Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 49
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Nat Immunol ; 15(3): 231-8, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24464131

RESUMEN

Although interleukin 1 (IL-1) induces expression of the transcription factor IRF1 (interferon-regulatory factor 1), the roles of IRF1 in immune and inflammatory responses and mechanisms of its activation remain elusive. Here we found that IRF1 was essential for IL-1-induced expression of the chemokines CXCL10 and CCL5, which recruit mononuclear cells into sites of sterile inflammation. Newly synthesized IRF1 acquired Lys63 (K63)-linked polyubiquitination mediated by the apoptosis inhibitor cIAP2 that was enhanced by the bioactive lipid S1P. In response to IL-1, cIAP2 and the sphingosine kinase SphK1 (the enzyme that generates S1P) formed a complex with IRF1, which led to its activation. Thus, IL-1 triggered a hitherto unknown signaling cascade that controlled the induction of IRF1-dependent genes that encode molecules important for sterile inflammation.


Asunto(s)
Quimiocina CCL5/biosíntesis , Quimiocina CXCL10/biosíntesis , Factor 1 Regulador del Interferón/metabolismo , Interleucina-1/metabolismo , Transducción de Señal/inmunología , Animales , Quimiocina CCL5/inmunología , Quimiocina CXCL10/inmunología , Quimiotaxis de Leucocito/inmunología , Ensayo de Inmunoadsorción Enzimática , Humanos , Immunoblotting , Inmunoprecipitación , Inflamación/inmunología , Inflamación/metabolismo , Factor 1 Regulador del Interferón/inmunología , Interleucina-1/inmunología , Leucocitos Mononucleares/inmunología , Leucocitos Mononucleares/metabolismo , Lisina , Ratones , Ratones Noqueados , Reacción en Cadena en Tiempo Real de la Polimerasa , Ubiquitinación
2.
Glycobiology ; 32(8): 701-711, 2022 07 13.
Artículo en Inglés | MEDLINE | ID: mdl-35661210

RESUMEN

Interaction of immune cells with the systemic environment is necessary for the coordinated development and execution of immune responses. Monocyte-macrophage lineage cells reside at the junction of innate and adaptive immunity. Previously we reported that the sialyltransferase ST6GAL1 in the extracellular milieu modulates B cell development and IgG production, granulocyte production, and attenuates acute airway inflammation to bacterial challenge in mouse models. Here, we report that extracellular ST6GAL1 also elicits profound responses in monocyte-macrophage lineage cells. We show that recombinant ST6GAL1 adheres to subsets of thioglycolate-elicited inflammatory cells in the mouse peritoneum and to cultured human monocyte THP-1 cells. Exposure of the inflammatory cells to recombinant ST6GAL1 elicited wholesale changes in the gene expression profile of primary mouse myeloid cells; most notable was the striking up-regulation of monocyte-macrophage and monocyte-derived dendritic cell development pathway signature genes and transcription factors PU.1, NFκB and their target genes, driving increased monocyte-macrophage population and survival ex vivo. In the cultured human monocyte cells, the essential cell surface receptor of the monocyte-macrophage lineage, the M-CSF receptor (M-CSF-R, Csfr1) was a target of extracellular ST6GAL1 catalytic activity. Extracellular ST6GAL1 activated the M-CSF-R and initiated intracellular signaling events, namely, the nuclear translocation of NFκB subunit p65, and phosphorylation of ERK 1/2 and AKT. The findings implicate extracellular ST6GAL1 in monocyte development by a mechanism initiated at the cell surface and support an emerging paradigm of an extracellular glycan-modifying enzyme as a central regulator coordinating immune hematopoietic cell development and function.


Asunto(s)
Factor Estimulante de Colonias de Macrófagos , Monocitos , Animales , Antígenos CD/metabolismo , Diferenciación Celular , Humanos , Factor Estimulante de Colonias de Macrófagos/metabolismo , Macrófagos/metabolismo , Ratones , Monocitos/metabolismo , Fosforilación , Sialiltransferasas/genética , Sialiltransferasas/metabolismo , Transducción de Señal , Células THP-1
3.
FASEB J ; 34(3): 4293-4310, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-32017264

RESUMEN

Sphingosine kinase 2 (SphK2) is known to phosphorylate the nuclear sphingolipid metabolite to generate sphingosine-1-phosphate (S1P). Nuclear S1P is involved in epigenetic regulation of gene expression; however, the underlying mechanisms are not well understood. In this work, we have identified the role of nuclear S1P and SphK2 in regulating hypoxia-responsive master transcription factors hypoxia-inducible factor (HIF)-1α/2α, and their functions in breast cancer, with a focus on triple-negative breast cancer (TNBC). We have shown SphK2 is associated with HIF-1α in protein complexes, and is enriched at the promoters of HIF target genes, including vascular endothelial growth factor (VEGF), where it enhances local histone H3 acetylation and transcription. S1P specifically binds to the PAS domains of HIF-1α. SphK2, and HIF-1α expression levels are elevated in metastatic estrogen receptor-positive (ER+) and TNBC clinical tissue specimens compared to healthy breast tissue samples. To determine if S1P formation in the nucleus by SphK2 is a key regulator of HIF functions, we found using a preclinical TNBC xenograft mouse model, and an existing selective SphK2 inhibitor K-145, that nuclear S1P, histone acetylation, HIF-1α expression, and TNBC tumor growth were all reduced in vivo. Our results suggest that S1P and SphK2 in the nucleus are linked to the regulation of HIF-1α/2α functions associated with breast cancer progression, and may provide potential therapeutic targets.


Asunto(s)
Núcleo Celular/metabolismo , Lisofosfolípidos/metabolismo , Receptor de Adenosina A2B/metabolismo , Esfingosina/análogos & derivados , Receptor 1 de Factores de Crecimiento Endotelial Vascular/metabolismo , 5'-Nucleotidasa/genética , 5'-Nucleotidasa/metabolismo , Acetilación , Adenosina/metabolismo , Animales , Presión Sanguínea/genética , Presión Sanguínea/fisiología , Ensayo de Inmunoadsorción Enzimática , Epigénesis Genética/genética , Epigénesis Genética/fisiología , Femenino , Humanos , Immunoblotting , Inmunohistoquímica , Ratones , Ratones Endogámicos C57BL , Embarazo , Receptor de Adenosina A2B/genética , Esfingosina/metabolismo , Miembro 14 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/genética , Miembro 14 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/metabolismo , Receptor 1 de Factores de Crecimiento Endotelial Vascular/genética
4.
Int J Mol Sci ; 22(24)2021 Dec 13.
Artículo en Inglés | MEDLINE | ID: mdl-34948163

RESUMEN

Although numerous experiments revealed an essential role of a lipid mediator, sphingosine-1-phosphate (S1P), in breast cancer (BC) progression, the clinical significance of S1P remains unclear due to the difficulty of measuring lipids in patients. The aim of this study was to determine the plasma concentration of S1P in estrogen receptor (ER)-positive BC patients, as well as to investigate its clinical significance. We further explored the possibility of a treatment strategy targeting S1P in ER-positive BC patients by examining the effect of FTY720, a functional antagonist of S1P receptors, on hormone therapy-resistant cells. Plasma S1P levels were significantly higher in patients negative for progesterone receptor (PgR) expression than in those positive for expression (p = 0.003). Plasma S1P levels were also significantly higher in patients with larger tumor size (p = 0.012), lymph node metastasis (p = 0.014), and advanced cancer stage (p = 0.003), suggesting that higher levels of plasma S1P are associated with cancer progression. FTY720 suppressed the viability of not only wildtype MCF-7 cells, but also hormone therapy-resistant MCF-7 cells. Targeting S1P signaling in ER-positive BC appears to be a possible new treatment strategy, even for hormone therapy-resistant patients.


Asunto(s)
Neoplasias de la Mama/metabolismo , Lisofosfolípidos/análisis , Esfingosina/análogos & derivados , Adulto , Anciano , Biomarcadores de Tumor/sangre , Neoplasias de la Mama/genética , Línea Celular Tumoral , Progresión de la Enfermedad , Femenino , Clorhidrato de Fingolimod/farmacología , Expresión Génica/genética , Humanos , Metástasis Linfática , Lisofosfolípidos/sangre , Lisofosfolípidos/metabolismo , Células MCF-7 , Persona de Mediana Edad , Plasma/química , Receptores de Estrógenos/metabolismo , Receptores de Lisoesfingolípidos/metabolismo , Transducción de Señal , Esfingosina/análisis , Esfingosina/sangre , Esfingosina/metabolismo , Receptores de Esfingosina-1-Fosfato/efectos de los fármacos , Receptores de Esfingosina-1-Fosfato/metabolismo
5.
FASEB J ; 31(4): 1719-1730, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28082351

RESUMEN

Niemann-Pick type C (NPC) disease is a fatal neurodegenerative disorder caused by mutations in NPC1 or NPC2 with decreased functions leading to lysosomal accumulation of cholesterol and sphingolipids. FTY720/fingolimod, used for treatment of multiple sclerosis, is phosphorylated by nuclear sphingosine kinase 2, and its active phosphorylated form (FTY720-P) is an inhibitor of class I histone deacetylases. In this study, administration of clinically relevant doses of FTY720 to mice increased expression of NPC1 and -2 in brain and liver and decreased cholesterol in an SphK2-dependent manner. FTY720 greatly increased expression of NPC1 and -2 in human NPC1 mutant fibroblasts that correlated with formation of FTY720-P and significantly reduced the accumulation of cholesterol and glycosphingolipids. In agreement with this finding, FTY720 pretreatment of human NPC1 mutant fibroblasts restored transport of the cholera toxin B subunit, which binds ganglioside GM1, to the Golgi apparatus. Together, these findings suggest that FTY720 administration can ameliorate cholesterol and sphingolipid storage and trafficking defects in NPC1 mutant fibroblasts. Because neurodegeneration is the main clinical feature of NPC disease, and FTY720 accumulates in the CNS and has several advantages over available histone deacetylase inhibitors now in clinical trials, our work provides a potential opportunity for treatment of this incurable disease.-Newton, J., Hait, N. C., Maceyka, M., Colaco, A., Maczis, M., Wassif, C. A., Cougnoux, A., Porter, F. D., Milstien, S., Platt, N., Platt, F. M., Spiegel, S. FTY720/fingolimod increases NPC1 and NPC2 expression and reduces cholesterol and sphingolipid accumulation in Niemann-Pick type C mutant fibroblasts.


Asunto(s)
Colesterol/metabolismo , Clorhidrato de Fingolimod/farmacología , Inhibidores de Histona Desacetilasas/farmacología , Enfermedad de Niemann-Pick Tipo C/metabolismo , Proteínas/metabolismo , Esfingolípidos/metabolismo , Proteínas de Transporte Vesicular/metabolismo , Células 3T3 , Animales , Células Cultivadas , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Humanos , Péptidos y Proteínas de Señalización Intracelular , Masculino , Ratones , Ratones Endogámicos C57BL , Proteína Niemann-Pick C1 , Enfermedad de Niemann-Pick Tipo C/genética , Transporte de Proteínas , Proteínas/genética , Proteínas de Transporte Vesicular/genética
6.
J Surg Res ; 219: 202-213, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-29078883

RESUMEN

BACKGROUND: Doxorubicin is one of the most commonly used chemotherapeutic drugs for breast cancer; however, its use is limited by drug resistance and side effects. We hypothesized that adding FTY720, a sphingosine-1-phosphate (S1P) receptor functional antagonist, to doxorubicin would potentiate its effects by suppression of drug-induced inflammation. MATERIALS AND METHODS: The Cancer Genome Atlas, Gene Expression Omnibus data sets, and National Cancer Institute-60 panel were used for gene expressions and gene set enrichment analysis. E0771 syngeneic mammary tumor cells were used. OB/OB mice fed with western high-fat diet were used as an obesity model. RESULTS: STAT3 expression was significantly increased after doxorubicin treatment in human breast cancer that implicates that doxorubicin evokes inflammation. Expression of sphingosine kinase 1, the enzyme that produces S1P and links inflammation and cancer, tended to be higher in doxorubicin-resistant human cancer and cell lines. In a murine breast cancer model, sphingosine kinase 1, S1P receptor 1, interleukin 6, and STAT3 were overexpressed in the doxorubicin-treated group, whereas all of them were significantly suppressed with addition of FTY720. Combination therapy synergistically suppressed cancer growth both in vitro and in vivo. Furthermore, combination therapy showed higher efficacy in an obesity breast cancer model, where high body mass index demonstrated trends toward worse disease-free and overall survival, and high-serum S1P levels in human patients and volunteers. CONCLUSIONS: We found that FTY720 enhanced the efficacy of doxorubicin by suppression of drug-induced inflammation, and combination therapy showed stronger effect in obesity-related breast cancer.


Asunto(s)
Antibióticos Antineoplásicos/uso terapéutico , Neoplasias de la Mama/tratamiento farmacológico , Doxorrubicina/uso terapéutico , Clorhidrato de Fingolimod/uso terapéutico , Inmunosupresores/uso terapéutico , Neoplasias Mamarias Experimentales/tratamiento farmacológico , Animales , Protocolos de Quimioterapia Combinada Antineoplásica , Índice de Masa Corporal , Neoplasias de la Mama/etiología , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/mortalidad , Línea Celular Tumoral , Resistencia a Antineoplásicos , Femenino , Clorhidrato de Fingolimod/farmacología , Humanos , Inmunosupresores/farmacología , Lisofosfolípidos/sangre , Ratones , Obesidad/sangre , Obesidad/complicaciones , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Receptores de Lisoesfingolípidos/antagonistas & inhibidores , Estudios Retrospectivos , Factor de Transcripción STAT3/metabolismo , Esfingosina/análogos & derivados , Esfingosina/sangre
7.
Mediators Inflamm ; 2017: 4806541, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29269995

RESUMEN

Inflammation is part of our body's response to tissue injury and pathogens. It helps to recruit various immune cells to the site of inflammation and activates the production of mediators to mobilize systemic protective processes. However, chronic inflammation can increase the risk of diseases like cancer. Apart from cytokines and chemokines, lipid mediators, particularly sphingosine-1-phosphate (S1P) and ceramide-1-phosphate (C1P), contribute to inflammation and cancer. S1P is an important player in inflammation-associated colon cancer progression. On the other hand, C1P has been recognized to be involved in cancer cell growth, migration, survival, and inflammation. However, whether C1P is involved in inflammation-associated cancer is not yet established. In contrast, few studies have also suggested that S1P and C1P are involved in anti-inflammatory pathways regulated in certain cell types. Ceramide is the substrate for ceramide kinase (CERK) to yield C1P, and sphingosine is phosphorylated to S1P by sphingosine kinases (SphKs). Biological functions of sphingolipid metabolites have been studied extensively. Ceramide is associated with cell growth inhibition and enhancement of apoptosis while S1P and C1P are associated with enhancement of cell growth and survival. Altogether, S1P and C1P are important regulators of ceramide level and cell fate. This review focuses on S1P and C1P involvement in inflammation and cancer with emphasis on recent progress in the field.


Asunto(s)
Ceramidas/metabolismo , Inflamación/metabolismo , Lisofosfolípidos/metabolismo , Neoplasias/metabolismo , Esfingosina/análogos & derivados , Animales , Biomarcadores de Tumor/metabolismo , Humanos , Inflamación/etiología , Mediadores de Inflamación/metabolismo , Modelos Biológicos , Neoplasias/etiología , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Transducción de Señal , Esfingosina/metabolismo
8.
Mediators Inflamm ; 2017: 5984819, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29147072

RESUMEN

Sphingosine-1-phosphate (S1P) is a bioactive lipid mediator that has been shown to serve an important regulatory function in breast cancer progression. This study analyzes plasma S1P levels in breast cancer patients undergoing adjuvant therapy as compared to healthy control volunteers. 452 plasma S1P samples among 158 breast cancer patients, along with 20 healthy control volunteers, were analyzed. Mean S1P levels did not significantly differ between cancer patients and controls. Smoking was associated with higher S1P levels in cancer patients. Baseline S1P levels had weak inverse correlation with levels of the inflammatory mediator interleukin- (IL-) 17 and CCL-2 and positive correlation with tumor necrosis factor alpha (TNF-α). Midpoint S1P levels during adjuvant therapy were lower than baseline, with near return to baseline after completion, indicating a relationship between chemotherapy and circulating S1P. While stage of disease did not correlate with plasma S1P levels, they were lower among patients with Her2-enriched and triple-negative breast cancer as compared to luminal-type breast cancer. Plasma S1P levels are paradoxically suppressed in aggressive breast cancer and during adjuvant chemotherapy, which raises the possibility that postoperative plasma S1P levels do not reflect S1P secretion from resected breast cancer.


Asunto(s)
Neoplasias de la Mama/sangre , Lisofosfolípidos/sangre , Esfingosina/análogos & derivados , Adulto , Índice de Masa Corporal , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/patología , Quimiocina CCL2/sangre , Femenino , Humanos , Interleucina-17/sangre , Persona de Mediana Edad , Periodo Posoperatorio , Fumar/efectos adversos , Esfingosina/sangre , Factor de Necrosis Tumoral alfa/sangre
9.
J Lipid Res ; 57(9): 1636-43, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27459945

RESUMEN

Based on research carried out over the last decade, it has become increasingly evident that bile acids act not only as detergents, but also as important signaling molecules that exert various biological effects via activation of specific nuclear receptors and cell signaling pathways. Bile acids also regulate the expression of numerous genes encoding enzymes and proteins involved in the synthesis and metabolism of bile acids, glucose, fatty acids, and lipoproteins, as well as energy metabolism. Receptors activated by bile acids include, farnesoid X receptor α, pregnane X receptor, vitamin D receptor, and G protein-coupled receptors, TGR5, muscarinic receptor 2, and sphingosine-1-phosphate receptor (S1PR)2. The ligand of S1PR2, sphingosine-1-phosphate (S1P), is a bioactive lipid mediator that regulates various physiological and pathophysiological cellular processes. We have recently reported that conjugated bile acids, via S1PR2, activate and upregulate nuclear sphingosine kinase 2, increase nuclear S1P, and induce genes encoding enzymes and transporters involved in lipid and sterol metabolism in the liver. Here, we discuss the role of bile acids and S1P signaling in the regulation of hepatic lipid metabolism and in hepatobiliary diseases.


Asunto(s)
Ácidos y Sales Biliares/metabolismo , Metabolismo Energético/genética , Metabolismo de los Lípidos/genética , Lisofosfolípidos/metabolismo , Esfingosina/análogos & derivados , Ácidos Grasos/biosíntesis , Ácidos Grasos/metabolismo , Glucosa/biosíntesis , Glucosa/metabolismo , Humanos , Lipoproteínas/biosíntesis , Lipoproteínas/metabolismo , Receptores Citoplasmáticos y Nucleares/genética , Receptores Citoplasmáticos y Nucleares/metabolismo , Transducción de Señal , Esfingosina/metabolismo
10.
Hepatology ; 61(4): 1216-26, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25363242

RESUMEN

UNLABELLED: Bile acids are important hormones during the feed/fast cycle, allowing the liver to coordinately regulate nutrient metabolism. How they accomplish this has not been fully elucidated. Conjugated bile acids activate both the ERK1/2 and AKT signaling pathways via sphingosine 1-phosphate receptor 2 (S1PR2) in rodent hepatocytes and in vivo. Here, we report that feeding mice a high-fat diet, infusion of taurocholate into the chronic bile fistula rat, or overexpression of the gene encoding S1PR2 in mouse hepatocytes significantly upregulated hepatic sphingosine kinase 2 (SphK2) but not SphK1. Key genes encoding nuclear receptors/enzymes involved in nutrient metabolism were significantly downregulated in livers of S1PR2(-/-) and SphK2(-/-) mice. In contrast, overexpression of the gene encoding S1PR2 in primary mouse hepatocytes differentially increased SphK2, but not SphK1, and mRNA levels of key genes involved in nutrient metabolism. Nuclear levels of sphingosine-1-phosphate, an endogenous inhibitor of histone deacetylases 1 and 2, as well as the acetylation of histones H3K9, H4K5, and H2BK12 were significantly decreased in hepatocytes prepared from S1PR2(-/-) and SphK2(-/-) mice. CONCLUSION: Both S1PR2(-/-) and SphK2(-/-) mice rapidly developed fatty livers on a high-fat diet, suggesting the importance of conjugated bile acids, S1PR2, and SphK2 in regulating hepatic lipid metabolism.


Asunto(s)
Ácidos y Sales Biliares/fisiología , Regulación de la Expresión Génica , Hígado/metabolismo , Fosfotransferasas (Aceptor de Grupo Alcohol)/fisiología , Receptores de Lisoesfingolípidos/fisiología , Animales , Hepatocitos , Masculino , Ratones , Ratas , Ratas Sprague-Dawley , Receptores de Lisoesfingolípidos/genética
11.
FASEB J ; 29(12): 5018-28, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26324848

RESUMEN

Sphingosine 1-phosphate (S1P) is a pleiotropic bioactive sphingolipid metabolite that regulates numerous processes important for immune responses. S1P is made within cells and must be transported out of cells to exert its effects through activation of 5 specific cell surface GPCRs in an autocrine or paracrine fashion. Spinster 2 (Spns2) transports S1P out of cells, and its deletion in mice reduces circulating levels of S1P, alters immune cell trafficking, and induces lymphopenia. Here we examined the effects of Spns2 deletion on adaptive immune responses and in autoimmune disease models. Airway inflammation and hypersensitivity as well as delayed-type contact hypersensitivity were attenuated in Spns2(-/-) mice. Similarly, Spns2 deletion reduced dextran sodium sulfate- and oxazolone-induced colitis. Intriguingly, Spns2(-/-) mice were protected from the development of experimental autoimmune encephalopathy, a model of the autoimmune disease multiple sclerosis. Deletion of Spns2 also strongly alleviated disease development in collagen-induced arthritis. These results point to a broad role for Spns2-mediated S1P transport in the initiation and development of adaptive immune related disorders.


Asunto(s)
Proteínas de Transporte de Anión/fisiología , Enfermedades Autoinmunes/fisiopatología , Inflamación/fisiopatología , Animales , Proteínas de Transporte de Anión/genética , Modelos Animales de Enfermedad , Ratones , Ratones Noqueados
12.
Nature ; 465(7301): 1084-8, 2010 Jun 24.
Artículo en Inglés | MEDLINE | ID: mdl-20577214

RESUMEN

Tumour-necrosis factor (TNF) receptor-associated factor 2 (TRAF2) is a key component in NF-kappaB signalling triggered by TNF-alpha. Genetic evidence indicates that TRAF2 is necessary for the polyubiquitination of receptor interacting protein 1 (RIP1) that then serves as a platform for recruitment and stimulation of IkappaB kinase, leading to activation of the transcription factor NF-kappaB. Although TRAF2 is a RING domain ubiquitin ligase, direct evidence that TRAF2 catalyses the ubiquitination of RIP1 is lacking. TRAF2 binds to sphingosine kinase 1 (SphK1), one of the isoenzymes that generates the pro-survival lipid mediator sphingosine-1-phosphate (S1P) inside cells. Here we show that SphK1 and the production of S1P is necessary for lysine-63-linked polyubiquitination of RIP1, phosphorylation of IkappaB kinase and IkappaBalpha, and IkappaBalpha degradation, leading to NF-kappaB activation. These responses were mediated by intracellular S1P independently of its cell surface G-protein-coupled receptors. S1P specifically binds to TRAF2 at the amino-terminal RING domain and stimulates its E3 ligase activity. S1P, but not dihydro-S1P, markedly increased recombinant TRAF2-catalysed lysine-63-linked, but not lysine-48-linked, polyubiquitination of RIP1 in vitro in the presence of the ubiquitin conjugating enzymes (E2) UbcH13 or UbcH5a. Our data show that TRAF2 is a novel intracellular target of S1P, and that S1P is the missing cofactor for TRAF2 E3 ubiquitin ligase activity, indicating a new paradigm for the regulation of lysine-63-linked polyubiquitination. These results also highlight the key role of SphK1 and its product S1P in TNF-alpha signalling and the canonical NF-kappaB activation pathway important in inflammatory, antiapoptotic and immune processes.


Asunto(s)
Lisofosfolípidos/metabolismo , Esfingosina/análogos & derivados , Factor 2 Asociado a Receptor de TNF/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Animales , Biocatálisis , Línea Celular , Activación Enzimática , Humanos , Quinasa I-kappa B/metabolismo , Proteínas I-kappa B/metabolismo , Lisina/metabolismo , Lisofosfolípidos/biosíntesis , Lisofosfolípidos/química , Ratones , Modelos Moleculares , Inhibidor NF-kappaB alfa , FN-kappa B/metabolismo , Fosforilación , Fosfotransferasas (Aceptor de Grupo Alcohol)/genética , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Unión Proteica , Estructura Terciaria de Proteína , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Esfingosina/biosíntesis , Esfingosina/química , Esfingosina/metabolismo , Especificidad por Sustrato , Factor 2 Asociado a Receptor de TNF/química , Factor de Necrosis Tumoral alfa/farmacología , Enzimas Ubiquitina-Conjugadoras/metabolismo , Ubiquitinación/efectos de los fármacos
13.
Mediators Inflamm ; 2016: 2856829, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27843199

RESUMEN

[This corrects the article DOI: 10.1155/2016/8606878.].

14.
Mediators Inflamm ; 2016: 8606878, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26966342

RESUMEN

Sphingosine-1-phosphate (S1P) is a bioactive sphingolipid metabolite involved in many critical cell processes. It is produced by the phosphorylation of sphingosine by sphingosine kinases (SphKs) and exported out of cells via transporters such as spinster homolog 2 (Spns2). S1P regulates diverse physiological processes by binding to specific G protein-binding receptors, S1P receptors (S1PRs) 1-5, through a process coined as "inside-out signaling." The S1P concentration gradient between various tissues promotes S1PR1-dependent migration of T cells from secondary lymphoid organs into the lymphatic and blood circulation. S1P suppresses T cell egress from and promotes retention in inflamed peripheral tissues. S1PR1 in T and B cells as well as Spns2 in endothelial cells contributes to lymphocyte trafficking. FTY720 (Fingolimod) is a functional antagonist of S1PRs that induces systemic lymphopenia by suppression of lymphocyte egress from lymphoid organs. In this review, we summarize previous findings and new discoveries about the importance of S1P and S1PR signaling in the recruitment of immune cells and lymphocyte retention in inflamed tissues. We also discuss the role of S1P-S1PR1 axis in inflammatory diseases and wound healing.


Asunto(s)
Inflamación/metabolismo , Lisofosfolípidos/metabolismo , Esfingosina/análogos & derivados , Animales , Humanos , Modelos Biológicos , Transducción de Señal/fisiología , Esfingosina/metabolismo
15.
J Allergy Clin Immunol ; 136(4): 1035-46.e6, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25842287

RESUMEN

BACKGROUND: Asthma, a chronic inflammatory condition defined by episodic shortness of breath with expiratory wheezing and cough, is a serious health concern affecting more than 250 million persons. Genome-wide association studies have identified ORM (yeast)-like protein isoform 3 (ORMDL3) as a gene associated with susceptibility to asthma. Although its yeast ortholog is a negative regulator of de novo ceramide biosynthesis, how ORMDL3 contributes to asthma pathogenesis is not known. OBJECTIVES: We sought to decipher the molecular mechanism for the pathologic functions of ORMDL3 in asthma and the relationship to its evolutionarily conserved role in regulation of ceramide homeostasis. METHODS: We determined the relationship between expression of ORMDL3 and ceramide in epithelial and inflammatory cells and in asthma pathogenesis in mice. RESULTS: Although small increases in ORMDL3 expression decrease ceramide levels, remarkably, higher expression in lung epithelial cells and macrophages in vitro and in vivo increased ceramide production, which promoted chronic inflammation, airway hyperresponsiveness, and mucus production during house dust mite-induced allergic asthma. Moreover, nasal administration of the immunosuppressant drug FTY720/fingolimod reduced ORMDL3 expression and ceramide levels and mitigated airway inflammation and hyperreactivity and mucus hypersecretion in house dust mite-challenged mice. CONCLUSIONS: Our findings demonstrate that overexpression of ORMDL3 regulates ceramide homeostasis in cells in a complex manner and suggest that local FTY720 administration might be a useful therapeutic intervention for the control of allergic asthma.


Asunto(s)
Asma/inmunología , Ceramidas/inmunología , Regulación de la Expresión Génica/inmunología , Homeostasis/inmunología , Proteínas de la Membrana/inmunología , Animales , Asma/tratamiento farmacológico , Asma/genética , Asma/patología , Línea Celular Tumoral , Ceramidas/genética , Células Epiteliales/inmunología , Células Epiteliales/patología , Femenino , Clorhidrato de Fingolimod/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Homeostasis/efectos de los fármacos , Homeostasis/genética , Humanos , Inmunosupresores/farmacología , Macrófagos/inmunología , Macrófagos/patología , Proteínas de la Membrana/genética , Ratones , Mucosa Respiratoria/inmunología , Mucosa Respiratoria/patología
16.
J Allergy Clin Immunol ; 135(4): 1008-1018.e1, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25512083

RESUMEN

BACKGROUND: Sphingosine-1-phosphate (S1P) is a bioactive sphingolipid produced by mast cells (MCs) on cross-linking of their high-affinity receptors for IgE by antigen that can amplify MC responses by binding to its S1P receptors. An acute MC-dependent allergic reaction can lead to systemic shock, but the early events of its development in lung tissues have not been investigated, and S1P functions in the onset of allergic processes remain to be examined. OBJECTIVE: We used a highly specific neutralizing anti-S1P antibody (mAb) and the sphingosine-1-phosphate receptor 2 (S1PR2) antagonist JTE-013 to study the signaling contributions of S1P and S1PR2 to MC- and IgE-dependent airway allergic responses in mice within minutes after antigen challenge. METHODS: Allergic reaction was triggered by a single intraperitoneal dose of antigen in sensitized mice pretreated intraperitoneally with anti-S1P, isotype control mAb, JTE-013, or vehicle before antigen challenge. RESULTS: Kinetics experiments revealed early pulmonary infiltration of mostly T cells around blood vessels of sensitized mice 20 minutes after antigen exposure. Pretreatment with anti-S1P mAb inhibited in vitro MC activation, as well as in vivo development of airway infiltration and MC activation, reducing serum levels of histamine, cytokines, and the chemokines monocyte chemoattractant protein 1/CCL2, macrophage inflammatory protein 1α/CCL3, and RANTES/CCL5. S1PR2 antagonism or deficiency or MC deficiency recapitulated these results. Both in vitro and in vivo experiments demonstrated MC S1PR2 dependency for chemokine release and the necessity for signal transducer and activator of transcription 3 activation. CONCLUSION: Activation of S1PR2 by S1P and downstream signal transducer and activator of transcription 3 signaling in MCs regulate early T-cell recruitment to antigen-challenged lungs through chemokine production.


Asunto(s)
Hipersensibilidad/inmunología , Hipersensibilidad/metabolismo , Lisofosfolípidos/metabolismo , Mastocitos/inmunología , Mastocitos/metabolismo , Receptores de Lisoesfingolípidos/metabolismo , Esfingosina/análogos & derivados , Linfocitos T/inmunología , Linfocitos T/metabolismo , Traslado Adoptivo , Animales , Antígenos/inmunología , Degranulación de la Célula/efectos de los fármacos , Degranulación de la Célula/inmunología , Quimiocinas/metabolismo , Citocinas/metabolismo , Modelos Animales de Enfermedad , Femenino , Humanos , Pulmón/inmunología , Pulmón/metabolismo , Pulmón/patología , Lisofosfolípidos/antagonistas & inhibidores , Activación de Macrófagos/inmunología , Macrófagos/inmunología , Macrófagos/metabolismo , Macrófagos/patología , Ratones , Ratones Transgénicos , Pirazoles/farmacología , Piridinas/farmacología , Receptores de Lisoesfingolípidos/antagonistas & inhibidores , Factor de Transcripción STAT3/metabolismo , Esfingosina/antagonistas & inhibidores , Esfingosina/metabolismo , Receptores de Esfingosina-1-Fosfato
17.
FASEB J ; 28(10): 4347-58, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25002116

RESUMEN

The tumor necrosis factor (TNF) receptor family member CD40 plays an essential role in the activation of antigen-presenting cells, B cell maturation, and immunoglobulin (Ig) class switching critical for adaptive immunity. Although the bioactive sphingolipid metabolite sphingosine-1-phosphate (S1P) and the kinase that produces it, sphingosine kinase 1 (SphK1), have long been implicated in the actions of TNF mediated by engagement of TNFR1, nothing is yet known of their role in CD40-mediated events. We have now found that ligation of CD40 activates and translocates SphK1 to the plasma membrane, leading to generation of S1P. SphK1 inhibition in human tonsil B cells, as well as inhibition or deletion of SphK1 in mouse splenic B cells, significantly reduced CD40-mediated Ig class switching and plasma cell differentiation ex vivo. Optimal activation of downstream CD40 signaling pathways, including NF-κB, p38, and JNK, also required SphK1. In mice treated with a SphK1 inhibitor or in SphK1(-/-) mice, isotype switching to antigen-specific IgE was decreased in vivo by 70 and 55%, respectively. Our results indicate that SphK1 is important for CD40-mediated B cell activation and regulation of humoral responses and suggest that targeting SphK1 might be a useful therapeutic approach to control antigen-specific IgE production.


Asunto(s)
Antígenos CD40/metabolismo , Cambio de Clase de Inmunoglobulina , Inmunoglobulina E/genética , Lisofosfolípidos/metabolismo , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Esfingosina/análogos & derivados , Animales , Linfocitos B/citología , Linfocitos B/metabolismo , Antígenos CD40/genética , Diferenciación Celular , Membrana Celular/metabolismo , Células HEK293 , Humanos , Inmunoglobulina E/metabolismo , MAP Quinasa Quinasa 4/genética , MAP Quinasa Quinasa 4/metabolismo , Ratones , Ratones Endogámicos C57BL , FN-kappa B/genética , FN-kappa B/metabolismo , Fosfotransferasas (Aceptor de Grupo Alcohol)/genética , Transporte de Proteínas , Esfingosina/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/genética , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
18.
FASEB J ; 27(3): 1001-11, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23180825

RESUMEN

Sphingosine-1-phosphate (S1P), a ligand for 5 specific receptors, is a potent lipid mediator that plays important roles in lymphocyte trafficking and immune responses. S1P is produced inside cells and therefore must be secreted to exert its effects through these receptors. Spinster 2 (Spns2) is one of the cell surface transporters thought to secrete S1P. We have shown that Spns2 can export endogenous S1P from cells and also dihydro-S1P, which is active at all cell surface S1P receptors. Moreover, Spns2 mice have decreased levels of both of these phosphorylated sphingoid bases in blood, accompanied by increases in very long chain ceramide species, and have defective lymphocyte trafficking. Surprisingly, levels of S1P and dihydro-S1P were increased in lymph from Spns2 mice as well as in specific tissues, including lymph nodes, and interstitial fluid. Moreover, lymph nodes from Spns2 mice have aberrant lymphatic sinus that appeared collapsed, with reduced numbers of lymphocytes. Our data suggest that Spns2 is an S1P transporter in vivo that plays a role in regulation not only of blood S1P but also lymph node and lymph S1P levels and consequently influences lymphocyte trafficking and lymphatic vessel network organization.


Asunto(s)
Proteínas de Transporte de Anión/metabolismo , Ganglios Linfáticos/metabolismo , Vasos Linfáticos/metabolismo , Lisofosfolípidos/metabolismo , Esfingosina/análogos & derivados , Animales , Proteínas de Transporte de Anión/genética , Células HEK293 , Humanos , Ganglios Linfáticos/citología , Vasos Linfáticos/citología , Linfocitos/citología , Linfocitos/metabolismo , Lisofosfolípidos/genética , Ratones , Ratones Noqueados , Esfingosina/genética , Esfingosina/metabolismo
19.
FASEB J ; 25(2): 600-12, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20959514

RESUMEN

The potent lipid mediator sphingosine-1-phosphate (S1P) regulates diverse physiological processes by binding to 5 specific GPCRs, although it also has intracellular targets. Here, we demonstrate that S1P, produced in the mitochondria mainly by sphingosine kinase 2 (SphK2), binds with high affinity and specificity to prohibitin 2 (PHB2), a highly conserved protein that regulates mitochondrial assembly and function. In contrast, S1P did not bind to the closely related protein PHB1, which forms large, multimeric complexes with PHB2. In mitochondria from SphK2-null mice, a new aberrant band of cytochrome-c oxidase was detected by blue native PAGE, and interaction between subunit IV of cytochrome-c oxidase and PHB2 was greatly reduced. Moreover, depletion of SphK2 or PHB2 led to a dysfunction in mitochondrial respiration through cytochrome-c oxidase. Our data point to a new action of S1P in mitochondria and suggest that interaction of S1P with homomeric PHB2 is important for cytochrome-c oxidase assembly and mitochondrial respiration.


Asunto(s)
Complejo IV de Transporte de Electrones/metabolismo , Regulación Enzimológica de la Expresión Génica/fisiología , Lisofosfolípidos/biosíntesis , Mitocondrias Cardíacas/enzimología , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Proteínas Represoras/metabolismo , Esfingosina/análogos & derivados , Secuencia de Aminoácidos , Animales , Línea Celular , Complejo IV de Transporte de Electrones/genética , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Miocitos Cardíacos/enzimología , Miocitos Cardíacos/metabolismo , Consumo de Oxígeno/fisiología , Fosfotransferasas (Aceptor de Grupo Alcohol)/genética , Prohibitinas , Proteínas Represoras/genética , Esfingosina/biosíntesis
20.
Cancer Gene Ther ; 29(11): 1662-1675, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-35676533

RESUMEN

The sialyltransferase ST6GAL1 that adds α2-6 linked sialic acids to N-glycans of cell surface and secreted glycoproteins is prominently associated with many human cancers. Tumor-native ST6GAL1 promotes tumor cell behaviors such as invasion and resistance to cell stress and chemo- and radio-treatments. Canonically, ST6GAL1 resides in the intracellular secretory apparatus and glycosylates nascent glycoproteins in biosynthetic transit. However, ST6GAL1 is also released into the extracellular milieu and extracellularly remodels cell surface and secreted glycans. The impact of this non-canonical extrinsic mechanism of ST6GAL1 on tumor cell pathobiology is not known. We hypothesize that ST6GAL1 action is the combined effect of natively expressed sialyltransferase acting cell-autonomously within the ER-Golgi complex and sialyltransferase from extracellular origins acting extrinsically to remodel cell-surface glycans. We found that shRNA knockdown of intrinsic ST6GAL1 expression resulted in decreased ST6GAL1 cargo in the exosome-like vesicles as well as decreased breast tumor cell growth and invasive behavior in 3D in vitro cultures. Extracellular ST6GAL1, present in cancer exosomes or the freely soluble recombinant sialyltransferase, compensates for insufficient intrinsic ST6GAL1 by boosting cancer cell proliferation and increasing invasiveness. Moreover, we present evidence supporting the existence novel but yet uncharacterized cofactors in the exosome-like particles that potently amplify extrinsic ST6GAL1 action, highlighting a previously unknown mechanism linking this enzyme and cancer pathobiology. Our data indicate that extracellular ST6GAL1 from remote sources can compensate for cellular ST6GAL1-mediated aggressive tumor cell proliferation and invasive behavior and has great clinical potential for extracellular ST6GAL1 as these molecules are in the extracellular space should be easily accessible targets.


Asunto(s)
Neoplasias de la Mama , Sialiltransferasas , Humanos , Femenino , Sialiltransferasas/genética , Sialiltransferasas/metabolismo , Neoplasias de la Mama/genética , Glicoproteínas , Polisacáridos/metabolismo , Proliferación Celular , Antígenos CD/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA