUMEN

Spondyloarthritis (SpA) is a common rheumatic disease characterized by enthesis inflammation (enthesitis) and ectopic ossification (enthesophytes). The current pathogenesis model suggests that inflammation and mechanical stress are both strongly involved in SpA pathophysiology. We have previously observed that the levels of sphingosine 1-phosphate (S1P), a bone anabolic molecule, were particularly high in SpA patients' serum compared to healthy donors. Therefore, we wondered how this deregulation was related to SpA molecular mechanisms. Mouse primary osteoblasts, chondrocytes, and tenocytes were used as cell culture models. The sphingosine kinase 1 (Sphk1) gene expression and S1P secretion were significantly enhanced by cyclic stretch in osteoblasts and chondrocytes. Further, TNF-α and IL-17, cytokines implicated in enthesitis, increased Sphk1 mRNA in chondrocytes in an additive manner when combined to stretch. The immunochemistry on mouse ankles showed that sphingosine kinase 1 (SK1) was localized in some chondrocytes; the addition of a pro-inflammatory cocktail augmented Sphk1 expression in cultured ankles. Subsequently, fingolimod was used to block S1P metabolism in cell cultures. It inhibited S1P receptors (S1PRs) signaling and SK1 and SK2 activity in both osteoblasts and chondrocytes. Fingolimod also reduced S1PR-induced activation by SpA patients' synovial fluid (SF), demonstrating that the stimulation of chondrocytes by SFs from SpA patients involves S1P. In addition, when the osteogenic culture medium was supplemented with fingolimod, alkaline phosphatase activity, matrix mineralization, and bone formation markers were significantly reduced in osteoblasts and hypertrophic chondrocytes. Osteogenic differentiation was accompanied by an increase in S1prs mRNA, especially S1P1/3 , but their contribution to S1P-impact on mineralization seemed limited. Our results suggest that S1P might be overproduced in SpA enthesis in response to cytokines and mechanical stress, most likely by chondrocytes. Moreover, S1P could locally favor the abnormal ossification of the enthesis; therefore, blocking the S1P metabolic pathway could be a potential therapeutic approach for the treatment of SpA. © 2019 American Society for Bone and Mineral Research.


Asunto(s)
Citocinas/farmacología , Lisofosfolípidos/biosíntesis , Osteogénesis , Esfingosina/análogos & derivados , Espondiloartritis/patología , Espondiloartritis/fisiopatología , Estrés Mecánico , Adolescente , Adulto , Anciano , Animales , Calcificación Fisiológica/efectos de los fármacos , Células Cultivadas , Condrocitos/efectos de los fármacos , Condrocitos/metabolismo , Femenino , Clorhidrato de Fingolimod/farmacología , Humanos , Masculino , Redes y Vías Metabólicas/efectos de los fármacos , Ratones , Persona de Mediana Edad , Osteoblastos/efectos de los fármacos , Osteoblastos/metabolismo , Osteogénesis/efectos de los fármacos , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Receptores de Lisoesfingolípidos/metabolismo , Transducción de Señal , Esfingosina/biosíntesis , Líquido Sinovial/metabolismo , Tenocitos/efectos de los fármacos , Tenocitos/metabolismo , Regulación hacia Arriba/efectos de los fármacos , Adulto Joven
2.
J Cell Physiol ; 233(12): 9426-9436, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30063084

RESUMEN

Hypothalamic sphingosine-1-phosphate receptor 1 (S1PR1), the G protein-coupled receptor 1 of sphingosine-1-phosphate, has been described as a modulator in the control of energy homeostasis in rodents. However, this mechanism is still unclear. Here, we evaluate the role of interleukin 6 (IL-6) associated with acute physical exercise in the control of the hypothalamic S1PR1-signal transducer and activator of transcription 3 (STAT3) axis. Acute exercise session and an intracerebroventricular IL-6 injection increased S1PR1 protein content and STAT3 phosphorylation in the hypothalamus of lean and obese mice accompanied by a reduction in food consumption. Transcriptome analysis indicated a strong positive correlation between Il-6 and S1pr1 messenger RNA in several tissues of genetically diverse BXD mice strains and humans, including in the hypothalamus. Interestingly, exercise failed to stimulate the S1PR1-STAT3 axis in IL-6 knockout mice and the disruption of hypothalamic-specific IL-6 action blocked the anorexigenic effects of exercise. Taken together, our results indicate that physical exercise modulates the S1PR1 protein content in the hypothalamus, through the central action of IL-6.


Asunto(s)
Hipotálamo/metabolismo , Interleucina-6/metabolismo , Condicionamiento Físico Animal , Receptores de Lisoesfingolípidos/metabolismo , Factor de Transcripción STAT3/metabolismo , Transducción de Señal , Animales , Humanos , Inyecciones Intraventriculares , Interleucina-6/administración & dosificación , Interleucina-6/genética , Masculino , Ratones Endogámicos C57BL , Ratones Obesos , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores de Lisoesfingolípidos/genética , Receptores de Esfingosina-1-Fosfato
3.
Biol Pharm Bull ; 41(4): 592-596, 2018 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-29415945

RESUMEN

Psoriasis is a chronic inflammatory skin disease characterized by red, scaly and raised plaques. Thus far, T-cell infiltration is one of the most prominent pathogenic triggers, however, the exact molecular mechanisms underlying psoriasis have not been clearly established. Sphingolipid sphingosine-1-phosphate (S1P) is a lysophospholipid regulator modulating a variety of immune cell trafficking via interactions with its cognate receptors, S1P1-5. Activation of S1P signaling has recently emerged as a novel therapeutic avenue for psoriasis treatment. Here, we test a newly developed selective S1P1 modulator, Syl930, in four different psoriasis animal models. Our data reveals that oral administration of Syl930 can induce strong anti-proliferative and anti-inflammatory effects. Specifically, Syl930 decreases the pathological thickening of back skin induced by sodium lauryl sulfate (SLS), inhibits the proliferation of basal cells in a vaginal epithelium model and increases the granular layer scales in a mouse tail assay. Moreover, Syl930 can ameliorate the parakeratosis and acanthosis as well as improve granular layer composition and decrease the thickening of epidermis in a propranolol-induced guinea pig psoriasis model. Therefore, we demonstrate that Syl930 is a promising candidate for psoriasis therapy in clinical.


Asunto(s)
Antiinflamatorios no Esteroideos/uso terapéutico , Modelos Animales de Enfermedad , Oxazoles/uso terapéutico , Propanolaminas/uso terapéutico , Psoriasis/tratamiento farmacológico , Receptores de Lisoesfingolípidos/agonistas , Piel/efectos de los fármacos , Administración Oral , Animales , Animales no Consanguíneos , Antiinflamatorios no Esteroideos/administración & dosificación , Proliferación Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Evaluación Preclínica de Medicamentos , Femenino , Clorhidrato de Fingolimod/administración & dosificación , Clorhidrato de Fingolimod/uso terapéutico , Cobayas , Ratones , Índice Mitótico , Membrana Mucosa/efectos de los fármacos , Membrana Mucosa/inmunología , Membrana Mucosa/metabolismo , Membrana Mucosa/patología , Oxazoles/administración & dosificación , Propanolaminas/administración & dosificación , Psoriasis/inmunología , Psoriasis/metabolismo , Psoriasis/patología , Distribución Aleatoria , Receptores de Lisoesfingolípidos/metabolismo , Reproducibilidad de los Resultados , Piel/inmunología , Piel/metabolismo , Piel/patología , Organismos Libres de Patógenos Específicos , Receptores de Esfingosina-1-Fosfato , Vagina/efectos de los fármacos , Vagina/inmunología , Vagina/metabolismo , Vagina/patología
4.
Artículo en Inglés | MEDLINE | ID: mdl-29226621

RESUMEN

Sphingosine-1-phosphate receptor 1 (S1P1 ) modulators sequester circulating lymphocytes within lymph nodes, thereby preventing potentially pathogenic autoimmune cells from exiting into the blood stream and reaching inflamed tissues. S1P1 receptor modulation may thus offer potential to treat various autoimmune diseases. The first nonselective S1P1-5 receptor modulator FTY720/fingolimod/Gilenya® has successfully demonstrated clinical efficacy in relapsing forms of multiple sclerosis. However, cardiovascular, hepatic, and respiratory side-effects were reported and there is a need for novel S1P1 receptor modulators with better safety profiles. Here, we describe the discovery of cenerimod, a novel, potent and selective S1P1 receptor modulator with unique S1P1 receptor signaling properties and absence of broncho- and vasoconstrictor effects ex vivo and in vivo. Cenerimod dose-dependently lowered circulating lymphocyte counts in rats and mice after oral administration and effectively attenuated disease parameters in a mouse experimental autoimmune encephalitis (EAE) model. Cenerimod has potential as novel therapy with improved safety profile for autoimmune diseases with high unmet medical need.


Asunto(s)
Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Inmunosupresores/administración & dosificación , Linfocitos/efectos de los fármacos , Oxadiazoles/administración & dosificación , Piridinas/administración & dosificación , Receptores de Lisoesfingolípidos/agonistas , Administración Oral , Animales , Relación Dosis-Respuesta a Droga , Evaluación Preclínica de Medicamentos , Encefalomielitis Autoinmune Experimental/inmunología , Encefalomielitis Autoinmune Experimental/metabolismo , Células Endoteliales de la Vena Umbilical Humana , Humanos , Inmunosupresores/química , Inmunosupresores/farmacología , Recuento de Linfocitos , Ratones , Oxadiazoles/química , Oxadiazoles/farmacología , Piridinas/química , Piridinas/farmacología , Ratas , Receptores de Lisoesfingolípidos/metabolismo , Transducción de Señal/efectos de los fármacos
5.
Brain Res ; 1658: 51-59, 2017 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-28104351

RESUMEN

Recent studies indicate that Tau phosphorylation can be modulated by the compound FTY720-P, a global sphingosine-1-phosphate receptor (S1PR) agonist. The present work compared the effects of more selective S1PR agonists on Tau properties, using rat hippocampal slices as model system. Whereas Tau phosphorylation was not modified by the S1PR3 agonist CYM5541, Tau-Ser262 phosphorylation was significantly decreased by treatment with the S1PR1 agonist SEW2871. This effect appears to be quite restricted, as no changes in phosphorylation were elicited by the agonist on Tau-Ser199/202, Tau-Ser396 and Tau-Ser404 residues. In terms of molecular mechanisms, it is proposed that SEW2871-induced reduction of Tau-Ser262 phosphorylation depends on AMP-activated protein kinase alpha (AMPKα) inactivation via a pathway requiring AMPKα dephosphorylation at Thr172 by the protein phosphatase 2A (PP2A).


Asunto(s)
Fármacos del Sistema Nervioso Central/farmacología , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Oxadiazoles/farmacología , Receptores de Lisoesfingolípidos/agonistas , Tiofenos/farmacología , Proteínas tau/metabolismo , Proteínas Quinasas Activadas por AMP/metabolismo , Secuencia de Aminoácidos , Animales , Western Blotting , Evaluación Preclínica de Medicamentos , Isoxazoles/farmacología , Masculino , Estructura Molecular , Oxadiazoles/química , Fosforilación/efectos de los fármacos , Proteína Fosfatasa 2/metabolismo , Ratas Sprague-Dawley , Receptores de Lisoesfingolípidos/metabolismo , Receptores de Esfingosina-1-Fosfato , Tiofenos/química , Técnicas de Cultivo de Tejidos , Proteínas tau/genética
6.
J Lipid Res ; 58(2): 325-338, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27881715

RESUMEN

HDL normally transports about 50-70% of plasma sphingosine 1-phosphate (S1P), and the S1P in HDL reportedly mediates several HDL-associated biological effects and signaling pathways. The HDL receptor, SR-BI, as well as the cell surface receptors for S1P (S1PRs) may be involved partially and/or completely in these HDL-induced processes. Here we investigate the nature of the HDL-stimulated interaction between the HDL receptor, SR-BI, and S1PR1 using a protein-fragment complementation assay and confocal microscopy. In both primary rat aortic vascular smooth muscle cells and HEK293 cells, the S1P content in HDL particles increased intracellular calcium concentration, which was mediated by S1PR1. Mechanistic studies performed in HEK293 cells showed that incubation of cells with HDL led to an increase in the physical interaction between the SR-BI and S1PR1 receptors that mainly occurred on the plasma membrane. Model recombinant HDL (rHDL) particles formed in vitro with S1P incorporated into the particle initiated the internalization of S1PR1, whereas rHDL without supplemented S1P did not, suggesting that S1P transported in HDL can selectively activate S1PR1. In conclusion, these data suggest that S1P in HDL stimulates the transient interaction between SR-BI and S1PRs that can activate S1PRs and induce an elevation in intracellular calcium concentration.


Asunto(s)
Lipoproteínas HDL/metabolismo , Lisofosfolípidos/metabolismo , Receptores de Lisoesfingolípidos/metabolismo , Receptores Depuradores de Clase B/metabolismo , Esfingosina/análogos & derivados , Animales , Aorta/metabolismo , Transporte Biológico/genética , Calcio/metabolismo , Células HEK293 , Humanos , Lipoproteínas HDL/genética , Técnicas de Cultivo de Órganos , Ratas , Receptores de Lisoesfingolípidos/genética , Receptores Depuradores de Clase B/genética , Transducción de Señal , Esfingosina/metabolismo , Receptores de Esfingosina-1-Fosfato
7.
Zhonghua Nan Ke Xue ; 22(5): 393-400, 2016 May.
Artículo en Chino | MEDLINE | ID: mdl-27416661

RESUMEN

OBJECTIVE: To investigate the expressions of sphingosine-1-phosphate receptors 1-3 (S1P1- 3) in the corpus cavernosum of castrated male rats and its relationship with the NOS/NO/cGMP and RhoA/Rho kinase signaling pathways. METHODS: We equally randomized 18 eight-week-old healthy male SD rats into a sham-operation control, a castration, and a testosterone replacement (TR) group and harvested the bilateral testes and epididymides from the rats in the latter two groups, followed by 4 weeks of subcutaneous injection of testosterone propionate at 3 mg per kilogram of the body weight per day for those in the TR group and that of plant oil for those in the control and castration groups. At the age of 12 weeks, we measured the serum testosterone (T) level and maximum intracavernous pressure/mean arterial pressure (ICPmax/MAP) of the animals and determined the expressions of SlP1-3, eNOS, P-eNOS, ROCK1, and ROCK2 in the corpus cavernosum by Western blot and immunohistochemistry. RESULTS: The serum T level was significantly decreased in the rats of the castration group as compared with those of the control and TR groups ([0.41 ± 0.04] vs [16.01 ± 1.02] and [15.84 ± 1.32] nmol/L, P < 0.01), with no statistically significant difference between the latter two groups. The ICPmax/MAP at 0 V, 3 V, and 5 V electric stimulation was remarkably lower in the rats of the castration group (0.088 ± 0.014, 0.323 ± 0.014, and 0.432 ± 0.012) than in those of the control group (0.155 ± 0.011, 0.711 ± 0. 010, and 0.819 ± 0.024) and TR group (0.153 ± 0.012, 0.696 ± 0.017, and 0.763 ± 0.027) (P < 0.01), with no significant difference between the latter two groups. With GAPDH as internal control, the animals of the castration group showed markedly reduced expressions of S1P1 ([49.99 ± 3.39]%), eNOS ([46.82 ± 3.81]%) , and P-eNOS ([45.42 ± 4.35]%) in comparison with those in the control group ([72.57 ± 3.06], [89.76 ± 3.98], and [82.53 ± 8.92] and TR group ([71.77 ± 4.43], [87.19 ± 4.23], and [79.82 ± 7.38]%) (P < 0.01) , while the expressions of S1P2, S1P3, ROCK1, and ROCK2 were significantly upregulated in the castration group ([82.35 ± 4.13], [61.03 ± 5.14], [74.50 ± 4.02], and [69.83 ± 5.75]%) as compared with those in the control group ([41.67 ± 1.68], [31.66 ± 2.67], [35.69 ± 5.56], and [39.85 ± 7.17]%) and TR group ([42.80 ± 3.87], [32.25 ± 4.22], 38.06 ± 5.21], and [42.36 ± 4.44]%) (P < 0.01). CONCLUSION: Androgen deficiency induces significant reduction of ICPmax/ MAP in male rats, which is possibly associated with the decline of S1P1 in the corpus cavernosum, inhibition of the eNOS/NO/cGMP signaling pathway, increased expressions of S1P2 and S1P3, and activation of the RhoA/Rho kinase signaling pathway.


Asunto(s)
Orquiectomía , Pene/metabolismo , Receptores de Lisoesfingolípidos/metabolismo , Testosterona/farmacología , Animales , Masculino , Óxido Nítrico Sintasa de Tipo III/metabolismo , Distribución Aleatoria , Ratas , Ratas Sprague-Dawley , Testosterona/sangre , Quinasas Asociadas a rho/metabolismo
8.
Am J Physiol Gastrointest Liver Physiol ; 310(11): G1028-35, 2016 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-27079615

RESUMEN

SLC26A3 or Downregulated in adenoma (DRA) is the major Cl(-)/HCO3 (-) exchanger involved in electroneutral NaCl absorption in the mammalian intestine. Alterations in DRA function and expression have been implicated in diarrheal diseases associated with inflammation or infection. Therefore, agents that upregulate DRA activity may serve as potential antidiarrheals. In this regard, sphingosine-1-phosphate (S1P), a member of the bioactive sphingolipid family, has been shown to modulate various cellular processes including improvement of intestinal barrier function. However, the role of S1P in modulating intestinal chloride absorption by regulating DRA is not known. Therefore, the present studies were designed to examine the direct effects of S1P on apical Cl(-)/HCO3 (-) exchange activity and DRA expression. S1P significantly increased Cl(-)/HCO3 (-) exchange activity and also significantly increased DRA mRNA and protein expression. Increased DRA mRNA by S1P was accompanied by enhanced DRA promoter activity, indicating involvement of transcriptional mechanisms. The specific S1P receptor subtype-2 (S1PR2) antagonist JTE-013 blocked the stimulatory effects of S1P on DRA promoter activity, indicating the involvement of S1PR2 S1P-mediated increase in DRA promoter activity involved PI3K/Akt pathway. Progressive deletions of the DRA promoter indicated that the putative S1P-responsive elements are present in the -790/-398 region of the DRA promoter. Furthermore, results obtained from electrophoretic mobility shift assay showed that S1P stimulated DRA promoter activity via increased binding of Ying-Yang1 (YY1) in the S1P-responsive region. In conclusion, transcriptional modulation of DRA expression and function in response to S1P through a PI3/Akt pathway represents a novel role of S1P as a potential proabsorptive agent.


Asunto(s)
Antiportadores de Cloruro-Bicarbonato/metabolismo , Lisofosfolípidos/farmacología , Regiones Promotoras Genéticas , Esfingosina/análogos & derivados , Bicarbonatos/metabolismo , Células CACO-2 , Antiportadores de Cloruro-Bicarbonato/genética , Cloruros/metabolismo , Humanos , Transporte Iónico/efectos de los fármacos , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores de Lisoesfingolípidos/metabolismo , Esfingosina/farmacología , Transportadores de Sulfato , Factor de Transcripción YY1/metabolismo
9.
Am J Clin Dermatol ; 17(3): 191-200, 2016 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-26923915

RESUMEN

Several classes of new oral therapy are in use or in development for the treatment of psoriasis. Despite the high efficacy of biologics, new oral therapies remain important as patients generally prefer this mode of administration and they offer an alternative risk-benefit profile. In this review, we discuss the novel modes of action of these drugs, including modulation of cellular pathways involving diverse targets such as Janus kinase, phosphodiesterase 4, sphingosine 1-phosphate, A3 adenosine receptor and rho-associated kinase 2. We review the available evidence around licensed drugs (apremilast) and drugs that are advanced (tofacitinib) or early (ponesimod, baricitinib, peficitinib, INCB039110, CF101, KD025) in the development pipeline. The key limitations of these oral therapies are their modest efficacy profile (apremilast, ponesimod) and the limitations of their safety profile (tofacitinib, ponesimod), while the evidence for the early pipeline drugs are at phase II level only. Potential niches of current unmet needs include apremilast for patients with concomitant psoriatic arthritis, as combination treatments with biologic therapies, and/or for patients in whom multiple biologic therapies have failed due to immunogenicity and secondary inefficacy. The present knowledge gap regarding these novel drugs includes the need for longer clinical trials or observational studies to evaluate safety, and randomised phase III trials for the early pipeline drugs. We conclude that further research and data are necessary to conclusively establish the role of these agents in the current psoriasis treatment paradigm.


Asunto(s)
Artritis Psoriásica/tratamiento farmacológico , Inhibidores de Fosfodiesterasa 4/uso terapéutico , Piperidinas/uso terapéutico , Inhibidores de Proteínas Quinasas/uso terapéutico , Psoriasis/tratamiento farmacológico , Pirimidinas/uso terapéutico , Pirroles/uso terapéutico , Talidomida/análogos & derivados , Tiazoles/uso terapéutico , Adamantano/administración & dosificación , Adamantano/efectos adversos , Adamantano/análogos & derivados , Adamantano/uso terapéutico , Adenosina/administración & dosificación , Adenosina/efectos adversos , Adenosina/análogos & derivados , Adenosina/uso terapéutico , Antagonistas del Receptor de Adenosina A3/administración & dosificación , Antagonistas del Receptor de Adenosina A3/efectos adversos , Antagonistas del Receptor de Adenosina A3/uso terapéutico , Administración Oral , Azetidinas/administración & dosificación , Azetidinas/efectos adversos , Azetidinas/uso terapéutico , Factores Biológicos/uso terapéutico , Terapia Biológica , Ensayos Clínicos como Asunto , Humanos , Ácidos Isonicotínicos/administración & dosificación , Ácidos Isonicotínicos/efectos adversos , Ácidos Isonicotínicos/uso terapéutico , Quinasas Janus/antagonistas & inhibidores , Niacinamida/administración & dosificación , Niacinamida/efectos adversos , Niacinamida/análogos & derivados , Niacinamida/uso terapéutico , Inhibidores de Fosfodiesterasa 4/administración & dosificación , Inhibidores de Fosfodiesterasa 4/efectos adversos , Piperidinas/administración & dosificación , Piperidinas/efectos adversos , Inhibidores de Proteínas Quinasas/administración & dosificación , Inhibidores de Proteínas Quinasas/efectos adversos , Purinas , Pirazoles , Pirimidinas/administración & dosificación , Pirimidinas/efectos adversos , Pirroles/administración & dosificación , Pirroles/efectos adversos , Receptores de Lisoesfingolípidos/metabolismo , Sulfonamidas/administración & dosificación , Sulfonamidas/efectos adversos , Sulfonamidas/uso terapéutico , Talidomida/administración & dosificación , Talidomida/efectos adversos , Talidomida/uso terapéutico , Tiazoles/administración & dosificación , Tiazoles/efectos adversos , Quinasas Asociadas a rho/antagonistas & inhibidores
10.
IUBMB Life ; 68(5): 376-87, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-27027402

RESUMEN

Hepatic stellate cells (HSCs) are universally acknowledged to play a stimulative role in the pathogenesis of hepatic fibrosis and portal hypertension. HSCs when activated in response to liver injury are characterized with many changes, with HSC contraction being the most common cause of portal hypertension. Previous studies have shown that dihydroartemisinine (DHA) is a potential antifibrotic natural product by inducing HSC apoptosis, whereas the role of DHA in regulating HSC contraction and the mechanisms involved remain a riddle. Recent studies have emphasized on the importance of farnesoid X receptor (FXR) and sphingosine-1-phosphate receptor 2 (S1PR2) in controlling cell contractility. This study showed that DHA strongly induced the mRNA and protein expression of FXR in LX-2 cells in a dose- and time-dependent manner and inhibited HSC activation, implying a conceivable impact of DHA on HSC contraction. The gel contraction assays and fluorescence staining of actin cytoskeleton verified that DHA dose-dependently limited contraction of collagen lattices and reorganization of actin stress fibers in LX-2 cells. DHA also decreased the phosphorylation of myosin light chain that is responsible for the contractile force of HSCs. Furthermore, gain- or loss-of-function analyses exhibited a FXR- and S1PR2-dependent mechanism of inhibiting HSC contraction by DHA, and DHA decreased S1PR2 expression by modulating FXR activation. Subsequent work revealed that inhibition of both Ca(2+) -dependent and Ca(2+) -sensitization signaling transductions contributed to DHA-induced HSC relaxation. In summary, these findings suggest that DHA could restrict HSC contraction through modulating FXR/S1PR2 pathway-mediated Ca(2+) -dependent and Ca(2+) -sensitization signaling. Our discoveries make DHA a potential candidate for portal hypertension. © 2016 IUBMB Life 68(5):376-387, 2016.


Asunto(s)
Artemisininas/farmacología , Células Estrelladas Hepáticas/fisiología , Receptores Citoplasmáticos y Nucleares/metabolismo , Receptores de Lisoesfingolípidos/metabolismo , Línea Celular , Evaluación Preclínica de Medicamentos , Células Estrelladas Hepáticas/efectos de los fármacos , Humanos , Cirrosis Hepática/tratamiento farmacológico , Cirrosis Hepática/metabolismo , Cirrosis Hepática/patología , Transducción de Señal
11.
Bioorg Med Chem Lett ; 26(4): 1209-13, 2016 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-26794040

RESUMEN

The structure of the S1P2 antagonist 1 has been modified with the aim of improving its oral bioavailability. The chemical modification of the alkyl chain and carboxylic acid moieties of 1 led to significant improvements in the oral exposure of compounds belonging to this series. The optimization of the ring size of the urea portion of these molecules also led to remarkable improvements in the oral exposure. Based on these changes, the pyrrolidine derivative 16 was identified as a suitable candidate compound and showed excellent pharmacokinetic profiles in rat and dog, while maintaining high levels of potency and selective antagonistic activity toward S1P2.


Asunto(s)
Derivados del Benceno/química , Receptores de Lisoesfingolípidos/antagonistas & inhibidores , Administración Oral , Animales , Derivados del Benceno/síntesis química , Derivados del Benceno/farmacocinética , Disponibilidad Biológica , Perros , Evaluación Preclínica de Medicamentos , Semivida , Haplorrinos , Pirrolidinas/síntesis química , Pirrolidinas/química , Pirrolidinas/farmacocinética , Ratas , Receptores de Lisoesfingolípidos/metabolismo , Receptores de Esfingosina-1-Fosfato , Relación Estructura-Actividad
12.
J Appl Physiol (1985) ; 120(11): 1288-300, 2016 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-26718782

RESUMEN

To examine the role of sphingosine 1-phosphate (S1P) receptor 3 (S1P3) in modulating muscle properties, we utilized transgenic mice depleted of the receptor. Morphological analyses of extensor digitorum longus (EDL) muscle did not show evident differences between wild-type and S1P3-null mice. The body weight of 3-mo-old S1P3-null mice and the mean cross-sectional area of transgenic EDL muscle fibers were similar to those of wild-type. S1P3 deficiency enhanced the expression level of S1P1 and S1P2 receptors mRNA in S1P3-null EDL muscle. The contractile properties of S1P3-null EDL diverge from those of wild-type, largely more fatigable and less able to recover. The absence of S1P3 appears responsible for a lower availability of calcium during fatigue. S1P supplementation, expected to stimulate residual S1P receptors and signaling, reduced fatigue development of S1P3-null muscle. Moreover, in the absence of S1P3, denervated EDL atrophies less than wild-type. The analysis of atrophy-related proteins in S1P3-null EDL evidences high levels of the endogenous regulator of mitochondria biogenesis peroxisome proliferative-activated receptor-γ coactivator 1α (PGC-1α); preserving mitochondria could protect the muscle from disuse atrophy. In conclusion, the absence of S1P3 makes the muscle more sensitive to fatigue and slows down atrophy development after denervation, indicating that S1P3 is involved in the modulation of key physiological properties of the fast-twitch EDL muscle.


Asunto(s)
Fibras Musculares de Contracción Rápida/metabolismo , Fibras Musculares de Contracción Rápida/fisiología , Receptores de Lisoesfingolípidos/metabolismo , Animales , Atrofia/metabolismo , Atrofia/fisiopatología , Calcio/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos/metabolismo , Ratones Transgénicos/fisiología , Mitocondrias/metabolismo , Mitocondrias/fisiología , Fatiga Muscular/fisiología , Enfermedades Musculares/metabolismo , Enfermedades Musculares/fisiopatología , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/metabolismo , ARN Mensajero/metabolismo , Receptores de Esfingosina-1-Fosfato
13.
Aging (Albany NY) ; 9(1): 142-155, 2016 12 26.
Artículo en Inglés | MEDLINE | ID: mdl-28039439

RESUMEN

Recently, we demonstrated that the hypothalamic S1PR1/STAT3 axis plays a critical role in the control of food consumption and energy expenditure in rodents. Here, we found that reduction of hypothalamic S1PR1 expression occurs in an age-dependent manner, and was associated with defective thermogenic signaling and weight gain. To address the physiological relevance of these findings, we investigated the effects of chronic and acute exercise on the hypothalamic S1PR1/STAT3 axis. Chronic exercise increased S1PR1 expression and STAT3 phosphorylation in the hypothalamus, restoring the anorexigenic and thermogenic signals in middle-aged mice. Acutely, exercise increased sphingosine-1-phosphate (S1P) levels in the cerebrospinal fluid (CSF) of young rats, whereas the administration of CSF from exercised young rats into the hypothalamus of middle-aged rats at rest was sufficient to reduce the food intake. Finally, the intracerebroventricular (ICV) administration of S1PR1 activators, including the bioactive lipid molecule S1P, and pharmacological S1PR1 activator, SEW2871, induced a potent STAT3 phosphorylation and anorexigenic response in middle-aged rats. Overall, these results suggest that hypothalamic S1PR1 is important for the maintenance of energy balance and provide new insights into the mechanism by which exercise controls the anorexigenic and thermogenic signals in the central nervous system during the aging process.


Asunto(s)
Metabolismo Energético/fisiología , Hipotálamo/metabolismo , Lisofosfolípidos/metabolismo , Condicionamiento Físico Animal/fisiología , Receptores de Lisoesfingolípidos/metabolismo , Transducción de Señal/fisiología , Esfingosina/análogos & derivados , Absorciometría de Fotón , Tejido Adiposo Pardo/diagnóstico por imagen , Envejecimiento/fisiología , Animales , Homeostasis/fisiología , Interleucina-6/sangre , Masculino , Ratones , Consumo de Oxígeno/fisiología , Ratas , Ratas Wistar , Esfingosina/metabolismo , Receptores de Esfingosina-1-Fosfato , Proteína Desacopladora 1/metabolismo
14.
ACS Chem Biol ; 11(2): 409-14, 2016 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-26653336

RESUMEN

FTY720 sequesters lymphocytes in secondary lymphoid organs through effects on sphingosine-1-phosphate (S1P) receptors. However, at higher doses than are required for immunosuppression, FTY720 also functions as an anticancer agent in multiple animal models. Our published work indicates that the anticancer effects of FTY720 do not depend on actions at S1P receptors but instead stem from FTY720s ability to restrict access to extracellular nutrients by down-regulating nutrient transporter proteins. This result was significant because S1P receptor activation is responsible for FTY720s dose-limiting toxicity, bradycardia, that prevents its use in cancer patients. Here, we describe diastereomeric and enantiomeric 3- and 4-C-aryl 2-hydroxymethyl pyrrolidines that are more active than the previously known analogues. Of importance is that these compounds fail to activate S1P1 or S1P3 receptors in vivo but retain inhibitory effects on nutrient transporter proteins and anticancer activity in solid tumor xenograft models. Our studies reaffirm that the anticancer activity of FTY720 does not depend upon S1P receptor activation and uphold the promise of using S1P receptor-inactive azacyclic FTY720 analogues in human cancer patients.


Asunto(s)
Antineoplásicos/química , Antineoplásicos/uso terapéutico , Clorhidrato de Fingolimod/análogos & derivados , Clorhidrato de Fingolimod/uso terapéutico , Neoplasias/tratamiento farmacológico , Pirrolidinas/química , Pirrolidinas/uso terapéutico , Animales , Antineoplásicos/farmacología , Proteínas Portadoras/metabolismo , Línea Celular Tumoral , Clorhidrato de Fingolimod/farmacología , Humanos , Inmunosupresores/química , Inmunosupresores/farmacología , Inmunosupresores/uso terapéutico , Ratones Desnudos , Neoplasias/metabolismo , Neoplasias/patología , Pirrolidinas/farmacología , Receptores de Lisoesfingolípidos/metabolismo
15.
Immunity ; 42(2): 252-264, 2015 Feb 17.
Artículo en Inglés | MEDLINE | ID: mdl-25692701

RESUMEN

T follicular helper (Tfh) cells are essential for efficient B cell responses, yet the factors that regulate differentiation of this CD4(+) T cell subset are incompletely understood. Here we found that the KLF2 transcription factor serves to restrain Tfh cell generation. Induced KLF2 deficiency in activated CD4(+) T cells led to increased Tfh cell generation and B cell priming, whereas KLF2 overexpression prevented Tfh cell production. KLF2 promotes expression of the trafficking receptor S1PR1, and S1PR1 downregulation is essential for efficient Tfh cell production. However, KLF2 also induced expression of the transcription factor Blimp-1, which repressed transcription factor Bcl-6 and thereby impaired Tfh cell differentiation. Furthermore, KLF2 induced expression of the transcription factors T-bet and GATA3 and enhanced Th1 differentiation. Hence, our data indicate KLF2 is pivotal for coordinating CD4(+) T cell differentiation through two distinct and complementary mechanisms: via control of T cell localization and by regulation of lineage-defining transcription factors.


Asunto(s)
Diferenciación Celular/inmunología , Factores de Transcripción de Tipo Kruppel/inmunología , Células TH1/citología , Células TH1/inmunología , Traslado Adoptivo , Animales , Antígenos CD/biosíntesis , Antígenos de Diferenciación de Linfocitos T/biosíntesis , Linfocitos B/inmunología , Proteínas de Unión al ADN/biosíntesis , Regulación hacia Abajo , Factor de Transcripción GATA3/biosíntesis , Técnicas de Inactivación de Genes , Factores de Transcripción de Tipo Kruppel/biosíntesis , Factores de Transcripción de Tipo Kruppel/genética , Lectinas Tipo C/biosíntesis , Activación de Linfocitos/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Factor 1 de Unión al Dominio 1 de Regulación Positiva , Proteínas Proto-Oncogénicas c-bcl-6 , Receptores de Lisoesfingolípidos/biosíntesis , Receptores de Lisoesfingolípidos/metabolismo , Receptores de Esfingosina-1-Fosfato , Proteínas de Dominio T Box/biosíntesis , Factores de Transcripción/biosíntesis , Factores de Transcripción/metabolismo
16.
Exp Physiol ; 100(1): 95-107, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25557733

RESUMEN

NEW FINDINGS: What is the central question of this study? Why do different doses of sphingosine-1-phosphate (S1P) induce distinct biological effects in endothelial cells? What is the main finding and its importance? S1P at physiological concentrations preserved endothelial barrier function by binding to S1P receptor 1, then triggering Ca(2+) release from endoplasmic reticulum through phosphoinositide phospholipase C and inositol triphosphate, and consequently strengthening tight junction and F-actin assembly through Rac1 activation. Excessive S1P induced endothelial malfunction by activating S1P receptor 2 and RhoA/ROCK pathway, causing F-actin and tight junction disorganisation. Extracellular Ca(2+) influx was involved in this process. Sphingosine-1-phosphate (S1P) is a bioactive sphingolipid in plasma, and its plasma concentration can be adjusted through a complex metabolic process. The alterations in S1P levels and the activation of receptors collaboratively regulate distinct biological effects. This study was performed to investigate comparatively the effect of different concentrations of S1P on endothelial barrier function and to explore the roles of S1P receptors (S1PRs), Rho GTPases and calcium in S1P-induced endothelial responses. Endothelial barrier function was studied using transendothelial electric resistance and a resistance meter in human umbilical vein endothelial cells. Specific agonists or antagonists were applied to control the activation of S1P receptors and the release of calcium from different cellular compartments. The results indicated that at physiological concentrations, S1P preserved endothelial barrier function by binding with S1PR1. The activation of S1PR1 triggered the release of intracellular Ca(2+) from the endoplasmic reticulum through the PI-phospholipase C and inositol trisphosphate pathways. Consequently, the Rho GTPase Rac1 was activated, strengthening the assembly of tight junction proteins and F-actin. However, excessive S1P induced endothelial barrier dysfunction by activating S1PR2 followed by the RhoA/RhoA kinase pathway, causing the disorganization of F-actin and the disassembly of the tight junction protein ZO-1. An influx of extracellular Ca(2+) was involved in this process. These data suggest that physiological and excessive amounts of S1P induce different responses in human umbilical vein endothelial cells; the activation of the 1PR1-PLC-IP3 R-Ca(2+) -Rac1 pathway governs the low-dose S1P-enhanced endothelial barrier integrity, and the activation of S1PR2-calcium influx-RhoA/ROCK dominates the high-dose S1P-induced endothelial monolayer hyperpermeability response.


Asunto(s)
Señalización del Calcio/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Lisofosfolípidos/farmacología , Receptores de Lisoesfingolípidos/agonistas , Esfingosina/análogos & derivados , Permeabilidad Capilar/efectos de los fármacos , Forma de la Célula/efectos de los fármacos , Células Cultivadas , Relación Dosis-Respuesta a Droga , Impedancia Eléctrica , Retículo Endoplásmico/efectos de los fármacos , Retículo Endoplásmico/metabolismo , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Receptores de Lisoesfingolípidos/metabolismo , Esfingosina/farmacología , Receptores de Esfingosina-1-Fosfato , Factores de Tiempo , Proteína de Unión al GTP rac1/metabolismo , Quinasas Asociadas a rho/metabolismo , Proteína de Unión al GTP rhoA/metabolismo
17.
PLoS One ; 10(1): e0116873, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25633986

RESUMEN

Nephrokeli (NPKL) is a Chinese herbal formula that has been used to treat patients with IgA nephropathy (IgAN) for improvement of proteinuria and kidney injury. However, the mechanism remains unclear. Sphingosine-1-phosphate (S1P) and its receptors S1PR2 and S1PR3 are known to play an important role in kidney disease. Here, we tested whether NPKL is able to regulate the S1P pathway in the kidney of IgAN rats. Four groups of rats were included in the study: Control, IgAN, IgAN treated with losartan, and IgAN treated with NPKL. The IgAN model was generated by injection of bovine serum albumin and staphylococcus enterotoxin B. We found that IgAN rats had increased staining for proliferating cell nuclear antigen (PCNA) in the mesangial area and increased mRNA and protein levels of S1PR2 and S1PR3 in the kidney compared to control rats. Connective tissue growth factor (CTGF), a downstream growth factor in the S1P pathway, was also elevated in the kidney of IgAN rats. Treatment with either NPKL or losartan was able to reduce PCNA staining and the expression of both S1PR2 and S1PR3 in the kidney of IgAN rats. However, NPKL (but not losartan treatment) reduced the expression of CTGF in the kidney of IgAN rats. In addition, we treated rat mesangial cells with sera collected from either NPKL-treated rats or control rats and found that NPKL-serum was able to reduce S1P-induced mesangial cell proliferation and the expression of S1PR2/S1PR3 and CTGF. NPKL also attenuates expression of fibrosis, inflammation, and oxidative stress markers in the kidney of IgAN rats. Our studies provide the mechanism by which NPKL attenuates kidney injury in IgAN rats.


Asunto(s)
Medicamentos Herbarios Chinos/uso terapéutico , Glomerulonefritis por IGA/tratamiento farmacológico , Riñón/efectos de los fármacos , Lisofosfolípidos/metabolismo , Transducción de Señal/efectos de los fármacos , Esfingosina/análogos & derivados , Animales , Medicamentos Herbarios Chinos/farmacología , Femenino , Glomerulonefritis por IGA/metabolismo , Glomerulonefritis por IGA/patología , Riñón/metabolismo , Ratas , Ratas Sprague-Dawley , Receptores de Lisoesfingolípidos/metabolismo , Esfingosina/metabolismo , Receptores de Esfingosina-1-Fosfato
18.
J Am Soc Nephrol ; 26(4): 908-25, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25145931

RESUMEN

Sphingosine 1-phosphate (S1P), the natural sphingolipid ligand for a family of five G protein- coupled receptors (S1P1-S1P5Rs), regulates cell survival and lymphocyte circulation. We have shown that the pan-S1PR agonist, FTY720, attenuates kidney ischemia-reperfusion injury by directly activating S1P1 on proximal tubule (PT) cells, independent of the canonical lymphopenic effects of S1P1 activation on B and T cells. FTY720 also reduces cisplatin-induced AKI. Therefore, in this study, we used conditional PT-S1P1-null (PepckCreS1pr1(fl/fl)) and control (PepckCreS1pr1(w/wt)) mice to determine whether the protective effect of FTY720 in AKI is mediated by PT-S1P1. Cisplatin induced more renal injury in PT-S1P1-null mice than in controls. Although FTY720 produced lymphopenia in both control and PT-S1P1-null mice, it reduced injury only in control mice. Furthermore, the increase in proinflammatory cytokine (CXCL1, MCP-1, TNF-α, and IL-6) expression and infiltration of neutrophils and macrophages induced by cisplatin treatment was attenuated by FTY720 in control mice but not in PT-S1P1-null mice. Similarly, S1P1 deletion rendered cultured PT cells more susceptible to cisplatin-induced injury, whereas S1P1 overexpression protected PT cells from injury and preserved mitochondrial function. We conclude that S1P1 may have an important role in stabilizing mitochondrial function and that FTY720 administration represents a novel strategy in the prevention of cisplatin-induced AKI.


Asunto(s)
Lesión Renal Aguda/prevención & control , Túbulos Renales Proximales/efectos de los fármacos , Mitocondrias/efectos de los fármacos , Glicoles de Propileno/uso terapéutico , Receptores de Lisoesfingolípidos/agonistas , Esfingosina/análogos & derivados , Lesión Renal Aguda/inducido químicamente , Animales , Apoptosis/efectos de los fármacos , Respiración de la Célula , Cisplatino , Evaluación Preclínica de Medicamentos , Células Epiteliales/efectos de los fármacos , Clorhidrato de Fingolimod , Masculino , Ratones Endogámicos C57BL , Dinámicas Mitocondriales , Glicoles de Propileno/farmacología , Receptores de Lisoesfingolípidos/metabolismo , Esfingosina/farmacología , Esfingosina/uso terapéutico
19.
Nat Commun ; 5: 4859, 2014 Sep 25.
Artículo en Inglés | MEDLINE | ID: mdl-25255053

RESUMEN

Sphingosine 1-phosphate receptor 1 (S1PR1) is a G-protein-coupled receptor for sphingosine-1-phosphate (S1P) that has a role in many physiological and pathophysiological processes. Here we show that the S1P/S1PR1 signalling pathway in hypothalamic neurons regulates energy homeostasis in rodents. We demonstrate that S1PR1 protein is highly enriched in hypothalamic POMC neurons of rats. Intracerebroventricular injections of the bioactive lipid, S1P, reduce food consumption and increase rat energy expenditure through persistent activation of STAT3 and the melanocortin system. Similarly, the selective disruption of hypothalamic S1PR1 increases food intake and reduces the respiratory exchange ratio. We further show that STAT3 controls S1PR1 expression in neurons via a positive feedback mechanism. Interestingly, several models of obesity and cancer anorexia display an imbalance of hypothalamic S1P/S1PR1/STAT3 axis, whereas pharmacological intervention ameliorates these phenotypes. Taken together, our data demonstrate that the neuronal S1P/S1PR1/STAT3 signalling axis plays a critical role in the control of energy homeostasis in rats.


Asunto(s)
Metabolismo Energético , Hipotálamo/metabolismo , Lisofosfolípidos/metabolismo , Receptores de Lisoesfingolípidos/metabolismo , Esfingosina/análogos & derivados , Animales , Homeostasis , Masculino , Ratones , Ratones Endogámicos C57BL , Neuronas/metabolismo , Proopiomelanocortina/metabolismo , Ratas , Ratas Wistar , Receptores de Lisoesfingolípidos/genética , Factor de Transcripción STAT3/metabolismo , Transducción de Señal , Esfingosina/metabolismo , Receptores de Esfingosina-1-Fosfato
20.
Int J Mol Med ; 30(6): 1451-8, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22992945

RESUMEN

Sphingosine-1-phosphate (S1P) is a pluripotent lipid mediator that transmits signals through a family of G-protein-coupled receptors (GPCRs) to control diverse biological processes including inflammation and wound-healing. In this study, a novel biological activity of S1P in articular chondrocytes was identified. Human primary chondrocytes were cultured in a monolayer. Reverse transcription-polymerase chain reaction (RT-PCR) and western blotting were performed to detect genes and proteins involved in inflammation and cartilage degradation when human primary chondrocytes were stimulated by interleukin (IL)-1ß. Matrix metalloproteinase (MMP)-2 and MMP-9 activity was evaluated by gelatin zymography. Glycosaminoglycan (GAG) degradation was evaluated using the dimethylene blue method. Prostaglandin E2 (PGE2) was measured by enzyme-linked immunosorbent assay (ELISA). By using the S1P1 receptor agonist and antagonist, we discovered the key role played by S1P1 in the S1P-dependent inhibition of IL-1ß-induced inflammation in human chondrocytes. S1P dose-dependently inhibited IL-1ß-induced NF-κB p65, cyclooxygenase (COX)-2, MMP-1, MMP-3, MMP-13 and MMP-14 mRNA expression in human chondrocytes and IL-1ß-induced PGE2 synthesis and GAG degradation in human cartilage explants. W146, a known S1P1 receptor antagonist, inhibited the active form of NF-κB p65 and COX-2 expression induced by IL-1ß. The anti-inflammatory action of the S1P1 receptor agonist SEW2871 was similar to that of S1P. This study demonstrates that S1P has anti-inflammatory effects on chondrocytes via the S1P1 receptor. Our data suggest that targeting S1P and S1P1 may be a potential therapy for arthritis.


Asunto(s)
Antiinflamatorios/farmacología , Cartílago Articular/patología , Condrocitos/metabolismo , Interleucina-1beta/fisiología , Lisofosfolípidos/farmacología , Esfingosina/análogos & derivados , Células Cultivadas , Condrocitos/efectos de los fármacos , Condrocitos/patología , Ciclooxigenasa 2/genética , Ciclooxigenasa 2/metabolismo , Evaluación Preclínica de Medicamentos , Inducción Enzimática/efectos de los fármacos , Matriz Extracelular , Humanos , Articulación de la Rodilla/patología , Metaloproteinasas de la Matriz Secretadas/genética , Metaloproteinasas de la Matriz Secretadas/metabolismo , Osteoartritis de la Rodilla/tratamiento farmacológico , Osteoartritis de la Rodilla/metabolismo , Osteoartritis de la Rodilla/patología , Proteolisis , Receptores de Lisoesfingolípidos/metabolismo , Esfingosina/farmacología , Factor de Transcripción ReIA/genética , Factor de Transcripción ReIA/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA