Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Biochim Biophys Acta ; 1849(7): 830-5, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25252159

RESUMEN

Here we discuss the function of eukaryotic initiation factor 6 (eIF6; Tif6 in yeast). eIF6 binds 60S ribosomal subunits and blocks their joining to 40S. In this context, we propose that eIF6 impedes unproductive 80S formation, namely, the formation of 80S subunits without mRNA. Genetic evidence shows that eIF6 has a dual function: in yeast and mammals, nucleolar eIF6 is necessary for the biogenesis of 60S subunits. In mammals, cytoplasmic eIF6 is required for insulin and growth factor-stimulated translation. In contrast to other translation factors, eIF6 activity is not under mTOR control. The physiological significance of eIF6 impacts on cancer and on inherited Shwachman-Bodian-Diamond syndrome. eIF6 is overexpressed in specific human tumors. In a murine model of lymphomagenesis, eIF6 depletion leads to a striking increase of survival, without adverse effects. Shwachman-Bodian-Diamond syndrome is caused by loss of function of SBDS protein. In yeast, point mutations of Tif6, the yeast homolog of eIF6, rescue the quasi-lethal effect due to the loss of the SBDS homolog, Sdo1. We propose that eIF6 is a node regulator of ribosomal function and predict that prioritizing its pharmacological targeting will be of benefit in cancer and Shwachman-Bodian-Diamond syndrome. This article is part of a Special Issue entitled: Translation and Cancer.


Asunto(s)
Factores Eucarióticos de Iniciación/metabolismo , Linfoma/metabolismo , Proteínas de Neoplasias/metabolismo , Factores de Iniciación de Péptidos/metabolismo , Biosíntesis de Proteínas , Subunidades Ribosómicas Grandes de Eucariotas/metabolismo , Animales , Enfermedades de la Médula Ósea/genética , Enfermedades de la Médula Ósea/metabolismo , Enfermedades de la Médula Ósea/patología , Factores Eucarióticos de Iniciación/genética , Insuficiencia Pancreática Exocrina/genética , Insuficiencia Pancreática Exocrina/metabolismo , Insuficiencia Pancreática Exocrina/patología , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Lipomatosis/genética , Lipomatosis/metabolismo , Lipomatosis/patología , Linfoma/genética , Linfoma/patología , Ratones , Proteínas de Neoplasias/genética , Neoplasias Experimentales/genética , Neoplasias Experimentales/metabolismo , Neoplasias Experimentales/patología , Factores de Iniciación de Péptidos/genética , Subunidades Ribosómicas Grandes de Eucariotas/genética , Síndrome de Shwachman-Diamond , Serina-Treonina Quinasas TOR/genética , Serina-Treonina Quinasas TOR/metabolismo , Transactivadores
2.
Biochem Soc Trans ; 44(6): 1667-1673, 2016 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-27913676

RESUMEN

Over the past few years, there has been a growing interest in the interconnection between translation and metabolism. Important oncogenic pathways, like those elicited by c-Myc transcription factor and mTOR kinase, couple the activation of the translational machinery with glycolysis and fatty acid synthesis. Eukaryotic initiation factor 6 (eIF6) is a factor necessary for 60S ribosome maturation. eIF6 acts also as a cytoplasmic translation initiation factor, downstream of growth factor stimulation. eIF6 is up-regulated in several tumor types. Data on mice models have demonstrated that eIF6 cytoplasmic activity is rate-limiting for Myc-induced lymphomagenesis. In spite of this, eIF6 is neither transcriptionally regulated by Myc, nor post-transcriptionally regulated by mTOR. eIF6 stimulates a glycolytic and fatty acid synthesis program necessary for tumor growth. eIF6 increases the translation of transcription factors necessary for lipogenesis, such as CEBP/ß, ATF4 and CEBP/δ. Insulin stimulation leads to an increase in translation and fat synthesis blunted by eIF6 deficiency. Paradoxycally, long-term inhibition of eIF6 activity increases insulin sensitivity, suggesting that the translational activation observed upon insulin and growth factors stimulation acts as a feed-forward mechanism regulating lipid synthesis. The data on the role that eIF6 plays in cancer and in insulin sensitivity make it a tempting pharmacological target for cancers and metabolic diseases. We speculate that eIF6 inhibition will be particularly effective especially when mTOR sensitivity to rapamycin is abrogated by RAS mutations.


Asunto(s)
Factores de Iniciación de Péptidos/genética , Biosíntesis de Proteínas , Serina-Treonina Quinasas TOR/genética , Factores de Transcripción/genética , Factor de Transcripción Activador 4/genética , Factor de Transcripción Activador 4/metabolismo , Animales , Proteína beta Potenciadora de Unión a CCAAT/genética , Proteína beta Potenciadora de Unión a CCAAT/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Hipoglucemiantes/farmacología , Insulina/farmacología , Lipogénesis/efectos de los fármacos , Lipogénesis/genética , Enfermedades Metabólicas/genética , Enfermedades Metabólicas/metabolismo , Ratones , Modelos Genéticos , Neoplasias/genética , Neoplasias/metabolismo , Factores de Iniciación de Péptidos/metabolismo , Proteínas Proto-Oncogénicas c-myc/genética , Proteínas Proto-Oncogénicas c-myc/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Factores de Transcripción/metabolismo
3.
Int J Cancer ; 136(5): E272-81, 2015 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-25273595

RESUMEN

Tumors characterized by an intense ribosome biogenesis often display a more aggressive behavior. Ribosomal RNA (rRNA) synthesis is controlled at several levels, including the epigenetic regulation of the condensation of chromatin portions containing rRNA genes. JHDM1B (Jumonji C histone demethylase 1B) is a histone demethylase able to regulate the accessibility of rRNA genes. In this study, we aimed to define the contribution of JHDM1B expression to the features of breast cancer, a tumor type whose behavior is related to the rate of ribosome biogenesis. We show that, in breast cancer-derived cell lines, the increase in rRNA transcription that follows JHDM1B knock-down is mirrored by an augmented cell proliferation only in p53 compromised cells, while p53 competent cells undergo cellular senescence and death. The latter effect appears to be mediated by a p38-dependent phosphorylation of p53, inducing the expression of p15(Ink4b) and p21(Waf1). In breast cancers, lower JHDM1B expression correlates with an increased size of specifically stained nucleolar organized regions, a morphological parameter directly related to the rate of ribosome biogenesis and with a poorer prognosis. In addition, in tumors lacking the controller function of p53, a lower expression of JHDM1B is associated with an increased tumor size at diagnosis. Altogether, our data indicate that epigenetic activation of rDNA genes induced by JHDM1B depletion is associated with a p53-dependent growth arrest, but may promote cancer cell growth when p53 is lacking.


Asunto(s)
Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Proliferación Celular , Proteínas F-Box/metabolismo , Histona Demetilasas con Dominio de Jumonji/metabolismo , Ribosomas/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Western Blotting , Neoplasias de la Mama/mortalidad , Senescencia Celular , Proteínas F-Box/antagonistas & inhibidores , Proteínas F-Box/genética , Femenino , Humanos , Técnicas para Inmunoenzimas , Histona Demetilasas con Dominio de Jumonji/antagonistas & inhibidores , Histona Demetilasas con Dominio de Jumonji/genética , ARN Mensajero/genética , ARN Ribosómico/genética , ARN Ribosómico/metabolismo , ARN Interferente Pequeño/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células Tumorales Cultivadas , Proteína p53 Supresora de Tumor/genética
4.
Cell Mol Life Sci ; 70(8): 1439-50, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23212600

RESUMEN

The receptor for activated C-kinase 1 (RACK1) is a conserved structural protein of 40S ribosomes. Strikingly, deletion of RACK1 in yeast homolog Asc1 is not lethal. Mammalian RACK1 also interacts with many nonribosomal proteins, hinting at several extraribosomal functions. A knockout mouse for RACK1 has not previously been described. We produced the first RACK1 mutant mouse, in which both alleles of RACK1 gene are defective in RACK1 expression (ΔF/ΔF), in a pure C57 Black/6 background. In a sample of 287 pups, we observed no ΔF/ΔF mice (72 expected). Dissection and genotyping of embryos at various stages showed that lethality occurs at gastrulation. Heterozygotes (ΔF/+) have skin pigmentation defects with a white belly spot and hypopigmented tail and paws. ΔF/+ have a transient growth deficit (shown by measuring pup size at P11). The pigmentation deficit is partly reverted by p53 deletion, whereas the lethality is not. ΔF/+ livers have mild accumulation of inactive 80S ribosomal subunits by polysomal profile analysis. In ΔF/+ fibroblasts, protein synthesis response to extracellular and pharmacological stimuli is reduced. These results highlight the role of RACK1 as a ribosomal protein converging signaling to the translational apparatus.


Asunto(s)
Neuropéptidos/genética , Pigmentación , Biosíntesis de Proteínas , Animales , Células Cultivadas , Pérdida del Embrión/genética , Embrión de Mamíferos/metabolismo , Embrión de Mamíferos/ultraestructura , Técnicas de Inactivación de Genes , Células HEK293 , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neuropéptidos/metabolismo , Fenotipo , Interferencia de ARN , ARN Interferente Pequeño/genética , Receptores de Cinasa C Activada , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo
5.
Nat Aging ; 2024 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-38951692

RESUMEN

Accumulating senescent cells within tissues contribute to the progression of aging and age-related diseases. Botanical extracts, rich in phytoconstituents, present a useful resource for discovering therapies that could target senescence and thus improve healthspan. Here, we show that daily oral administration of a standardized extract of Salvia haenkei (Haenkenium (HK)) extended lifespan and healthspan of naturally aged mice. HK treatment inhibited age-induced inflammation, fibrosis and senescence markers across several tissues, as well as increased muscle strength and fur thickness compared with age-matched controls. We also found that HK treatment reduced acutely induced senescence by the chemotherapeutic agent doxorubicin, using p16LUC reporter mice. We profiled the constituent components of HK by mass spectrometry, and identified luteolin-the most concentrated flavonoid in HK-as a senomorphic compound. Mechanistically, by performing surface plasmon resonance and in situ proximity ligation assay, we found that luteolin disrupted the p16-CDK6 interaction. This work demonstrates that administration of HK promotes longevity in mice, possibly by modulating cellular senescence and by disrupting the p16-CDK6 interaction.

6.
Cancer Cell ; 41(3): 602-619.e11, 2023 03 13.
Artículo en Inglés | MEDLINE | ID: mdl-36868226

RESUMEN

Tumor cells promote the recruitment of immunosuppressive neutrophils, a subset of myeloid cells driving immune suppression, tumor proliferation, and treatment resistance. Physiologically, neutrophils are known to have a short half-life. Here, we report the identification of a subset of neutrophils that have upregulated expression of cellular senescence markers and persist in the tumor microenvironment. Senescent-like neutrophils express the triggering receptor expressed on myeloid cells 2 (TREM2) and are more immunosuppressive and tumor-promoting than canonical immunosuppressive neutrophils. Genetic and pharmacological elimination of senescent-like neutrophils decreases tumor progression in different mouse models of prostate cancer. Mechanistically, we have found that apolipoprotein E (APOE) secreted by prostate tumor cells binds TREM2 on neutrophils, promoting their senescence. APOE and TREM2 expression increases in prostate cancers and correlates with poor prognosis. Collectively, these results reveal an alternative mechanism of tumor immune evasion and support the development of immune senolytics targeting senescent-like neutrophils for cancer therapy.


Asunto(s)
Apolipoproteínas E , Neoplasias de la Próstata , Animales , Humanos , Masculino , Ratones , Apolipoproteínas E/metabolismo , Senescencia Celular/genética , Glicoproteínas de Membrana/genética , Células Mieloides/metabolismo , Neoplasias de la Próstata/metabolismo , Receptores Inmunológicos/metabolismo , Microambiente Tumoral
7.
Nat Cancer ; 4(8): 1102-1121, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37460872

RESUMEN

Cancer is highly infiltrated by myeloid-derived suppressor cells (MDSCs). Currently available immunotherapies do not completely eradicate MDSCs. Through a genome-wide analysis of the translatome of prostate cancers driven by different genetic alterations, we demonstrate that prostate cancer rewires its secretome at the translational level to recruit MDSCs. Among different secreted proteins released by prostate tumor cells, we identified Hgf, Spp1 and Bgn as the key factors that regulate MDSC migration. Mechanistically, we found that the coordinated loss of Pdcd4 and activation of the MNK/eIF4E pathways regulate the mRNAs translation of Hgf, Spp1 and Bgn. MDSC infiltration and tumor growth were dampened in prostate cancer treated with the MNK1/2 inhibitor eFT508 and/or the AKT inhibitor ipatasertib, either alone or in combination with a clinically available MDSC-targeting immunotherapy. This work provides a therapeutic strategy that combines translation inhibition with available immunotherapies to restore immune surveillance in prostate cancer.


Asunto(s)
Neoplasias de la Próstata , Proteínas Serina-Treonina Quinasas , Masculino , Humanos , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Fosforilación , Factor 4E Eucariótico de Iniciación/genética , Factor 4E Eucariótico de Iniciación/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Neoplasias de la Próstata/genética , Células Mieloides/metabolismo , Factor de Crecimiento de Hepatocito/metabolismo , Osteopontina/metabolismo , Biglicano/metabolismo
8.
Biochim Biophys Acta ; 1803(7): 826-31, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20359507

RESUMEN

Adhesion to fibronectin stimulates protein synthesis (translation) of fibroblasts. Protein synthesis stimulation is dependent from the activation of beta(1)-integrin. beta(1)-Integrin elicits a PI3K cascade that modulates eIF4F (eukaryotic initiation factor 4F) complex formation. In the attempt to further dissect elements of the PI3K cascade that might be responsible for fibronectin-stimulated translation, we used pharmacological inhibitors of known kinases. We found that JNK inhibition, by SP600125 treatment, increased (35)S-methionine incorporation. Paradoxically, the increase in methionine incorporation was associated to a reduction of initiation of translation. These data imply that, during the adhesion of fibroblasts to fibronectin, conspicuous protein degradation occurs. Indeed, we found that inhibition of the proteasome by MG132 also increased methionine incorporation. Cotranslational degradation depended on PI3K activation. In spite of this, serum promoted translation, but not cotranslational degradation. The crosstalk between translation and degradation was further analyzed by studying the phosphorylation of initiation factors. Briefly, inhibition of JNK leads to eIF2alpha phosphorylation, which may account for the decrease in initiation of translation. In conclusion, beta(1)-integrin-activated translation causes the synthesis of short-lived proteins, whose degradation is controlled by the JNK pathway. We hypothesize that JNK is a general regulator of cotranslational degradation.


Asunto(s)
Adhesión Celular/fisiología , Proteínas Quinasas JNK Activadas por Mitógenos/antagonistas & inhibidores , Biosíntesis de Proteínas , Animales , Antracenos/metabolismo , Línea Celular , Cromonas/metabolismo , Citoesqueleto/metabolismo , Inhibidores Enzimáticos/metabolismo , Fibroblastos/citología , Fibroblastos/fisiología , Fibronectinas/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/genética , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Ratones , Morfolinas/metabolismo , Células 3T3 NIH , Fosfatidilinositol 3-Quinasas/metabolismo
9.
Nat Commun ; 12(1): 4878, 2021 08 12.
Artículo en Inglés | MEDLINE | ID: mdl-34385447

RESUMEN

A postprandial increase of translation mediated by eukaryotic Initiation Factor 6 (eIF6) occurs in the liver. Its contribution to steatosis and disease is unknown. In this study we address whether eIF6-driven translation contributes to disease progression. eIF6 levels increase throughout the progression from Non-Alcoholic Fatty Liver Disease (NAFLD) to hepatocellular carcinoma. Reduction of eIF6 levels protects the liver from disease progression. eIF6 depletion blunts lipid accumulation, increases fatty acid oxidation (FAO) and reduces oncogenic transformation in vitro. In addition, eIF6 depletion delays the progression from NAFLD to hepatocellular carcinoma, in vivo. Mechanistically, eIF6 depletion reduces the translation of transcription factor C/EBPß, leading to a drop in biomarkers associated with NAFLD progression to hepatocellular carcinoma and preserves mitochondrial respiration due to the maintenance of an alternative mTORC1-eIF4F translational branch that increases the expression of transcription factor YY1. We provide proof-of-concept that in vitro pharmacological inhibition of eIF6 activity recapitulates the protective effects of eIF6 depletion. We hypothesize the existence of a targetable, evolutionarily conserved translation circuit optimized for lipid accumulation and tumor progression.


Asunto(s)
Carcinoma Hepatocelular/genética , Neoplasias Hepáticas/genética , Enfermedad del Hígado Graso no Alcohólico/genética , Factores de Iniciación de Péptidos/genética , Biosíntesis de Proteínas/genética , Animales , Proteína beta Potenciadora de Unión a CCAAT/genética , Proteína beta Potenciadora de Unión a CCAAT/metabolismo , Carcinoma Hepatocelular/metabolismo , Línea Celular , Transformación Celular Neoplásica/efectos de los fármacos , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Clofazimina/farmacología , Dieta Alta en Grasa/efectos adversos , Progresión de la Enfermedad , Silenciador del Gen , Humanos , Lipogénesis/efectos de los fármacos , Lipogénesis/genética , Neoplasias Hepáticas/metabolismo , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Obesidad/etiología , Obesidad/genética , Obesidad/metabolismo , Factores de Iniciación de Péptidos/antagonistas & inhibidores , Factores de Iniciación de Péptidos/metabolismo
10.
Cancer Cell ; 39(1): 68-82.e9, 2021 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-33186519

RESUMEN

Metastases account for most cancer-related deaths, yet the mechanisms underlying metastatic spread remain poorly understood. Recent evidence demonstrates that senescent cells, while initially restricting tumorigenesis, can induce tumor progression. Here, we identify the metalloproteinase inhibitor TIMP1 as a molecular switch that determines the effects of senescence in prostate cancer. Senescence driven either by PTEN deficiency or chemotherapy limits the progression of prostate cancer in mice. TIMP1 deletion allows senescence to promote metastasis, and elimination of senescent cells with a senolytic BCL-2 inhibitor impairs metastasis. Mechanistically, TIMP1 loss reprograms the senescence-associated secretory phenotype (SASP) of senescent tumor cells through activation of matrix metalloproteinases (MMPs). Loss of PTEN and TIMP1 in prostate cancer is frequent and correlates with resistance to docetaxel and worst clinical outcomes in patients treated in an adjuvant setting. Altogether, these findings provide insights into the dual roles of tumor-associated senescence and can potentially impact the treatment of prostate cancer.


Asunto(s)
Docetaxel/administración & dosificación , Eliminación de Gen , Fosfohidrolasa PTEN/genética , Neoplasias de la Próstata/patología , Inhibidor Tisular de Metaloproteinasa-1/genética , Animales , Senescencia Celular/efectos de los fármacos , Docetaxel/farmacología , Regulación Neoplásica de la Expresión Génica , Humanos , Masculino , Metaloproteinasas de la Matriz/metabolismo , Ratones , Metástasis de la Neoplasia , Trasplante de Neoplasias , Células PC-3 , Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/metabolismo
11.
Cancer Res ; 81(24): 6207-6218, 2021 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-34753775

RESUMEN

It has been recognized for decades that ERBB signaling is important in prostate cancer, but targeting ERBB receptors as a therapeutic strategy for prostate cancer has been ineffective clinically. However, we show here that membranous HER3 protein is commonly highly expressed in lethal prostate cancer, associating with reduced time to castration resistance (CR) and survival. Multiplex immunofluorescence indicated that the HER3 ligand NRG1 is detectable primarily in tumor-infiltrating myelomonocytic cells in human prostate cancer; this observation was confirmed using single-cell RNA sequencing of human prostate cancer biopsies and murine transgenic prostate cancer models. In castration-resistant prostate cancer (CRPC) patient-derived xenograft organoids with high HER3 expression as well as mouse prostate cancer organoids, recombinant NRG1 enhanced proliferation and survival. Supernatant from murine bone marrow-derived macrophages and myeloid-derived suppressor cells promoted murine prostate cancer organoid growth in vitro, which could be reversed by a neutralizing anti-NRG1 antibody and ERBB inhibition. Targeting HER3, especially with the HER3-directed antibody-drug conjugate U3-1402, exhibited antitumor activity against HER3-expressing prostate cancer. Overall, these data indicate that HER3 is commonly overexpressed in lethal prostate cancer and can be activated by NRG1 secreted by myelomonocytic cells in the tumor microenvironment, supporting HER3-targeted therapeutic strategies for treating HER3-expressing advanced CRPC. SIGNIFICANCE: HER3 is an actionable target in prostate cancer, especially with anti-HER3 immunoconjugates, and targeting HER3 warrants clinical evaluation in prospective trials.


Asunto(s)
Anticuerpos Monoclonales Humanizados/farmacología , Biomarcadores de Tumor/metabolismo , Camptotecina/análogos & derivados , Neurregulina-1/metabolismo , Organoides/patología , Neoplasias de la Próstata/patología , Receptor ErbB-3/antagonistas & inhibidores , Animales , Antineoplásicos Inmunológicos/farmacología , Apoptosis , Biomarcadores de Tumor/genética , Camptotecina/farmacología , Proliferación Celular , Estudios de Seguimiento , Humanos , Masculino , Ratones Endogámicos NOD , Ratones SCID , Neurregulina-1/genética , Organoides/efectos de los fármacos , Organoides/metabolismo , Pronóstico , Estudios Prospectivos , Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/metabolismo , Receptor ErbB-3/genética , Receptor ErbB-3/metabolismo , Tasa de Supervivencia , Células Tumorales Cultivadas , Microambiente Tumoral , Ensayos Antitumor por Modelo de Xenoinjerto
12.
J Clin Psychopharmacol ; 29(6): 576-83, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19910724

RESUMEN

The aim of this study was to model in mice the association between metabolic syndrome and the administration of atypical antipsychotic (AAP). Two dosages (4 and 8 mg/kg per day) of olanzapine (OL) were infused in 36 female mice for 30 days by osmotic mini-pumps. This study was also designed to further extend the implications raised in other experiments by our model of AAP-induced metabolic dysregulation. Through the use of the osmotic mini-pumps, this model is aimed to circumvent the shorter (than in humans) half-life of AAPs in rodents and to chronically administer OL by a reliable and less disturbing method. Indirect calorimetry was used to evaluate metabolic rate (MR) and respiratory exchange ratio together with weight and caloric intake. Serum insulin, leptin, and glucose tolerance (oral glucose tolerance test) were assessed. Pancreatic beta cells insulin levels, periuterine and liver fat content were also analyzed. Olanzapine-infused mice exhibited a reduction of overall MR (kilojoule per hour) and resting MR and respiratory exchange ratio, with periuterine fat significantly enlarged. All metabolic alterations were detected at the highest dose, with major effects found on weight gain and hyperphagia. Impaired glucose metabolism, associated with hyperinsulinemia and hyperleptinemia were found. Insulin resistance was evidenced by the raise of HOMA-IR index. Increased insulin and lipid storage were detected at pancreatic and hepatic levels respectively. These findings illustrate the development of a cluster of risk factors (metabolic syndrome) and, for the first time, a decrease of energy expenditure (MR) due to chronic OL infusion.


Asunto(s)
Benzodiazepinas/administración & dosificación , Benzodiazepinas/efectos adversos , Dislipidemias/sangre , Metabolismo Energético/fisiología , Intolerancia a la Glucosa/sangre , Resistencia a la Insulina/fisiología , Animales , Peso Corporal/efectos de los fármacos , Peso Corporal/fisiología , Dislipidemias/inducido químicamente , Metabolismo Energético/efectos de los fármacos , Femenino , Intolerancia a la Glucosa/inducido químicamente , Bombas de Infusión Implantables , Infusiones Intravenosas , Síndrome Metabólico/sangre , Síndrome Metabólico/inducido químicamente , Ratones , Olanzapina , Factores de Tiempo
13.
Nat Genet ; 50(2): 219-228, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29335542

RESUMEN

The mechanisms by which mitochondrial metabolism supports cancer anabolism remain unclear. Here, we found that genetic and pharmacological inactivation of pyruvate dehydrogenase A1 (PDHA1), a subunit of the pyruvate dehydrogenase complex (PDC), inhibits prostate cancer development in mouse and human xenograft tumor models by affecting lipid biosynthesis. Mechanistically, we show that in prostate cancer, PDC localizes in both the mitochondria and the nucleus. Whereas nuclear PDC controls the expression of sterol regulatory element-binding transcription factor (SREBF)-target genes by mediating histone acetylation, mitochondrial PDC provides cytosolic citrate for lipid synthesis in a coordinated manner, thereby sustaining anabolism. Additionally, we found that PDHA1 and the PDC activator pyruvate dehydrogenase phosphatase 1 (PDP1) are frequently amplified and overexpressed at both the gene and protein levels in prostate tumors. Together, these findings demonstrate that both mitochondrial and nuclear PDC sustain prostate tumorigenesis by controlling lipid biosynthesis, thus suggesting this complex as a potential target for cancer therapy.


Asunto(s)
Compartimento Celular/fisiología , Lipogénesis , Neoplasias de la Próstata/metabolismo , Piruvato Deshidrogenasa (Lipoamida)/genética , Complejo Piruvato Deshidrogenasa/fisiología , Animales , Línea Celular Tumoral , Núcleo Celular/genética , Núcleo Celular/metabolismo , Núcleo Celular/patología , Células Cultivadas , Citoplasma/genética , Citoplasma/metabolismo , Citoplasma/patología , Humanos , Lipogénesis/genética , Masculino , Ratones , Ratones Noqueados , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/patología , Procesamiento Proteico-Postraduccional/genética , Piruvato Deshidrogenasa (Lipoamida)/metabolismo , Complejo Piruvato Deshidrogenasa/metabolismo
15.
Dev Comp Immunol ; 77: 69-76, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-28743432

RESUMEN

Eukaryotic Initiation Factor 6 (eIF6) is required for 60S ribosomal subunit biogenesis and efficient initiation of translation. Intriguingly, in both mice and humans, endogenous levels of eIF6 are detrimental as they act as tumor and obesity facilitators, raising the question on the evolutionary pressure that maintains high eIF6 levels. Here we show that, in mice and humans, high levels of eIF6 are required for proper immune functions. First, eIF6 heterozygous (het) mice show an increased mortality during viral infection and a reduction of peripheral blood CD4+ Effector Memory T cells. In human CD4+ T cells, eIF6 levels rapidly increase upon T-cell receptor activation and drive the glycolytic switch and the acquisition of effector functions. Importantly, in CD4+ T cells, eIF6 levels control interferon-γ (IFN-γ) secretion without affecting proliferation. In conclusion, the immune system has a high evolutionary pressure for the maintenance of a dynamic and powerful regulation of the translational machinery.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Factores Eucarióticos de Iniciación/metabolismo , Factores de Iniciación de Péptidos/metabolismo , Receptores de Antígenos de Linfocitos T/metabolismo , Virosis/inmunología , Animales , Células Cultivadas , Factores Eucarióticos de Iniciación/genética , Glucólisis , Homeostasis , Humanos , Sistema Inmunológico , Memoria Inmunológica , Interferón gamma/metabolismo , Activación de Linfocitos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Factores de Iniciación de Péptidos/genética , Transducción de Señal
16.
Cell Rep ; 21(6): 1507-1520, 2017 Nov 07.
Artículo en Inglés | MEDLINE | ID: mdl-29117557

RESUMEN

Regular endurance training improves muscle oxidative capacity and reduces the risk of age-related disorders. Understanding the molecular networks underlying this phenomenon is crucial. Here, by exploiting the power of computational modeling, we show that endurance training induces profound changes in gene regulatory networks linking signaling and selective control of translation to energy metabolism and tissue remodeling. We discovered that knockdown of the mTOR-independent factor Eif6, which we predicted to be a key regulator of this process, affects mitochondrial respiration efficiency, ROS production, and exercise performance. Our work demonstrates the validity of a data-driven approach to understanding muscle homeostasis.


Asunto(s)
Factores Eucarióticos de Iniciación/metabolismo , Ejercicio Físico , Músculo Esquelético/metabolismo , Acetilación , Animales , Calorimetría , Cromatografía Líquida de Alta Presión , Regulación hacia Abajo , Metabolismo Energético/fisiología , Factores Eucarióticos de Iniciación/deficiencia , Factores Eucarióticos de Iniciación/genética , Redes Reguladoras de Genes , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mitocondrias/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Oxígeno/metabolismo , Condicionamiento Físico Animal , Proteoma/análisis , Especies Reactivas de Oxígeno/metabolismo , Ribosomas/metabolismo , Espectrometría de Masas en Tándem , Transcripción Genética , Regulación hacia Arriba
17.
Oncotarget ; 6(35): 37471-85, 2015 Nov 10.
Artículo en Inglés | MEDLINE | ID: mdl-26462016

RESUMEN

eIF6 is an antiassociation factor that regulates the availability of active 80S. Its activation is driven by the RACK1/PKCß axis, in a mTORc1 independent manner. We previously described that eIF6 haploinsufficiency causes a striking survival in the Eµ-Myc mouse lymphoma model, with lifespans extended up to 18 months. Here we screen for eIF6 expression in human cancers. We show that Malignant Pleural Mesothelioma tumors (MPM) and a MPM cell line (REN cells) contain high levels of hyperphosphorylated eIF6. Enzastaurin is a PKC beta inhibitor used in clinical trials. We prove that Enzastaurin treatment decreases eIF6 phosphorylation rate, but not eIF6 protein stability. The growth of REN, in vivo, and metastasis are reduced by either Enzastaurin treatment or eIF6 shRNA. Molecular analysis reveals that eIF6 manipulation affects the metabolic status of malignant mesothelioma cells. Less glycolysis and less ATP content are evident in REN cells depleted for eIF6 or treated with Enzastaurin (Anti-Warburg effect). We propose that eIF6 is necessary for malignant mesothelioma growth, in vivo, and can be targeted by kinase inhibitors.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Proliferación Celular , Factores Eucarióticos de Iniciación/metabolismo , Mesotelioma/metabolismo , Neoplasias Pleurales/metabolismo , Adenosina Trifosfato/metabolismo , Animales , Biomarcadores de Tumor/genética , Línea Celular Tumoral , Factores Eucarióticos de Iniciación/genética , Regulación Neoplásica de la Expresión Génica , Glucólisis , Humanos , Indoles/farmacología , Mesotelioma/genética , Mesotelioma/patología , Mesotelioma/terapia , Ratones Endogámicos NOD , Ratones SCID , Fosforilación , Neoplasias Pleurales/genética , Neoplasias Pleurales/patología , Neoplasias Pleurales/terapia , Proteína Quinasa C beta/antagonistas & inhibidores , Proteína Quinasa C beta/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Estabilidad Proteica , Interferencia de ARN , Tratamiento con ARN de Interferencia , Transducción de Señal , Factores de Tiempo , Transfección , Carga Tumoral , Regulación hacia Arriba , Ensayos Antitumor por Modelo de Xenoinjerto
18.
Nat Commun ; 6: 8261, 2015 Sep 18.
Artículo en Inglés | MEDLINE | ID: mdl-26383020

RESUMEN

Insulin regulates glycaemia, lipogenesis and increases mRNA translation. Cells with reduced eukaryotic initiation factor 6 (eIF6) do not increase translation in response to insulin. The role of insulin-regulated translation is unknown. Here we show that reduction of insulin-regulated translation in mice heterozygous for eIF6 results in normal glycaemia, but less blood cholesterol and triglycerides. eIF6 controls fatty acid synthesis and glycolysis in a cell autonomous fashion. eIF6 acts by exerting translational control of adipogenic transcription factors like C/EBPß, C/EBPδ and ATF4 that have G/C rich or uORF sequences in their 5' UTR. The outcome of the translational activation by eIF6 is a reshaping of gene expression with increased levels of lipogenic and glycolytic enzymes. Finally, eIF6 levels modulate histone acetylation and amounts of rate-limiting fatty acid synthase (Fasn) mRNA. Since obesity, type 2 diabetes, and cancer require a Fasn-driven lipogenic state, we propose that eIF6 could be a therapeutic target for these diseases.


Asunto(s)
Resistencia a la Insulina/genética , Metabolismo de los Lípidos/genética , Factores de Iniciación de Péptidos/genética , Biosíntesis de Proteínas/genética , ARN Mensajero/metabolismo , Transcripción Genética/genética , Células 3T3 , Acetilación , Factor de Transcripción Activador 4/genética , Factor de Transcripción Activador 4/metabolismo , Adipocitos/metabolismo , Adipogénesis/genética , Animales , Western Blotting , Proteína beta Potenciadora de Unión a CCAAT/genética , Proteína beta Potenciadora de Unión a CCAAT/metabolismo , Proteína delta de Unión al Potenciador CCAAT/genética , Proteína delta de Unión al Potenciador CCAAT/metabolismo , Electroforesis en Gel de Poliacrilamida , Ácido Graso Sintasas/genética , Ácido Graso Sintasas/metabolismo , Ácidos Grasos , Regulación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Glucosa/metabolismo , Prueba de Tolerancia a la Glucosa , Glucógeno/metabolismo , Glucólisis/genética , Células HEK293 , Hepatocitos/metabolismo , Código de Histonas , Humanos , Ácido Láctico/metabolismo , Lipogénesis/genética , Hígado/diagnóstico por imagen , Hígado/metabolismo , Células Madre Mesenquimatosas , Ratones , Oxidación-Reducción , Factores de Iniciación de Péptidos/metabolismo , Radiografía , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
19.
Cell Cycle ; 10(20): 3441-6, 2011 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-22031223

RESUMEN

Ribosome biogenesis and translation can be simplified as the processes of generating ribosomes and their use for decoding mRNA into a protein. Ribosome biogenesis has been efficiently studied in unicellular organisms like the budding yeast, allowing us a deep and basic knowledge of this process in growing cells. Translation has been modeled in vitro and in unicellular organisms. These studies have given us an important insight into the mechanisms and evolutionarily conserved aspects of ribosome biology. However, we advocate the need of the direct study of these processes in multicellular organisms. Analysis of ribosome biogenesis and translation in vivo in Metazoa and mammalian models is emerging and unveils the unexpected consequences of perturbed ribosome biogenesis and translation. Here, we will describe how one factor, eIF6, plays a crucial role both in the generation of the large ribosomal subunit and its availability for translation. From there, we will make specific conclusions on the physiological relevance of eIF6 in 80S formation, cell cycle progression and disease, raising the point that the control of gene expression may occur at the unexpected level of the large ribosomal subunit. In the future, the modulation of eIF6 binding to the 60S may be pharmacologically exploited to reduce the growth of cancer cells or ameliorate the phenotype of SDS syndrome.


Asunto(s)
Enfermedades de la Médula Ósea/metabolismo , Ciclo Celular/fisiología , Transformación Celular Neoplásica/metabolismo , Factores Eucarióticos de Iniciación/metabolismo , Insuficiencia Pancreática Exocrina/metabolismo , Regulación de la Expresión Génica/fisiología , Lipomatosis/metabolismo , Biosíntesis de Proteínas/fisiología , Subunidades Ribosómicas Grandes/fisiología , Animales , Humanos , Modelos Biológicos , Subunidades Ribosómicas Grandes/metabolismo , Síndrome de Shwachman-Diamond , Transducción de Señal/fisiología
20.
PLoS One ; 6(12): e29136, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-22216185

RESUMEN

Initiation is the rate-limiting phase of protein synthesis, controlled by signaling pathways regulating the phosphorylation of translation factors. Initiation has three steps, 43S, 48S and 80S formation. 43S formation is repressed by eIF2α phosphorylation. The subsequent steps, 48S and 80S formation are enabled by growth factors. 48S relies on eIF4E-mediated assembly of eIF4F complex; 4E-BPs competitively displace eIF4E from eIF4F. Two pathways control eIF4F: 1) mTORc1 phosphorylates and inactivates 4E-BPs, leading to eIF4F formation; 2) the Ras-Mnk cascade phosphorylates eIF4E. We show that REN and NCI-H28 mesothelioma cells have constitutive activation of both pathways and maximal translation rate, in the absence of exogenous growth factors. Translation is rapidly abrogated by phosphorylation of eIF2α. Surprisingly, pharmacological inhibition of mTORc1 leads to the complete dephosphorylation of downstream targets, without changes in methionine incorporation. In addition, the combined administration of mTORc1 and MAPK/Mnk inhibitors has no additive effect. The inhibition of both mTORc1 and mTORc2 does not affect the metabolic rate. In spite of this, mTORc1 inhibition reduces eIF4F complex formation, and depresses translocation of TOP mRNAs on polysomes. Downregulation of eIF4E and overexpression of 4E-BP1 induce rapamycin sensitivity, suggesting that disruption of eIF4F complex, due to eIF4E modulation, competes with its recycling to ribosomes. These data suggest the existence of a dynamic equilibrium in which eIF4F is not essential for all mRNAs and is not displaced from translated mRNAs, before recycling to the next.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Factor 4E Eucariótico de Iniciación/metabolismo , Fosfoproteínas/metabolismo , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Proteínas de Ciclo Celular , Línea Celular Tumoral , Humanos , Metionina/metabolismo , Fosforilación , Unión Proteica , ARN Mensajero/metabolismo , Sirolimus/farmacología , Serina-Treonina Quinasas TOR/metabolismo , Transcripción Genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA