Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Carcinogenesis ; 39(1): 47-55, 2018 01 12.
Artículo en Inglés | MEDLINE | ID: mdl-28968647

RESUMEN

Aberrant sphingolipid metabolism has been reported to promote breast cancer progression. Sphingosine kinase 1 (SphK1) is a key metabolic enzyme for the formation of pro-survival S1P from pro-apoptotic ceramide. The role of SphK1 in breast cancer has been well studied in estrogen receptor (ER)-positive breast cancer; however, its role in human epidermal growth factor 2 (HER2)-positive breast cancer remains unclear. Here, we show that genetic deletion of SphK1 significantly reduced mammary tumor development with reduced tumor incidence and multiplicity in the MMTV-neu transgenic mouse model. Gene expression analysis revealed significant reduction of claudin-2 (CLDN2) expression in tumors from SphK1 deficient mice, suggesting that CLDN2 may mediate SphK1's function. It is remarkable that SphK1 deficiency in HER2-positive breast cancer model inhibited tumor formation by the different mechanism from ER-positive breast cancer. In vitro experiments demonstrated that overexpression of SphK1 in ER-/PR-/HER2+ human breast cancer cells enhanced cell proliferation, colony formation, migration and invasion. Furthermore, immunostaining of SphK1 and CLDN2 in HER2-positive human breast tumors revealed a correlation in high-grade disease. Taken together, these findings suggest that SphK1 may play a pivotal role in HER2-positive breast carcinogenesis. Targeting SphK1 may represent a novel approach for HER2-positive breast cancer chemoprevention and/or treatment.


Asunto(s)
Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Fosfotransferasas (Aceptor de Grupo Alcohol)/genética , Receptor ErbB-2/genética , Animales , Neoplasias de la Mama/metabolismo , Modelos Animales de Enfermedad , Femenino , Humanos , Ratones , Ratones Transgénicos
2.
Carcinogenesis ; 38(12): 1218-1227, 2017 12 07.
Artículo en Inglés | MEDLINE | ID: mdl-29028945

RESUMEN

Accumulating evidence suggests that the sphingosine kinase 1 (SphK1)/sphingosine 1-phosphate (S1P) pathway plays a pivotal role in colon carcinogenesis. Our previous studies indicate that the SphK1/S1P pathway mediates colon carcinogenesis at least by regulating cyclooxygenase 2 (COX-2) expression and prostaglandin E2 (PGE2) production. However, the mechanisms by which this pathway regulates colon carcinogenesis are still unclear. First, we show that SphK1 deficient mice significantly attenuated azoxymethane (AOM)-induced colon carcinogenesis as measured by colon tumor incidence, multiplicity, and volume. We found that AOM activates peritoneal macrophages to induce SphK1, COX-2, and tumor necrosis factor (TNF)-α expression in WT mice. Interestingly, SphK1 knockout (KO) mice revealed significant reduction of COX-2 and TNF-α expression from AOM-activated peritoneal macrophages, suggesting that SphK1 regulates COX-2 and TNF-α expression in peritoneal macrophages. We found that inoculation of WT peritoneal macrophages restored the carcinogenic effect of AOM in Sphk1 KO mice as measured by aberrant crypt foci (ACF) formation, preneoplastic lesions of colon cancer. In addition, downregulation of SphK1 only in peritoneal macrophage by short hairpin RNA (shRNA) reduced the number of ACF per colon induced by AOM. Intraperitoneal injection of sphingolipids demonstrates that S1P enhanced AOM-induced ACF formation, while ceramide inhibited. Finally, we show that SphK inhibitor SKI-II significantly reduced the number of ACF per colon. These results suggest that SphK1 expression plays a pivotal role in the early stages of colon carcinogenesis through regulating COX-2 and TNF-α expression from activated peritoneal macrophages.


Asunto(s)
Carcinogénesis/metabolismo , Neoplasias del Colon/patología , Macrófagos Peritoneales/enzimología , Fosfotransferasas (Aceptor de Grupo Alcohol)/biosíntesis , Animales , Neoplasias del Colon/enzimología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
4.
Cell Commun Signal ; 15(1): 30, 2017 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-28810912

RESUMEN

BACKGROUND: Magnesium (Mg2+) is an essential cation implicated in carcinogenesis, solid tumor progression and metastatic potential. The Transient Receptor Potential Melastatin Member 7 (TRPM7) is a divalent ion channel involved in cellular and systemic Mg2+ homeostasis. Abnormal expression of TRPM7 is found in numerous cancers, including colon, implicating TRPM7 in this process. METHODS: To establish a possible link between systemic magnesium (Mg2+) status, the Mg2+ conducting channel TRPM7 in colon epithelial cells, and colon carcinogenesis, in vitro whole-cell patch clamp electrophysiology, qPCR, and pharmacological tools were used probing human colorectal adenocarcinoma HT-29 as well as normal primary mouse colon epithelial cells. This was extended to and combined with aberrant crypt foci development in an azoxymethane-induced colorectal cancer mouse model under hypomagnesemia induced by diet or pharmacologic intervention. RESULTS: We find that TRPM7 drives colon cancer cell proliferation in human HT-29 and expresses in normal primary mouse colon epithelia. This is linked to TRPM7's dominant role as Mg2+ transporter, since high extracellular Mg2+ supplementation cannot rescue inhibition of cell proliferation caused by suppressing TRPM7 either genetically or pharmacologically. In vivo experiments in mice provide evidence that the specific TRPM7 inhibitor waixenicin A, given as a single bolus injection, induces transient hypomagnesemia and increases intestinal absorption of calcium. Repeated injections of waixenicin A over 3 weeks cause hypomagnesemia via insufficient Mg2+ absorption by the colon. However, neither waixenicin A, nor a diet low in Mg2+, affect aberrant crypt foci development in an azoxymethane-induced colorectal cancer mouse model. CONCLUSION: Early stage colon cancer proceeds independent of systemic Mg2+ status and TRPM7, and waixenicin A is a useful pharmacological tool to study of TRPM7 in vitro and in vivo.


Asunto(s)
Adenocarcinoma/metabolismo , Proliferación Celular/efectos de los fármacos , Neoplasias del Colon/metabolismo , Deficiencia de Magnesio/metabolismo , Canales Catiónicos TRPM/antagonistas & inhibidores , Acetatos/farmacología , Adenocarcinoma/etiología , Animales , Azoximetano/toxicidad , Calcio/metabolismo , Células Cultivadas , Neoplasias del Colon/etiología , Diterpenos/farmacología , Células HT29 , Humanos , Absorción Intestinal , Deficiencia de Magnesio/sangre , Deficiencia de Magnesio/etiología , Masculino , Ratones , Ratones Endogámicos C57BL
5.
J Transl Med ; 15(1): 120, 2017 06 06.
Artículo en Inglés | MEDLINE | ID: mdl-28583134

RESUMEN

BACKGROUND: Accumulating evidence suggests that sphingosine kinase 1 (SphK1)/sphingosine 1-phosphate pathway plays a pivotal role in colon carcinogenesis. METHODS: To further support the evidence, we investigated the effects of SphK1 using three separate animal models: SphK1 knockout mice, SphK1 overexpressing transgenic mice, and SphK1 overexpression in human colon cancer xenografts. Using azoxymethane (AOM, colon carcinogen), we analyzed colon tumor development in SphK1 KO and SphK1 overexpression in intestinal epithelial cells regulated by a tet-on system. Then, we analyzed subcutaneous tumor growth using xenografts of HT-29 human colon cancer cell. Finally, immunohistochemical analyses for SphK1 and COX-2 were performed on human colon cancer tissue microarray. RESULTS: SphK1 KO mice, compared to wild-type mice, demonstrated a significant inhibition in colon cancer development induced by AOM (58.6% vs. 96.4%, respectively, P < 0.005). Tumor multiplicity (1.00 vs. 1.64 per colon, respectively, P < 0.05) and tumor volume (14.82 mm3 vs. 29.10 mm3, P < 0.05) were both significantly reduced in SphK1 KO mice compared to wild-type mice. Next, SphK1 overexpression in HT-29 enhanced tumor growth as compared to GFP control in nude mice (229.5 mm3 vs. 90.9 mm3, respectively, P < 0.05). Furthermore, overexpression of SphK1 in intestinal epithelial cells significantly enhances AOM-induced colon tumor formation (P < 0.05). Lastly, SphK1 and COX-2 intensity tended to reduce overall survival of late stage colon cancer patients. CONCLUSIONS: SphK1 expression regulates the early stage of colon carcinogenesis and tumor growth, thus inhibition of SphK1 may be an effective strategy for colon cancer chemoprevention.


Asunto(s)
Neoplasias del Colon/enzimología , Neoplasias del Colon/patología , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Anciano , Animales , Azoximetano , Carcinogénesis/patología , Proliferación Celular , Ciclooxigenasa 2/metabolismo , Enterocitos/metabolismo , Enterocitos/patología , Femenino , Células HT29 , Humanos , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Persona de Mediana Edad , Estadificación de Neoplasias
6.
FASEB J ; 30(12): 4159-4171, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27609772

RESUMEN

Alterations in sphingolipid metabolism, especially ceramide and sphingosine 1-phosphate, have been linked to colon cancer, suggesting that enzymes of sphingolipid metabolism may emerge as novel regulators and targets in colon cancer. Neutral ceramidase (nCDase), a key enzyme in sphingolipid metabolism that hydrolyzes ceramide into sphingosine, is highly expressed in the intestine; however, its role in colon cancer has not been defined. Here we show that molecular and pharmacological inhibition of nCDase in colon cancer cells increases ceramide, and this is accompanied by decreased cell survival and increased apoptosis and autophagy, with minimal effects on noncancerous cells. Inhibition of nCDase resulted in loss of ß-catenin and inhibition of ERK, components of pathways relevant for colon cancer development. Furthermore, inhibition of nCDase in a xenograft model delayed tumor growth and increased ceramide while decreasing proliferation. It is noteworthy that mice lacking nCDase treated with azoxymethane were protected from tumor formation. Taken together, these studies show that nCDase is pivotal for regulating initiation and development of colon cancer, and these data suggest that this enzyme is a suitable and novel target for colon cancer therapy.-García-Barros, M., Coant, N., Kawamori, T., Wada, M., Snider, A. J., Truman, J.-P., Wu, B. X., Furuya, H., Clarke, C. J., Bialkowska, A. B., Ghaleb, A., Yang, V. W., Obeid, L. M., Hannun, Y. A. Role of neutral ceramidase in colon cancer.


Asunto(s)
Ceramidas/metabolismo , Neoplasias del Colon/enzimología , Metabolismo de los Lípidos/fisiología , Ceramidasa Neutra/metabolismo , Animales , Colon/metabolismo , Humanos , Masculino , Ratones Noqueados , Esfingolípidos/metabolismo , beta Catenina/metabolismo
7.
PLoS One ; 9(12): e113998, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25460165

RESUMEN

Sphingosine kinase 1 (SK1), one of two SK enzymes, is highly regulated and has been shown to act as a focal point for the action of many growth factors and cytokines. SK1 leads to generation of sphingosine-1-phosphate (S1P) and potentially the activation of S1P receptors to mediate biologic effects. Our previous studies implicated SK1/S1P in the regulation of inflammatory processes, specifically in inflammatory bowel disease (IBD). These studies were conducted using a total body knockout mouse for SK1 and were unable to determine the source of SK1/S1P (hematopoietic or extra-hematopoietic) involved in the inflammatory responses. Therefore, bone marrow transplants were performed with wild-type (WT) and SK1-/- mice and colitis induced with dextran sulfate sodium (DSS). Irrespective of the source of SK1/S1P, bone marrow or tissue, DSS induced colitis in all mice; however, mice lacking SK1 in both hematopoietic and extra-hematopoietic compartments exhibited decreased crypt damage. Systemic inflammation was assessed, and mice with WT bone marrow demonstrated significant neutrophilia in response to DSS. In the local inflammatory response, mice lacking SK1/S1P in either bone marrow or tissue exhibited decreased induction of cytokines and less activation of STAT3 (signal transducer and activator of transcription 3). Interestingly, we determined that extra-hematopoietic SK1 is necessary for the induction of cyclooxygenase 2 (COX2) in colon epithelium in response to DSS-induced colitis. Taken together our data suggest that hematopoietic-derived SK1/S1P regulates specific aspects of the systemic inflammatory response, while extra-hematopoietic SK1 in the colon epithelium is necessary for the autocrine induction of COX2 in DSS-induced colitis.


Asunto(s)
Sistema Hematopoyético/metabolismo , Enfermedades Inflamatorias del Intestino/metabolismo , Fosfotransferasas (Aceptor de Grupo Alcohol)/fisiología , Animales , Colitis/inducido químicamente , Colitis/patología , Inflamación/genética , Enfermedades Inflamatorias del Intestino/patología , Lisofosfolípidos/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fosfotransferasas (Aceptor de Grupo Alcohol)/genética , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Receptores de Lisoesfingolípidos/metabolismo , Esfingosina/análogos & derivados , Esfingosina/metabolismo
8.
Cancer Cell Int ; 14(1): 76, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25197261

RESUMEN

BACKGROUND: Head and neck squamous cell carcinoma (HNSCC) is characterized by aggressive loco-regional invasion. Sphingosine kinase1 (SphK1), an enzyme in sphingolipid metabolism, is emerging as a key player in HNSCC pathology. The observation that SphK1 is overexpressed in all HNSCC stages and is associated with depth of tumor invasion, metastasis and clinical failure underscores the importance of SphK1 in HNSCC pathology. Still, the mechanisms underlying SphK1 regulation of invasion have not been delineated. Therefore, we sought to mechanistically describe how SphK1 regulates invasion in HNSCC. METHODS: Invasion assays were used to measure invasive ability of SphK1 overexpressing human tongue squamous cell carcinoma (SCC-25 cells). Western blotting, quantitative qPCR, ELISA and zymography were used to measure the effect of SphK1 and sphingosine 1-phoshate receptor 1 (S1P1) on invasion measures, MMP-2/9, E-cadherin, EGFR, IL-6/STAT3, in SCC-25 cells. RESULTS: SphK1 expression is elevated in cells with an invasive phenotype as compared to non-invasive phenotype. We show SphK1 overexpression increased EGF-induced EGFR/ERK and AKT activity, increased matrix metalloproteinase (MMP)-2/9 mRNA and reduced E-cadherin. SphK1 overexpression also increased IL-6 concentration and EGF-induced STAT3 phosphorylation, exemplifying that SphK1 modulates IL-6/STAT3 signaling. Notably, we show that S1P1 knockdown reduced IL-6/STAT3 signaling, representing another pathway by which SphK1/S1P regulates invasion. CONCLUSIONS: Taken together, our data suggest that SphK1 sits at the hub of multiple key signaling cascades, all which have been implicated in the regulation of invasiveness, making SphK1 an attractive target for the development of HNSCC therapies.

9.
Nutr Cancer ; 65 Suppl 1: 54-60, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23682783

RESUMEN

The present study explored the preventive effects of perilla oil, rich in α-linolenic acid, in rodent models of colon tumorigenesis. Six-week-old male F344 rats were fed diets containing 5% corn oil or 10 or 20% perilla oil. Colonic aberrant crypt foci (ACF) were induced by azoxymethane (AOM) and colonic ACF were evaluated. In familial adenomatous polyposis mode, APC(min) mice fed with 20% corn oil or perilla oil for 80 days and intestines were evaluated for polyps. Multiple colonic mucosal and polyp samples were assayed for the expression and activity of cyclooxygenase COX-isoforms. Dietary perilla oil produced a dose-dependent inhibition of AOM-induced colonic ACF formation (by 35-53%, P < 0.01-0.005) and reduced the number of foci with ≥ 4 crypts/focus (by 38-50%, P < 0.01-0.001) in F344 rats. Dietary perilla oil significantly inhibited development of small intestinal (>69%, P < 0.0001) and colon tumors (>52%, P < 0.03) in APC(min) mice. Administration of perilla oil produced lower levels of type-2 prostaglandins (38-53%) from COX-activities in polyps of APC(min) mice. These observations demonstrate that dietary perilla oil rich in ω-3 fatty acids possesses preventive activity against intestinal neoplastic lesions, both in FAP in genetically-predisposed tissues, as well as against chemically induced preneoplastic lesions in the colon.


Asunto(s)
Focos de Criptas Aberrantes/patología , Poliposis Adenomatosa del Colon/tratamiento farmacológico , Anticarcinógenos/farmacología , Carcinogénesis/efectos de los fármacos , Neoplasias del Colon/tratamiento farmacológico , Ácido alfa-Linolénico/farmacología , Focos de Criptas Aberrantes/inducido químicamente , Poliposis Adenomatosa del Colon/inducido químicamente , Poliposis Adenomatosa del Colon/patología , Animales , Azoximetano/toxicidad , Carcinogénesis/inducido químicamente , Neoplasias del Colon/inducido químicamente , Neoplasias del Colon/patología , Aceite de Maíz/administración & dosificación , Ciclooxigenasa 1/genética , Ciclooxigenasa 1/metabolismo , Ciclooxigenasa 2/genética , Ciclooxigenasa 2/metabolismo , Relación Dosis-Respuesta a Droga , Femenino , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/patología , Masculino , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Aceites de Plantas/farmacología , Ratas , Ratas Endogámicas F344
10.
FASEB J ; 27(2): 656-64, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23109673

RESUMEN

Accumulating evidence suggests that sphingosine kinase 1 (SphK1) plays a key role in carcinogenesis by regulating cyclooxygenase-2 (COX-2) expression. Recent clinical studies have revealed that COX-2 inhibitors cause adverse cardiovascular side effects, likely due to inhibition of prostacyclin (PGI(2)). In this work, we investigated the roles of SphK1 inhibition on blood pressure (BP). The results show that lack of SphK1 expression did not exacerbate angiotensin II (Ang II)-induced acute hypertension, whereas celecoxib, a COX-2 inhibitor, augmented and sustained higher BP in mice. Interestingly, SphK1-knockout mice inhibited prostaglandin E(2) (PGE(2)) but not PGI(2) production in response to Ang II, whereas celecoxib blocked both PGE(2) and PGI(2) production. Mechanistically, SphK1 down-regulation by siRNA in human umbilical vein endothelial cells decreased cytokine-induced PGE(2) production primarily through inhibition of microsomal PGE synthase-1 (mPGES-1), not COX-2. SphK1 down-regulation also decreased MKK6 expression, which phosphorylates and activates P38 MAPK, which, in turn, regulates early growth response-1 (Egr-1), a transcription factor of mPGES-1. Together, these data indicate that SphK1 regulates PGE(2) production by mPGES-1 expression via the p38 MAPK pathway, independent of COX-2 signaling, in endothelial cells, suggesting that SphK1 inhibition may be a promising strategy for cancer chemoprevention with lack of the adverse cardiovascular side effects associated with coxibs.


Asunto(s)
Presión Sanguínea/fisiología , Fosfotransferasas (Aceptor de Grupo Alcohol)/antagonistas & inhibidores , Animales , Secuencia de Bases , Celecoxib , Ciclooxigenasa 2/metabolismo , Inhibidores de la Ciclooxigenasa 2/farmacología , Dinoprostona/biosíntesis , Epoprostenol/biosíntesis , Técnicas de Silenciamiento del Gen , Células Endoteliales de la Vena Umbilical Humana , Humanos , Hipertensión/tratamiento farmacológico , Hipertensión/fisiopatología , Oxidorreductasas Intramoleculares/metabolismo , Sistema de Señalización de MAP Quinasas , Masculino , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Proteínas Mitocondriales , Fosfotransferasas (Aceptor de Grupo Alcohol)/deficiencia , Fosfotransferasas (Aceptor de Grupo Alcohol)/genética , Fosfotransferasas (Aceptor de Grupo Alcohol)/fisiología , Prostaglandina-E Sintasas , Pirazoles/farmacología , ARN Interferente Pequeño/genética , Sulfonamidas/farmacología
11.
Biomolecules ; 3(3): 481-513, 2013 Aug 12.
Artículo en Inglés | MEDLINE | ID: mdl-24970177

RESUMEN

Head and neck squamous cell carcinoma (HNSCC) has a high reoccurrence rate and an extremely low survival rate. There is limited availability of effective therapies to reduce the rate of recurrence, resulting in high morbidity and mortality of advanced cases. Late presentation, delay in detection of lesions, and a high rate of metastasis make HNSCC a devastating disease. This review offers insight into the role of sphingosine kinase-1 (SphK1), a key enzyme in sphingolipid metabolism, in HNSCC. Sphingolipids not only play a structural role in cellular membranes, but also modulate cell signal transduction pathways to influence biological outcomes such as senescence, differentiation, apoptosis, migration, proliferation, and angiogenesis. SphK1 is a critical regulator of the delicate balance between proliferation and apoptosis. The highest expression of SphK1 is found in the advanced stage of disease, and there is a positive correlation between SphK1 expression and recurrent tumors. On the other hand, silencing SphK1 reduces HNSCC tumor growth and sensitizes tumors to radiation-induced death.  Thus, SphK1 plays an important and influential role in determining HNSCC proliferation and metastasis. We discuss roles of SphK1 and other sphingolipids in HNSCC development and therapeutic strategies against HNSCC.

12.
Prostaglandins Other Lipid Mediat ; 99(3-4): 124-30, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22940715

RESUMEN

Sphingolipids are emerging as important mediators of immune and inflammatory responses. We have previously demonstrated that sphingosine-1-phosphate (S1P) and its synthetic enzyme sphingosine kinase-1 (SK1) play an important role in inflammatory bowel disease. S1P generation is dependent on SK phosphorylation of sphingosine. Generation of sphingosine results only from the breakdown of ceramide by ceramidases (CDase). In this study, we set out to determine the role of neutral CDase (nCDase) in S1P generation and inflammatory bowel disease. To this end, we established nCDase expression is increased in patients with ulcerative colitis. Using the dextran sulfate sodium (DSS)-induced colitis model, we determined nCDase activity increased in colon epithelium, but not submucosa, in wild-type (WT) mice. Following DSS, ceramide levels were elevated in colon epithelium from WT and nCDase(-/-) mice, while S1P levels were significantly elevated only in the epithelium of nCDase(-/-) mice. Similarly, cyclooxygenase-2 (Cox-2) levels were significantly elevated only in the epithelium of nCDase(-/-) mice. Neutral CDase(-/-) mice also exhibited higher endotoxin levels in circulation, as well as higher circulating levels of S1P. This increase in S1P in nCDase(-/-) mice was accompanied by a marked leukocytosis, most notably circulating neutrophils and lymphocytes. Taken together these data demonstrate that loss of nCDase results in an unexpected increase in S1P generation in inflammation, and suggests that nCDase may actually protect against inflammation.


Asunto(s)
Colitis Ulcerosa/metabolismo , Colon/metabolismo , Inflamación/metabolismo , Mucosa Intestinal/metabolismo , Leucocitosis/metabolismo , Lisofosfolípidos/metabolismo , Ceramidasa Neutra/deficiencia , Esfingosina/análogos & derivados , Animales , Colitis Ulcerosa/inducido químicamente , Colitis Ulcerosa/complicaciones , Colitis Ulcerosa/patología , Colon/patología , Sulfato de Dextran , Modelos Animales de Enfermedad , Endotoxinas/sangre , Humanos , Inflamación/complicaciones , Inflamación/patología , Mucosa Intestinal/patología , Leucocitosis/complicaciones , Leucocitosis/patología , Ratones , Ratones Noqueados , Ceramidasa Neutra/genética , Transducción de Señal , Esfingosina/metabolismo
13.
EMBO Mol Med ; 4(8): 761-75, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22707406

RESUMEN

Mechanisms by which cancer cells communicate with the host organism to regulate lung colonization/metastasis are unclear. We show that this communication occurs via sphingosine 1-phosphate (S1P) generated systemically by sphingosine kinase 1 (SK1), rather than via tumour-derived S1P. Modulation of systemic, but not tumour SK1, prevented S1P elevation, and inhibited TRAMP-induced prostate cancer growth in TRAMP(+/+) SK1(-/-) mice, or lung metastasis of multiple cancer cells in SK1(-/-) animals. Genetic loss of SK1 activated a master metastasis suppressor, Brms1 (breast carcinoma metastasis suppressor 1), via modulation of S1P receptor 2 (S1PR2) in cancer cells. Alterations of S1PR2 using pharmacologic and genetic tools enhanced Brms1. Moreover, Brms1 in S1PR2(-/-) MEFs was modulated by serum S1P alterations. Accordingly, ectopic Brms1 in MB49 bladder cancer cells suppressed lung metastasis, and stable knockdown of Brms1 prevented this process. Importantly, inhibition of systemic S1P signalling using a novel anti-S1P monoclonal antibody (mAb), Sphingomab, attenuated lung metastasis, which was prevented by Brms1 knockdown in MB49 cells. Thus, these data suggest that systemic SK1/S1P regulates metastatic potential via regulation of tumour S1PR2/Brms1 axis.


Asunto(s)
Neoplasias Pulmonares/patología , Neoplasias Pulmonares/secundario , Lisofosfolípidos/metabolismo , Metástasis de la Neoplasia/patología , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Neoplasias de la Próstata/patología , Transducción de Señal , Esfingosina/análogos & derivados , Animales , Modelos Animales de Enfermedad , Humanos , Masculino , Ratones , Ratones Noqueados , Receptores de Lisoesfingolípidos/metabolismo , Proteínas Represoras/metabolismo , Esfingosina/metabolismo , Receptores de Esfingosina-1-Fosfato , Neoplasias de la Vejiga Urinaria/patología
14.
Cancer Metastasis Rev ; 30(3-4): 567-76, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22005951

RESUMEN

The bioactive sphingolipids including, ceramide, sphingosine, and sphingosine-1-phosphate (S1P) have important roles in several types of signaling and regulation of many cellular processes including cell proliferation, apoptosis, senescence, angiogenesis, and transformation. Recent accumulating evidence suggests that ceramide- and S1P-mediated pathways have been implicated in cancer development, progression, and chemotherapy. Ceramide mediates numerous cell-stress responses, such as induction of apoptosis and cell senescence, whereas S1P plays pivotal roles in cell survival, migration, and inflammation. These sphingolipids with opposing roles can be interconverted within cells, suggesting that the balance between them is related to cell fate. Importantly, these sphingolipids are metabolically related through actions of enzymes including ceramidases, ceramide synthases, sphingosine kinases, and S1P phosphatases thereby forming a network of metabolically interrelated bioactive lipid mediators whose importance in normal cellular function and diseases is gaining appreciation. In this review, we summarize involvement of sphingolipids and their related enzymes in pathogenesis and therapy of cancer and discuss future directions of sphingolipid field in cancer research.


Asunto(s)
Neoplasias de la Mama/metabolismo , Neoplasias del Colon/metabolismo , Neoplasias de Cabeza y Cuello/metabolismo , Neoplasias de la Próstata/metabolismo , Esfingolípidos/metabolismo , Animales , Neoplasias de la Mama/patología , Transformación Celular Neoplásica , Neoplasias del Colon/patología , Femenino , Neoplasias de Cabeza y Cuello/patología , Humanos , Inflamación/metabolismo , Masculino , Neoplasias de la Próstata/patología
15.
Cancer Prev Res (Phila) ; 4(3): 454-62, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21209394

RESUMEN

It is important to identify novel and effective targets for cancer prevention and therapy against head and neck squamous cell carcinoma (HNSCC), one of the most lethal cancers. Accumulating evidence suggests that the bioactive sphingolipids, such as sphingosine-1-phosphate (S1P) and its generating enzyme, sphingosine kinase 1 (SphK1) play pivotal roles in several important biological functions including promoting tumor growth and carcinogenesis. However, roles of SphK1/S1P in HNSCC development and/or progression have not been defined previously. Therefore, in this study, we first analyzed the expression of SphK1 in human HNSCC tumor samples and normal head & neck tissues (n = 78 and 17, respectively) using immunohistochemistry. The data showed that SphK1 is overexpressed in all of the HNSCC tumors tested (stages I-IV). We next investigated whether SphK1 is necessary for HNSCC development. To define the role of SphK1/S1P in HNSCC development, we utilized 4-nitroquinoline-1-oxide (4-NQO)-induced HNSCC model in wild-type mice compared with SphK1(-/-) knockout (KO) mice. Remarkably, we found that the genetic loss of SphK1, which reduced S1P generation, significantly prevented 4-NQO-induced HNSCC carcinogenesis, with decreased tumor incidence, multiplicity, and volume when compared with controls. Moreover, our data indicated that prevention of 4-NQO-induced HNSCC development in SphK1(-/-) KO mice might be associated with decreased cell proliferation, increased levels of cleaved (active) caspase 3, and downregulation of phospho (active) AKT expression. Thus, these novel data suggest that SphK1/S1P signaling may play important roles in HNSCC carcinogenesis, and that targeting SphK1/S1P might provide a novel strategy for chemoprevention and treatment against HNSCC.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Neoplasias de Cabeza y Cuello/enzimología , Fosfotransferasas (Aceptor de Grupo Alcohol)/fisiología , Animales , Proliferación Celular , Progresión de la Enfermedad , Neoplasias de Cabeza y Cuello/metabolismo , Humanos , Inmunohistoquímica/métodos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Modelos Genéticos , Análisis de Secuencia por Matrices de Oligonucleótidos , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo
16.
Adv Exp Med Biol ; 688: 109-17, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20919649

RESUMEN

Bioactive sphingolipids play key roles in the regulation of several fundamental biological processes such as proliferation, apoptosis and transformation. The recent development of genetically engineered mouse (GEM) models has enabled the study of functional roles of sphingolipids in normal development and disease. In this chapter, we review the phenotypes of GEM models (knockout mice) that lack sphingolipid metabolism-related enzymes, discuss what we have learned from animal models and describe future directions of animal models in sphingolipid research.


Asunto(s)
Ceramidas/fisiología , Animales , Ceramidasas/deficiencia , Ceramidasas/fisiología , Ratones , Ratones Noqueados , Modelos Animales , Fosfotransferasas (Aceptor de Grupo Alcohol)/deficiencia , Fosfotransferasas (Aceptor de Grupo Alcohol)/fisiología , Esfingomielina Fosfodiesterasa/deficiencia , Esfingomielina Fosfodiesterasa/fisiología , Transferasas (Grupos de Otros Fosfatos Sustitutos)/deficiencia , Transferasas (Grupos de Otros Fosfatos Sustitutos)/fisiología
17.
Prostaglandins Other Lipid Mediat ; 90(1-2): 31-6, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19647091

RESUMEN

Accumulating evidence suggests that cyclooxygenase-2 (COX-2) and prostaglandin E(2) (PGE(2)) may play an important role in colon carcinogenesis. Thus, blockage of this pathway may be a suitable strategy for colon cancer chemoprevention. Recent clinical studies suggest that COX-2 inhibitors cause adverse cardiovascular effects due to prostacyclin (PGI(2)) inhibition. To test our hypothesis that inhibition of PGE(2) signaling through E-prostanoid (EP) receptors may offer a safer cardiovascular profile than COX-2 inhibition, we analyzed expression of 6-keto PGF(1alpha), a hydrated form of PGI(2) and PGI(2) synthase, which was stimulated with cytokines in human umbilical vein endothelial cells (HUVECs) treated with the EP(1) receptor antagonist ONO-8711 or the COX-2 inhibitor celecoxib. ONO-8711 did not inhibit both 6-keto PGF(1alpha) production and PGIS expression, whereas celecoxib did in HUVECs. ONO-8711 also inhibited cytokine-induced tissue factor expression in HUVECs. These results suggest that ONO-8711 may be a safer chemopreventive agent with respect to cardiovascular events.


Asunto(s)
Dinoprostona/metabolismo , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Epoprostenol/biosíntesis , Receptores de Prostaglandina E/metabolismo , Transducción de Señal/efectos de los fármacos , Coagulación Sanguínea/efectos de los fármacos , Compuestos Bicíclicos con Puentes/farmacología , Caproatos/farmacología , Enfermedades Cardiovasculares/tratamiento farmacológico , Celecoxib , Línea Celular , Citocinas/farmacología , Fibrinolíticos/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Receptores Activados del Proliferador del Peroxisoma/genética , Pirazoles/farmacología , Receptores de Prostaglandina E/antagonistas & inhibidores , Receptores de Prostaglandina E/genética , Elementos de Respuesta , Sulfonamidas/farmacología , Tromboplastina/genética
18.
FASEB J ; 23(2): 405-14, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18824518

RESUMEN

Sphingosine kinase 1 (SphK1) phosphorylates sphingosine to form sphingosine-1-phosphate (S1P) and is a critical regulator of sphingolipid-mediated functions. Cell-based studies suggest a tumor-promoting function for the SphK1/S1P pathway. Also, our previous studies implicated the SphK1/S1P pathway in the induction of the arachidonic acid cascade, a major inflammatory pathway involved in colon carcinogenesis. Therefore, we investigated whether the SphK1/S1P pathway is necessary for mediating carcinogenesis in vivo. Here, we report that 89% (42/47) of human colon cancer samples stained positively for SphK1, whereas normal colon mucosa had negative or weak staining. Adenomas had higher expression of SphK1 vs. normal mucosa, and colon cancers with metastasis had higher expression of SphK1 than those without metastasis. In the azoxymethane (AOM) murine model of colon cancer, SphK1 and S1P were significantly elevated in colon cancer tissues compared to normal mucosa. Moreover, blood levels of S1P were higher in mice with colon cancers than in those without cancers. Notably, SphK1(-/-) mice subjected to AOM had significantly less aberrant crypt foci (ACF) formation and significantly reduced colon cancer development. These results are the first in vivo evidence that the SphK1/S1P pathway contributes to colon carcinogenesis and that inhibition of this pathway is a potential target for chemoprevention.


Asunto(s)
Transformación Celular Neoplásica/metabolismo , Transformación Celular Neoplásica/patología , Neoplasias del Colon/enzimología , Neoplasias del Colon/patología , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Animales , Azoximetano/farmacología , Transformación Celular Neoplásica/genética , Neoplasias del Colon/inducido químicamente , Ciclooxigenasa 2/metabolismo , Sulfato de Dextran/farmacología , Regulación Enzimológica de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Humanos , Inflamación/genética , Inflamación/metabolismo , Inflamación/patología , Lisofosfolípidos/biosíntesis , Ratones , Ratones Noqueados , Fosfotransferasas (Aceptor de Grupo Alcohol)/deficiencia , Fosfotransferasas (Aceptor de Grupo Alcohol)/genética , Esfingosina/análogos & derivados , Esfingosina/biosíntesis
19.
FASEB J ; 23(1): 143-52, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18815359

RESUMEN

The bioactive lipid sphingosine-1-phosphate (S1P) is emerging as an important mediator of immune and inflammatory responses. S1P formation is catalyzed by sphingosine kinase (SK), of which the SK1 isoenzyme is activated by tumor necrosis alpha (TNF-alpha). SK1 has been shown to be required for mediating TNF-alpha inflammatory responses in cells, including induction of cyclooxygenase 2 (COX-2). Because TNF-alpha and COX-2 are increased in patients with inflammatory bowel disease (IBD), we investigated the role of SK1 in a murine model of colitis. SK1(-/-) mice treated with dextran sulfate sodium (DSS) had significantly less blood loss, weight loss, colon shortening, colon histological damage, and splenomegaly than did wild-type (WT) mice. In addition, SK1(-/-) mice had no systemic inflammatory response. Moreover, WT but not SK1(-/-) mice treated with dextran sulfate sodium had significant increases in blood S1P levels, colon SK1 message and activity, and colon neutrophilic infiltrate. Unlike WT mice, SK1(-/-) mice failed to show colonic COX-2 induction despite an exaggerated TNF-alpha response; thus implicating for the first time SK1 in TNF-alpha-mediated COX-2 induction in vivo. Inhibition of SK1 may prove to be a valuable therapeutic target by inhibiting systemic and local inflammation in IBD.


Asunto(s)
Colitis/inducido químicamente , Colitis/metabolismo , Sulfato de Dextran/toxicidad , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Animales , Peso Corporal , Colitis Ulcerosa/metabolismo , Colitis Ulcerosa/patología , Colon/patología , Ciclooxigenasa 2/metabolismo , Eritrocitos/metabolismo , Regulación de la Expresión Génica/fisiología , Humanos , Lisofosfolípidos/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Tamaño de los Órganos , Fosfotransferasas (Aceptor de Grupo Alcohol)/genética , Esfingosina/análogos & derivados , Esfingosina/metabolismo , Bazo/patología , Factor de Necrosis Tumoral alfa/metabolismo
20.
Biochem Pharmacol ; 73(9): 1288-96, 2007 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-17250812

RESUMEN

Poor oral bioavailability has been a major limitation for the successful use of dietary flavonoids as cancer chemopreventive agents. In this study, we examined fully methylated flavones as promising improved agents. In the human oral SCC-9 cancer cells, 5,7-dimethoxyflavone and 5,7,4'-trimethoxyflavone were both 10 times more potent inhibitors of cell proliferation (IC(50) values 5-8 microM) than the corresponding unmethylated analogs chrysin and apigenin. Flow cytometry indicated that both methylated flavones arrested the SCC-9 cells in the G1 phase with a concomitant decrease in the S phase, dramatically different from the unmethylated analogs, which promoted G2/M phase arrest. Both methylated compounds inhibited the proliferation of two other cancer cell lines with very little effect on two immortalized normal cell lines. Examination of additional flavone structures indicated that methylated flavones in general have antiproliferative properties. Finally, we demonstrated that 5,7-dimethoxyflavone, in contrast to its unmethylated analog chrysin, was well absorbed and had high oral bioavailability as well as tissue accumulation in vivo in the rat. Thus, fully methylated flavones appear to have great potential as cancer chemopreventive/chemotherapeutic agents, in particular in oral cancer.


Asunto(s)
Proliferación Celular/efectos de los fármacos , Flavonas/farmacología , Flavonas/farmacocinética , Metilación , Animales , Apigenina/farmacocinética , Apigenina/farmacología , Disponibilidad Biológica , Ciclo Celular/efectos de los fármacos , Ciclo Celular/fisiología , Ensayos de Selección de Medicamentos Antitumorales , Flavonoides/farmacocinética , Flavonoides/farmacología , Masculino , Ratas , Ratas Endogámicas F344 , Pruebas de Toxicidad , Células Tumorales Cultivadas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA