Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 225
Filtrar
1.
Handb Exp Pharmacol ; 256: 19-49, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31302759

RESUMO

The BA-responsive GPCRs S1PR2 and TGR5 are almost ubiquitously expressed in human and rodent tissues. In the liver, S1PR2 is expressed in all cell types, while TGR5 is predominately found in non-parenchymal cells. In contrast to S1PR2, which is mainly activated by conjugated bile acids (BAs), all BAs serve as ligands for TGR5 irrespective of their conjugation state and substitution pattern.Mice with targeted deletion of either S1PR2 or TGR5 are viable and develop no overt phenotype. In liver injury models, S1PR2 exerts pro-inflammatory and pro-fibrotic effects and thus aggravates liver damage, while TGR5 mediates anti-inflammatory, anti-cholestatic, and anti-fibrotic effects. Thus, inhibitors of S1PR2 signaling and agonists for TGR5 have been employed to attenuate liver injury in rodent models for cholestasis, nonalcoholic steatohepatitis, and fibrosis/cirrhosis.In biliary epithelial cells, both receptors activate a similar signaling cascade resulting in ERK1/2 phosphorylation and cell proliferation. Overexpression of both S1PR2 and TGR5 was found in human cholangiocarcinoma tissue as well as in CCA cell lines, where stimulation of both GPCRs resulted in transactivation of the epidermal growth factor receptor and triggered cell proliferation as well as increased cell migration and invasiveness.This chapter will focus on the function of S1PR2 and TGR5 in different liver cell types and summarizes current knowledge on the role of these receptors in liver disease models.


Assuntos
Ácidos e Sais Biliares/fisiologia , Receptores Acoplados a Proteínas G/fisiologia , Animais , Neoplasias dos Ductos Biliares , Colangiocarcinoma , Humanos , Fígado , Hepatopatias , Camundongos , Receptores de Lisoesfingolipídeo/fisiologia , Receptores de Esfingosina-1-Fosfato
2.
Biochim Biophys Acta Mol Cell Res ; 1866(4): 554-565, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30611767

RESUMO

Hearing loss is among the most prevalent sensory impairments in humans. Cochlear implantable devices represent the current therapies for hearing loss but have various shortcomings. ERM (ezrin- radixin -moesin) are a family of adaptor proteins that link plasma membrane with actin cytoskeleton, playing a crucial role in cell morphology and in the formation of membrane protrusions. Recently, bioactive sphingolipids have emerged as regulators of ERM proteins. Sphingosine 1-phosphate (S1P) is a pleiotropic sphingolipid which regulates fundamental cellular functions such as proliferation, survival, migration as well as processes such as development and inflammation mainly via ligation to its specific receptors S1PR (S1P1-5). Experimental findings demonstrate a key role for S1P signaling axis in the maintenance of auditory function. Preservation of cellular junctions is a fundamental function both for S1P and ERM proteins, crucial for the maintenance of cochlear integrity. In the present work, S1P was found to activate ERM in a S1P2-dependent manner in murine auditory epithelial progenitors US/VOT-E36. S1P-induced ERM activation potently contributed to actin cytoskeletal remodeling and to the appearance of ionic currents and membrane passive properties changes typical of more differentiated cells. Moreover, PKC and Akt activation was found to mediate S1P-induced ERM phosphorylation. The obtained findings contribute to demonstrate the role of S1P signaling pathway in inner ear biology and to disclose potential innovative therapeutical approaches in the field of hearing loss prevention and treatment.


Assuntos
Citoesqueleto de Actina/metabolismo , Cóclea/citologia , Proteínas do Citoesqueleto/metabolismo , Células Epiteliais/fisiologia , Lisofosfolipídeos/fisiologia , Esfingosina/análogos & derivados , Animais , Linhagem Celular , Membrana Celular/fisiologia , Fenômenos Eletrofisiológicos , Células Epiteliais/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Proteínas de Membrana/metabolismo , Camundongos , Proteínas dos Microfilamentos/metabolismo , Proteína Quinase C/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptores de Lisoesfingolipídeo/fisiologia , Esfingosina/fisiologia , Receptores de Esfingosina-1-Fosfato , Células-Tronco/fisiologia , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
3.
FASEB J ; 33(2): 1711-1726, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30188757

RESUMO

Sphingosine-1-phosphate (S1P) is involved in the regulation of important cellular processes, including immune-cell trafficking and proliferation. Altered S1P signaling is strongly associated with inflammation, cancer progression, and atherosclerosis; however, the mechanisms underlying its pathophysiologic effects are only partially understood. This study evaluated the effects of S1P in vitro and in vivo on the biosynthesis of leukotrienes (LTs), which form a class of lipid mediators involved in the pathogenesis of inflammatory diseases. Here, we report for the first time that S1P potently suppresses LT biosynthesis in Ca2+-ionophore-stimulated intact human neutrophils. S1P treatment resulted in intracellular Ca2+ mobilization, perinuclear translocation, and finally irreversible suicide inactivation of the LT biosynthesis key enzyme 5-lipoxygenase (5-LO). Agonist studies and S1P receptor mRNA expression analysis provided evidence for a S1P receptor 4-mediated effect, which was confirmed by a functional knockout of S1P4 in HL60 cells. Systemic administration of S1P in wild-type mice decreased both macrophage and neutrophil migration in the lungs in response to LPS and significantly attenuated 5-LO product formation, whereas these effects were abrogated in 5-LO or S1P4 knockout mice. In summary, targeting the 5-LO pathway is an important mechanism to explain S1P-mediated pathophysiologic effects. Furthermore, agonism at S1P4 represents a novel effective strategy in pharmacotherapy of inflammation.-Fettel, J., Kühn, B., Guillen, N. A., Sürün, D., Peters, M., Bauer, R., Angioni, C., Geisslinger, G., Schnütgen, F., Meyer zu Heringdorf, D., Werz, O., Meybohm, P., Zacharowski, K., Steinhilber, D., Roos, J., Maier, T. J. Sphingosine-1-phosphate (S1P) induces potent anti-inflammatory effects in vitro and in vivo by S1P receptor 4-mediated suppression of 5-lipoxygenase activity.


Assuntos
Anti-Inflamatórios/farmacologia , Araquidonato 5-Lipoxigenase/efeitos dos fármacos , Lisofosfolipídeos/farmacologia , Receptores de Lisoesfingolipídeo/fisiologia , Esfingosina/análogos & derivados , Animais , Araquidonato 5-Lipoxigenase/biossíntese , Araquidonato 5-Lipoxigenase/metabolismo , Ácido Araquidônico/metabolismo , Cálcio/metabolismo , Linhagem Celular , Feminino , Humanos , Lisofosfolipídeos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Neutrófilos/enzimologia , Neutrófilos/metabolismo , Pneumonia/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Receptores de Lisoesfingolipídeo/genética , Receptores de Lisoesfingolipídeo/metabolismo , Transdução de Sinais , Esfingosina/metabolismo , Esfingosina/farmacologia , Especificidade por Substrato
4.
Am J Pathol ; 188(12): 2811-2825, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30273600

RESUMO

Colonic inflammation, a hallmark of inflammatory bowel disease, can be influenced by host intrinsic and extrinsic factors. There continues to be a need for models of colonic inflammation that can both provide insights into disease pathogenesis and be used to investigate potential therapies. Herein, we tested the utility of colonoscopic-guided pinch biopsies in mice for studying colonic inflammation and its treatment. Gene expression profiling of colonic wound beds after injury showed marked changes, including increased expression of genes important for the inflammatory response. Interestingly, many of these gene expression changes mimicked those alterations found in inflammatory bowel disease patients. Biopsy-induced inflammation was associated with increases in neutrophils, macrophages, and natural killer cells. Injury also led to elevated levels of sphingosine-1-phosphate (S1P), a bioactive lipid that is an important mediator of inflammation mainly through its receptor, S1P1. Genetic deletion of S1P1 in the endothelium did not alter the inflammatory response but led to increased colonic bleeding. Bacteria invaded into the wound beds, raising the possibility that microbes contributed to the observed changes in mucosal gene expression. In support of this, reducing bacterial abundance markedly attenuated the inflammatory response to wounding. Taken together, this study demonstrates the utility of the pinch biopsy model of colonic injury to elucidate the molecular underpinnings of colonic inflammation and its treatment.


Assuntos
Colo/imunologia , Modelos Animais de Doenças , Inflamação/imunologia , Mucosa Intestinal/imunologia , Microbiota , Receptores de Lisoesfingolipídeo/fisiologia , Cirurgia Assistida por Computador/métodos , Animais , Antibacterianos/farmacologia , Biópsia , Células Cultivadas , Colo/lesões , Colo/cirurgia , Colonoscopia/métodos , Feminino , Perfilação da Expressão Gênica , Inflamação/metabolismo , Inflamação/microbiologia , Doenças Inflamatórias Intestinais , Mucosa Intestinal/efeitos dos fármacos , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiologia , Lisofosfolipídeos/metabolismo , Masculino , Camundongos , Camundongos Knockout , Esfingosina/análogos & derivados , Esfingosina/metabolismo , Receptores de Esfingosina-1-Fosfato
5.
Circ Res ; 122(8): 1069-1083, 2018 04 13.
Artigo em Inglês | MEDLINE | ID: mdl-29475983

RESUMO

RATIONALE: Multilineage-differentiating stress enduring (Muse) cells, pluripotent marker stage-specific embryonic antigen-3+ cells, are nontumorigenic endogenous pluripotent-like stem cells obtainable from various tissues including the bone marrow. Their therapeutic efficiency has not been validated in acute myocardial infarction. OBJECTIVE: The main objective of this study is to clarify the efficiency of intravenously infused rabbit autograft, allograft, and xenograft (human) bone marrow-Muse cells in a rabbit acute myocardial infarction model and their mechanisms of tissue repair. METHODS AND RESULTS: In vivo dynamics of Nano-lantern-labeled Muse cells showed preferential homing of the cells to the postinfarct heart at 3 days and 2 weeks, with ≈14.5% of injected GFP (green fluorescent protein)-Muse cells estimated to be engrafted into the heart at 3 days. The migration and homing of the Muse cells was confirmed pharmacologically (S1PR2 [sphingosine monophosphate receptor 2]-specific antagonist JTE-013 coinjection) and genetically (S1PR2-siRNA [small interfering ribonucleic acid]-introduced Muse cells) to be mediated through the S1P (sphingosine monophosphate)-S1PR2 axis. They spontaneously differentiated into cells positive for cardiac markers, such as cardiac troponin-I, sarcomeric α-actinin, and connexin-43, and vascular markers. GCaMP3 (GFP-based Ca calmodulin probe)-labeled Muse cells that engrafted into the ischemic region exhibited increased GCaMP3 fluorescence during systole and decreased fluorescence during diastole. Infarct size was reduced by ≈52%, and the ejection fraction was increased by ≈38% compared with vehicle injection at 2 months, ≈2.5 and ≈2.1 times higher, respectively, than that induced by mesenchymal stem cells. These effects were partially attenuated by the administration of GATA4-gene-silenced Muse cells. Muse cell allografts and xenografts efficiently engrafted and recovered functions, and allografts remained in the tissue and sustained functional recovery for up to 6 months without immunosuppression. CONCLUSIONS: Muse cells may provide reparative effects and robust functional recovery and may, thus, provide a novel strategy for the treatment of acute myocardial infarction.


Assuntos
Lisofosfolipídeos/fisiologia , Infarto do Miocárdio/cirurgia , Células-Tronco Pluripotentes/transplante , Receptores de Lisoesfingolipídeo/fisiologia , Esfingosina/análogos & derivados , Aloenxertos , Animais , Autoenxertos , Diferenciação Celular , Movimento Celular/fisiologia , Fator de Transcrição GATA4/antagonistas & inibidores , Fator de Transcrição GATA4/genética , Fator de Transcrição GATA4/fisiologia , Sobrevivência de Enxerto , Proteínas de Fluorescência Verde/análise , Xenoenxertos , Humanos , Luciferases/análise , Proteínas Luminescentes/análise , Masculino , Infarto do Miocárdio/patologia , Células-Tronco Pluripotentes/citologia , Células-Tronco Pluripotentes/metabolismo , Pirazóis/farmacologia , Piridinas/farmacologia , Interferência de RNA , RNA Interferente Pequeno/farmacologia , Coelhos , Receptores de Lisoesfingolipídeo/antagonistas & inibidores , Receptores de Lisoesfingolipídeo/genética , Proteínas Recombinantes de Fusão/análise , Especificidade da Espécie , Esfingosina/fisiologia , Receptores de Esfingosina-1-Fosfato
6.
Int J Mol Sci ; 19(2)2018 Jan 31.
Artigo em Inglês | MEDLINE | ID: mdl-29385066

RESUMO

Sphingolipids, sphingolipid metabolizing enzymes, and their receptors network are being recognized as part of the signaling mechanisms, which govern breast cancer cell growth, migration, and survival during chemotherapy treatment. Approximately 70% of breast cancers are estrogen receptor (ER) positive and, thus, rely on estrogen signaling. Estrogen activates an intracellular network composed of many cytoplasmic and nuclear mediators. Some estrogen effects can be mediated by sphingolipids. Estrogen activates sphingosine kinase 1 (SphK1) and amplifies the intracellular concentration of sphingosine-1-phosphate (S1P) in breast cancer cells during stimulation of proliferation and survival. Specifically, Estrogen activates S1P receptors (S1PR) and induces growth factor receptor transactivation. SphK, S1P, and S1PR expression are causally associated with endocrine resistance and progression to advanced tumor stages in ER-positive breast cancers in vivo. Recently, the network of SphK/S1PR was shown to promote the development of ER-negative cancers and breast cancer stem cells, as well as stimulating angiogenesis. Novel findings confirm and broaden our knowledge about the cross-talk between sphingolipids and estrogen network in normal and malignant cells. Current S1PRs therapeutic inhibition was indicated as a promising chemotherapy approach in non-responsive and advanced malignancies. Considering that sphingolipid signaling has a prominent role in terminally differentiated cells, the impact should be considered when designing specific SphK/S1PR inhibitors. This study analyzes the dynamic of the transformation of sphingolipid axis during a transition from normal to pathological condition on the level of the whole organism. The sphingolipid-based mediation and facilitation of global effects of estrogen were critically accented as a bridging mechanism that should be explored in cancer prevention.


Assuntos
Neoplasias da Mama/metabolismo , Estrogênios/metabolismo , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Receptores de Lisoesfingolipídeo/metabolismo , Transdução de Sinais , Animais , Neoplasias da Mama/fisiopatologia , Feminino , Humanos , Masculino , Camundongos , Fosfotransferases (Aceptor do Grupo Álcool)/fisiologia , Receptores de Lisoesfingolipídeo/fisiologia , Células-Tronco/metabolismo , Células-Tronco/fisiologia
7.
Clin Pharmacol Drug Dev ; 7(3): 263-276, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-28783871

RESUMO

Ozanimod is a novel, selective, oral sphingosine-1-phosphate (1 and 5) receptor modulator in development for multiple sclerosis and inflammatory bowel disease. This randomized, double-blind, placebo-controlled, positive-controlled, parallel-group thorough QT study characterized the effects of ozanimod on cardiac repolarization in healthy subjects. Eligible subjects were randomized to 1 of 2 groups: ozanimod (escalated from 0.25 to 2 mg over 14 days) or placebo (for 14 days). A single dose of moxifloxacin 400 mg or placebo was administered on days 2 and 17. The primary end point was the time-matched, placebo-corrected, baseline-adjusted mean QTcF (ΔΔQTcF). A total of 113/124 (91.1%) subjects completed the study. The upper limits of the 2-sided 90% confidence intervals for ΔΔQTcF for both ozanimod 1 and 2 mg were below the 10-millisecond regulatory threshold. No QTcF >480 milliseconds or postdose change in QTcF of >60 milliseconds was observed. There was no evidence of a positive relationship between concentrations of ozanimod and its active metabolites and ΔΔQTcF. Although ozanimod blunted the observed diurnal increase in heart rate, excursions below predose heart rates were no greater than with placebo. Results demonstrate that ozanimod does not prolong the QTc interval or cause clinically significant bradycardia, supporting ozanimod's evolving favorable cardiac safety profile.


Assuntos
Eletrocardiografia/efeitos dos fármacos , Frequência Cardíaca/efeitos dos fármacos , Indanos/farmacologia , Oxidiazóis/farmacologia , Receptores de Lisoesfingolipídeo/fisiologia , Adulto , Método Duplo-Cego , Eletrocardiografia/métodos , Feminino , Frequência Cardíaca/fisiologia , Humanos , Indanos/efeitos adversos , Masculino , Oxidiazóis/efeitos adversos
8.
Pharmacology ; 101(1-2): 72-75, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29131082

RESUMO

BACKGROUND: Aspirin plays a crucial role in the prevention of cardiovascular diseases. We previously described that aspirin has effects beyond inhibition of platelet aggregation, as it inhibited thrombin-mediated release of sphingosine-1-phosphate (S1P) from human platelets. S1P is a bioactive lipid with important functions on inflammation and apoptosis. In endothelial cells (EC), S1P is a key regulator of cell migration. In this study, we aimed to analyze the effects of aspirin on platelet-induced EC migration. METHODS: Human umbilical EC migration was measured by Boyden chamber assay. EC migration was induced by platelet supernatants of thrombin receptor-activating peptide-1 (AP1) stimulated platelets. To investigate the S1P receptor subtype that promotes EC migration, specific inhibitors of S1P receptor subtypes were applied. RESULTS: S1P induced EC migration in a concentration-dependent manner. EC migration induced by AP1-stimulated platelet supernatants was reduced by aspirin. S1P1 receptor inhibition almost completely abolished EC migration induced by activated platelets. The inhibition of S1P2 or S1P3 receptor had no effect. CONCLUSION: Aspirin inhibits EC migration induced by activated platelets that is in part due to S1P and mediated by the endothelial S1P1 receptor. The clinical significance of this novel mechanism of aspirin action has to be investigated in future studies.


Assuntos
Aspirina/farmacologia , Plaquetas/fisiologia , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Lisofosfolipídeos/fisiologia , Receptores de Lisoesfingolipídeo/fisiologia , Esfingosina/análogos & derivados , Anilidas/farmacologia , Movimento Celular , Células Cultivadas , Células Endoteliais da Veia Umbilical Humana/fisiologia , Humanos , Organofosfonatos/farmacologia , Receptores de Lisoesfingolipídeo/antagonistas & inibidores , Esfingosina/fisiologia
10.
J Endocrinol ; 235(3): 251-265, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-28970286

RESUMO

Aldosterone has been identified as an important factor in obesity-associated hypertension. Here, we investigated whether sphingosine-1-phosphate (S1P), which has previously been linked to obesity, increases aldosterone release. S1P-induced aldosterone release was determined in NCI H295R cells in the presence of S1P receptor (S1PR) antagonists. In vivo release of S1P (100-300 µg/kgbw) was investigated in pithed, lean Sprague Dawley (SD) rats, diet-obese spontaneous hypertensive rats (SHRs), as well as in lean or obese Zucker rats. Aldosterone secretion was increased in NCI H295R cells by S1P, the selective S1PR1 agonist SEW2871 and the selective S1PR2 antagonist JTE013. Treatment with the S1PR1 antagonist W146 or fingolimod and the S1PR1/3 antagonist VPbib2319 decreased baseline and/or S1P-stimulated aldosterone release. Compared to saline-treated SD rats, plasma aldosterone increased by ~50 pg/mL after infusing S1P. Baseline levels of S1P and aldosterone were higher in obese than in lean SHRs. Adrenal S1PR expression did not differ between chow- or CD-fed rats that had the highest S1PR1 and lowest S1PR4 levels. S1P induced a short-lasting increase in plasma aldosterone in obese, but not in lean SHRs. However, 2-ANOVA did not demonstrate any difference between lean and obese rats. S1P-induced aldosterone release was also similar between obese and lean Zucker rats. We conclude that S1P is a local regulator of aldosterone production. S1PR1 agonism induces an increase in aldosterone secretion, while stimulating adrenal S1PR2 receptor suppresses aldosterone production. A significant role of S1P in influencing aldosterone secretion in states of obesity seems unlikely.


Assuntos
Aldosterona/metabolismo , Lisofosfolipídeos/fisiologia , Obesidade/metabolismo , Receptores de Lisoesfingolipídeo/fisiologia , Esfingosina/análogos & derivados , Animais , Dieta Hiperlipídica , Humanos , Lisofosfolipídeos/metabolismo , Masculino , Obesidade/sangue , Obesidade/etiologia , Ratos , Ratos Endogâmicos SHR , Ratos Sprague-Dawley , Ratos Zucker , Receptores de Lisoesfingolipídeo/metabolismo , Transdução de Sinais/fisiologia , Esfingosina/metabolismo , Esfingosina/fisiologia , Células Tumorais Cultivadas
11.
J Exp Med ; 214(9): 2695-2713, 2017 Sep 04.
Artigo em Inglês | MEDLINE | ID: mdl-28739604

RESUMO

Metastasis is the primary cause of cancer death. The inflammatory tumor microenvironment contributes to metastasis, for instance, by recruiting blood and lymph vessels. Among tumor-infiltrating immune cells, tumor-associated macrophages (TAMs) take a center stage in promoting both tumor angiogenesis and metastatic spread. We found that genetic deletion of the S1P receptor 1 (S1pr1) alone in CD11bhi CD206+ TAMs infiltrating mouse breast tumors prevents pulmonary metastasis and tumor lymphangiogenesis. Reduced lymphangiogenesis was also observed in the nonrelated methylcholanthrene-induced fibrosarcoma model. Transcriptome analysis of isolated TAMs from both entities revealed reduced expression of the inflammasome component Nlrp3 in S1PR1-deficient TAMs. Macrophage-dependent lymphangiogenesis in vitro was triggered upon inflammasome activation and required both S1PR1 signaling and IL-1ß production. Finally, NLRP3 expression in tumor-infiltrating macrophages correlated with survival, lymph node invasion, and metastasis of mammary carcinoma patients. Conceptually, our study indicates an unappreciated role of the NLRP3 inflammasome in promoting metastasis via the lymphatics downstream of S1PR1 signaling in macrophages.


Assuntos
Interleucina-1beta/fisiologia , Linfangiogênese/fisiologia , Macrófagos/fisiologia , Proteína 3 que Contém Domínio de Pirina da Família NLR/fisiologia , Metástase Neoplásica/fisiopatologia , Receptores de Lisoesfingolipídeo/fisiologia , Animais , Neoplasias da Mama/patologia , Neoplasias da Mama/fisiopatologia , Feminino , Fibrossarcoma/fisiopatologia , Humanos , Metástase Linfática , Neoplasias Mamárias Experimentais/fisiopatologia , Camundongos , Camundongos Endogâmicos C57BL , Receptores de Esfingosina-1-Fosfato
12.
J Pathol ; 242(1): 62-72, 2017 05.
Artigo em Inglês | MEDLINE | ID: mdl-28240350

RESUMO

Undifferentiated nasopharyngeal carcinoma (NPC) is a cancer with high metastatic potential that is consistently associated with Epstein-Barr virus (EBV) infection. In this study, we have investigated the functional contribution of sphingosine-1-phosphate (S1P) signalling to the pathogenesis of NPC. We show that EBV infection or ectopic expression of the EBV-encoded latent genes (EBNA1, LMP1, and LMP2A) can up-regulate sphingosine kinase 1 (SPHK1), the key enzyme that produces S1P, in NPC cell lines. Exogenous addition of S1P promotes the migration of NPC cells through the activation of AKT; shRNA knockdown of SPHK1 resulted in a reduction in the levels of activated AKT and inhibition of cell migration. We also show that S1P receptor 3 (S1PR3) mRNA is overexpressed in EBV-positive NPC patient-derived xenografts and a subset of primary NPC tissues, and that knockdown of S1PR3 suppressed the activation of AKT and the S1P-induced migration of NPC cells. Taken together, our data point to a central role for EBV in mediating the oncogenic effects of S1P in NPC and identify S1P signalling as a potential therapeutic target in this disease. Copyright © 2017 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Assuntos
Infecções por Vírus Epstein-Barr/complicações , Lisofosfolipídeos/fisiologia , Neoplasias Nasofaríngeas/virologia , Proteína Oncogênica v-akt/metabolismo , Receptores de Lisoesfingolipídeo/metabolismo , Esfingosina/análogos & derivados , Adulto , Idoso , Animais , Carcinoma , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Movimento Celular/fisiologia , Infecções por Vírus Epstein-Barr/genética , Infecções por Vírus Epstein-Barr/metabolismo , Infecções por Vírus Epstein-Barr/patologia , Feminino , Regulação Neoplásica da Expressão Gênica/fisiologia , Técnicas de Silenciamento de Genes , Xenoenxertos , Humanos , Lisofosfolipídeos/farmacologia , Masculino , Pessoa de Meia-Idade , Carcinoma Nasofaríngeo , Neoplasias Nasofaríngeas/genética , Neoplasias Nasofaríngeas/metabolismo , Neoplasias Nasofaríngeas/patologia , RNA Mensageiro/genética , Receptores de Lisoesfingolipídeo/genética , Receptores de Lisoesfingolipídeo/fisiologia , Transdução de Sinais/fisiologia , Esfingosina/farmacologia , Esfingosina/fisiologia , Receptores de Esfingosina-1-Fosfato , Regulação para Cima
13.
Clin Pharmacokinet ; 56(4): 395-408, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-27638335

RESUMO

BACKGROUND: Ponesimod is a selective, orally active sphingosine-1-phosphate receptor 1 modulator currently undergoing clinical evaluation for the treatment of multiple sclerosis (MS) in phase III clinical trials. Ponesimod dose-dependently reduces peripheral blood lymphocyte counts by blocking the egress of lymphocytes from lymphoid organs. METHODS: A population pharmacokinetic (PK) analysis was performed based on pooled data from 13 clinical studies. Interindividual variability (IIV) and the impact of key demographic variables and other covariates on ponesimod exposure were assessed quantitatively. RESULTS: A two-compartment model with sequential zero/first-order absorption, including lag time, intercompartmental drug flow, and first-order clearance, adequately described the PK of ponesimod. Body weight, race, MS, psoriasis, hepatic impairment, drug formulation, and food were identified to significantly affect the concentration-time profile. The inclusion of these covariates into the model explained approximately 25 % of the IIV in the PK of ponesimod. Model predictions indicated that the impact of the identified covariates on ponesimod steady-state exposure is within 20 % of exposure, and thus within the margins of the IIV, with the exception of hepatic impairment. Changes up to threefold were predicted for severe cases of liver dysfunction. CONCLUSION: The rich data set enabled building a comprehensive population PK model that accurately predicts the concentration-time data of ponesimod. Covariates other than hepatic impairment were considered not clinically relevant and thus do not require dose adjustment. A potential dose adaptation can be conducted based on the final model.


Assuntos
Ensaios Clínicos como Assunto , Interações Alimento-Droga/fisiologia , Modelos Biológicos , Receptores de Lisoesfingolipídeo/fisiologia , Tiazóis/farmacocinética , Ensaios Clínicos como Assunto/métodos , Composição de Medicamentos , Humanos , Tiazóis/química
14.
Beijing Da Xue Xue Bao Yi Xue Ban ; 48(6): 987-993, 2016 12 18.
Artigo em Chinês | MEDLINE | ID: mdl-27987502

RESUMO

OBJECTIVE: To construct sphingosine 1-phosphate receptor-1 (S1P1)-small interfering RNA (siRNA) lentiviral vectors and infect human salivary gland cells (HSG), and to investigate its possible therapy on Sjogren's syndrome. METHODS: HSG cells were divided into blank group, empty vector group, scramble-siRNA group and S1P1-siRNA group. The lentiviral vectors expressing siRNA against S1P1 and the pLL3.7 were respectively transfected into 293T cells with pMD2.G, pMDL g/p RRE, pRSV-REV to produce virus, and then infect HSG cells. The efficiency was observed by flow cytometry after the transfection for 48 h. The expression levels of S1P1 mRNA of HSG were detected by real-time RT-PCR and the expression of S1P1 protein was detected by immunohistochemistry method. The expression levels of interferon-γ (IFN-γ) and interleukin (IL)-17 in the supernatant of the cells were detected by ELISA method. RESULTS: (1) The scramble-siRNA, S1P1-siRNA lentiviral vector was successfully constructed, and the lentivirus titer was about 3.5×108 TU/mL. (2) The level of S1P1 mRNA was lower in S1P1-siRNA group than those in the blank group, empty vector group, and scramble-siRNA group 48 h after infection, there were significant differences between them (P<0.05). (3) The expression of S1P1 protein was lower in S1P1-siRNA group than those in blank group, empty vector group, and scramble-siRNA group 48 h after transfection, there were significant differences between them (P<0.05). (4) The levels of IL-17 were lower in S1P1-siRNA group than those in blank group, empty vector group, and scramble-siRNA group 48 h after transfection, there were significant differences between them (P<0.05). (5) The levels of IFN-γ in S1P1-siRNA group were lower than those in blank group, empty vector group, and scramble-siRNA group 48 h after transfection, there were significant differences between them (P<0.05). CONCLUSION: The lentiviral vector targeting S1P1 was successfully constructed. S1P1 siRNA could suppress the levels of S1P1 mRNA and protein, and decrease the expression of IL-17 and IFN-γ. S1P1 siRNA could infect HSG cells stably and inhibit the expression of S1P1 gene specifically and efficiently, and reduce the levels of inflammatory cytokines.


Assuntos
Regulação da Expressão Gênica/efeitos dos fármacos , Vetores Genéticos/farmacologia , RNA Interferente Pequeno/biossíntese , RNA Interferente Pequeno/farmacologia , Receptores de Lisoesfingolipídeo/efeitos dos fármacos , Receptores de Lisoesfingolipídeo/genética , Receptores de Lisoesfingolipídeo/fisiologia , Transfecção/métodos , Citocinas , Vetores Genéticos/administração & dosagem , Vetores Genéticos/biossíntese , Humanos , Técnicas In Vitro/métodos , Interferon gama/efeitos dos fármacos , Interferon gama/genética , Interleucina-17/genética , Lentivirus , RNA Mensageiro , Glândulas Salivares/efeitos dos fármacos
15.
Oncotarget ; 7(44): 71873-71886, 2016 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-27708249

RESUMO

The infiltration of melanoma tumors by macrophages is often correlated with poor prognosis. However, the molecular signals that regulate the dialogue between malignant cells and the inflammatory microenvironment remain poorly understood. We previously reported an increased expression of sphingosine kinase-1 (SK1), which produces the bioactive lipid sphingosine 1-phosphate (S1P), in melanoma. The present study aimed at defining the role of tumor SK1 in the recruitment and differentiation of macrophages in melanoma. Herein, we show that downregulation of SK1 in melanoma cells causes a reduction in the percentage of CD206highMHCIIlow M2 macrophages in favor of an increased proportion of CD206lowMHCIIhigh M1 macrophages into the tumor. This macrophage differentiation orchestrates T lymphocyte recruitment as well as tumor rejection through the expression of Th1 cytokines and chemokines. In vitro experiments indicated that macrophage migration is triggered by the binding of tumor S1P to S1PR1 receptors present on macrophages whereas macrophage differentiation is stimulated by SK1-induced secretion of TGF-ß1. Finally, RNA-seq analysis of human melanoma tumors revealed a positive correlation between SK1 and TGF-ß1 expression. Altogether, our findings demonstrate that melanoma SK1 plays a key role in the recruitment and phenotypic shift of the tumor macrophages that promote melanoma growth.


Assuntos
Macrófagos/fisiologia , Melanoma/imunologia , Fosfotransferases (Aceptor do Grupo Álcool)/fisiologia , Animais , Linhagem Celular Tumoral , Movimento Celular , Polaridade Celular , Proliferação de Células , Regulação para Baixo , Humanos , Melanoma/patologia , Camundongos , Camundongos Endogâmicos C57BL , Receptores de Lisoesfingolipídeo/fisiologia , Receptores de Esfingosina-1-Fosfato , Fator de Crescimento Transformador beta1/fisiologia
16.
Biopharm Drug Dispos ; 37(9): 561-573, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-27764535

RESUMO

Pharmacokinetic (PK) and pharmacodynamic (PD) modeling was conducted for the reduction of peripheral lymphocytes after oral administration of CS-0777 to healthy rats, monkeys and experimental autoimmune encephalomyelitis (EAE) induced rats. The phosphorylated active metabolite of CS-0777, M1, is a selective sphingosine 1-phosphate receptor-1 modulator. A linear one- and two-compartment model with a reversible metabolism process characterized the time courses of CS-0777 and M1 concentrations in rats and monkeys, respectively. The relationship between lymphocyte counts and M1 concentrations in blood was well described by an indirect response model in all animals examined. An Imax of 0.815 and an IC50 of 6.58 nM in healthy rats, an Imax of 0.807 and an IC50 of 5.09 nM in the EAE rats, an Imax of 0.789 and an IC50 of 0.484 nM in monkeys were estimated by the indirect PD model. Since the IC50 values calculated in terms of the unbound plasma concentration in rats and monkeys were within a similar range, after correction of the IC50 in blood described above with the blood to plasma concentration ratio and the plasma free fraction of M1, it was revealed that there is no species difference in the essential activity of M1 against lymphocyte reduction. The sensitivity of the lymphocytes to M1 was not affected by the EAE status. Comparison of the simulated lymphocyte reduction in EAE rats after multiple dosing with CS-0777 and the actual EAE clinical scores implies that the significant suppressive effect on EAE did not require the elimination of all lymphocytes from the blood. Copyright © 2016 John Wiley & Sons, Ltd.


Assuntos
Amino Álcoois/administração & dosagem , Amino Álcoois/sangue , Linfócitos/efeitos dos fármacos , Linfócitos/metabolismo , Pirróis/administração & dosagem , Pirróis/sangue , Receptores de Lisoesfingolipídeo/fisiologia , Administração Oral , Animais , Relação Dose-Resposta a Droga , Feminino , Macaca fascicularis , Masculino , Ratos , Ratos Endogâmicos Lew , Ratos Sprague-Dawley , Ratos Wistar , Especificidade da Espécie
17.
Front Biosci (Landmark Ed) ; 21(7): 1296-313, 2016 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-27100508

RESUMO

Sphingosine-1-phosphate (S1P) is a bioactive sphingolipid metabolite generated by phosphorylation of sphingosine catalyzed by sphingosine kinase. S1P acts mainly through its high affinity G-protein-coupled receptors and participates in the regulation of multiple systems, including cardiovascular system. It has been shown that S1P signaling is involved in the regulation of cardiac chronotropy and inotropy and contributes to cardioprotection as well as cardiac remodeling; S1P signaling regulates vascular function, such as vascular tone and endothelial barrier, and possesses an anti-atherosclerotic effect; S1P signaling is also implicated in the regulation of blood pressure. Therefore, manipulation of S1P signaling may offer novel therapeutic approaches to cardiovascular diseases. As several S1P receptor modulators and sphingosine kinase inhibitors have been approved or under clinical trials for the treatment of other diseases, it may expedite the test and implementation of these S1P-based drugs in cardiovascular diseases.


Assuntos
Fenômenos Fisiológicos Cardiovasculares , Lisofosfolipídeos/fisiologia , Esfingosina/análogos & derivados , Animais , Pressão Sanguínea/fisiologia , Doença da Artéria Coronariana/fisiopatologia , Endotélio Vascular/fisiologia , Humanos , Natriurese/fisiologia , Receptores de Lisoesfingolipídeo/fisiologia , Transdução de Sinais , Esfingosina/fisiologia
18.
Blood ; 127(11): 1380-1, 2016 Mar 17.
Artigo em Inglês | MEDLINE | ID: mdl-26989186

RESUMO

Inactivating mutations in the sphingosine-1-phosphate (S1P) receptor 2 (S1PR2) promoter have been associated with the germinal center (GC) B-cell diffuse large B-cell lymphoma (GCB-DLBCL) subtype. In this issue of Blood, Flori et al have now identified S1PR2 as a tumor suppressor that is transcriptionally silenced by forkhead box protein 1 (FOXP1) in the aggressive, activated B-cell (ABC-DLBCL) subtype.


Assuntos
Fatores de Transcrição Forkhead/fisiologia , Linfoma Difuso de Grandes Células B/patologia , Proteínas de Neoplasias/fisiologia , Receptores de Lisoesfingolipídeo/fisiologia , Proteínas Repressoras/fisiologia , Transdução de Sinais/fisiologia , Animais , Humanos
19.
J Am Soc Nephrol ; 27(11): 3383-3393, 2016 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26961351

RESUMO

Epithelial and endothelial injury and a cascade of immune and interstitial cell activation in the kidney lead to AKI. After mild to moderate AKI, the epithelium can regenerate and restore kidney function, yet little is known about the endothelium during these repair processes. Sphingosine 1-phosphate receptor 1 (S1P1), a G protein-coupled receptor, is necessary for vascular homeostasis. Here, we used an inducible genetic approach in a mouse model of AKI, ischemia-reperfusion injury (IRI), to determine the temporal effects of endothelial S1P1 during AKI. Deletion of endothelial S1P1 before IRI exacerbated kidney injury and inflammation, and the delayed deletion of S1P1 after IRI prevented kidney recovery, resulting in chronic inflammation and progressive fibrosis. Specifically, S1P1 directly suppressed endothelial activation of leukocyte adhesion molecule expression and inflammation. Altogether, the data indicate activation of endothelial S1P1 is necessary to protect from IRI and permit recovery from AKI. Endothelial S1P1 may be a therapeutic target for the prevention of early injury as well as prevention of progressive kidney fibrosis after AKI.


Assuntos
Injúria Renal Aguda/prevenção & controle , Receptores de Lisoesfingolipídeo/fisiologia , Receptores de Lisoesfingolipídeo/uso terapêutico , Animais , Endotélio , Fibrose/prevenção & controle , Rim/irrigação sanguínea , Rim/patologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Recuperação de Função Fisiológica , Traumatismo por Reperfusão/prevenção & controle
20.
Proc Natl Acad Sci U S A ; 113(5): 1351-6, 2016 Feb 02.
Artigo em Inglês | MEDLINE | ID: mdl-26787880

RESUMO

Blunting immunopathology without abolishing host defense is the foundation for safe and effective modulation of infectious and autoimmune diseases. Sphingosine 1-phosphate receptor 1 (S1PR1) agonists are effective in treating infectious and multiple autoimmune pathologies; however, mechanisms underlying their clinical efficacy are yet to be fully elucidated. Here, we uncover an unexpected mechanism of convergence between S1PR1 and interferon alpha receptor 1 (IFNAR1) signaling pathways. Activation of S1PR1 signaling by pharmacological tools or endogenous ligand sphingosine-1 phosphate (S1P) inhibits type 1 IFN responses that exacerbate numerous pathogenic conditions. Mechanistically, S1PR1 selectively suppresses the type I IFN autoamplification loop in plasmacytoid dendritic cells (pDCs), a specialized DC subset, for robust type I IFN release. S1PR1 agonist suppression is pertussis toxin-resistant, but inhibited by an S1PR1 C-terminal-derived transactivating transcriptional activator (Tat)-fusion peptide that blocks receptor internalization. S1PR1 agonist treatment accelerates turnover of IFNAR1, suppresses signal transducer and activator of transcription 1 (STAT1) phosphorylation, and down-modulates total STAT1 levels, thereby inactivating the autoamplification loop. Inhibition of S1P-S1PR1 signaling in vivo using the selective antagonist Ex26 significantly elevates IFN-α production in response to CpG-A. Thus, multiple lines of evidence demonstrate that S1PR1 signaling sets the sensitivity of pDC amplification of IFN responses, thereby blunting pathogenic immune responses. These data illustrate a lipid G-protein coupled receptor (GPCR)-IFNAR1 regulatory loop that balances effective and detrimental immune responses and elevated endogenous S1PR1 signaling. This mechanism will likely be advantageous in individuals subject to a range of inflammatory conditions.


Assuntos
Células Dendríticas/metabolismo , Interferon-alfa/metabolismo , Receptor de Interferon alfa e beta/metabolismo , Receptores de Lisoesfingolipídeo/fisiologia , Animais , Camundongos , Camundongos Knockout , Proteólise , Receptor de Interferon alfa e beta/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...