Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
Int J Mol Sci ; 23(10)2022 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-35628321

RESUMEN

Extracellular vesicles (EVs) are membranous particles released by all cell types. Their role as functional carrier of bioactive molecules is boosted by cells that actively secrete them in biological fluids or in the intercellular space (interstitial EVs, iEVs). Here we have optimised a method for the isolation and characterization of zebrafish iEVs from whole melanoma tissues. Zebrafish melanoma iEVs are around 140 nm in diameter, as determined by nanoparticle tracking and transmission electron microscopy (TEM) analysis. Western blot analysis shows enrichment for CD63 and Alix in the iEV fraction, but not in melanoma cell lysates. Super resolution and confocal microscopy reveal that purified zebrafish iEVs are green fluorescent protein positive (GFP+), indicating that they integrate the oncogene GFP-HRASV12G used to induce melanoma in this model within their vesicular membrane or luminal content. Analysis of RNA-Seq data found 118 non-coding (nc)RNAs differentially distributed between zebrafish melanoma and their iEVs, with only 17 of them being selectively enriched in iEVs. Among these, the RNA components of RNAses P and MRP, which process ribosomal RNA precursors, mitochondrial RNAs, and some mRNAs, were enriched in zebrafish and human melanoma EVs, but not in iEVs extracted from brain tumours. We found that melanoma iEVs induce an inflammatory response when injected in larvae, with increased expression of interferon responsive genes, and this effect is reproduced by MRP- or P-RNAs injected into circulation. This suggests that zebrafish melanoma iEVs are a source of MRP- and P-RNAs that can trigger inflammation in cells of the innate immune system.


Asunto(s)
Vesículas Extracelulares , Melanoma , Animales , Vesículas Extracelulares/metabolismo , Inflamación/genética , Inflamación/metabolismo , Melanoma/genética , Melanoma/metabolismo , ARN no Traducido/metabolismo , Pez Cebra/genética
2.
Dev Biol ; 457(2): 215-225, 2020 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-30998907

RESUMEN

Therapeutic approaches for cutaneous melanoma are flourishing, but despite promising results, there is an increasing number of reported primary or secondary resistance to the growing sets of drugs approved for therapy in the clinics. Combinatorial approaches may overcome resistance, as they may tackle specific weaknesses of melanoma cells, not sufficient on their own, but effective in combination with other therapies. The transgenic zebrafish line kita:ras develops melanoma with high frequency. At 3 dpf, transgenic kita:ras larvae show a hyperpigmentation phenotype as earliest evidence of abnormal melanocyte growth. Using this model, we performed a chemical screen based on automated detection of a reduction of melanocyte number caused by any of 1280 FDA or EMA approved drugs of the library. The analysis showed that 55 molecules were able to reduce by 60% or more the number of melanocytes per embryo. We further tested two compounds for each of the 5 classes, and a farnesyltransferase inhibitor (Lonafarnib), that inhibits an essential post-translational modification of HRAS and suppresses the hyperpigmentation phenotype. Combinations of Clotrimazole and Lonafarnib showed the most promising results in zebrafish embryos, allowing a dose reduction of both drugs. We performed validation of these observations in the metastatic human melanoma cell line A375M, and in normal human epithelial melanocytes (NHEM) in order to investigate the mechanism of action of Clotrimazole in blocking the proliferation of transformed melanocytes. Viability assay and analysis of energy metabolism in Clotrimazole treated cells show that this drug specifically affects melanoma cells in vitro and transformed melanocytes in vivo, having no effects on NHEM or wild type larvae. Similar effects were observed with another hit of the same class, Miconazole. Furthermore, we show that the effects of Clotrimazole are mediated by the inhibition of hexokinase activity, which is lethal to the abnormal metabolic profile of melanoma cells in vitro and in vivo. Thus, our study shows that the zebrafish can provide a phenotype-rich assay for fully automated screening approaches to identify drugs for synthetic lethal treatment in melanoma and suggest further testing of Clotrimazole in combinatorial treatments.


Asunto(s)
Antineoplásicos/farmacología , Clotrimazol/farmacología , Melanoma/tratamiento farmacológico , Piperidinas/farmacología , Piridinas/farmacología , Neoplasias Cutáneas/tratamiento farmacológico , Animales , Animales Modificados Genéticamente , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Ensayos de Selección de Medicamentos Antitumorales/métodos , Farnesiltransferasa/antagonistas & inhibidores , Humanos , Melanocitos/metabolismo , Melanoma/metabolismo , Miconazol/farmacología , Neoplasias Cutáneas/metabolismo , Pez Cebra , Melanoma Cutáneo Maligno
3.
Nature ; 488(7410): 231-5, 2012 Aug 09.
Artículo en Inglés | MEDLINE | ID: mdl-22722852

RESUMEN

Non-coding RNAs (ncRNAs) are involved in an increasingly recognized number of cellular events. Some ncRNAs are processed by DICER and DROSHA RNases to give rise to small double-stranded RNAs involved in RNA interference (RNAi). The DNA-damage response (DDR) is a signalling pathway that originates from a DNA lesion and arrests cell proliferation3. So far, DICER and DROSHA RNA products have not been reported to control DDR activation. Here we show, in human, mouse and zebrafish, that DICER and DROSHA, but not downstream elements of the RNAi pathway, are necessary to activate the DDR upon exogenous DNA damage and oncogene-induced genotoxic stress, as studied by DDR foci formation and by checkpoint assays. DDR foci are sensitive to RNase A treatment, and DICER- and DROSHA-dependent RNA products are required to restore DDR foci in RNase-A-treated cells. Through RNA deep sequencing and the study of DDR activation at a single inducible DNA double-strand break, we demonstrate that DDR foci formation requires site-specific DICER- and DROSHA-dependent small RNAs, named DDRNAs, which act in a MRE11­RAD50­NBS1-complex-dependent manner (MRE11 also known as MRE11A; NBS1 also known as NBN). DDRNAs, either chemically synthesized or in vitro generated by DICER cleavage, are sufficient to restore the DDR in RNase-A-treated cells, also in the absence of other cellular RNAs. Our results describe an unanticipated direct role of a novel class of ncRNAs in the control of DDR activation at sites of DNA damage.


Asunto(s)
Daño del ADN/genética , ARN no Traducido/genética , Ribonucleasa III/genética , Pez Cebra/genética , Animales , Proteínas de Ciclo Celular/metabolismo , Línea Celular , Roturas del ADN de Doble Cadena , Enzimas Reparadoras del ADN/metabolismo , Proteínas de Unión al ADN/metabolismo , Activación Enzimática , Células HEK293 , Células HeLa , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Ratones , Proteínas Nucleares/metabolismo , Interferencia de ARN , ARN no Traducido/biosíntesis , Ribonucleasa Pancreática/metabolismo , Análisis de Secuencia de ARN , Especificidad por Sustrato/genética
4.
J Exp Med ; 219(6)2022 06 06.
Artículo en Inglés | MEDLINE | ID: mdl-35510953

RESUMEN

Genetic alterations in RET lead to activation of ERK and AKT signaling and are associated with hereditary and sporadic thyroid cancer and lung cancer. Highly selective RET inhibitors have recently entered clinical use after demonstrating efficacy in treating patients with diverse tumor types harboring RET gene rearrangements or activating mutations. In order to understand resistance mechanisms arising after treatment with RET inhibitors, we performed a comprehensive molecular and genomic analysis of a patient with RET-rearranged thyroid cancer. Using a combination of drug screening and proteomic and biochemical profiling, we identified an adaptive resistance to RET inhibitors that reactivates ERK signaling within hours of drug exposure. We found that activation of FGFR signaling is a mechanism of adaptive resistance to RET inhibitors that activates ERK signaling. Combined inhibition of FGFR and RET prevented the development of adaptive resistance to RET inhibitors, reduced cell viability, and decreased tumor growth in cellular and animal models of CCDC6-RET-rearranged thyroid cancer.


Asunto(s)
Neoplasias Pulmonares , Neoplasias de la Tiroides , Animales , Proteínas del Citoesqueleto/genética , Humanos , Neoplasias Pulmonares/patología , Proteómica , Proteínas Proto-Oncogénicas c-ret/genética , Receptores de Factores de Crecimiento de Fibroblastos , Neoplasias de la Tiroides/tratamiento farmacológico , Neoplasias de la Tiroides/genética
5.
FASEB J ; 24(8): 2727-38, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20335228

RESUMEN

Sphingosine-1-phosphate (S1P) is an important bioactive sphingolipid involved in angiogenesis and lymphangiogenesis, 2 important processes that influence the growth, survival, and spread of tumors. S1P acts as an extracellular mediator through binding to 5 highly specific S1P receptors, S1P(1-5). Sphingosine kinase-1 (SK1), one of 2 known sphingosine kinase enzymes responsible for S1P production, appears to be overexpressed in many tumors. Although a role for S1P in angiogenesis and lymphangiogenesis has been established, it is unclear whether S1P secreted from cancer cells has a paracrine function in a tumor environment. Here we investigated whether modulation of cellular SK1 could initiate a paracrine angiogenic and lymphangiogenic switch. We found that SK1 overexpression in HEK cells or its down-regulation in glioma or breast cancer cells modulated extracellular S1P levels accordingly, which in turn increased or decreased both migration and tube formation in cocultured vascular or lymphatic endothelial cells. In contrast, down-regulation of sphingosine kinase 2 in both glioma and breast cancer cells had no appreciable effect on cellular or secreted S1P levels. In addition, vascular endothelial growth factors VEGF and VEGF-C down-regulation in cancer cells appeared insufficient to block the angiogenic and lymphangiogenic switch triggered by these cells. Moreover, S1P initiated endothelial cell sprouting in 3-dimensional collagen matrices, which is representative of a multistep angiogenic process. Our data collectively demonstrate for the first time that SK1 plays an essential role in regulating in vitro paracrine angiogenesis and lymphangiogenesis.


Asunto(s)
Linfangiogénesis , Neovascularización Patológica , Neovascularización Fisiológica , Comunicación Paracrina/fisiología , Fosfotransferasas (Aceptor de Grupo Alcohol)/fisiología , Neoplasias de la Mama/irrigación sanguínea , Neoplasias de la Mama/patología , Línea Celular , Línea Celular Tumoral , Técnicas de Cocultivo , Células Endoteliales/citología , Femenino , Regulación de la Expresión Génica/fisiología , Glioma/irrigación sanguínea , Glioma/patología , Humanos , Fosfotransferasas (Aceptor de Grupo Alcohol)/genética
6.
Elife ; 102021 08 31.
Artículo en Inglés | MEDLINE | ID: mdl-34463618

RESUMEN

Although virtually all gene networks are predicted to be controlled by miRNAs, the contribution of this important layer of gene regulation to tissue homeostasis in adult animals remains unclear. Gain and loss-of-function experiments have provided key insights into the specific function of individual miRNAs, but effective genetic tools to study the functional consequences of global inhibition of miRNA activity in vivo are lacking. Here we report the generation and characterization of a genetically engineered mouse strain in which miRNA-mediated gene repression can be reversibly inhibited without affecting miRNA biogenesis or abundance. We demonstrate the usefulness of this strategy by investigating the consequences of acute inhibition of miRNA function in adult animals. We find that different tissues and organs respond differently to global loss of miRNA function. While miRNA-mediated gene repression is essential for the homeostasis of the heart and the skeletal muscle, it is largely dispensable in the majority of other organs. Even in tissues where it is not required for homeostasis, such as the intestine and hematopoietic system, miRNA activity can become essential during regeneration following acute injury. These data support a model where many metazoan tissues primarily rely on miRNA function to respond to potentially pathogenic events.


Asunto(s)
Redes Reguladoras de Genes , MicroARNs/genética , Complejo Silenciador Inducido por ARN/genética , Animales , Femenino , Homeostasis , Ratones , Ratones Transgénicos , Péptidos/metabolismo , Embarazo , Regeneración/genética , Transgenes
7.
Prostaglandins Other Lipid Mediat ; 85(3-4): 107-14, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18166496

RESUMEN

Sphingosine kinase 1 (SK1) and its product sphingosine-1-phosphate (S1P) have been implicated in the regulation of many cellular processes including growth regulation, protection from apoptosis, stimulation of angiogenesis, and most recently as mediators of the TNF-alpha inflammatory response. In this study we set out to examine the role of SK1/S1P in the RAW macrophage response to the potent inflammatory stimulus lipopolysaccharide (LPS). We show that LPS increases cellular levels of SK1 message and protein. This increase is at the transcriptional level and is accompanied by increased SK activity and generation of S1P. S1P is able to cause increases in COX-2 and PGE2 levels in RAW cells. Knockdown of SK1 using siRNA is able to inhibit the TNF but not the LPS inflammatory response. Moreover, knockdown of SK1 enhances both TNF- and LPS-induced apoptosis. These data indicate that there is a dual and distinct role for SK1 and S1P in the TNF and the LPS inflammatory pathways.


Asunto(s)
Inflamación/fisiopatología , Lisofosfolípidos/fisiología , Fosfotransferasas (Aceptor de Grupo Alcohol)/fisiología , Esfingosina/análogos & derivados , Animales , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Lipopolisacáridos , Macrófagos , Ratones , Esfingosina/fisiología , Factor de Necrosis Tumoral alfa/farmacología , Regulación hacia Arriba
8.
Front Genet ; 9: 675, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30619488

RESUMEN

Ras genes are among the most commonly mutated genes in human cancer; yet our understanding of their oncogenic activity at the molecular mechanistic level is incomplete. To identify downstream events that mediate ras-induced cellular transformation in vivo, we analyzed global microRNA expression in three different models of Ras-induction and tumor formation in zebrafish. Six microRNAs were found increased in Ras-induced melanoma, glioma and in an inducible model of ubiquitous Ras expression. The upregulation of the microRNAs depended on the activation of the ERK and AKT pathways and to a lesser extent, on mTOR signaling. Two Ras-induced microRNAs (miR-146a and 193a) target Jmjd6, inducing downregulation of its mRNA and protein levels at the onset of Ras expression during melanoma development. However, at later stages of melanoma progression, jmjd6 levels were found elevated. The dynamic of Jmjd6 levels during progression of melanoma in the zebrafish model suggests that upregulation of the microRNAs targeting Jmjd6 may be part of an anti-cancer response. Indeed, triple transgenic fish engineered to express a microRNA-resistant Jmjd6 from the onset of melanoma have increased tumor burden, higher infiltration of leukocytes and shorter melanoma-free survival. Increased JMJD6 expression is found in several human cancers, including melanoma, suggesting that the up-regulation of Jmjd6 is a critical event in tumor progression. The following link has been created to allow review of record GSE37015: http://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?token=jjcrbiuicyyqgpc&acc=GSE37015.

9.
Biochim Biophys Acta Mol Cell Biol Lipids ; 1863(10): 1179-1192, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30056170

RESUMEN

Glioblastoma is one of the most malignant, angiogenic, and incurable tumors in humans. The aberrant communication between glioblastoma cells and tumor microenvironment represents one of the major factors regulating glioblastoma malignancy and angiogenic properties. Emerging evidence implicates sphingosine-1-phosphate signaling in the pathobiology of glioblastoma and angiogenesis, but its role in glioblastoma-endothelial crosstalk remains largely unknown. In this study, we sought to determine whether the crosstalk between glioblastoma cells and brain endothelial cells regulates sphingosine-1-phosphate signaling in the tumor microenvironment. Using human glioblastoma and brain endothelial cell lines, as well as primary brain endothelial cells derived from human glioblastoma, we report that glioblastoma-co-culture promotes the expression, activity, and plasma membrane enrichment of sphingosine kinase 2 in brain endothelial cells, leading to increased cellular level of sphingosine-1-phosphate, and significant potentiation of its secretion. In turn, extracellular sphingosine-1-phosphate stimulates glioblastoma cell proliferation, and brain endothelial cells migration and angiogenesis. We also show that, after co-culture, glioblastoma cells exhibit enhanced expression of S1P1 and S1P3, the sphingosine-1-phosphate receptors that are of paramount importance for cell growth and invasivity. Collectively, our results envision glioblastoma-endothelial crosstalk as a multi-compartmental strategy to enforce pro-tumoral sphingosine-1-phosphate signaling in the glioblastoma microenvironment.


Asunto(s)
Encéfalo/patología , Células Endoteliales/metabolismo , Glioblastoma/patología , Lisofosfolípidos/metabolismo , Neovascularización Patológica/metabolismo , Transducción de Señal , Esfingosina/análogos & derivados , Microambiente Tumoral , Animales , Línea Celular Tumoral , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Técnicas de Cocultivo , Células Endoteliales/efectos de los fármacos , Células Endoteliales/patología , Glioblastoma/metabolismo , Humanos , Neovascularización Patológica/patología , Fosfotransferasas (Aceptor de Grupo Alcohol)/antagonistas & inhibidores , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Ratas , Receptores de Lisoesfingolípidos/metabolismo , Transducción de Señal/efectos de los fármacos , Esfingosina/metabolismo , Microambiente Tumoral/efectos de los fármacos
10.
Methods Enzymol ; 434: 233-41, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17954250

RESUMEN

In recent years, sphingolipid metabolites ceramide, sphingosine, and sphingosine-1-phosphate have emerged as important second messengers in addition to their role as precursors of biomembrane components. The investigation of these sphingolipid metabolites requires the development of new, more sensitive methods for assaying the enzymes involved in their production. This chapter describes the utilization of mass spectrometry technology in combination with nonnaturally occurring C(17) sphingoid bases in the in vitro assays of two of the enzymes of the sphingolipid pathway, ceramide synthase and sphingosine kinase. These new in vitro methods provide high sensitivity and extreme accuracy even when crude extracts are used as enzyme sources.


Asunto(s)
Oxidorreductasas/metabolismo , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Esfingolípidos/aislamiento & purificación , Esfingolípidos/metabolismo , Animales , Cromatografía Líquida de Alta Presión/métodos , Ácidos Grasos/aislamiento & purificación , Ácidos Grasos/metabolismo , Cinética , Espectrometría de Masas/métodos , Oxidorreductasas/aislamiento & purificación , Fosfotransferasas (Aceptor de Grupo Alcohol)/aislamiento & purificación
11.
Genes (Basel) ; 8(9)2017 Sep 20.
Artículo en Inglés | MEDLINE | ID: mdl-28930163

RESUMEN

Over the past 15 years, zebrafish have emerged as a powerful tool for studying human cancers. Transgenic techniques have been employed to model different types of tumors, including leukemia, melanoma, glioblastoma and endocrine tumors. These models present histopathological and molecular conservation with their human cancer counterparts and have been fundamental for understanding mechanisms of tumor initiation and progression. Moreover, xenotransplantation of human cancer cells in embryos or adult zebrafish offers the advantage of studying the behavior of human cancer cells in a live organism. Chemical-genetic screens using zebrafish embryos have uncovered novel druggable pathways and new therapeutic strategies, some of which are now tested in clinical trials. In this review, we will report on recent advances in using zebrafish as a model in cancer studies-with specific focus on four cancer types-where zebrafish has contributed to novel discoveries or approaches to novel therapies.

12.
Elife ; 62017 03 28.
Artículo en Inglés | MEDLINE | ID: mdl-28350298

RESUMEN

Thyroid cancer is common, yet the sequence of alterations that promote tumor formation are incompletely understood. Here, we describe a novel model of thyroid carcinoma in zebrafish that reveals temporal changes due to BRAFV600E. Through the use of real-time in vivo imaging, we observe disruption in thyroid follicle structure that occurs early in thyroid development. Combinatorial treatment using BRAF and MEK inhibitors reversed the developmental effects induced by BRAFV600E. Adult zebrafish expressing BRAFV600E in thyrocytes developed invasive carcinoma. We identified a gene expression signature from zebrafish thyroid cancer that is predictive of disease-free survival in patients with papillary thyroid cancer. Gene expression studies nominated TWIST2 as a key effector downstream of BRAF. Using CRISPR/Cas9 to genetically inactivate a TWIST2 orthologue, we suppressed the effects of BRAFV600E and restored thyroid morphology and hormone synthesis. These data suggest that expression of TWIST2 plays a role in an early step of BRAFV600E-mediated transformation.


Asunto(s)
Morfogénesis , Proteínas Proto-Oncogénicas B-raf/metabolismo , Glándula Tiroides/embriología , Glándula Tiroides/fisiología , Hormonas Tiroideas/metabolismo , Neoplasias de la Tiroides/patología , Proteína Relacionada con Twist 2/biosíntesis , Animales , Modelos Animales de Enfermedad , Eliminación de Gen , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Mutación Missense , Proteínas Proto-Oncogénicas B-raf/genética , Proteína Relacionada con Twist 2/genética , Pez Cebra
14.
Mol Endocrinol ; 25(12): 2144-56, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22016563

RESUMEN

We recently reported that IGF-II binding to the IGF-II/mannose-6-phosphate (M6P) receptor activates the ERK1/2 cascade by triggering sphingosine kinase 1 (SK1)-dependent transactivation of G protein-coupled sphingosine 1-phosphate (S1P) receptors. Here, we investigated the mechanism of IGF-II/M6P receptor-dependent sphingosine kinase 1 (SK1) activation in human embryonic kidney 293 cells. Pretreating cells with protein kinase C (PKC) inhibitor, bisindolylmaleimide-I, abolished IGF-II-stimulated translocation of green fluorescent protein (GFP)-tagged SK1 to the plasma membrane and activation of endogenous SK1, implicating PKC as an upstream regulator of SK1. Using confocal microscopy to examine membrane translocation of GFP-tagged PKCα, ß1, ß2, δ, and ζ, we found that IGF-II induced rapid, transient, and isoform-specific translocation of GFP-PKCß2 to the plasma membrane. Immunoblotting of endogenous PKC phosphorylation confirmed PKCß2 activation in response to IGF-II. Similarly, IGF-II stimulation caused persistent membrane translocation of the kinase-deficient GFP-PKCß2 (K371R) mutant, which does not dissociate from the membrane after translocation. IGF-II stimulation increased diacylglycerol (DAG) levels, the established activator of classical PKC. Interestingly, the polyunsaturated fraction of DAG was increased, indicating involvement of phosphatidyl inositol/phospholipase C (PLC). Pretreating cells with the PLC inhibitor, U73122, attenuated IGF-II-dependent DAG production and PKCß2 phosphorylation, blocked membrane translocation of the kinase-deficient GFP-PKCß2 (K371R) mutant, and reduced sphingosine 1-phosphate production, suggesting that PLC/PKCß2 are upstream regulators of SK1 in the pathway. Taken together, these data provide evidence that activation of PLC and PKCß2 by the IGF-II/M6P receptor are required for the activation of SK1.


Asunto(s)
Activación Enzimática , Factor II del Crecimiento Similar a la Insulina/fisiología , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Proteína Quinasa C/antagonistas & inhibidores , Fosfolipasas de Tipo C/antagonistas & inhibidores , Proliferación Celular , Diglicéridos/metabolismo , Activadores de Enzimas/farmacología , Técnicas de Silenciamiento del Gen , Proteínas Fluorescentes Verdes/metabolismo , Células HEK293 , Humanos , Indoles/farmacología , Factor II del Crecimiento Similar a la Insulina/farmacología , Lisofosfolípidos/metabolismo , Maleimidas/farmacología , Células Mesangiales/efectos de los fármacos , Células Mesangiales/fisiología , Fosfotransferasas (Aceptor de Grupo Alcohol)/antagonistas & inhibidores , Proteína Quinasa C/metabolismo , Proteína Quinasa C beta , Transporte de Proteínas , Interferencia de ARN , Receptor IGF Tipo 1/metabolismo , Receptor IGF Tipo 2/genética , Receptor IGF Tipo 2/metabolismo , Proteínas Recombinantes de Fusión/metabolismo , Transducción de Señal , Esfingosina/análogos & derivados , Esfingosina/metabolismo , Esfingosina/farmacología , Acetato de Tetradecanoilforbol/farmacología , Fosfolipasas de Tipo C/metabolismo
15.
PLoS One ; 5(12): e15170, 2010 Dec 10.
Artículo en Inglés | MEDLINE | ID: mdl-21170325

RESUMEN

BACKGROUND: Melanoma is the most aggressive and lethal form of skin cancer. Because of the increasing incidence and high lethality of melanoma, animal models for continuously observing melanoma formation and progression as well as for testing pharmacological agents are needed. METHODOLOGY AND PRINCIPAL FINDINGS: Using the combinatorial Gal4-UAS system, we have developed a zebrafish transgenic line that expresses oncogenic HRAS under the kita promoter. Already at 3 days transgenic kita-GFP-RAS larvae show a hyper-pigmentation phenotype as earliest evidence of abnormal melanocyte growth. By 2-4 weeks, masses of transformed melanocytes form in the tail stalk of the majority of kita-GFP-RAS transgenic fish. The adult tumors evident between 1-3 months of age faithfully reproduce the immunological, histological and molecular phenotypes of human melanoma, but on a condensed time-line. Furthermore, they show transplantability, dependence on mitfa expression and do not require additional mutations in tumor suppressors. In contrast to kita expressing melanocyte progenitors that efficiently develop melanoma, mitfa expressing progenitors in a second Gal4-driver line were 4 times less efficient in developing melanoma during the three months observation period. CONCLUSIONS AND SIGNIFICANCE: This indicates that zebrafish kita promoter is a powerful tool for driving oncogene expression in the right cells and at the right level to induce early onset melanoma in the presence of tumor suppressors. Thus our zebrafish model provides a link between kita expressing melanocyte progenitors and melanoma and offers the advantage of a larval phenotype suitable for large scale drug and genetic modifier screens.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Melanoma/metabolismo , Proteínas Proto-Oncogénicas c-kit/metabolismo , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Neoplasias Cutáneas/metabolismo , Animales , Animales Modificados Genéticamente , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Genes p53 , Melanocitos/citología , Melanocitos/metabolismo , Melanoma/genética , Modelos Genéticos , Fosfohidrolasa PTEN/metabolismo , Fenotipo , Regiones Promotoras Genéticas , Neoplasias Cutáneas/genética , Transgenes , Pez Cebra
16.
Zebrafish ; 6(4): 417-24, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20047469

RESUMEN

We have established a model of melanoma progression in zebrafish through the generation of transgenic lines specifically expressing oncogenic human HRAS in the melanocytic lineage. In these tumors we have carried out quantitative expression analysis of several putative cancer genes, from known and predicted cancer gene lists. In particular, we analyzed 39 out of 101 putative cancer genes identified with a bioinformatics approach and selected for the low frequency of duplication and the high connectivity in protein networks. Data obtained by real-time polymerase chain reaction analysis from zebrafish melanoma tissue shows that the expression of many cancer genes is downregulated in zebrafish melanomas, whereas only cell cycle genes are upregulated. To understand whether this trend is due to global repression of gene expression associated to a repressive chromatin state, we investigated whether changes of histone methylation were detectable in our melanoma model. We found substantial differences in the levels of H3K9me3, H4K20me2, H3K27me3, H3K4me3, and H3R2me2a immunostaining in melanoma tissue when compared with normal skin. Thus our analysis suggests that in our model, like in human melanoma, important changes occur to the methylation status of histones. Although the outcome of these changes is still unknown, they could be responsible for the global repression of gene expression through epigenetic regulation shown in this study.


Asunto(s)
Modelos Animales de Enfermedad , Regulación hacia Abajo , Epigénesis Genética , Regulación Neoplásica de la Expresión Génica , Melanoma/genética , Neoplasias Cutáneas/genética , Pez Cebra/genética , Animales , Animales Modificados Genéticamente , Biomarcadores de Tumor/genética , Histonas/metabolismo , Humanos , Melanoma/metabolismo , Neoplasias Cutáneas/metabolismo , Pez Cebra/metabolismo
17.
J Biol Chem ; 284(19): 12979-88, 2009 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-19279008

RESUMEN

Activation of protein kinase C (PKC) by the phorbol ester (phorbol 12-myristate 13-acetate) induces ceramide formation through the salvage pathway involving, in part, acid beta-glucosidase 1 (GBA1), which cleaves glucosylceramide to ceramide. Here, we examine the role of the GBA1-ceramide pathway, in regulating a pro-inflammatory pathway initiated by PKC and leading to activation of p38 and induction of interleukin 6 (IL-6). Inhibition of ceramide formation by fumonisin B1 or down-regulation of PKCdelta potentiated PMA-induced activation of p38 in human breast cancer MCF-7 cells. Similarly, knockdown of GBA1 by small interfering RNAs or pharmacological inhibition of GBA1 promoted further activation of p38 after PMA treatment, implicating the GBA1-ceramide pathway in the termination of p38 activation. Knockdown of GBA1 also evoked the hyperproduction of IL-6 in response to 4beta phorbol 12-myristate 13-acetate. On the other hand, increasing cellular ceramide with cell-permeable ceramide treatment resulted in attenuation of the IL-6 response. Importantly, silencing the delta isoform of the p38 family significantly attenuated the hyperproduction of IL-6. Reciprocally, p38delta overexpression induced IL-6 biosynthesis. Thus, the GBA1-ceramide pathway is suggested to play an important role in terminating p38delta activation responsible for IL-6 biosynthesis. Furthermore, the p38delta isoform was identified as a novel and predominant target of ceramide signaling as well as a regulator of IL-6 biosynthesis.


Asunto(s)
Neoplasias de la Mama/metabolismo , Ceramidas/metabolismo , Glucosilceramidasa/fisiología , Interleucina-6/metabolismo , Proteína Quinasa 13 Activada por Mitógenos/metabolismo , Western Blotting , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Carcinógenos/farmacología , Línea Celular Tumoral , Ensayo de Inmunoadsorción Enzimática , Femenino , Silenciador del Gen/fisiología , Humanos , Inositol/análogos & derivados , Inositol/metabolismo , Fosforilación , Proteína Quinasa C/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Interferente Pequeño/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Acetato de Tetradecanoilforbol/farmacología , Factor de Necrosis Tumoral alfa/farmacología
18.
J Biol Chem ; 284(19): 12972-8, 2009 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-19279011

RESUMEN

Activation of protein kinase C (PKC) promotes the salvage pathway of ceramide formation, and acid sphingomyelinase has been implicated, in part, in providing substrate for this pathway (Zeidan, Y. H., and Hannun, Y. A. (2007) J. Biol. Chem. 282, 11549-11561). In the present study, we examined whether acid beta-glucosidase 1 (GBA1), which hydrolyzes glucosylceramide to form lysosomal ceramide, was involved in PKC-regulated formation of ceramide from recycled sphingosine. Glucosylceramide levels declined after treatment of MCF-7 cells with a potent PKC activator, phorbol 12-myristate 13-acetate (PMA). Silencing GBA1 by small interfering RNAs significantly attenuated acid glucocerebrosidase activity and decreased PMA-induced formation of ceramide by 50%. Silencing GBA1 blocked PMA-induced degradation of glucosylceramide and generation of sphingosine, the source for ceramide biosynthesis. Reciprocally, forced expression of GBA1 increased ceramide levels. These observations indicate that GBA1 activation can generate the source (sphingosine) for PMA-induced formation of ceramide through the salvage pathway. Next, the role of PKCdelta, a direct effector of PMA, in the formation of ceramide was determined. By attenuating expression of PKCdelta, cells failed to trigger PMA-induced alterations in levels of ceramide, sphingomyelin, and glucosylceramide. Thus, PKCdelta activation is suggested to stimulate the degradation of both sphingomyelin and glucosylceramide leading to the salvage pathway of ceramide formation. Collectively, GBA1 is identified as a novel source of regulated formation of ceramide, and PKCdelta is an upstream regulator of this pathway.


Asunto(s)
Neoplasias de la Mama/metabolismo , Ceramidas/biosíntesis , Glucosilceramidasa/fisiología , Glucosilceramidas/metabolismo , Western Blotting , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Carcinógenos/farmacología , Línea Celular Tumoral , Cromatografía Líquida de Alta Presión , Femenino , Silenciador del Gen/fisiología , Humanos , Lisosomas/metabolismo , Proteína Quinasa C/metabolismo , ARN Interferente Pequeño/genética , Esfingomielinas/metabolismo , Esfingosina/metabolismo , Espectrometría de Masas en Tándem , Acetato de Tetradecanoilforbol/farmacología
19.
J Biol Chem ; 283(6): 3365-3375, 2008 Feb 08.
Artículo en Inglés | MEDLINE | ID: mdl-18055454

RESUMEN

Sphingosine 1-phosphate (S1P), a sphingolipid metabolite that plays an important role in the regulation of cell survival, growth, migration, and angiogenesis, acts both inside the cells and as an extracellular mediator through binding to five G protein-coupled receptors (S1P(1-5)). Sphingosine kinase 1 (SK1), the enzyme responsible for S1P production, is overexpressed in many solid tumors, including gliomas. One common feature of these tumors is the presence of "hypoxic regions," characterized by cells expressing high levels of hypoxia-inducible factors HIF-1alpha and HIF-2alpha, two transcription regulators that modulate the levels of proteins with crucial roles in tumor progression. So far, nothing is known about the role and the regulation of SK1 during tumor-induced hypoxia or about SK1 regulation and HIFs. Here we investigated the role of HIF-1alpha and HIF-2alpha in the regulation of SK1 during hypoxic stress in glioma-derived U87MG cells. We report that hypoxia increases SK1 mRNA levels, protein expression, and enzyme activity, followed by intracellular S1P production and S1P release. Interestingly, knockdown of HIF-2alpha by small interfering RNA abolished the induction of SK1 and the production of extracellular S1P after CoCl(2) treatment, whereas HIF-1alpha small interfering RNA resulted in an increase of HIF-2alpha and of SK1 protein levels. Moreover, using chromatin immunoprecipitation analysis, we demonstrate that HIF-2alpha binds the SK1 promoter. Functionally, we demonstrate that conditioned medium from hypoxia-treated tumor cells results in neoangiogenesis in human umbilical vein endothelial cells in a S1P receptor-dependent manner. These studies provide evidence of a link between S1P production as a potent angiogenic agent and the hypoxic phenotype observed in many tumors.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/biosíntesis , Neoplasias Encefálicas/metabolismo , Regulación Enzimológica de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Glioma/metabolismo , Factor 1 Inducible por Hipoxia/biosíntesis , Fosfotransferasas (Aceptor de Grupo Alcohol)/biosíntesis , Regiones Promotoras Genéticas , Progresión de la Enfermedad , Humanos , ARN Mensajero/metabolismo , ARN Interferente Pequeño/metabolismo , Transcripción Genética , Regulación hacia Arriba
20.
J Neurooncol ; 87(1): 23-33, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17975708

RESUMEN

HMGB1 (high mobility group box 1 protein) is a nuclear protein that can also act as an extracellular trigger of inflammation, proliferation and migration, mainly through RAGE (the receptor for advanced glycation end products); HMGB1-RAGE interactions have been found to be important in a number of cancers. We investigated whether HMGB1 is an autocrine factor in human glioma cells. Western blots showed HMGB1 and RAGE expression in human malignant glioma cell lines. HMGB1 induced a dose-dependent increase in cell proliferation, which was found to be RAGE-mediated and involved the MAPK/ERK pathway. Moreover, in a wounding model, it induced a significant increase in cell migration, and RAGE-dependent activation of Rac1 was crucial in giving the tumour cells a motile phenotype. The fact that blocking DNA replication with anti-mitotic agents did not reduce the distance migrated suggests the independence of the proliferative and migratory effects. We also found that glioma cells contain HMGB1 predominantly in the nucleus, and cannot secrete it constitutively or upon stimulation; however, necrotic glioma cells can release HMGB1 after it has translocated from the nucleus to cytosol. These findings provide the first evidence supporting the existence of HMGB1/RAGE signalling pathways in human glioblastoma cells, and suggest that HMGB1 may play an important role in the relationship between necrosis and malignancy in glioma tumours by acting as an autocrine factor that is capable of promoting the growth and migration of tumour cells.


Asunto(s)
Neoplasias Encefálicas/metabolismo , Movimiento Celular/fisiología , Glioblastoma/metabolismo , Proteína HMGB1/metabolismo , Transducción de Señal/fisiología , Western Blotting , Línea Celular Tumoral , Proliferación Celular , Productos Finales de Glicación Avanzada , Humanos , Proteínas Quinasas Activadas por Mitógenos/biosíntesis , Necrosis/fisiopatología , Receptor para Productos Finales de Glicación Avanzada , Receptores Inmunológicos/biosíntesis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA