Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Más filtros

Banco de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Lab Invest ; 97(1): 24-33, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27869796

RESUMEN

With more patients dying from metastasis than from primary cancers, metastasis is a very important area in cancer research. Investigators thereby heavily rely on animal models of metastasis to common organs such as the lung to improve our insight into the pathogenesis and to research novel therapeutic approaches to combat metastasis. In this experimental context, novel tools that allow longitudinal monitoring of lung metastasis in individual animals are highly needed. We have therefore evaluated for the first time microcomputed tomography (µCT) as a very efficient and crossvalidated means to noninvasively and repeatedly monitor metastasis to the lung in individual, free-breathing syngeneic mice. Two individual clones of KLN205 cancer cells were intravenously injected in syngeneic DBA/2 mice and lung metastasis was monitored weekly during 3 weeks using µCT, and was compared with the current gold standard histology and bioluminescence imaging (BLI). µCT enabled us to visualize diffuse tumor morphology and also to extract four different biomarkers that quantify not only tumor load but also aerated space in the lung as a marker of vital lung capacity and potential compensatory mechanisms. Complementary to BLI, applying this novel µCT-based approach enabled us to unravel sensitively and efficiently differences in metastatic potential between two cellular clones. In conclusion, µCT and BLI offer biomarkers that describe different and complementary aspects of lung metastasis, underlining the importance of multimodality follow-up. The added value of µCT findings is important to better assess lung metastasis and host/lung response in preclinical studies, which will be valuable for translational applications.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Mediciones Luminiscentes/métodos , Neoplasias Pulmonares/diagnóstico por imagen , Microtomografía por Rayos X/métodos , Animales , Línea Celular Tumoral , Modelos Animales de Enfermedad , Femenino , Humanos , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Ratones , Ratones Endogámicos DBA , Metástasis de la Neoplasia , Reproducibilidad de los Resultados , Sensibilidad y Especificidad , Factores de Tiempo , Carga Tumoral
2.
J Exp Clin Cancer Res ; 42(1): 92, 2023 Apr 19.
Artículo en Inglés | MEDLINE | ID: mdl-37072838

RESUMEN

BACKGROUND: One of the key limitations of targeted cancer therapies is the rapid onset of therapy resistance. Taking BRAF-mutant melanoma as paradigm, we previously identified the lipogenic regulator SREBP-1 as a central mediator of resistance to MAPK-targeted therapy. Reasoning that lipogenesis-mediated alterations in membrane lipid poly-unsaturation lie at the basis of therapy resistance, we targeted fatty acid synthase (FASN) as key player in this pathway to evoke an exquisite vulnerability to clinical inducers of reactive oxygen species (ROS), thereby rationalizing a novel clinically actionable combination therapy to overcome therapy resistance. METHODS: Using gene expression analysis and mass spectrometry-based lipidomics of BRAF-mutant melanoma cell lines, melanoma PDX and clinical data sets, we explored the association of FASN expression with membrane lipid poly-unsaturation and therapy-resistance. Next, we treated therapy-resistant models with a preclinical FASN inhibitor TVB-3664 and a panel of ROS inducers and performed ROS analysis, lipid peroxidation tests and real-time cell proliferation assays. Finally, we explored the combination of MAPK inhibitors, TVB-3664 and arsenic trioxide (ATO, as a clinically used ROS-inducer) in Mel006 BRAF mutant PDX as a gold model of therapy resistance and assessed the effect on tumor growth, survival and systemic toxicity. RESULTS: We found that FASN expression is consistently increased upon the onset of therapy resistance in clinical melanoma samples, in cell lines and in Mel006 PDX and is associated with decreased lipid poly-unsaturation. Forcing lipid poly-unsaturation in therapy-resistant models by combining MAPK inhibition with FASN inhibition attenuated cell proliferation and rendered cells exquisitely sensitive to a host of ROS inducers. In particular, the triple combination of MAPK inhibition, FASN inhibition, and the clinical ROS-inducing compound ATO dramatically increased survival of Mel006 PDX models from 15 to 72% with no associated signs of toxicity. CONCLUSIONS: We conclude that under MAPK inhibition the direct pharmacological inhibition of FASN evokes an exquisite vulnerability to inducers of ROS by increasing membrane lipid poly-unsaturation. The exploitation of this vulnerability by combining MAPK and/or FASN inhibitors with inducers of ROS greatly delays the onset of therapy resistance and increases survival. Our work identifies a clinically actionable combinatorial treatment for therapy-resistant cancer.


Asunto(s)
Melanoma , Proteínas Proto-Oncogénicas B-raf , Humanos , Especies Reactivas de Oxígeno/metabolismo , Proteínas Proto-Oncogénicas B-raf/genética , Lípidos de la Membrana/farmacología , Lípidos de la Membrana/uso terapéutico , Melanoma/tratamiento farmacológico , Melanoma/genética , Melanoma/patología , Inhibidores de Proteínas Quinasas/farmacología , Línea Celular Tumoral , Resistencia a Antineoplásicos
3.
Eur J Nucl Med Mol Imaging ; 38(10): 1842-53, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21732108

RESUMEN

PURPOSE: The aim of this study was to evaluate the impact of androgen ablation therapy in different prostate cancer (PCa) cell lines--reflecting different stages of the disease--on (18)F-fluorodeoxyglucose (FDG), 11C-choline and 11C-acetate uptake. METHODS: Uptake experiments were performed in androgen-sensitive (LNCaP, PC346C) and independent cell lines (22Rv1, PC346DCC, PC-3) as well as in a benign prostatic hyperplasia (BPH-1) cell line. Tracer uptake was assessed under androgen ablation. Results of the cancer cell lines were normalized to those of BPH-1. To evaluate the effect of androgen on the uptake of 18F-FDG, 11C-choline and 11C-acetate in PCa cell lines, 10(-8) M R1881, 10(-10) M R1881, the combination of 10(-10) M R1881 plus 10(-6) M Casodex or 10(-6) M Casodex alone were added in parallel cell cultures 1 day before uptake experiments. Uptake in androgen-supplemented cell cultures was compared to the uptake under androgen deprivation. Uptake was corrected for cell number using protein content. RESULTS: Compared to BPH-1, a higher 18F-FDG uptake was observed only in PC346C cells, whereas a higher 11C-choline and markedly increased 11C-acetate uptake was seen in all cancer cell lines. Androgens significantly modulated the uptake of 18F-FDG in LNCaP, PC346C and 22Rv1 cells, and of 11C-choline in the PC346C and 22Rv1 cell line. No androgenic effect on 11C-choline and 18F-FDG uptake was observed in PC-3 and PC346DCC cells. 11C-Acetate uptake was independent of androgen status in all PCa cell lines studied. CONCLUSION: 18F-FDG uptake in PCa cell lines showed the highest variability and strongest androgen effect, suggesting its poor potential for metabolic imaging of advanced PCa. In contrast to 18F-FDG and 11C-choline, 11C-acetate uptake was unaffected by androgens and thus 11C-acetate seems best for monitoring PCa progression.


Asunto(s)
Acetatos/metabolismo , Andrógenos/farmacología , Colina/metabolismo , Fluorodesoxiglucosa F18/metabolismo , Neoplasias de la Próstata/patología , Transporte Biológico/efectos de los fármacos , Radioisótopos de Carbono , Línea Celular Tumoral , Humanos , Masculino , Estadificación de Neoplasias
4.
Viruses ; 13(12)2021 11 25.
Artículo en Inglés | MEDLINE | ID: mdl-34960632

RESUMEN

To date, no vaccines or antivirals are available against Zika virus (ZIKV). In addition, the mechanisms underlying ZIKV-associated pathogenesis of the central nervous system (CNS) are largely unexplored. Getting more insight into the cellular pathways that ZIKV recruits to facilitate infection of susceptible cells will be crucial for establishing an effective treatment strategy. In general, cells secrete a number of vesicles, known as extracellular vesicles (EVs), in response to viral infections. These EVs serve as intercellular communicators. Here, we investigated the role of EVs derived from ZIKV-infected human brain microvascular endothelial cells on the blood-brain barrier (BBB) system. We demonstrated that ZIKV-infected EVs (IEVs) can incorporate viral components, including ZIKV RNA, NS1, and E-protein, and further transfer them to several types of CNS cells. Using label-free impedance-based biosensing, we observed that ZIKV and IEVs can temporally disturb the monolayer integrity of BBB-mimicking cells, possibly by inducing structural rearrangements of the adherent protein VE-cadherin (immunofluorescence staining). Finally, differences in the lipidomic profile between EVs and their parental cells possibly suggest a preferential sorting mechanism of specific lipid species into the vesicles. To conclude, these data suggest that IEVs could be postulated as vehicles (Trojan horse) for ZIKV transmission via the BBB.


Asunto(s)
Barrera Hematoencefálica/metabolismo , Vesículas Extracelulares/metabolismo , Infección por el Virus Zika/transmisión , Virus Zika/fisiología , Barrera Hematoencefálica/virología , Células Cultivadas , Sistema Nervioso Central/virología , Células Endoteliales/virología , Vesículas Extracelulares/virología , Humanos , Lipidómica , ARN Viral/metabolismo , Proteínas no Estructurales Virales/metabolismo , Proteínas Virales/genética , Proteínas Virales/metabolismo , Infección por el Virus Zika/virología
5.
Cancer Res ; 81(19): 4981-4993, 2021 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-34362796

RESUMEN

Dysregulated lipid metabolism is a prominent feature of prostate cancer that is driven by androgen receptor (AR) signaling. Here we used quantitative mass spectrometry to define the "lipidome" in prostate tumors with matched benign tissues (n = 21), independent unmatched tissues (n = 47), and primary prostate explants cultured with the clinical AR antagonist enzalutamide (n = 43). Significant differences in lipid composition were detected and spatially visualized in tumors compared with matched benign samples. Notably, tumors featured higher proportions of monounsaturated lipids overall and elongated fatty acid chains in phosphatidylinositol and phosphatidylserine lipids. Significant associations between lipid profile and malignancy were validated in unmatched samples, and phospholipid composition was characteristically altered in patient tissues that responded to AR inhibition. Importantly, targeting tumor-related lipid features via inhibition of acetyl-CoA carboxylase 1 significantly reduced cellular proliferation and induced apoptosis in tissue explants. This characterization of the prostate cancer lipidome in clinical tissues reveals enhanced fatty acid synthesis, elongation, and desaturation as tumor-defining features, with potential for therapeutic targeting. SIGNIFICANCE: This study identifies malignancy and treatment-associated changes in lipid composition of clinical prostate cancer tissues, suggesting that mediators of these lipidomic changes could be targeted using existing metabolic agents.


Asunto(s)
Metabolismo de los Lípidos , Lipidómica , Lípidos de la Membrana/metabolismo , Neoplasias de la Próstata/metabolismo , Biomarcadores , Biología Computacional/métodos , Metabolismo Energético , Humanos , Metabolismo de los Lípidos/efectos de los fármacos , Lipidómica/métodos , Masculino , Metabolómica/métodos , Terapia Molecular Dirigida , Clasificación del Tumor , Estadificación de Neoplasias , Neoplasias de la Próstata/diagnóstico , Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/etiología , Espectrometría de Masa por Ionización de Electrospray , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción , Espectrometría de Masas en Tándem , Regulador Transcripcional ERG/genética , Regulador Transcripcional ERG/metabolismo
6.
Nat Commun ; 9(1): 2500, 2018 06 27.
Artículo en Inglés | MEDLINE | ID: mdl-29950559

RESUMEN

Whereas significant anti-tumor responses are observed in most BRAFV600E-mutant melanoma patients exposed to MAPK-targeting agents, resistance almost invariably develops. Here, we show that in therapy-responsive cells BRAF inhibition induces downregulation of the processing of Sterol Regulator Element Binding (SREBP-1) and thereby lipogenesis. Irrespective of the escape mechanism, therapy-resistant cells invariably restore this process to promote lipid saturation and protect melanoma from ROS-induced damage and lipid peroxidation. Importantly, pharmacological SREBP-1 inhibition sensitizes BRAFV600E-mutant therapy-resistant melanoma to BRAFV600E inhibitors both in vitro and in a pre-clinical PDX in vivo model. Together, these data indicate that targeting SREBP-1-induced lipogenesis may offer a new avenue to overcome acquisition of resistance to BRAF-targeted therapy. This work also provides evidence that targeting vulnerabilities downstream of oncogenic signaling offers new possibilities in overcoming resistance to targeted therapies.


Asunto(s)
Antineoplásicos/farmacología , Resistencia a Antineoplásicos/genética , Lipogénesis/genética , Melanoma/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas B-raf/antagonistas & inhibidores , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/metabolismo , Animales , Antineoplásicos/uso terapéutico , Línea Celular Tumoral , Regulación hacia Abajo , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Técnicas de Inactivación de Genes , Humanos , Lipogénesis/efectos de los fármacos , Masculino , Melanocitos , Melanoma/genética , Ratones , Ratones Desnudos , Ratones SCID , Mutación , Inhibidores de Proteínas Quinasas/uso terapéutico , Proteínas Proto-Oncogénicas B-raf/genética , Piridinas/farmacología , Transducción de Señal/genética , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/antagonistas & inhibidores , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/genética , Tiazoles/farmacología , Vemurafenib , Ensayos Antitumor por Modelo de Xenoinjerto
7.
Cancer Res ; 65(6): 2441-8, 2005 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-15781660

RESUMEN

Aggressive cancer cells typically show a high rate of energy-consuming anabolic processes driving the synthesis of lipids, proteins, and DNA. Here, we took advantage of the ability of the cell-permeable nucleoside 5-aminoimidazole-4-carboxamide (AICA) riboside to increase the intracellular levels of AICA ribotide, an AMP analogue, mimicking a low energy status of the cell. Treatment of cancer cells with AICA riboside impeded lipogenesis, decreased protein translation, and blocked DNA synthesis. Cells treated with AICA riboside stopped proliferating and lost their invasive properties and their ability to form colonies. When administered in vivo, AICA riboside attenuated the growth of MDA-MB-231 tumors in nude mice. These findings point toward a central tie between energy, anabolism, and cancer and suggest that the cellular energy sensing machinery in cancer cells is an exploitable target for cancer prevention and/or therapy.


Asunto(s)
Aminoimidazol Carboxamida/análogos & derivados , Aminoimidazol Carboxamida/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Próstata/tratamiento farmacológico , Ribonucleósidos/farmacología , Adenosina Monofosfato/metabolismo , Adenilato Quinasa/metabolismo , Aminoimidazol Carboxamida/metabolismo , Animales , Materiales Biomiméticos/metabolismo , Materiales Biomiméticos/farmacología , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Procesos de Crecimiento Celular/efectos de los fármacos , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Metabolismo Energético , Femenino , Humanos , Masculino , Ratones , Ratones Desnudos , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , Ribonucleósidos/metabolismo , Ribonucleótidos/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
8.
Mol Cancer Ther ; 5(9): 2211-7, 2006 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16985054

RESUMEN

Because of its ability to mimic a low energy status of the cell, the cell-permeable nucleoside 5-aminoimidazole-4-carboxamide (AICA) riboside was proposed as an antineoplastic agent switching off major energy-consuming processes associated with the malignant phenotype (lipid production, DNA synthesis, cell proliferation, cell migration, etc.). Key to the antineoplastic action of AICA riboside is its conversion to ZMP, an AMP mimetic that at high concentrations activates the AMP-activated protein kinase (AMPK). Here, in an attempt to increase the efficacy of AICA riboside, we pretreated cancer cells with methotrexate, an antimetabolite blocking the metabolism of ZMP. Methotrexate enhanced the AICA riboside-induced accumulation of ZMP and led to a decrease in the levels of ATP, which functions as an intrasteric inhibitor of AMPK. Consequently, methotrexate markedly sensitized AMPK for activation by AICA riboside and potentiated the inhibitory effects of AICA riboside on tumor-associated processes. As cotreatment elicited antiproliferative effects already at concentrations of compounds that were only marginally effective when used alone, our findings on the cooperation between methotrexate and AICA riboside provide new opportunities both for the application of classic antimetabolic chemotherapeutics, such as methotrexate, and for the exploitation of the energy-sensing machinery as a target for cancer intervention.


Asunto(s)
Aminoimidazol Carboxamida/análogos & derivados , Neoplasias de la Mama/tratamiento farmacológico , Carcinoma de Células Escamosas/tratamiento farmacológico , Metotrexato/farmacología , Ribonucleósidos/farmacología , Proteínas Quinasas Activadas por AMP , Adenosina Trifosfato/metabolismo , Aminoimidazol Carboxamida/antagonistas & inhibidores , Aminoimidazol Carboxamida/metabolismo , Aminoimidazol Carboxamida/farmacocinética , Aminoimidazol Carboxamida/farmacología , Neoplasias de la Mama/enzimología , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Carcinoma de Células Escamosas/enzimología , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patología , Línea Celular Tumoral , ADN de Neoplasias/antagonistas & inhibidores , ADN de Neoplasias/biosíntesis , Sinergismo Farmacológico , Activación Enzimática/efectos de los fármacos , Humanos , Lípidos/biosíntesis , Complejos Multienzimáticos/metabolismo , Nucleótido Desaminasas/antagonistas & inhibidores , Nucleótido Desaminasas/genética , Nucleótido Desaminasas/metabolismo , Fosforribosilaminoimidazolcarboxamida-Formiltransferasa/antagonistas & inhibidores , Fosforribosilaminoimidazolcarboxamida-Formiltransferasa/genética , Fosforribosilaminoimidazolcarboxamida-Formiltransferasa/metabolismo , Fosforribosilglicinamida-Formiltransferasa/antagonistas & inhibidores , Fosforribosilglicinamida-Formiltransferasa/genética , Fosforribosilglicinamida-Formiltransferasa/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Purinas/antagonistas & inhibidores , Purinas/biosíntesis , Interferencia de ARN , Ribonucleósidos/farmacocinética , Ribonucleótidos/antagonistas & inhibidores , Ribonucleótidos/metabolismo
9.
Oncotarget ; 7(9): 9975-92, 2016 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-26862738

RESUMEN

The development of cancer is often accompanied by a loss of the primary cilium, a microtubule-based cellular protrusion that functions as a cellular antenna and that puts a break on cell proliferation. Hence, restoration of the primary cilium in cancer cells may represent a novel promising approach to attenuate tumor growth. Using a high content analysis-based approach we screened a library of clinically evaluated compounds and marketed drugs for their ability to restore primary cilium expression in pancreatic ductal cancer cells. A diverse set of 118 compounds stimulating cilium expression was identified. These included glucocorticoids, fibrates and other nuclear receptor modulators, neurotransmitter regulators, ion channel modulators, tyrosine kinase inhibitors, DNA gyrase/topoisomerase inhibitors, antibacterial compounds, protein inhibitors, microtubule modulators, and COX inhibitors. Certain compounds also dramatically affected the length of the cilium. For a selection of compounds (Clofibrate, Gefitinib, Sirolimus, Imexon and Dexamethasone) their ability to restore ciliogenesis was confirmed in a panel of human cancer cell line models representing different cancer types (pancreas, lung, kidney, breast). Most compounds attenuated cell proliferation, at least in part through induction of the primary cilium, as demonstrated by cilium removal using chloral hydrate. These findings reveal that several commonly used drugs restore ciliogenesis in cancer cells, and warrant further investigation of their antineoplastic properties.


Asunto(s)
Antineoplásicos/farmacología , Proliferación Celular/efectos de los fármacos , Cilios/efectos de los fármacos , Ensayos de Selección de Medicamentos Antitumorales/métodos , Células A549 , Antineoplásicos/clasificación , Carcinoma Ductal Pancreático/metabolismo , Carcinoma Ductal Pancreático/patología , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Cilios/metabolismo , Gefitinib , Humanos , Microscopía Confocal , Neoplasias/metabolismo , Neoplasias/patología , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patología , Quinazolinas/farmacología , Reproducibilidad de los Resultados
10.
Oncotarget ; 7(11): 12582-97, 2016 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-26862848

RESUMEN

Lung cancer is the leading cause of cancer death. Beyond first line treatment, few therapeutic options are available, particularly for squamous cell carcinoma (SCC). Here, we have explored the phospholipidomes of 30 human SCCs and found that they almost invariably (in 96.7% of cases) contain phospholipids with longer acyl chains compared to matched normal tissues. This trait was confirmed using in situ 2D-imaging MS on tissue sections and by phospholipidomics of tumor and normal lung tissue of the L-IkkαKA/KA mouse model of lung SCC. In both human and mouse, the increase in acyl chain length in cancer tissue was accompanied by significant changes in the expression of acyl chain elongases (ELOVLs). Functional screening of differentially expressed ELOVLs by selective gene knockdown in SCC cell lines followed by phospholipidomics revealed ELOVL6 as the main elongation enzyme responsible for acyl chain elongation in cancer cells. Interestingly, inhibition of ELOVL6 drastically reduced colony formation of multiple SCC cell lines in vitro and significantly attenuated their growth as xenografts in vivo in mouse models. These findings identify acyl chain elongation as one of the most common traits of lung SCC discovered so far and pinpoint ELOVL6 as a novel potential target for cancer intervention.


Asunto(s)
Acetiltransferasas/metabolismo , Carcinoma de Células Escamosas , Neoplasias Pulmonares , Fosfolípidos/química , Animales , Carcinoma de Células Escamosas/química , Elongasas de Ácidos Grasos , Xenoinjertos , Humanos , Neoplasias Pulmonares/química , Ratones
11.
Mol Biol Cell ; 26(12): 2321-32, 2015 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-25904332

RESUMEN

Distortion of primary cilium formation is increasingly recognized as a key event in many human pathologies. One of the underlying mechanisms involves aberrant activation of the lipogenic transcription factor sterol regulatory element-binding protein 1c (SREBP1c), as observed in cancer cells. To gain more insight into the molecular pathways by which SREBP1c suppresses primary ciliogenesis, we searched for overlap between known ciliogenesis regulators and targets of SREBP1. One of the candidate genes that was consistently up-regulated in cellular models of SREBP1c-induced cilium repression was phospholipase A2 group III (PLA2G3), a phospholipase that hydrolyzes the sn-2 position of glycerophospholipids. Use of RNA interference and a chemical inhibitor of PLA2G3 rescued SREBP1c-induced cilium repression. Cilium repression by SREBP1c and PLA2G3 involved alterations in endosomal recycling and vesicular transport toward the cilium, as revealed by aberrant transferrin and Rab11 localization, and was largely mediated by an increase in lysophosphatidylcholine and lysophosphatidylethanolamine levels. Together these findings indicate that aberrant activation of SREBP1c suppresses primary ciliogenesis by PLA2G3-mediated distortion of vesicular trafficking and suggest that PLA2G3 is a novel potential target to normalize ciliogenesis in SREBP1c-overexpressing cells, including cancer cells.


Asunto(s)
Cilios/fisiología , Fosfolipasas A2 Grupo III/fisiología , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/fisiología , Vesículas Transportadoras/fisiología , Animales , Secuencia de Bases , Células Cultivadas , Cilios/metabolismo , Perros , Femenino , Fosfolipasas A2 Grupo III/genética , Humanos , Ratones , Datos de Secuencia Molecular , Transporte de Proteínas , Alineación de Secuencia , Sus scrofa
12.
J Clin Endocrinol Metab ; 89(1): 219-26, 2004 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-14715853

RESUMEN

Compared with the conventional approach, which recommended only insulin therapy when blood glucose levels exceeded 12 mmol/liter, strict maintenance of blood glucose levels less than 6.1 mmol/liter with intensive insulin therapy has shown to reduce intensive care mortality, acute renal failure, critical illness polyneuropathy, and bloodstream infections in critically ill patients by about 40%. This study of 363 patients, requiring intensive care for more than 7 d and randomly assigned to either conventional or intensive insulin therapy, examines the effects of intensive insulin therapy on glucose and lipid homeostasis and their respective impact on the improved outcome. Intensive insulin therapy effectively normalized blood glucose levels within 24 h, both in survivors and nonsurvivors. Intensive insulin therapy also increased serum levels of low-density lipoprotein (P = 0.007) and high-density lipoprotein (P = 0.005), whereas it suppressed the elevated serum triglyceride concentrations (P < 0.0001). Multivariate logistic regression analysis, corrected for baseline univariate risk factors and the effect on inflammation, indicated that lipid rather than glucose control independently determined the beneficial effects of intensive insulin therapy on morbidity and mortality. In postmortem biopsies obtained from 74 patients who died in the intensive care unit, intensive insulin therapy increased mRNA levels of skeletal muscle glucose transporter 4 (P = 0.02) and hexokinase (P = 0.03), unlike those of hepatic glucokinase. In conclusion, our data suggest that intensive insulin therapy normalizes blood glucose levels through stimulation of peripheral glucose uptake and concomitantly partially restores the abnormalities in the serum lipid profile, which may have contributed significantly to the improved outcome of protracted critical illness.


Asunto(s)
Glucemia/análisis , Enfermedad Crítica , Insulina/administración & dosificación , Lípidos/sangre , Proteínas Musculares , Factores de Transcripción , Resultado del Tratamiento , APACHE , Lesión Renal Aguda/prevención & control , Bacteriemia/prevención & control , Proteínas Potenciadoras de Unión a CCAAT/genética , Colesterol/sangre , HDL-Colesterol/sangre , LDL-Colesterol/sangre , Proteínas de Unión al ADN/genética , Glucoquinasa/genética , Transportador de Glucosa de Tipo 4 , Hexoquinasa/genética , Homeostasis , Humanos , Lipoproteína Lipasa/genética , Lipoproteínas HDL/sangre , Lipoproteínas LDL/sangre , Hígado/enzimología , Modelos Logísticos , Proteínas de Transporte de Monosacáridos/genética , Músculo Esquelético/química , Polineuropatías/prevención & control , ARN Mensajero/análisis , Proteína 1 de Unión a los Elementos Reguladores de Esteroles , Triglicéridos/sangre
13.
J Clin Endocrinol Metab ; 87(12): 5516-23, 2002 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-12466347

RESUMEN

IGF binding protein-1 (IGFBP-1), an important regulator of IGF bioavailability, has been shown to correlate with mortality in critically ill patients. In the liver, IGFBP-1 is transcriptionally repressed by insulin, and it is therefore a potential marker of hepatic insulin sensitivity. We have recently shown that, compared with conventional treatment, maintenance of normoglycemia with intensive insulin therapy decreased morbidity and mortality of continuously fed critically ill patients. This study compares the effect of conventional and intensive insulin therapy on IGFBP-1 and assesses its predictive value for mortality. In 363 patients who were dependent on intensive care for more than 7 d and were randomly assigned to either conventional or intensive insulin therapy, serum IGFBP-1 levels were measured on admission, on d 1, 8, 15, 22, and 29, and on the day of intensive care unit discharge or death. In addition, IGFBP-1 and phosphoenolpyruvate carboxykinase mRNA levels were measured by real-time RT-PCR in postmortem liver biopsies obtained from 74 patients who died in the intensive care unit. Although intensive insulin treatment lowered glycemia, it had no effect on IGFBP-1 serum levels. Instead, serum IGFBP-1 concentration was significantly higher in patients who ultimately died, and it differentiated nonsurvivors from survivors 3 wk before death. The predictive value of serum IGFBP-1 for mortality was similar to that of the APACHE-II score. Like circulating IGFBP-1, hepatic mRNA levels of IGFBP-1 and the similarly insulin-regulated gene, phosphoenolpyruvate carboxykinase, were not significantly different between conventional and intensive insulin therapy groups. These data suggest that hepatic insulin resistance in prolonged critically ill patients, reflected by high serum IGFBP-1 levels, is not overcome by intensive insulin therapy, and that this may affect patient outcome.


Asunto(s)
Enfermedad Crítica , Proteína 1 de Unión a Factor de Crecimiento Similar a la Insulina/sangre , Anciano , Glucemia/análisis , Carboxiliasas/genética , Enfermedad Crónica , Enfermedad Crítica/mortalidad , Femenino , Predicción , Expresión Génica , Humanos , Insulina/uso terapéutico , Proteína 1 de Unión a Factor de Crecimiento Similar a la Insulina/genética , Unidades de Cuidados Intensivos , Tiempo de Internación , Hígado/fisiopatología , Masculino , Persona de Mediana Edad
14.
Mol Cell Endocrinol ; 205(1-2): 21-31, 2003 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-12890564

RESUMEN

In prostate cancer cell lines in culture androgens cause a marked and coordinated upregulation of the expression of several lipogenic genes. Here, using castrated male Wistar rats as an experimental paradigm, we investigated whether coordinated androgen stimulation of lipogenic gene expression represents a more general physiological regulation in non-cancerous androgen-responsive cells as well. In typical target tissues for androgen action such as the ventral prostate and the lacrimal gland, androgen deprivation resulted in a marked reduction in the mRNA and protein levels of genes involved in fatty acid (fatty acid synthase and acetyl-CoA-carboxylase) and cholesterol synthesis (HMG-CoA-reductase and farnesyl diphosphate synthase). Readministration of testosterone immediately following orchidectomy restored the expression of all four genes. Substitution of testosterone by the non-aromatizable androgen dihydrotestosterone gave rise to comparable changes in the mRNA and protein levels of the lipogenic genes under investigation, confirming the involvement of the androgen receptor in the observed effects. In support of the coordinate nature of this regulation, androgen-induced upregulation of lipogenic gene expression is accompanied by an increase in the nuclear content of SREBP, a key lipogenic transcription factor. Taken together, these findings provide evidence for a coordinate regulation of lipogenic gene expression not only in prostate cancer cell lines in culture but also in non-cancerous androgen-responsive tissues in vivo.


Asunto(s)
Andrógenos/farmacología , Ácido Graso Sintasas/metabolismo , Metabolismo de los Lípidos , Próstata/metabolismo , Factores de Transcripción , Transferasas Alquil y Aril/genética , Transferasas Alquil y Aril/metabolismo , Animales , Secuencia de Bases , Proteínas Potenciadoras de Unión a CCAAT/análisis , Proteínas de Unión al ADN/análisis , Ácido Graso Sintasas/genética , Expresión Génica , Geraniltranstransferasa , Masculino , Ratones , Datos de Secuencia Molecular , Próstata/efectos de los fármacos , Próstata/enzimología , ARN Mensajero/metabolismo , Ratas , Ratas Wistar , Proteína 1 de Unión a los Elementos Reguladores de Esteroles , Regulación hacia Arriba
15.
Cancer Res ; 70(22): 9453-62, 2010 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-20889723

RESUMEN

Aberrant activation of fatty acid synthesis is a key feature of many advanced human cancers. Unlike in classical lipogenic tissues, this process has been implicated in membrane production required for rapid cell proliferation. Here, to gain further insight into the consequences of tumor-associated fatty acid synthesis, we have mimicked the lipogenic phenotype of cancer cells in Xenopus embryos by microinjection of RNA encoding the lipogenic transcription factor sterol regulatory element binding protein 1c (SREBP1c). Dramatic morphologic changes were observed that could be linked to alterations in Wnt and Hedgehog signaling, and ultimately to a distortion of the primary cilium. This is a sophisticated microtubular sensory organelle that is expressed on the surface of nearly every cell type and that is lost in many cancers. SREBP1c-induced loss of the primary cilium could be confirmed in mammalian Madin-Darby canine kidney (MDCK) cells and was mediated by changes in the supply of fatty acids. Conversely, inhibition of fatty acid synthesis in highly lipogenic human prostate cancer cells restored the formation of the primary cilium. Lipid-induced ciliary loss was associated with mislocalization of apical proteins, distortion of cell polarization, and aberrant epithelial tissue development as revealed in three-dimensional cultures of MDCK cells and in the developing mouse prostate. These data imply that tumor-associated lipogenesis, in addition to rendering cells more autonomous in terms of lipid supply, disturbs cilium formation and contributes to impaired environmental sensing, aberrant signaling, and distortion of polarized tissue architecture, which are all hallmarks of cancer.


Asunto(s)
Cilios/metabolismo , Embrión no Mamífero/metabolismo , Ácidos Grasos/biosíntesis , ARN/metabolismo , Animales , Técnicas de Cultivo de Célula , Línea Celular , Línea Celular Tumoral , Cilios/genética , Embrión no Mamífero/embriología , Femenino , Proteínas Hedgehog/genética , Proteínas Hedgehog/metabolismo , Humanos , Lipogénesis/genética , Masculino , Ratones , Ratones de la Cepa 129 , Microinyecciones , Microscopía Confocal , Microscopía Electrónica de Rastreo , Próstata/crecimiento & desarrollo , Próstata/metabolismo , ARN/administración & dosificación , ARN/genética , Interferencia de ARN , Transducción de Señal/genética , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/genética , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/metabolismo , Proteínas Wnt/genética , Proteínas Wnt/metabolismo , Xenopus , Proteínas de Xenopus/genética , Proteínas de Xenopus/metabolismo
16.
Cancer Res ; 70(20): 8117-26, 2010 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-20876798

RESUMEN

Activation of de novo lipogenesis in cancer cells is increasingly recognized as a hallmark of aggressive cancers and has been implicated in the production of membranes for rapid cell proliferation. In the current report, we provide evidence that this activation has a more profound role. Using a mass spectrometry-based phospholipid analysis approach, we show that clinical tumor tissues that display the lipogenic phenotype show an increase in the degree of lipid saturation compared with nonlipogenic tumors. Reversal of the lipogenic switch in cancer cells by treatment with the lipogenesis inhibitor soraphen A or by targeting lipogenic enzymes with small interfering RNA leads to a marked decrease in saturated and mono-unsaturated phospholipid species and increases the relative degree of polyunsaturation. Because polyunsaturated acyl chains are more susceptible to peroxidation, inhibition of lipogenesis increases the levels of peroxidation end products and renders cells more susceptible to oxidative stress-induced cell death. As saturated lipids pack more densely, modulation of lipogenesis also alters lateral and transversal membrane dynamics as revealed by diffusion of membrane-targeted green fluorescent protein and by the uptake and response to doxorubicin. These data show that shifting lipid acquisition from lipid uptake toward de novo lipogenesis dramatically changes membrane properties and protects cells from both endogenous and exogenous insults. These findings provide important new insights into the role of de novo lipogenesis in cancer cells, and they provide a rationale for the use of lipogenesis inhibitors as antineoplastic agents and as chemotherapeutic sensitizers.


Asunto(s)
Radicales Libres/farmacología , Lipogénesis/fisiología , Lípidos de la Membrana/metabolismo , Neoplasias/metabolismo , Antibióticos Antineoplásicos/metabolismo , División Celular , Membrana Celular/efectos de los fármacos , Membrana Celular/fisiología , Colesterol/metabolismo , Doxorrubicina/metabolismo , Células HCT116/efectos de los fármacos , Células HCT116/metabolismo , Humanos , Immunoblotting , Peroxidación de Lípido , Masculino , Neoplasias/patología , Fosfolípidos/metabolismo , Próstata/metabolismo , Próstata/patología , Prostatectomía , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , ARN Interferente Pequeño/genética , Espectrometría de Masa por Ionización de Electrospray , Transfección , Triglicéridos/metabolismo
17.
Biochem Biophys Res Commun ; 302(4): 898-903, 2003 Mar 21.
Artículo en Inglés | MEDLINE | ID: mdl-12646257

RESUMEN

Fatty acid synthase (FAS) is a key metabolic enzyme catalyzing the synthesis of long-chain saturated fatty acids. It plays a central role in the production of surfactant in fetal lungs, in the supply of fatty components of milk, and in the conversion and storage of energy in liver and adipose tissue. Remarkably high levels of FAS expression are found in the majority of human epithelial cancers. As the role of FAS in cancer cells remains largely unknown, we have initiated studies to assess the fate of newly synthesized lipids in cancer cells and have estimated the contribution of FAS to the synthesis of specific lipid classes by treating the cells with small interfering RNAs targeting FAS. Here, we show that in cancer cells FAS plays a major role in the synthesis of phospholipids partitioning into detergent-resistant membrane microdomains. These are raft-aggregates implicated in key cellular processes including signal transduction, intracellular trafficking, cell polarization, and cell migration. These findings reveal a novel role for FAS, provide important new insights into the otherwise poorly understood mechanisms underlying the control of lipid composition of membrane microdomains, and point to a link between FAS overexpression and dysregulation of membrane composition and functioning in tumor cells.


Asunto(s)
Detergentes/química , Ácido Graso Sintasas/metabolismo , Microdominios de Membrana/metabolismo , Fosfolípidos/metabolismo , Regulación hacia Abajo/fisiología , Humanos , Masculino , Microdominios de Membrana/química , Neoplasias de la Próstata , ARN Interferente Pequeño/metabolismo , Células Tumorales Cultivadas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA