Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 137
Filtrar
1.
Science ; 384(6691): 66-73, 2024 Apr 05.
Artículo en Inglés | MEDLINE | ID: mdl-38574138

RESUMEN

Asthma is deemed an inflammatory disease, yet the defining diagnostic feature is mechanical bronchoconstriction. We previously discovered a conserved process called cell extrusion that drives homeostatic epithelial cell death when cells become too crowded. In this work, we show that the pathological crowding of a bronchoconstrictive attack causes so much epithelial cell extrusion that it damages the airways, resulting in inflammation and mucus secretion in both mice and humans. Although relaxing the airways with the rescue treatment albuterol did not affect these responses, inhibiting live cell extrusion signaling during bronchoconstriction prevented all these features. Our findings show that bronchoconstriction causes epithelial damage and inflammation by excess crowding-induced cell extrusion and suggest that blocking epithelial extrusion, instead of the ensuing downstream inflammation, could prevent the feed-forward asthma inflammatory cycle.


Asunto(s)
Asma , Bronquios , Broncoconstricción , Animales , Humanos , Ratones , Asma/patología , Asma/fisiopatología , Broncoconstricción/efectos de los fármacos , Inflamación/patología , Transducción de Señal , Canales Iónicos/antagonistas & inhibidores , Lisofosfolípidos/antagonistas & inhibidores , Esfingosina/análogos & derivados , Esfingosina/antagonistas & inhibidores , Bronquios/patología , Bronquios/fisiopatología
2.
Molecules ; 26(17)2021 Aug 24.
Artículo en Inglés | MEDLINE | ID: mdl-34500564

RESUMEN

Sphingosine 1-phosphate (S1P) is an extensively studied signaling molecule that contributes to cell proliferation, survival, migration and other functions through binding to specific S1P receptors. The cycle of S1P1 internalization upon S1P binding and recycling to the cell surface when local S1P concentrations are low drives T cell trafficking. S1P1 modulators, such as fingolimod, disrupt this recycling by inducing persistent S1P1 internalization and receptor degradation, which results in blocked egress of T cells from the secondary lymphoid tissues. The approval of these compounds for the treatment of multiple sclerosis has placed the development of S1PR modulators in the focus of pharmacological research, mostly for autoimmune indications. Here, we report on a novel anellated bismorpholino derivative of oxy-fingolimod, named ST-2191, which exerts selective S1P1 agonist and functional antagonist potency. ST-2191 is also effective in reducing the lymphocyte number in mice, and this effect is not dependent on phosphorylation by sphingosine kinase 2 for activity. These data show that ST-2191 is a novel S1P1 modulator, but further experiments are needed to analyze the therapeutic impact of ST-2191 in animal models of autoimmune diseases.


Asunto(s)
Clorhidrato de Fingolimod/farmacología , Lisofosfolípidos/agonistas , Lisofosfolípidos/antagonistas & inhibidores , Esfingosina/análogos & derivados , Animales , Células CHO , Cricetulus , Humanos , Recuento de Linfocitos/métodos , Lisofosfolípidos/metabolismo , Ratones , Ratones Endogámicos C57BL , Esclerosis Múltiple/tratamiento farmacológico , Esclerosis Múltiple/metabolismo , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Receptores de Lisoesfingolípidos/metabolismo , Transducción de Señal/efectos de los fármacos , Esfingosina/agonistas , Esfingosina/antagonistas & inhibidores , Esfingosina/metabolismo , Linfocitos T/metabolismo
3.
Sci Rep ; 11(1): 15308, 2021 07 28.
Artículo en Inglés | MEDLINE | ID: mdl-34321503

RESUMEN

Sphingosine 1-phosphate (S1P) is a bioactive signalling sphingolipid that is increased in diseases such as obesity and diabetes. S1P can modulate platelet function, however the direction of effect and S1P receptors (S1PRs) involved are controversial. Here we describe the role of S1P in regulating human platelet function and identify the receptor subtypes responsible for S1P priming. Human platelets were treated with protease-activated receptor 1 (PAR-1)-activating peptide in the presence or absence of S1P, S1PR agonists or antagonists, and sphingosine kinases inhibitors. S1P alone did not induce platelet aggregation but at low concentrations S1P enhanced PAR1-mediated platelet responses, whereas PAR1 responses were inhibited by high concentrations of S1P. This biphasic effect was mimicked by pan-S1PR agonists. Specific agonists revealed that S1PR1 receptor activation has a positive priming effect, S1PR2 and S1PR3 have no effect on platelet function, whereas S1PR4 and S1PR5 receptor activation have an inhibitory effect on PAR-1 mediated platelet function. Although platelets express both sphingosine kinase 1/2, enzymes which phosphorylate sphingosine to produce S1P, only dual and SphK2 inhibition reduced platelet function. These results support a role for SphK2-mediated S1P generation in concentration-dependent positive and negative priming of platelet function, through S1PR1 and S1PR4/5 receptors, respectively.


Asunto(s)
Lisofosfolípidos/farmacología , Activación Plaquetaria/efectos de los fármacos , Receptores de Esfingosina-1-Fosfato/efectos de los fármacos , Esfingosina/análogos & derivados , Plaquetas/efectos de los fármacos , Plaquetas/ultraestructura , Proteínas Portadoras/farmacología , Forma de la Célula/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Humanos , Lisofosfolípidos/agonistas , Lisofosfolípidos/antagonistas & inhibidores , Fragmentos de Péptidos/farmacología , Péptidos/farmacología , Fosfotransferasas (Aceptor de Grupo Alcohol)/antagonistas & inhibidores , Fosfotransferasas (Aceptor de Grupo Alcohol)/fisiología , Agregación Plaquetaria/efectos de los fármacos , Receptor PAR-1/agonistas , Esfingosina/agonistas , Esfingosina/antagonistas & inhibidores , Esfingosina/farmacología , Receptores de Esfingosina-1-Fosfato/fisiología
4.
Osteoarthritis Cartilage ; 29(9): 1335-1345, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34144150

RESUMEN

OBJECTIVE: Cartilage loss observed in osteoarthritis (OA) is prevented when osteoclasts in the subchondral bone are inhibited in mice. Here, we investigated the role of the osteoclast secretome and of the lipid mediator sphingosine 1-phosphate (S1P) in chondrocyte metabolism and OA. MATERIALS AND METHODS: We used SphK1LysMCre and wild type mice to assess the effect of murine osteoclast secretome in chondrocyte metabolism. Gene and protein expressions of matrix metalloproteinase (Mmp) were quantified in chondrocytes and explants by RT-qPCR and Western blots. SphK1LysMCre mice or wild type mice treated with S1P2 receptor inhibitor JTE013 or anti-S1P neutralizing antibody sphingomab are analyzed by OA score and immunohistochemistry. RESULTS: The osteoclast secretome increased the expression of Mmp3 and Mmp13 in murine chondrocytes and cartilage explants and activated the JNK signaling pathway, which led to matrix degradation. JTE013 reversed the osteoclast-mediated chondrocyte catabolism and protected mice against OA, suggesting that osteoclastic S1P contributes to cartilage damage in OA via S1P/S1P2 signaling. The activity of sphingosine kinase 1 (SphK1) increased with osteoclast differentiation, and its expression was enhanced in subchondral bone of mice with OA. The expression of Mmp3 and Mmp13 in chondrocytes was low upon stimulation with the secretome of Sphk1-lacking osteoclasts. Cartilage damage was significantly reduced in SphK1LysMCre mice, but not the synovial inflammation. Finally, intra-articular administration of sphingomab inhibited the cartilage damage and synovial inflammation. CONCLUSIONS: Lack of S1P in myeloid cells and local S1P neutralization alleviates from osteoarthritis in mice. These data identify S1P as a therapeutic target in OA.


Asunto(s)
Condrocitos/metabolismo , Lisofosfolípidos/antagonistas & inhibidores , Osteoartritis/metabolismo , Osteoartritis/prevención & control , Osteoclastos/metabolismo , Secretoma/metabolismo , Esfingosina/análogos & derivados , Animales , Masculino , Ratones , Esfingosina/antagonistas & inhibidores
5.
Immunopharmacol Immunotoxicol ; 43(2): 192-202, 2021 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-33504231

RESUMEN

OBJECTIVES: Ulcerative colitis (UC) is a chronic inflammatory bowel disease that is associated with high sphingosine kinase 1(SPHK1) expression in the colon, however its role in pathogenesis of UC is not clearly understood so, the aim of the present study was to clarify the role of SPHK1 and investigate whether the anti-inflammatory effects of metformin in UC is mediated by Sphingosine kinase 1/sphingosine 1 phosphate (S1P) signaling pathway. MATERIAL AND METHODS: Colitis was induced in adult male wistar rats by intra rectal administration of oxazolone in the fifth and seventh days from initial presensitization. Oxazolone treated rats were divided into untreated oxazolone group, metformin and mesalazine treated groups both in a dose of 100 mg/kg/day orally for 21 days. Along with these groups normal control and saline groups were used .Colitis was assessed by colon length, disease activity index (DAI) and histological examination of colontissue. Plasma samples were used to measure S1P.SPHK1 activity, signal transducer and activator of transcription -3(STAT-3), interleukin-6 (IL-6), nitric oxide (NO), myeloperoxidase activity (MPO), reduced glutathione (GSH) and tissue expression of intracellular cell adhesion molecule -1(ICAM-1) and caspase-3 genes were measured in tissue. RESULTS: Metformin successfully attenuated oxazolone colitis by increasing colon length, decreasing DAI and improved colon histologic picture. Metformin also induced a significant decrease in Plasma SIP, SPHK1 activity, inflammatory, oxidative stress markers, ICAM-1 and Caspase-3 genes expression compared to oxazolone group. CONCLUSION: It is revealed that metformin alleviated inflammation and underlying mechanism may result from inhibition of SPHK1/S1P signaling pathway.


Asunto(s)
Colitis Ulcerosa/sangre , Colitis Ulcerosa/tratamiento farmacológico , Lisofosfolípidos/sangre , Metformina/uso terapéutico , Oxazolona/toxicidad , Fosfotransferasas (Aceptor de Grupo Alcohol)/sangre , Esfingosina/análogos & derivados , Animales , Colitis Ulcerosa/inducido químicamente , Colon/efectos de los fármacos , Colon/metabolismo , Colon/patología , Inflamación/sangre , Inflamación/inducido químicamente , Inflamación/tratamiento farmacológico , Lisofosfolípidos/antagonistas & inhibidores , Masculino , Metformina/farmacología , Fosfotransferasas (Aceptor de Grupo Alcohol)/antagonistas & inhibidores , Ratas , Ratas Wistar , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Esfingosina/antagonistas & inhibidores , Esfingosina/sangre
6.
Life Sci ; 256: 117988, 2020 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-32569777

RESUMEN

OBJECTIVE: Rheumatoid arthritis (RA) is a common inflammatory autoimmune disease characterized by the formation of joint synovitis and pannus. Sphingosine 1-phosphate (S1P) is an important mediator related to angiogenesis, inflammation and autoimmunity. As Geniposide (GE) has potent immuno-modulation function, we investigated the effects on the dynamic balance of angiogenesis-related factors and Sphingosine kinase 1 (SphK1)-S1P-S1P receptor 1 (S1PR1) signal transduction in adjuvant-induced arthritis (AA) rats. METHOD: The model evaluation was performed from paw swelling degree, arthritis index and movement score. The immunohistochemistry and enzyme-linked immunosorbent assay were used to study the microvascular density (MVD) and pro/anti-angiogenic factors levels. The cell viability was examined by cell counting kit-8 assay. SphK1, S1PR1 mRNA and protein levels in fibroblast-like synoviocytes (FLSs) were detected by quantitative real-time polymerase chain reaction and Western blotting. RESULTS: The results showed that GE can apparently suppressed the inflammatory pathological status. The arthritis index, paw swelling and MVD of AA rats were decreased with dose dependence (⁎P < 0.05, ⁎⁎P < 0.01). In addition, GE can reduce the secretion of vascular endothelial growth factor (VEGF) and angiopoietin-1 (Ang-1), promote the secretion of endostatin (ES) and inhibit excessive proliferation of FLSs (⁎P < 0.05, ⁎⁎P < 0.01). Importantly, GE can significantly inhibit the activity of SphK1, the level of S1P and the expression of SphK1 and S1PR1 in FLSs (⁎P < 0.05, ⁎⁎P < 0.01). CONCLUSION: It indicated that GE reduces the activity of SphK1 by restoring the dynamic balance between pro/anti-angiogenic factors, thereby interfering with SphK1-S1P-S1PR1 signal transduction, reducing the formation of synovial microvessels and exerting anti-angiogenesis effect of RA.


Asunto(s)
Sistemas de Liberación de Medicamentos/métodos , Iridoides/administración & dosificación , Lisofosfolípidos/metabolismo , Neovascularización Patológica/metabolismo , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Receptores de Esfingosina-1-Fosfato/metabolismo , Esfingosina/análogos & derivados , Animales , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Células Cultivadas , Lisofosfolípidos/antagonistas & inhibidores , Masculino , Neovascularización Patológica/tratamiento farmacológico , Fosfotransferasas (Aceptor de Grupo Alcohol)/antagonistas & inhibidores , Distribución Aleatoria , Ratas , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Esfingosina/antagonistas & inhibidores , Esfingosina/metabolismo , Receptores de Esfingosina-1-Fosfato/antagonistas & inhibidores
7.
Hypertension ; 75(2): 383-392, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31838904

RESUMEN

High blood pressure is a risk factor for cardiovascular diseases. Ang II (angiotensin II), a key pro-hypertensive hormone, mediates target organ consequences such as endothelial dysfunction and cardiac hypertrophy. S1P (sphingosine-1-phosphate), produced by Sphk1 (sphingosine kinase 1), plays a pivotal role in the pathogenesis of hypertension and downstream organ damage, as it controls vascular tone and regulates cardiac remodeling. Accordingly, we aimed to examine if pharmacological inhibition of Sphk1 using selective inhibitor PF543 can represent a useful vasoprotective and cardioprotective anti-hypertensive strategy in vivo. PF543 was administered intraperitoneally throughout a 14-day Ang II-infusion in C57BL6/J male mice. Pharmacological inhibition of Sphk1 improved endothelial function of arteries of hypertensive mice that could be mediated via decrease in eNOS (endothelial nitric oxide synthase) phosphorylation at T495. This effect was independent of blood pressure. Importantly, PF543 also reduced cardiac hypertrophy (heart to body weight ratio, 5.6±0.2 versus 6.4±0.1 versus 5.9±0.2 mg/g; P<0.05 for Sham, Ang II+placebo, and Ang II+PF543-treated mice, respectively). Mass spectrometry revealed that PF543 elevated cardiac sphingosine, that is, Sphk1 substrate, content in vivo. Mechanistically, RNA-Seq indicated a decreased expression of cardiac genes involved in actin/integrin organization, S1pr1 signaling, and tissue remodeling. Indeed, downregulation of Rock1 (Rho-associated coiled-coil containing protein kinase 1), Stat3 (signal transducer and activator of transcription 3), PKC (protein kinase C), and ERK1/2 (extracellular signal-regulated kinases 1/2) level/phosphorylation by PF543 was observed. In summary, pharmacological inhibition of Sphk1 partially protects against Ang II-induced cardiac hypertrophy and endothelial dysfunction. Therefore, it may represent a promising target for harnessing residual cardiovascular risk in hypertension.


Asunto(s)
Presión Sanguínea/fisiología , Hipertensión/tratamiento farmacológico , Lisofosfolípidos/genética , Metanol/análogos & derivados , Pirrolidinas/administración & dosificación , ARN/genética , Esfingosina/análogos & derivados , Remodelación Ventricular/fisiología , Animales , Modelos Animales de Enfermedad , Inhibidores Enzimáticos/administración & dosificación , Hipertensión/genética , Hipertensión/metabolismo , Inyecciones Intraperitoneales , Lisofosfolípidos/antagonistas & inhibidores , Lisofosfolípidos/metabolismo , Masculino , Metanol/administración & dosificación , Ratones , Ratones Endogámicos C57BL , Miocardio/metabolismo , Transducción de Señal , Esfingosina/antagonistas & inhibidores , Esfingosina/genética , Esfingosina/metabolismo , Sulfonas , Remodelación Ventricular/efectos de los fármacos
8.
Neurosci Lett ; 715: 134599, 2020 01 10.
Artículo en Inglés | MEDLINE | ID: mdl-31722234

RESUMEN

Expression of sphingosine/sphingosine 1-phosphate (SPH/S1P) in resident cells of the central nervous system plays an important role in the pathogenesis of multiple sclerosis (MS). Accumulated evidence has shown the protective effects of S1P receptor modulators on MS and its animal model, experimental autoimmune encephalomyelitis (EAE). However, effective therapies to regulate SPH/S1P molecules themselves have not been well addressed. Our previous studies showed that matrine (MAT), a natural alkaloid component extracted from the Sophora root, has beneficial effects in EAE through immunomodulation. Here we demonstrate that MAT alleviated astrogliosis in the CNS of EAE rats, and downregulated levels of SPH, S1P and S1P1 expression in CNS tissues and astrocytes. Expression of SPH kinases (SPHK) 1 and 2, which splice SPH into S1P, was also inhibited by MAT treatment. In vitro studies showed a direct inhibitory effect of MAT on S1P1 expression of activated astrocytes, suggesting that MAT could function as an S1PRs antagonist. Moreover, MAT upregulated the expression of plasma gelsolin, which combines with S1P to reduce its concentration. These findings indicate that MAT could alleviate astrogliosis in EAE through diminishing the SPH/SPHK/S1P1 pathway.


Asunto(s)
Alcaloides/farmacología , Encefalomielitis Autoinmune Experimental/metabolismo , Gliosis/metabolismo , Lisofosfolípidos/antagonistas & inhibidores , Lisofosfolípidos/metabolismo , Quinolizinas/farmacología , Transducción de Señal/fisiología , Esfingosina/análogos & derivados , Alcaloides/uso terapéutico , Animales , Células Cultivadas , Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Femenino , Gliosis/tratamiento farmacológico , Quinolizinas/uso terapéutico , Ratas , Ratas Wistar , Transducción de Señal/efectos de los fármacos , Esfingosina/antagonistas & inhibidores , Esfingosina/metabolismo , Matrinas
9.
Biochem Biophys Res Commun ; 516(3): 934-940, 2019 08 27.
Artículo en Inglés | MEDLINE | ID: mdl-31277939

RESUMEN

This study shows the effects of tamoxifen, a known estrogen receptor antagonist used in the treatment of breast cancer, on the sphingolipid pathway of Trypanosoma cruzi, searching for potential chemotherapeutic targets. A dose-dependent epimastigote growth inhibition at increasing concentration of tamoxifen was determined. In blood trypomastigotes, treatment with 10 µM showed 90% lysis, while 86% inhibition of intracellular amastigote development was obtained using 50 µM. Lipid extracts from treated and non-treated metabolically labelled epimastigotes evidenced by thin layer chromatography different levels of sphingolipids and MALDI-TOF mass spectrometry analysis assured the identity of the labelled species. Comparison by HPLC-ESI mass spectrometry of lipids, notably exhibited a dramatic increase in the level of ceramide in tamoxifen-treated parasites and a restrained increase of ceramide-1P and sphingosine, indicating that the drug is acting on the enzymes involved in the final breakdown of ceramide. The ultrastructural analysis of treated parasites revealed characteristic morphology of cells undergoing an apoptotic-like death process. Flow cytometry confirmed cell death by an apoptotic-like machinery indicating that tamoxifen triggers this process by acting on the parasitic sphingolipid pathway.


Asunto(s)
Antiprotozoarios/farmacología , Estadios del Ciclo de Vida/efectos de los fármacos , Metabolismo de los Lípidos/efectos de los fármacos , Esfingolípidos/antagonistas & inhibidores , Tamoxifeno/farmacología , Trypanosoma cruzi/efectos de los fármacos , Animales , Apoptosis/efectos de los fármacos , Ceramidas/antagonistas & inhibidores , Ceramidas/biosíntesis , Enfermedad de Chagas/tratamiento farmacológico , Enfermedad de Chagas/parasitología , Modelos Animales de Enfermedad , Reposicionamiento de Medicamentos , Antagonistas de Estrógenos/farmacología , Ratones , Ratones Endogámicos BALB C , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción , Esfingolípidos/biosíntesis , Esfingosina/antagonistas & inhibidores , Esfingosina/biosíntesis , Trypanosoma cruzi/crecimiento & desarrollo , Trypanosoma cruzi/metabolismo
10.
Biochem Biophys Res Commun ; 514(4): 1058-1065, 2019 07 05.
Artículo en Inglés | MEDLINE | ID: mdl-31097221

RESUMEN

Resolvins (Rvs) are endogenous lipid mediators that promote resolution of inflammation and return to homeostasis. We previously reported that RvD1 both facilitates M2 macrophage polarization of Kupffer cells (KCs) and efferocytosis and modulates thioredoxin 2-mediated mitochondrial quality control in liver ischemia/reperfusion (IR) injury. However, the specific cellular or molecular targets of RvD1 remain poorly understood. Sphingosine-1-phosphate (S1P), the natural sphingolipid ligand for a family of G protein-coupled receptors (S1P1-S1P5), regulates lymphocyte circulation and various immune responses. Here we investigated the role of RvD1 in IR-induced hepatocellular damage with a focus on S1P signaling. Male C57BL/6 mice were subjected to partial hepatic ischemia for 60 min, followed by reperfusion. Mice were pretreated with RvD1 (15 µg/kg, i.p.) 1 h prior to ischemia and immediately before reperfusion. To deplete KCs, liposome clodronate was administered (100 µL/mice, i.v.) 24 h prior to ischemia. Mice were pretreated with VPC23019 (100 µg/kg, i.p.), an antagonist for S1P1/S1P3 10 min prior to initial RvD1 treatment. Exogenous RvD1 attenuated IR-induced hepatocellular damage as evidenced by serum HMGB1 release. RvD1 attenuated the decrease in hepatic S1P concentration induced by IR. KC depletion by liposome clodronate did not alter the effect of RvD1 on sphingosine kinases (SKs) and S1P receptors, suggesting independency of KCs. Moreover, in purified hepatocytes of mice exposed to IR, mRNA expression of SK1, SK2, S1P1, and S1P3 decreased significantly, and this was attenuated by RvD1. Finally, VPC23019 pretreatment abolished the hepatoprotective effects of RvD1 in serum HMGB1 release. Our findings suggest that RvD1 protects the liver against IR injury by activating S1P signaling.


Asunto(s)
Ácidos Docosahexaenoicos/farmacología , Hígado/efectos de los fármacos , Lisofosfolípidos/metabolismo , Daño por Reperfusión/tratamiento farmacológico , Esfingosina/análogos & derivados , Animales , Rayos Infrarrojos , Hígado/metabolismo , Hígado/patología , Lisofosfolípidos/antagonistas & inhibidores , Masculino , Ratones , Ratones Endogámicos C57BL , Fosfoserina/análogos & derivados , Fosfoserina/farmacología , ARN Mensajero/antagonistas & inhibidores , ARN Mensajero/metabolismo , Daño por Reperfusión/metabolismo , Daño por Reperfusión/patología , Transducción de Señal/efectos de los fármacos , Esfingosina/antagonistas & inhibidores , Esfingosina/metabolismo
11.
Commun Biol ; 2: 59, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30775460

RESUMEN

Bacterial pore-forming toxin aerolysin-like proteins (ALPs) are widely distributed in animals and plants. However, functional studies on these ALPs remain in their infancy. ßγ-CAT is the first example of a secreted pore-forming protein that functions to modulate the endolysosome pathway via endocytosis and pore formation on endolysosomes. However, the specific cell surface molecules mediating the action of ßγ-CAT remain elusive. Here, the actions of ßγ-CAT were largely attenuated by either addition or elimination of acidic glycosphingolipids (AGSLs). Further study revealed that the ALP and trefoil factor (TFF) subunits of ßγ-CAT bind to gangliosides and sulfatides, respectively. Additionally, disruption of lipid rafts largely impaired the actions of ßγ-CAT. Finally, the ability of ßγ-CAT to clear pathogens was attenuated in AGSL-eliminated frogs. These findings revealed a previously unknown double binding pattern of an animal-secreted ALP in complex with TFF that initiates ALP-induced endolysosomal pathway regulation, ultimately leading to effective antimicrobial responses.


Asunto(s)
Glicoesfingolípidos Acídicos/química , Proteínas Anfibias/inmunología , Toxinas Bacterianas/inmunología , Infecciones por Bacterias Gramnegativas/inmunología , Lisosomas/inmunología , Complejos Multiproteicos/inmunología , Proteínas Citotóxicas Formadoras de Poros/inmunología , Factor Trefoil-3/inmunología , Glicoesfingolípidos Acídicos/antagonistas & inhibidores , Glicoesfingolípidos Acídicos/biosíntesis , Aeromonas hydrophila/crecimiento & desarrollo , Aeromonas hydrophila/patogenicidad , Proteínas Anfibias/genética , Proteínas Anfibias/metabolismo , Animales , Anuros , Toxinas Bacterianas/genética , Toxinas Bacterianas/metabolismo , Ceramidas/antagonistas & inhibidores , Ceramidas/biosíntesis , Ceramidas/química , Cerebrósidos/antagonistas & inhibidores , Cerebrósidos/biosíntesis , Cerebrósidos/química , Gangliósidos/antagonistas & inhibidores , Gangliósidos/biosíntesis , Gangliósidos/química , Expresión Génica , Infecciones por Bacterias Gramnegativas/genética , Infecciones por Bacterias Gramnegativas/microbiología , Humanos , Interleucina-1beta/biosíntesis , Lisosomas/efectos de los fármacos , Lisosomas/microbiología , Microdominios de Membrana/efectos de los fármacos , Microdominios de Membrana/inmunología , Microdominios de Membrana/microbiología , Meperidina/análogos & derivados , Meperidina/farmacología , Complejos Multiproteicos/genética , Complejos Multiproteicos/metabolismo , Proteínas Citotóxicas Formadoras de Poros/genética , Proteínas Citotóxicas Formadoras de Poros/metabolismo , Esfingosina/antagonistas & inhibidores , Esfingosina/biosíntesis , Esfingosina/química , Células THP-1 , Factor Trefoil-3/genética , Factor Trefoil-3/metabolismo
12.
Mol Cancer Ther ; 18(4): 856-867, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30787172

RESUMEN

Inhibition of VEGFR signaling is an effective treatment for renal cell carcinoma, but resistance continues to be a major problem. Recently, the sphingosine phosphate (S1P) signaling pathway has been implicated in tumor growth, angiogenesis, and resistance to antiangiogenic therapy. S1P is a bioactive lipid that serves an essential role in developmental and pathologic angiogenesis via activation of the S1P receptor 1 (S1P1). S1P1 signaling counteracts VEGF signaling and is required for vascular stabilization. We used in vivo and in vitro angiogenesis models including a postnatal retinal angiogenesis model and a renal cell carcinoma murine tumor model to test whether simultaneous inhibition of S1P1 and VEGF leads to improved angiogenic inhibition. Here, we show that inhibition of S1P signaling reduces the endothelial cell barrier and leads to excessive angiogenic sprouting. Simultaneous inhibition of S1P and VEGF signaling further disrupts the tumor vascular beds, decreases tumor volume, and increases tumor cell death compared with monotherapies. These studies suggest that inhibition of angiogenesis at two stages of the multistep process may maximize the effects of antiangiogenic therapy. Together, these data suggest that combination of S1P1 and VEGFR-targeted therapy may be a useful therapeutic strategy for the treatment of renal cell carcinoma and other tumor types.


Asunto(s)
Carcinoma de Células Renales/tratamiento farmacológico , Carcinoma de Células Renales/metabolismo , Neoplasias Renales/tratamiento farmacológico , Neoplasias Renales/metabolismo , Receptores de Esfingosina-1-Fosfato/antagonistas & inhibidores , Receptor 2 de Factores de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Inhibidores de la Angiogénesis/farmacología , Animales , Anticuerpos Monoclonales/farmacología , Carcinoma de Células Renales/irrigación sanguínea , Carcinoma de Células Renales/patología , Línea Celular Tumoral , Quimioterapia Combinada , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Femenino , Humanos , Neoplasias Renales/irrigación sanguínea , Neoplasias Renales/patología , Lisofosfolípidos/antagonistas & inhibidores , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Desnudos , Neovascularización Patológica/tratamiento farmacológico , Esfingosina/análogos & derivados , Esfingosina/antagonistas & inhibidores , Sunitinib/farmacología , Resultado del Tratamiento , Carga Tumoral/efectos de los fármacos , Factor A de Crecimiento Endotelial Vascular/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto
13.
Biochem Cell Biol ; 96(6): 742-751, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-29940125

RESUMEN

Idiopathic pulmonary fibrosis is an agnogenic chronic disorder with high morbidity and low survival rate. Quercetin is a flavonoid found in a variety of herbs with anti-fibrosis function. In this study, bleomycin was employed to induce a pulmonary fibrosis mouse model. The quercetin administration ameliorated bleomycin-induced pulmonary fibrosis, evidenced by the expression level changes of hydroxyproline, fibronectin, α-smooth muscle actin, Collagen I, and Collagen III. Similar results were observed in transforming growth factor (TGF)-ß-treated human embryonic lung fibroblast (HELF). The bleomycin or TGF-ß administration caused the increase of sphingosine-1-phosphate (S1P) level in pulmonary tissue and HELF cells, as well as its activation-required kinase, sphingosine kinase 1 (SphK1), and its degradation enzyme, sphinogosine-1-phosphate lyase (S1PL). However, the increase of S1P, SphK1, and S1PL was attenuated by application of quercetin. In addition, the effect of quercetin on fibrosis was abolished by the ectopic expression of SphK1. The colocalization of SphK1/S1PL and fibroblast specific protein 1 (FSP1) suggested the roles of fibroblasts in pulmonary fibrosis. In summary, we demonstrated that quercetin ameliorated pulmonary fibrosis in vivo and in vitro by inhibiting SphK1/S1P signaling.


Asunto(s)
Fibrosis Pulmonar Idiopática/tratamiento farmacológico , Lisofosfolípidos/antagonistas & inhibidores , Fosfotransferasas (Aceptor de Grupo Alcohol)/antagonistas & inhibidores , Quercetina/farmacología , Transducción de Señal/efectos de los fármacos , Esfingosina/análogos & derivados , Animales , Células Cultivadas , Humanos , Fibrosis Pulmonar Idiopática/metabolismo , Fibrosis Pulmonar Idiopática/patología , Lisofosfolípidos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Esfingosina/antagonistas & inhibidores , Esfingosina/metabolismo
14.
Curr Pharm Des ; 24(9): 983-988, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29384055

RESUMEN

BACKGROUND: Preterm birth (PTB), defined as birth before 37 completed weeks of gestation, occurs in up to 18 percent of births worldwide and accounts for the majority of perinatal morbidity and mortality. While the single most common cause of PTB has been identified as inflammation, safe and effective pharmacotherapy to prevent PTB has yet to be developed. METHODS: Our group has used an in vivo model of inflammation-driven PTB, biochemical methods, pharmacological approaches, a novel endothelin receptor antagonist that we synthesized and RNA knockdown to help establish the role of endothelin-1 (ET-1) in inflammation-associated PTB. Further, we have used our in vivo model to test whether sphingosine kinase, which acts downstream of ET-1, plays a role in PTB. RESULTS: We have shown that levels of endothelin converting enzyme-1 (ECE-1) and ET-1 are increased when PTB is induced in timed pregnant mice with lipopolysaccharide (LPS) and that blocking ET-1 action, pharmacologically or using ECE-1 RNA silencing, rescues LPS-induced mice from PTB. ET-1 activates the sphingosine kinase/sphingosine-1-phosphate (SphK/S1P) pathway. S1P, in turn, is an important signaling molecule in the proinflammatory response. Interestingly, we have shown that SphK inhibition also prevents LPS-induced PTB in timed pregnant mice. Further, we showed that SphK inhibition suppresses the ECE-1/ET-1 axis, implicating positive feedback regulation of the SphK/S1P/ECE-1/ET-1 axis. CONCLUSION: The ET-1/SphK/SIP pathway is a potential pharmacotherapeutic target for the prevention of PTB.


Asunto(s)
Endotelina-1/metabolismo , Inflamación/metabolismo , Lisofosfolípidos/metabolismo , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Nacimiento Prematuro/metabolismo , Esfingosina/análogos & derivados , Animales , Endotelina-1/antagonistas & inhibidores , Femenino , Humanos , Lipopolisacáridos/antagonistas & inhibidores , Lipopolisacáridos/farmacología , Lisofosfolípidos/antagonistas & inhibidores , Ratones , Fosfotransferasas (Aceptor de Grupo Alcohol)/antagonistas & inhibidores , Embarazo , Nacimiento Prematuro/prevención & control , Esfingosina/antagonistas & inhibidores , Esfingosina/metabolismo
15.
J Crohns Colitis ; 12(suppl_2): S678-S686, 2018 Aug 22.
Artículo en Inglés | MEDLINE | ID: mdl-28961752

RESUMEN

Sphingosine 1 phosphate [S1P] is a bioactive lipid mediator involved in the regulation of several cellular processes though the activation of a G protein-coupled receptor family known as the S1P receptors [S1PRs]. Advances in the understanding of the biological activities mediated by S1PRs have sparked great interest in the S1P/S1PRs axes as new therapeutic targets for the modulation of several cellular processes. In particular, the S1P/S1PR1 axis has been identified as key regulator for lymphocyte migration from lymph nodes. The blockade of this axis is emerging as a new therapeutic approach to control the aberrant leukocyte migration into the mucosa in inflammatory bowel disease [IBD]. This review briefly summarises the current evidence coming from clinical studies, and discusses the future prospects of S1P inhibitors for treatment of inflammatory bowel disease.


Asunto(s)
Inmunosupresores/uso terapéutico , Enfermedades Inflamatorias del Intestino/tratamiento farmacológico , Lisofosfolípidos/antagonistas & inhibidores , Esfingosina/análogos & derivados , Animales , Canfanos/uso terapéutico , Movimiento Celular/efectos de los fármacos , Clorhidrato de Fingolimod/uso terapéutico , Humanos , Inmunosupresores/efectos adversos , Indanos/uso terapéutico , Enfermedades Inflamatorias del Intestino/inmunología , Leucocitos/fisiología , Lisofosfolípidos/metabolismo , Terapia Molecular Dirigida , Oxadiazoles/uso terapéutico , Receptores de Lisoesfingolípidos/metabolismo , Transducción de Señal , Esfingosina/antagonistas & inhibidores , Esfingosina/metabolismo , Compuestos de Sulfhidrilo/uso terapéutico , Sulfonamidas/uso terapéutico
16.
Clin Pharmacol Ther ; 103(6): 1083-1092, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-28913826

RESUMEN

Ponesimod is a selective sphingosine-1-phosphate-1 (S1P1 ) receptor modulator currently under investigation for the treatment of multiple sclerosis. S1P receptor modulators reduce heart rate following treatment initiation. This effect disappears with repeated dosing, enabling development of innovative uptitration regimens to optimize patient safety. There are currently no published pharmacokinetic/pharmacodynamic models describing the heart rate reduction of S1P receptor modulators in humans. The model developed here provides quantification of this effect for ponesimod. A direct-effect Imax model with estimated maximum reduction of 45%, tolerance development, and circadian variation best described this effect. The pooled data from nine clinical studies enabled characterization of interindividual variability. The model was used to simulate different treatment regimens to compare the effect of high initial doses vs. gradual uptitration with respect to the occurrence of bradycardia. The results indicate a better safety profile when using gradual uptitration. The model allows studying dosing regimens not clinically tested in silico.


Asunto(s)
Tolerancia a Medicamentos/fisiología , Frecuencia Cardíaca/efectos de los fármacos , Lisofosfolípidos/antagonistas & inhibidores , Esfingosina/análogos & derivados , Tiazoles/farmacología , Adulto , Ritmo Circadiano , Relación Dosis-Respuesta a Droga , Monitoreo del Ambiente , Femenino , Humanos , Masculino , Esfingosina/antagonistas & inhibidores , Tiazoles/administración & dosificación , Tiazoles/farmacocinética
17.
Tumour Biol ; 39(4): 1010428317699133, 2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-28381169

RESUMEN

Elucidating the interaction between cancer and non-cancer cells, such as blood vessels, immune cells, and other stromal cells, in the tumor microenvironment is imperative in understanding the mechanisms underlying cancer progression and metastasis, which is expected to lead to the development of new therapeutics. Sphingosine-1-phosphate is a bioactive lipid mediator that promotes cell survival, proliferation, migration, angiogenesis/lymphangiogenesis, and immune responsiveness, which are all factors involved in cancer progression. Sphingosine-1-phosphate is generated inside cancer cells by sphingosine kinases and then exported into the tumor microenvironment. Although sphingosine-1-phosphate is anticipated to play an important role in the tumor microenvironment and cancer progression, determining sphingosine-1-phosphate levels in the tumor microenvironment has been difficult due to a lack of established methods. We have recently developed a method to measure sphingosine-1-phosphate levels in the interstitial fluid that bathes cancer cells in the tumor microenvironment, and reported that high levels of sphingosine-1-phosphate exist in the tumor interstitial fluid. Importantly, sphingosine-1-phosphate can be secreted from cancer cells and non-cancer components such as immune cells and vascular/lymphatic endothelial cells in the tumor microenvironment. Furthermore, sphingosine-1-phosphate affects both cancer and non-cancer cells in the tumor microenvironment promoting cancer progression. Here, we review the roles of sphingosine-1-phosphate in the interaction between cancer and non-cancer cells in tumor microenvironment, and discuss future possibilities for targeted therapies against sphingosine-1-phosphate signaling for cancer patients.


Asunto(s)
Lisofosfolípidos/fisiología , Neoplasias/etiología , Esfingosina/análogos & derivados , Microambiente Tumoral , Humanos , Lisofosfolípidos/análisis , Lisofosfolípidos/antagonistas & inhibidores , Neoplasias/terapia , Esfingosina/análisis , Esfingosina/antagonistas & inhibidores , Esfingosina/fisiología
18.
Gastroenterology ; 153(1): 233-248.e16, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28363640

RESUMEN

BACKGROUND & AIMS: There is growing interest in the use of bone marrow cells to treat liver fibrosis, however, little is known about their antifibrotic efficacy or the identity of their effector cell(s). Sphingosine-1-phosphate (S1P) mediates egress of immune cells from the lymphoid organs into the lymphatic vessels; we investigated its role in the response of hematopoietic stem cells (HSCs) to liver fibrosis in mice. METHODS: Purified (c-kit+/sca1+/lin-) HSCs were infused repeatedly into mice undergoing fibrotic liver injury. Chronic liver injury was induced in BoyJ mice by injection of carbon tetrachloride (CCl4) or placement on a methionine-choline-deficient diet. Some mice were irradiated and given transplants of bone marrow cells from C57BL6 mice, with or without the S1P antagonist FTY720; we then studied HSC mobilization and localization. Migration of HSC lines was quantified in Transwell assays. Levels of S1P in liver, bone marrow, and lymph fluid were measured using an enzyme-linked immunosorbent assay. Liver tissues were collected and analyzed by immunohistochemical quantitative polymerase chain reaction and sphingosine kinase activity assays. We performed quantitative polymerase chain reaction analyses of the expression of sphingosine kinase 1 and 2, sphingosine-1-phosphate lyase 1, and sphingosine-1-phosphate phosphatase 1 in normal human liver and cirrhotic liver from patients with alcohol-related liver disease (n = 6). RESULTS: Infusions of HSCs into mice with liver injury reduced liver scarring based on picrosirius red staining (49.7% reduction in mice given HSCs vs control mice; P < .001), and hepatic hydroxyproline content (328 mg/g in mice given HSCs vs 428 mg/g in control mice; P < .01). HSC infusion also reduced hepatic expression of α-smooth muscle actin (0.19 ± 0.007-fold compared with controls; P < .0001) and collagen type I α 1 chain (0.29 ± 0.17-fold compared with controls; P < .0001). These antifibrotic effects were maintained with infusion of lymphoid progenitors that lack myeloid potential and were associated with increased numbers of recipient neutrophils and macrophages in liver. In studies of HSC cell lines, we found HSCs to recruit monocytes, and this process to require C-C motif chemokine receptor 2. In fibrotic liver tissue from mice and patients, hepatic S1P levels increased owing to increased hepatic sphingosine kinase-1 expression, which contributed to a reduced liver:lymph S1P gradient and limited HSC egress from the liver. Mice given the S1P antagonist (FTY720) with HSCs had increased hepatic retention of HSCs (1697 ± 247 cells in mice given FTY720 vs 982 ± 110 cells in controls; P < .05), and further reductions in fibrosis. CONCLUSIONS: In studies of mice with chronic liver injury, we showed the antifibrotic effects of repeated infusions of purified HSCs. We found that HSCs promote recruitment of endogenous macrophages and neutrophils. Strategies to reduce SIP signaling and increase retention of HSCs in the liver could increase their antifibrotic activities and be developed for treatment of patients with liver fibrosis.


Asunto(s)
Movimiento Celular/efectos de los fármacos , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/fisiología , Cirrosis Hepática/prevención & control , Lisofosfolípidos/antagonistas & inhibidores , Lisofosfolípidos/metabolismo , Esfingosina/análogos & derivados , Actinas/metabolismo , Aldehído-Liasas/genética , Animales , Línea Celular , Enfermedad Hepática Crónica Inducida por Sustancias y Drogas/complicaciones , Colágeno Tipo I/metabolismo , Cadena alfa 1 del Colágeno Tipo I , Femenino , Clorhidrato de Fingolimod/uso terapéutico , Expresión Génica , Humanos , Inmunosupresores/uso terapéutico , Hígado/metabolismo , Hígado/patología , Cirrosis Hepática/etiología , Cirrosis Hepática/genética , Cirrosis Hepática/patología , Linfa/metabolismo , Macrófagos , Masculino , Proteínas de la Membrana/genética , Ratones , Monocitos , Neutrófilos , Monoéster Fosfórico Hidrolasas/genética , Fosfotransferasas (Aceptor de Grupo Alcohol)/genética , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Esfingosina/antagonistas & inhibidores , Esfingosina/metabolismo
19.
Invest Ophthalmol Vis Sci ; 58(4): 2258-2265, 2017 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-28418499

RESUMEN

Purpose: To investigate levels of sphingosine-1-phosphate (S1P) in aqueous fluid samples taken before and after filtration surgery and S1P-induced human conjunctival fibroblast (HCF) responses. Methods: Levels of S1P and its related sphingophospholipids in aqueous fluid obtained immediately before and after filtration surgery were determined by liquid chromatography-tandem mass spectrometry. HCFs were used for all in vitro experiments. The expression of five S1P receptor subtypes in HCFs was examined by quantitative real-time PCR. The effect of S1P and receptor-specific antagonists on HCF viability and cell migration was assessed by WST-1 assay and scratch migration assay, respectively. Differentiation to myofibroblasts and extracellular matrix production was evaluated by examining changes in F-actin, α-smooth muscle actin (αSMA), and collagen expression with immunocytochemistry, Western blotting, and collagen accumulation assay, respectively. Results: No significant S1P levels in the aqueous fluid samples were detectable immediately before surgery, but postoperative levels of several lysophospholipids, including S1P, dehydro-S1P, and sphingosine, were significantly increased to bioactive concentrations in aqueous fluid in the blebs (P < 0.0001). mRNA expression of the three main S1P receptor subtypes was detected in HCFs. Although S1P levels did not influence HCF proliferation, S1P enhanced cell migration, which could be inhibited by the S1P2 antagonist JTE 013. F-actin, αSMA, and collagen expression was significantly increased by S1P stimulation and was reduced by JTE 013. Conclusions: Bioactive S1P concentrations were present in the aqueous fluid at the end of filtration surgery. S1P activated HCFs via S1P2 receptors. These results revealed the potential of S1P2 antagonists in preventing scarring after glaucoma filtration surgery.


Asunto(s)
Conjuntiva/química , Fibroblastos/química , Cirugía Filtrante , Glaucoma/cirugía , Lisofosfolípidos/análisis , Esfingosina/análogos & derivados , Movimiento Celular/efectos de los fármacos , Supervivencia Celular , Células Cultivadas , Cromatografía Liquida , Colágeno/metabolismo , Conjuntiva/citología , Conjuntiva/metabolismo , Fibroblastos/metabolismo , Cirugía Filtrante/métodos , Glaucoma/metabolismo , Humanos , Lisofosfolípidos/antagonistas & inhibidores , Lisofosfolípidos/metabolismo , Esfingosina/análisis , Esfingosina/antagonistas & inhibidores , Esfingosina/metabolismo , Espectrometría de Masas en Tándem
20.
Cancer ; 123(4): 576-582, 2017 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-27727447

RESUMEN

BACKGROUND: Upregulation of sphingosine-1-phosphate (S1P) may mediate resistance to vascular endothelial growth factor (VEGF)-directed therapies and inhibit antitumor immunity. Antagonism of S1P in preclinical models appears to overcome this resistance. In this phase 2 study, the authors assessed the activity of sonepcizumab, a first-in-class inhibitor of S1P, in patients with metastatic renal cell carcinoma (mRCC) with a history of prior VEGF-directed therapy. METHODS: Patients were required to have clear cell mRCC and to have received treatment with at least 1 prior VEGF-directed agent. Prior treatment with immunotherapeutic agents and ≤1 mammalian target of rapamycin inhibitors was permitted. The primary endpoint of the study was progression-free survival. Additional endpoints included response rate and safety, and overall survival (OS) performed post hoc. RESULTS: A total of 40 patients were enrolled with a median of 3 prior therapies (range, 1-5 prior therapies), 78% of whom had intermediate-risk disease by second-line International Metastatic Renal Cell Carcinoma Database Consortium (IMDC) criteria. Although the current study did not achieve its primary endpoint based on the 2-month progression-free survival, a median OS of 21.7 months was observed. Four patients (10%) demonstrated a partial response, with a median duration of response of 5.9 months. No grade 3/4 treatment-related adverse events were observed in >5% of patients (adverse events were graded and recorded for each patient using Common Terminology Criteria for Adverse Events [version 4.0]); the most frequent grade 1/2 treatment-related adverse events were fatigue (30%), weight gain (18%), constipation (15%), and nausea (15%). Biomarker studies demonstrated an increase in S1P concentrations with therapy. Comprehensive genomic profiling of 3 patients with a clinical benefit of >24 months indicated von Hippel-Lindau (VHL) and polybromo-1 (PBRM1) alterations. CONCLUSIONS: The encouraging OS and favorable safety profile observed with sonepcizumab should prompt further investigation of the agent in combination with VEGF-directed agents or checkpoint inhibitors. Cancer 2017;123:576-582. © 2016 American Cancer Society.


Asunto(s)
Anticuerpos Monoclonales/administración & dosificación , Carcinoma de Células Renales/tratamiento farmacológico , Lisofosfolípidos/antagonistas & inhibidores , Esfingosina/análogos & derivados , Factor A de Crecimiento Endotelial Vascular/genética , Anciano , Anciano de 80 o más Años , Anticuerpos Monoclonales/efectos adversos , Biomarcadores de Tumor/genética , Carcinoma de Células Renales/genética , Carcinoma de Células Renales/inmunología , Carcinoma de Células Renales/patología , Proteínas de Unión al ADN , Supervivencia sin Enfermedad , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos/patología , Femenino , Humanos , Estimación de Kaplan-Meier , Lisofosfolípidos/inmunología , Masculino , Persona de Mediana Edad , Metástasis de la Neoplasia , Proteínas Nucleares/genética , Esfingosina/antagonistas & inhibidores , Esfingosina/inmunología , Factores de Transcripción/genética , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Proteína Supresora de Tumores del Síndrome de Von Hippel-Lindau/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA