Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 185
Filtrar
1.
Tohoku J Exp Med ; 262(3): 173-180, 2024 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-38123304

RESUMEN

SKI-349 is a novel sphingosine kinases (SPHK) inhibitor with anti-tumor effects. This study aimed to assess the effect of SKI-349 on cell biological behaviors, downstream pathways, and its synergistic effect with sorafenib in hepatocellular carcinoma (HCC). HCC cell lines (Huh7 and Hep3B) were treated with SKI-349 at concentrations of 1, 2, 4, or 8 µM. Then, SPHK1/2 activity, cell viability, proliferation, apoptosis, invasion, and protein expressions of phosphorylated-protein kinase B (p-AKT), AKT, phosphorylated-mammalian target of rapamycin (p-mTOR) and mTOR were detected. Combination index values of SKI-349 (0, 1, 2, 4, or 8 µM) and sorafenib (0, 2.5, 5, 10, or 20 µM) were calculated. SKI-349 decreased the relative SPHK1 and SPHK2 activity compared with blank control in a dose-dependent manner in the Huh7 and Hep3B cell lines. Meanwhile, SKI-349 reduced cell viability, 5-ethynyl-2'-deoxyuridine (EdU) positive cells, and invasive cells, while it increased apoptotic cells compared to blank control in a dose-dependent manner in Huh7 and Hep3B cell lines. Based on the western blot assay, SKI-349 decreased the ratio of p-AKT to AKT and that of p-mTOR to mTOR compared with blank control in a dose-dependent manner in the Huh7 and Hep3B cell lines. Additionally, SKI-349 combined with sorafenib declined cell viability with concentration gradient effects compared to SKI-349 sole treatment, and they had synergistic cytotoxic effects in Huh7 and Hep3B cell lines. SKI-349 suppresses SPHK1 and SPHK2 activity, cell viability, invasion, and AKT/mTOR signaling pathway, as well as exhibits a synergistic cytotoxic effect with sorafenib in HCC.


Asunto(s)
Antineoplásicos , Carcinoma Hepatocelular , Neoplasias Hepáticas , Humanos , Carcinoma Hepatocelular/tratamiento farmacológico , Carcinoma Hepatocelular/patología , Sorafenib/farmacología , Sorafenib/uso terapéutico , Esfingosina/farmacología , Esfingosina/uso terapéutico , Proteínas Proto-Oncogénicas c-akt/metabolismo , Supervivencia Celular , Neoplasias Hepáticas/tratamiento farmacológico , Neoplasias Hepáticas/patología , Niacinamida/farmacología , Compuestos de Fenilurea/farmacología , Compuestos de Fenilurea/uso terapéutico , Línea Celular Tumoral , Transducción de Señal , Antineoplásicos/farmacología , Serina-Treonina Quinasas TOR/metabolismo , Serina-Treonina Quinasas TOR/uso terapéutico , Apoptosis , Proliferación Celular
2.
Adv Sci (Weinh) ; 9(14): e2104452, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35289120

RESUMEN

SPHK1 (sphingosine kinase-1) catalyzes the phosphorylation of sphingosine to sphingosine-1-phosphate (S1P), is found to be highly expressed in solid tumors. Here, extracellular vesicles (EVs) are identified as the key transporters of SPHK1 to the tumor microenvironment. Consequently, SPHK1-packaged EVs elevate S1P levels in the tumor microenvironment, where S1P appears as an immunosuppressive agent. However, the exact mechanism of how S1P mediates its immunosuppressive effects in cancer is not understood. It is investigated that S1P can induce T cell exhaustion. S1P can also upregulate programmed death ligand-1 (PDL-1) expression through E2F1-mediated transcription. Notably, an SPHK1 inhibitor PF543 improves T cell-mediated cytotoxicity. Furthermore, combining PF543 with an anti-PD-1 antibody reduces tumor burden and metastasis more effectively than PF543 alone in vivo. These data demonstrate a previously unrecognized mechanism of how SPHK1-packaged EVs contribute to the progression of ovarian cancer and thus present the potential clinical application of inhibiting SPHK1/S1P signaling to improve immune checkpoint blockage (anti-PD-1 antibody) therapy in ovarian cancer.


Asunto(s)
Vesículas Extracelulares , Neoplasias Ováricas , Carcinoma Epitelial de Ovario , Vesículas Extracelulares/metabolismo , Femenino , Humanos , Inmunoterapia , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/metabolismo , Receptores de Lisoesfingolípidos/metabolismo , Receptores de Lisoesfingolípidos/uso terapéutico , Esfingosina/metabolismo , Esfingosina/uso terapéutico , Linfocitos T/metabolismo , Linfocitos T/patología , Microambiente Tumoral
3.
Cells ; 10(5)2021 05 04.
Artículo en Inglés | MEDLINE | ID: mdl-34064516

RESUMEN

Sphingolipids are important structural membrane components and, together with cholesterol, are often organized in lipid rafts, where they act as signaling molecules in many cellular functions. They play crucial roles in regulating pathobiological processes, such as cancer, inflammation, and infectious diseases. The bioactive metabolites ceramide, sphingosine-1-phosphate, and sphingosine have been shown to be involved in the pathogenesis of several microbes. In contrast to ceramide, which often promotes bacterial and viral infections (for instance, by mediating adhesion and internalization), sphingosine, which is released from ceramide by the activity of ceramidases, kills many bacterial, viral, and fungal pathogens. In particular, sphingosine is an important natural component of the defense against bacterial pathogens in the respiratory tract. Pathologically reduced sphingosine levels in cystic fibrosis airway epithelial cells are normalized by inhalation of sphingosine, and coating plastic implants with sphingosine prevents bacterial infections. Pretreatment of cells with exogenous sphingosine also prevents the viral spike protein of severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) from interacting with host cell receptors and inhibits the propagation of herpes simplex virus type 1 (HSV-1) in macrophages. Recent examinations reveal that the bactericidal effect of sphingosine might be due to bacterial membrane permeabilization and the subsequent death of the bacteria.


Asunto(s)
Infecciones Bacterianas/inmunología , Micosis/inmunología , Transducción de Señal/inmunología , Esfingosina/metabolismo , Virosis/inmunología , Animales , Infecciones Bacterianas/tratamiento farmacológico , Infecciones Bacterianas/metabolismo , Infecciones Bacterianas/microbiología , Pared Celular/efectos de los fármacos , Ceramidas/metabolismo , Modelos Animales de Enfermedad , Herpesvirus Humano 1/inmunología , Humanos , Lisofosfolípidos/metabolismo , Microdominios de Membrana/inmunología , Microdominios de Membrana/metabolismo , Micosis/tratamiento farmacológico , Micosis/metabolismo , Micosis/microbiología , SARS-CoV-2/inmunología , Esfingolípidos/metabolismo , Esfingosina/análogos & derivados , Esfingosina/farmacología , Esfingosina/uso terapéutico , Virosis/tratamiento farmacológico , Virosis/metabolismo , Virosis/virología
4.
FASEB J ; 33(4): 5181-5195, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30629456

RESUMEN

Because the association between sphingosine 1-phosphate (S1P)/apolipoprotein M (ApoM) and chronic kidney diseases has not been established, we investigated the involvement of S1P/ApoM in the phenotypes of IgA nephropathy in hyper-IgA (HIGA) mice. The overexpression of ApoM in adenoviral gene transfer ameliorated the phenotypes of IgA nephropathy in HIGA mice, whereas the knockdown of ApoM with siRNA caused deterioration. When ApoM-overexpressing HIGA mice were treated with VPC23019, an antagonist against S1P receptor 1 (S1P1) and 3 (S1P3), we observed that the protective effects of ApoM were reversed, whereas JTE013, an antagonist against S1P2, did not inhibit the effects. We also found that S1P bound to albumin accelerated the proliferation of MES13 cells and the fibrotic changes of HK2 cells, which were inhibited by JTE013, whereas S1P bound to ApoM suppressed these changes, which were inhibited by VPC23019. These results suggest that S1P bound to ApoM possesses properties protective against the phenotypes of IgA nephropathy through S1P1 and S1P3, whereas S1P bound to albumin exerts deteriorating effects through S1P2. ApoM may be useful as a therapeutic target to treat or retard the progression of IgA nephropathy.-Kurano, M., Tsuneyama, K., Morimoto, Y., Nishikawa, M., Yatomi, Y. Apolipoprotein M suppresses the phenotypes of IgA nephropathy in hyper-IgA mice.


Asunto(s)
Apolipoproteínas M/uso terapéutico , Glomerulonefritis por IGA/sangre , Glomerulonefritis por IGA/tratamiento farmacológico , Inmunoglobulina A/sangre , Lisofosfolípidos/uso terapéutico , Esfingosina/análogos & derivados , Animales , Western Blotting , Línea Celular , Creatinina/sangre , Creatinina/orina , Femenino , Glomerulonefritis por IGA/metabolismo , Glomerulonefritis por IGA/orina , Inmunoglobulina A/metabolismo , Ratones , Ratones Endogámicos BALB C , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Esfingosina/uso terapéutico , Receptores de Esfingosina-1-Fosfato/antagonistas & inhibidores , Receptores de Esfingosina-1-Fosfato/metabolismo
5.
Oxid Med Cell Longev ; 2018: 4397159, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29785244

RESUMEN

Fingolimod hydrochloride (FTY720) is a first-in-class of sphingosine-1-phosphate (S1P) receptor modulator approved to treat multiple sclerosis by its phosphorylated form (FTY720-P). Recently, a novel role of FTY720 as a potential anticancer drug has emerged. One of the anticancer mechanisms of FTY720 involves the induction of reactive oxygen species (ROS) and subsequent apoptosis, which is largely independent of its property as an S1P modulator. ROS have been considered as a double-edged sword in tumor initiation/progression. Intriguingly, prooxidant therapies have attracted much attention due to its efficacy in cancer treatment. These strategies include diverse chemotherapeutic agents and molecular targeted drugs such as sulfasalazine which inhibits the CD44v-xCT (cystine transporter) axis. In this review, we introduce our recent discoveries using a chemical genomics approach to uncover a signaling network relevant to FTY720-mediated ROS signaling and apoptosis, thereby proposing new potential targets for combination therapy as a means to enhance the antitumor efficacy of FTY720 as a ROS generator. We extend our knowledge by summarizing various measures targeting the vulnerability of cancer cells' defense mechanisms against oxidative stress. Future directions that may lead to the best use of FTY720 and ROS-targeted strategies as a promising cancer treatment are also discussed.


Asunto(s)
Apoptosis/efectos de los fármacos , Organofosfatos/farmacología , Especies Reactivas de Oxígeno/metabolismo , Esfingosina/análogos & derivados , Animales , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Humanos , Inmunosupresores/farmacología , Inmunosupresores/uso terapéutico , Organofosfatos/uso terapéutico , Esfingosina/farmacología , Esfingosina/uso terapéutico
6.
Arch Med Res ; 48(2): 147-155, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-28625317

RESUMEN

BACKGROUND AND AIMS: Although preconditioning of sphingosine 1-phosphate (S1P) has been shown to protect myocytes from hypoxia reoxgenation injury in vitro, the role of S1P postconditioning on myocardial ischemia reperfusion injury (MIRI) in vivo and its related mechanism are unknown. The aim of this study was to investigate the protective role of sphingosine 1-phosphate (S1P) postconditioning in MIRI via its effects on mitochondrial signaling and Akt/Gsk3ß phosphorylation. METHODS: Rats were subjected to MIRI, consisting of 30 min of ischemia followed by 120 min of reperfusion, with S1P administered at the beginning of the reperfusion. Myocardial infarct size and apoptotic index were measured by triphenyltetrazolium (TTC) and terminal deoxynucleotide transferase dUTP nick-end labeling (TUNEL) assays, respectively. Akt and Gsk3ß phosphorylation, caspase-3 cleavage, and cytochrome c translocation were assessed by western blot. Mitochondrial permeability transition pore (MPTP) opening and mitochondrial membrane potential (MMP, ΔΨ) were also examined to determine overall mitochondrial function. RESULTS: S1P postconditioning significantly decreased myocardial infarct size and apoptosis, as well as enhanced Akt and Gsk3ß phosphorylation, attenuated caspase-3 cleavage and cytosolic cytochrome c translocation, and inhibited MPTP opening, which subsequently preserved Δψ. Electron microscopy also confirmed that S1P helped maintain myocardial mitochondria integrity. Moreover, the protective effects of S1P treatment were blocked by cotreatment with a PI3K inhibitor, LY294002. CONCLUSIONS: These results suggest that S1P postconditioning protects against MIRI by regulating mitochondrial signaling and Akt/Gsk3ß phosphorylation.


Asunto(s)
Glucógeno Sintasa Quinasa 3 beta/metabolismo , Poscondicionamiento Isquémico , Lisofosfolípidos/farmacología , Mitocondrias Cardíacas/efectos de los fármacos , Daño por Reperfusión Miocárdica/prevención & control , Proteínas Proto-Oncogénicas c-akt/metabolismo , Esfingosina/análogos & derivados , Animales , Cromonas/farmacología , Citocromos c/metabolismo , Lisofosfolípidos/uso terapéutico , Masculino , Potencial de la Membrana Mitocondrial , Mitocondrias Cardíacas/metabolismo , Proteínas de Transporte de Membrana Mitocondrial/metabolismo , Poro de Transición de la Permeabilidad Mitocondrial , Morfolinas/farmacología , Infarto del Miocardio/metabolismo , Infarto del Miocardio/patología , Infarto del Miocardio/prevención & control , Daño por Reperfusión Miocárdica/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3 , Fosforilación , Transporte de Proteínas , Ratas Wistar , Transducción de Señal , Esfingosina/farmacología , Esfingosina/uso terapéutico
7.
Artículo en Inglés | MEDLINE | ID: mdl-28377281

RESUMEN

Sphingosine-1-phosphate (S1P) is a biologically active sphingolipid metabolite which has been implicated in many diseases including cancer and inflammatory diseases. Recently, sphingosine kinase 1 (SK1), one of the isozymes which generates S1P, has been implicated in the development and progression of inflammatory bowel disease (IBD). Based on our previous work, we set out to determine the efficacy of a novel SK1 selective inhibitor, LCL351, in a murine model of IBD. LCL351 selectively inhibits SK1 both in vitro and in cells. LCL351, which accumulates in relevant tissues such as colon, did not have any adverse side effects in vivo. In mice challenged with dextran sodium sulfate (DSS), a murine model for IBD, LCL351 treatment protected from blood loss and splenomegaly. Additionally, LCL351 treatment reduced the expression of pro-inflammatory markers, and reduced neutrophil infiltration in colon tissue. Our results suggest inflammation associated with IBD can be targeted pharmacologically through the inhibition and degradation of SK1. Furthermore, our data also identifies desirable properties of SK1 inhibitors.


Asunto(s)
Colitis/tratamiento farmacológico , Colitis/inmunología , Sulfato de Dextran/efectos adversos , Guanidinas/farmacología , Fosfotransferasas (Aceptor de Grupo Alcohol)/antagonistas & inhibidores , Esfingosina/farmacología , Células A549 , Quimiocina CXCL1/genética , Quimiocina CXCL2/genética , Colitis/inducido químicamente , Colitis/genética , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/inmunología , Guanidinas/uso terapéutico , Humanos , Esfingosina/uso terapéutico , Factor de Necrosis Tumoral alfa/genética
8.
Gynecol Endocrinol ; 33(6): 476-479, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28277139

RESUMEN

To investigate whether sphingosine-1-phosphate (S1P), an apoptosis-inhibitor would be able to inhibit chemotherapy induced human granulosa cell apoptosis. Cultures of primary granulosa cells were isolated from women undergoing in vitro fertilization (IVF). MTT assay was used to measure the optimum concentration of CTX and S1P acts on human granulosa cells. Granulosa cells were added with pertussis toxin (PTX), the PI3K inhibitor LY294002. Western blot analysis was used to analyze the signaling pathway of proteins and cell apoptosis. We found that S1P (10 mm) statistically significantly decreased granulosa cell apoptosis after cyclophosphamide (CTX) treatment. The decreased cell apoptosis induced by S1P was abolished after treatment with LY294002, PI3K inhibitor. CONCLUSIONS: Treatment with S1P can inhibit the CTX-induced granulosa cell apoptosis. The S1P protective effect is mediated by activating the PI3K/Akt pathway.


Asunto(s)
Antineoplásicos Alquilantes/efectos adversos , Ciclofosfamida/efectos adversos , Células de la Granulosa/efectos de los fármacos , Lisofosfolípidos/uso terapéutico , Insuficiencia Ovárica Primaria/prevención & control , Esfingosina/análogos & derivados , Apoptosis/efectos de los fármacos , Células Cultivadas , Evaluación Preclínica de Medicamentos , Femenino , Humanos , Lisofosfolípidos/farmacología , Fosforilación/efectos de los fármacos , Insuficiencia Ovárica Primaria/inducido químicamente , Proteínas Proto-Oncogénicas c-akt/metabolismo , Esfingosina/farmacología , Esfingosina/uso terapéutico
9.
Autoimmun Rev ; 16(5): 495-503, 2017 May.
Artículo en Inglés | MEDLINE | ID: mdl-28279838

RESUMEN

Inflammatory bowel diseases (IBD), including ulcerative colitis and Crohn's disease, involve an inappropriate immune reaction in the digestive tract, causing a variety of disabling symptoms. The advent of monoclonal antibodies (anti-tumor necrosis factor, anti-integrin, anti-interleukin -23) has revolutionized IBD management. Nevertheless, these agents, with potential for immunogenicity, are associated with high rates of response loss and disease relapse over time. They are also associated with high production costs. Sphingosine-1-phosphate (S1P), a membrane-derived lysophospholipid signaling molecule, is implicated in a vast array of physiological and pathophysiological processes, primarily via extracellular activation of S1P1-S1P5 receptors. S1P1, S1P4 and S1P5 are involved in regulation of the immune system, while S1P2 and S1P3 may be associated with cardiovascular, pulmonary, and theoretical cancer-related risks. Targeting S1P receptors for inflammatory conditions has been successful in clinical trials leading to approval of the non-selective S1P modulator, fingolimod, for relapsing forms of multiple sclerosis. However, the association of this non-selective S1P modulator with serious adverse events provides the rationale for developing more selective S1P receptor modulators. Until recently, three S1P modulators with differing selectivity for S1P receptors were in clinical development for IBD: ozanimod (RPC1063), etrasimod (APD334) and amiselimod (MT-1303). The development of amiselimod has been stopped as Biogen are currently focusing on other drugs in its portfolio. Following encouraging results from the Phase 2 TOUCHSTONE trial, a Phase 3 trial of the S1P modulator ozanimod in patients with moderate-to-severe ulcerative colitis is ongoing. Etrasimod is also being tested in a phase 2 trial in ulcerative colitis. These pipeline medications can be administered orally and may avoid the formation of anti-drug antibodies that can lead to treatment failure with injectable biologic therapies for IBD. Data from ongoing clinical trials will establish the relationship between the selectivity of S1P modulators and their safety and efficacy in IBD, as well as their potential place in the clinical armamentarium for IBD.


Asunto(s)
Enfermedades Inflamatorias del Intestino/tratamiento farmacológico , Lisofosfolípidos/uso terapéutico , Esfingosina/análogos & derivados , Humanos , Enfermedades Inflamatorias del Intestino/patología , Lisofosfolípidos/metabolismo , Esfingosina/metabolismo , Esfingosina/uso terapéutico
10.
Vet Comp Oncol ; 15(3): 1115-1118, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-27136276

RESUMEN

OSU-2S is a FTY720 (Fingolimod) derivative that lacks immunosuppressive properties but exhibits strong anti-tumour activity in several haematological and solid tumour models. We have recently shown OSU-2S to mediate potent cytotoxicity in human mantle cell lymphoma cell lines and primary cells. We report here the pre-clinical activity of OSU-2S in spontaneous B-cell lymphoma of dogs which shares many characteristics of human lymphoma. OSU-2S mediated apoptosis in canine B-cell lines and primary B-cell lymphoma cells obtained from spontaneous lymphoma bearing dogs. OSU-2S induced reactive oxygen species (ROS) in canine lymphoma cells and inhibition of ROS partially rescued OSU-2S-mediated cell death. These studies provide a rational basis for the use of spontaneous lymphoma in pet dogs as a preclinical large animal model for the development of OSU-2S as small molecule for treating people and dogs with lymphoma.


Asunto(s)
Antineoplásicos/uso terapéutico , Enfermedades de los Perros/tratamiento farmacológico , Linfoma de Células B/veterinaria , Glicoles de Propileno/uso terapéutico , Especies Reactivas de Oxígeno/antagonistas & inhibidores , Esfingosina/análogos & derivados , Animales , Apoptosis/efectos de los fármacos , Células Cultivadas , Perros , Linfoma de Células B/tratamiento farmacológico , Esfingosina/uso terapéutico
11.
Ren Fail ; 38(7): 1089-98, 2016 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-27309733

RESUMEN

Contrast induced nephropathy (CIN) is a major cause of morbidity, and increased costs as well as an increased risk of death. This study was evaluated effects of exogenous sphingosylphosphorylcholine (SPC) administration on CIN in rats. Eight animals were included in each of the following eight groups: control, control phosphate-buffered solution (PBS), control SPC 2, control SPC 10, CIN, CIN PBS, CIN SPC 2 and CIN SPC 10. The induced nephropathy was created by injected with 4 g iodine/kg body weight. SPC was administered 3 d at a daily two different doses of 2 µm/mL and 10 µm/mL intraperitoneally. The severity of renal injury score was determined by the histological and immunohistochemical changes in the kidney. Malondialdehyde (MDA), nitric oxide (NO) and superoxide dismutase (SOD) were determined to evaluate the oxidative status in the renal tissue. Treatment with 2 and 10 µM SPC inhibited the increase in renal MDA, NO levels significantly and also attenuated the depletion of SOD in the renal injuryCIN. These data were supported by histopathological findings. The inducible nitric oxide synthase positive cells and apoptotic cells in the renal tissue were observed to be reduced with the 2 and 10 µM SPC treatment. These findings suggested that 2 and 10 µM doses can attenuate renal damage in contrast nephropathy by prevention of oxidative stress and apoptosis. The low and high dose SPC may be a promising new therapeutic agent for CIN.


Asunto(s)
Lesión Renal Aguda/inducido químicamente , Lesión Renal Aguda/tratamiento farmacológico , Antioxidantes/uso terapéutico , Medios de Contraste/efectos adversos , Riñón/efectos de los fármacos , Fosforilcolina/análogos & derivados , Esfingosina/análogos & derivados , Animales , Antioxidantes/administración & dosificación , Apoptosis/efectos de los fármacos , Creatinina/sangre , Humanos , Inyecciones Intraperitoneales , Riñón/metabolismo , Riñón/patología , Malondialdehído/metabolismo , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa de Tipo II/metabolismo , Estrés Oxidativo/efectos de los fármacos , Fosforilcolina/administración & dosificación , Fosforilcolina/uso terapéutico , Ratas , Ratas Wistar , Esfingosina/administración & dosificación , Esfingosina/uso terapéutico , Superóxido Dismutasa/metabolismo
12.
Cell Physiol Biochem ; 38(6): 2094-102, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27184795

RESUMEN

BACKGROUND: Pulmonary Staphylococcus aureus (S. aureus) infections occur early in a high percentage of cystic fibrosis (CF) patients and it is believed that these infections facilitate further colonization of CF lungs with Pseudomonas aeruginosa (P. aeruginosa). Previous studies demonstrated a marked reduction of sphingosine in tracheal and bronchial epithelial cells in CF compared to wild type mice, while ceramide is massively increased in CF mice. METHODS: We investigated the effect of C18-sphingosine and C16-ceramide on S. aureus in vitro. Based on our results we performed pulmonary infections with S. aureus and tested the influence of sphingosine inhalation. RESULTS: In vitro incubation of S. aureus with C18-sphingosine rapidly killed S. aureus, while C16-ceramide did not affect bacterial survival, but abrogated the effect of C18-sphingosine when applied together. The in vivo infection experiments revealed a high susceptibility of CF mice to pulmonary infection with S. aureus. Inhalation of C18-sphingosine rescued CF mice from pulmonary infections with different clinical S. aureus isolates, including a methicillin-resistant S. aureus (MRSA) strain. CONCLUSIONS: Our data indicate that the imbalance between ceramide and sphingosine in the CF respiratory tract prevents killing of S. aureus and causes the high susceptibility of CF mice to pulmonary S. aureus infections.


Asunto(s)
Antibacterianos/uso terapéutico , Fibrosis Quística/complicaciones , Neumonía Estafilocócica/complicaciones , Neumonía Estafilocócica/tratamiento farmacológico , Esfingosina/uso terapéutico , Staphylococcus aureus/efectos de los fármacos , Animales , Antibacterianos/metabolismo , Antibacterianos/farmacología , Ceramidas/metabolismo , Ceramidas/farmacología , Ceramidas/uso terapéutico , Fibrosis Quística/metabolismo , Humanos , Pulmón/metabolismo , Pulmón/microbiología , Staphylococcus aureus Resistente a Meticilina/efectos de los fármacos , Ratones , Neumonía Estafilocócica/metabolismo , Esfingosina/metabolismo , Esfingosina/farmacología
13.
Toxicol Lett ; 236(1): 43-59, 2015 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-25939952

RESUMEN

FTY720 is a potent immunosuppressant which has preclinical antitumor efficacy in various cancer models. However, its role in glioblastoma remains unclear. In the present study, we found that FTY720 induced extrinsic apoptosis, necroptosis and autophagy in human glioblastoma cells. Inhibition of autophagy by either RNA interference or chemical inhibitors attenuated FTY720-induced apoptosis and necrosis. Furthermore, autophagy, apoptosis and necrosis induction were dependent on reactive oxygen species-c-Jun N-terminal kinase-protein 53 (ROS-JNK-p53) loop mediated phosphatidylinositide 3-kinases/protein kinase B/mammalian target of rapamycin/p70S6 kinase (PI3K/AKT/mTOR/p70S6K) pathway. In addition, receptor-interacting protein 1 and 3 (RIP1 and RIP3) served as an upstream of ROS-JNK-p53 loop. However, the phosphorylation form of FTY720 induced autophagy but not apoptosis and necroptosis. Finally, the in vitro results were validated in vivo in xenograft mouse of glioblastoma cells. In conclusion, the current study provided novel insights into understanding the mechanisms and functions of FTY720-induced apoptosis, necroptosis and autophagy in human glioblastoma cells.


Asunto(s)
Antineoplásicos/uso terapéutico , Apoptosis/efectos de los fármacos , Autofagia/efectos de los fármacos , Glioblastoma/tratamiento farmacológico , Glicoles de Propileno/uso terapéutico , Transducción de Señal/efectos de los fármacos , Esfingosina/análogos & derivados , Animales , Antineoplásicos/administración & dosificación , Antineoplásicos/farmacología , Proteínas Reguladoras de la Apoptosis/antagonistas & inhibidores , Proteínas Reguladoras de la Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/metabolismo , Beclina-1 , Biomarcadores/metabolismo , Línea Celular Tumoral , Relación Dosis-Respuesta a Droga , Clorhidrato de Fingolimod , Glioblastoma/metabolismo , Glioblastoma/patología , Humanos , Masculino , Proteínas de la Membrana/antagonistas & inhibidores , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones Desnudos , Necrosis , Proteínas de Neoplasias/antagonistas & inhibidores , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Glicoles de Propileno/administración & dosificación , Glicoles de Propileno/farmacología , Interferencia de ARN , Especies Reactivas de Oxígeno/metabolismo , Proteína Serina-Treonina Quinasas de Interacción con Receptores/antagonistas & inhibidores , Proteína Serina-Treonina Quinasas de Interacción con Receptores/genética , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Esfingosina/administración & dosificación , Esfingosina/farmacología , Esfingosina/uso terapéutico , Carga Tumoral/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
15.
J Clin Invest ; 125(4): 1379-87, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25831442

RESUMEN

Membrane sphingolipids are metabolized to sphingosine-1-phosphate (S1P), a bioactive lipid mediator that regulates many processes in vertebrate development, physiology, and pathology. Once exported out of cells by cell-specific transporters, chaperone-bound S1P is spatially compartmentalized in the circulatory system. Extracellular S1P interacts with five GPCRs that are widely expressed and transduce intracellular signals to regulate cellular behavior, such as migration, adhesion, survival, and proliferation. While many organ systems are affected, S1P signaling is essential for vascular development, neurogenesis, and lymphocyte trafficking. Recently, a pharmacological S1P receptor antagonist has won approval to control autoimmune neuroinflammation in multiple sclerosis. The availability of pharmacological tools as well as mouse genetic models has revealed several physiological actions of S1P and begun to shed light on its pathological roles. The unique mode of signaling of this lysophospholipid mediator is providing novel opportunities for therapeutic intervention, with possibilities to target not only GPCRs but also transporters, metabolic enzymes, and chaperones.


Asunto(s)
Lisofosfolípidos/fisiología , Esfingosina/análogos & derivados , Lesión Pulmonar Aguda/tratamiento farmacológico , Anemia de Células Falciformes/sangre , Animales , Enfermedades Autoinmunes/tratamiento farmacológico , Enfermedades Autoinmunes/metabolismo , Enfermedades Cardiovasculares/metabolismo , Fenómenos Fisiológicos Celulares/fisiología , Modelos Animales de Enfermedad , Clorhidrato de Fingolimod , Movilización de Célula Madre Hematopoyética , Humanos , Gripe Humana/metabolismo , Lisofosfolípidos/agonistas , Lisofosfolípidos/antagonistas & inhibidores , Lípidos de la Membrana/metabolismo , Ratones , Esclerosis Múltiple/tratamiento farmacológico , Neoplasias/sangre , Neovascularización Fisiológica/fisiología , Neurogénesis/fisiología , Glicoles de Propileno/uso terapéutico , Receptores de Lisoesfingolípidos/antagonistas & inhibidores , Receptores de Lisoesfingolípidos/fisiología , Esfingolípidos/metabolismo , Esfingosina/agonistas , Esfingosina/antagonistas & inhibidores , Esfingosina/fisiología , Esfingosina/uso terapéutico
16.
Clin Immunol ; 158(1): 103-13, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25829233

RESUMEN

The variable response to therapy in multiple sclerosis (MS) suggests a need for personalized approaches based on individual genetic differences. GWAS have linked CBLB gene polymorphisms with MS and recent evidence demonstrated that these polymorphisms can be associated with abnormalities in T cell function and response to interferon-ß therapy. Cbl-b is an E3 ubiquitin ligase that regulates T cell activation and Cbl-b-deficient (Cbl-b(-/-)) mice show T cell abnormalities described in MS patients. We now show that Cbl-b(-/-) T cells demonstrate significant lymph node trafficking abnormalities. We thus asked whether the MS-approved drug, FTY720, postulated to trap T cells in lymphoid tissues, is less effective in the context of Cbl-b dysfunction. We now report that FTY720 significantly inhibits EAE in Cbl-b(-/-) mice. Our results newly document a role for Cbl-b in T cell trafficking but suggest nevertheless that MS patients with Cbl-b abnormalities may still be excellent candidates for FTY720 treatment.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Movimiento Celular/genética , Encefalomielitis Autoinmune Experimental/inmunología , Inmunosupresores/farmacología , Ganglios Linfáticos/metabolismo , Esclerosis Múltiple/inmunología , Glicoles de Propileno/farmacología , Proteínas Proto-Oncogénicas c-cbl/genética , Esfingosina/análogos & derivados , Linfocitos T/metabolismo , Proteínas Adaptadoras Transductoras de Señales/inmunología , Traslado Adoptivo , Animales , Movimiento Celular/efectos de los fármacos , Movimiento Celular/inmunología , Clorhidrato de Fingolimod , Proteínas de Homeodominio/genética , Inmunosupresores/uso terapéutico , Ganglios Linfáticos/efectos de los fármacos , Ganglios Linfáticos/inmunología , Activación de Linfocitos/efectos de los fármacos , Activación de Linfocitos/genética , Ratones , Ratones Noqueados , Esclerosis Múltiple/tratamiento farmacológico , Glicoles de Propileno/uso terapéutico , Proteínas Proto-Oncogénicas c-cbl/inmunología , Esfingosina/farmacología , Esfingosina/uso terapéutico , Linfocitos T/efectos de los fármacos
17.
J Pediatr Surg ; 50(4): 591-7, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25840069

RESUMEN

BACKGROUND/PURPOSE: The goal of this study was to evaluate effects of exogenous sphingosylphosphorylcholine (SPC) administration on acute lung injury induced by pulmonary contusion in rats. METHODS: Eight animals were included in each of the following five groups: control, contusion, contusion phosphate-buffered solution (PBS), contusion SPC 2, contusion SPC 10. SPC was administered 3 days at a daily two different doses of 2 µm/ml and 10 µm/ml intraperitoneally. The severity of lung injury was determined by the neutrophil activation and histological and immunohistochemical changes in the lung. Malondialdehyde (MDA), nitric oxide (NO), superoxide dismutase (SOD), glutathione peroxidase (GPx) and glutathione (GSH) were determined to evaluate the oxidative status in the lung tissue. RESULTS: Treatment with 2 µM SPC inhibited the increase in lung MDA and NO levels significantly and also attenuated the depletion of SOD, GPx, and GSH in the lung injury induced by pulmonary contusion. These data were supported by histopathological findings. The inducible nitric oxide synthase (iNOS) positive cells and apoptotic cells in the lung tissue were observed to be reduced with the 2 µM SPC treatment. But, the 10 µM SPC treatment did not provide similar effects. CONCLUSIONS: In conclusion, these findings suggested that 2 µM SPC can attenuate lung damage in pulmonary contusion by prevention of oxidative stress, inflammatory process and apoptosis. All these findings suggest that low dose SPC may be a promising new therapeutic agent for acute lung injury.


Asunto(s)
Lesión Pulmonar Aguda/prevención & control , Contusiones/complicaciones , Estrés Oxidativo/efectos de los fármacos , Fosforilcolina/análogos & derivados , Sustancias Protectoras/uso terapéutico , Esfingosina/análogos & derivados , Lesión Pulmonar Aguda/etiología , Animales , Relación Dosis-Respuesta a Droga , Esquema de Medicación , Inyecciones Intraperitoneales , Fosforilcolina/farmacología , Fosforilcolina/uso terapéutico , Sustancias Protectoras/farmacología , Ratas , Ratas Wistar , Esfingosina/farmacología , Esfingosina/uso terapéutico , Resultado del Tratamiento
18.
PLoS One ; 10(3): e0119664, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25781943

RESUMEN

BACKGROUND: New evidence shows that high density lipoproteins (HDL) have protective effects beyond their role in reverse cholesterol transport. Reconstituted HDL (rHDL) offer an attractive means of clinically exploiting these novel effects including cardioprotection against ischemia reperfusion injury (IRI). However, basic rHDL composition is limited to apolipoprotein AI (apoAI) and phospholipids; addition of bioactive compound may enhance its beneficial effects. OBJECTIVE: The aim of this study was to investigate the role of rHDL in post-ischemic model, and to analyze the potential impact of sphingosine-1-phosphate (S1P) in rHDL formulations. METHODS AND RESULTS: The impact of HDL on IRI was investigated using complementary in vivo, ex vivo and in vitro IRI models. Acute post-ischemic treatment with native HDL significantly reduced infarct size and cell death in the ex vivo, isolated heart (Langendorff) model and the in vivo model (-48%, p<0.01). Treatment with rHDL of basic formulation (apoAI + phospholipids) had a non-significant impact on cell death in vitro and on the infarct size ex vivo and in vivo. In contrast, rHDL containing S1P had a highly significant, protective influence ex vivo, and in vivo (-50%, p<0.01). This impact was comparable with the effects observed with native HDL. Pro-survival signaling proteins, Akt, STAT3 and ERK1/2 were similarly activated by HDL and rHDL containing S1P both in vitro (isolated cardiomyocytes) and in vivo. CONCLUSION: HDL afford protection against IRI in a clinically relevant model (post-ischemia). rHDL is significantly protective if supplemented with S1P. The protective impact of HDL appears to target directly the cardiomyocyte.


Asunto(s)
Lipoproteínas HDL/farmacología , Daño por Reperfusión Miocárdica/tratamiento farmacológico , Miocitos Cardíacos/efectos de los fármacos , Animales , Apolipoproteína A-I/farmacología , Apolipoproteína A-I/uso terapéutico , Células Cultivadas , Lipoproteínas HDL/uso terapéutico , Lisofosfolípidos/farmacología , Lisofosfolípidos/uso terapéutico , Masculino , Ratones , Ratones Endogámicos C57BL , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Miocitos Cardíacos/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas , Ratas Wistar , Factor de Transcripción STAT3/metabolismo , Esfingosina/análogos & derivados , Esfingosina/farmacología , Esfingosina/uso terapéutico
19.
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
20.
Ideggyogy Sz ; 67(7-8): 220-8, 2014 Jul 30.
Artículo en Húngaro | MEDLINE | ID: mdl-25509362

RESUMEN

Multiple sclerosis (MS) is the most common chronic disease of the central nervous system in young adults. No curative therapy is known. Currently, six drugs are available that can reduce the activity of MS. The first-line drugs can completely reduce the activity of the disease in nearly two-thirds of the patients. In the remainder, who suffer from breakthrough disease, the condition of the patient worsens, and second-line therapies must be used. The second-line drug natalizumab exhibits almost double efficacy of the first-line drugs, but also have less favourable adverse effects. As a severe side-effect for instance, natalizumab carries the risk of the development of progressive multifocal leucoencephalopathy (PML), caused by a polyoma virus, the JC virus. There are three major risk factors for PML: an anti-JCV antibody status, a long duration of natalizumab treatment and prior immunosuppressant therapy. The lowest-risk group (1:14,286) comprises of patients who are anti-JCV antibody-negative, in whom the prior immunosuppressant use and duration of natalizumab therapy do not influence the risk of PML. With no prior immunosuppressant treatment, the incidence of PML increases to 1 in 192 patients after 2 years among those who are anti-JCV antibody-positive. These data may lead the physician to decide to discontinue natalizumab treatment. The half-life of natalizumab is three months; during this time other therapies can not be administered and the patients encounter the rebound effect: as the patients receiving natalizumab therapy displayed a high disease activity before treatment, the rebound effect can lead to relapses. After the termination of natalizumab second-line disease-modifying therapy with fingolimod may be introduced; no PML cases occur in response to fingolimod treatment. In the large majority of patients taking natalizumab who do not develop PML, this drug is highly effective and can prevent the progression of MS. The benefit of therapy and the risk of PML must be considered on an individual basis, with regard to the disease activity, the progression and the MRI activity, before natalizumab therapy is implemented.


Asunto(s)
Anticuerpos Monoclonales Humanizados/uso terapéutico , Huésped Inmunocomprometido , Inmunosupresores/uso terapéutico , Leucoencefalopatía Multifocal Progresiva/etiología , Esclerosis Múltiple/tratamiento farmacológico , Adulto , Anticuerpos Monoclonales Humanizados/administración & dosificación , Anticuerpos Monoclonales Humanizados/efectos adversos , Anticuerpos Monoclonales Humanizados/farmacología , Progresión de la Enfermedad , Clorhidrato de Fingolimod , Humanos , Inmunosupresores/administración & dosificación , Inmunosupresores/efectos adversos , Inmunosupresores/farmacología , Virus JC , Leucoencefalopatía Multifocal Progresiva/inmunología , Leucoencefalopatía Multifocal Progresiva/virología , Imagen por Resonancia Magnética , Esclerosis Múltiple/inmunología , Esclerosis Múltiple/patología , Natalizumab , Glicoles de Propileno/uso terapéutico , Esfingosina/análogos & derivados , Esfingosina/uso terapéutico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA