Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 83
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 154(3): 651-63, 2013 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-23911327

RESUMEN

Vessel sprouting by migrating tip and proliferating stalk endothelial cells (ECs) is controlled by genetic signals (such as Notch), but it is unknown whether metabolism also regulates this process. Here, we show that ECs relied on glycolysis rather than on oxidative phosphorylation for ATP production and that loss of the glycolytic activator PFKFB3 in ECs impaired vessel formation. Mechanistically, PFKFB3 not only regulated EC proliferation but also controlled the formation of filopodia/lamellipodia and directional migration, in part by compartmentalizing with F-actin in motile protrusions. Mosaic in vitro and in vivo sprouting assays further revealed that PFKFB3 overexpression overruled the pro-stalk activity of Notch, whereas PFKFB3 deficiency impaired tip cell formation upon Notch blockade, implying that glycolysis regulates vessel branching.


Asunto(s)
Células Endoteliales/metabolismo , Glucólisis , Neovascularización Fisiológica , Fosfofructoquinasa-2/metabolismo , Animales , Línea Celular Tumoral , Células Cultivadas , Células Endoteliales/citología , Femenino , Eliminación de Gen , Silenciador del Gen , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Fosfofructoquinasa-2/genética , Seudópodos/metabolismo , Pez Cebra
2.
Int J Cancer ; 153(9): 1671-1683, 2023 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-37497753

RESUMEN

Breast cancer is composed of metabolically coupled cellular compartments with upregulation of TP53 Induced Glycolysis and Apoptosis Regulator (TIGAR) in carcinoma cells and loss of caveolin 1 (CAV1) with upregulation of monocarboxylate transporter 4 (MCT4) in fibroblasts. The mechanisms that drive metabolic coupling are poorly characterized. The effects of TIGAR on fibroblast CAV1 and MCT4 expression and breast cancer aggressiveness was studied using coculture and conditioned media systems and in-vivo. Also, the role of cytokines in promoting tumor metabolic coupling via MCT4 on cancer aggressiveness was studied. TIGAR downregulation in breast carcinoma cells reduces tumor growth. TIGAR overexpression in carcinoma cells drives MCT4 expression and NFkB activation in fibroblasts. IL6 and TGFB drive TIGAR upregulation in carcinoma cells, reduce CAV1 and increase MCT4 expression in fibroblasts. Tumor growth is abrogated in the presence of MCT4 knockout fibroblasts and environment. We discovered coregulation of c-MYC and TIGAR in carcinoma cells driven by lactate. Metabolic coupling primes the tumor microenvironment allowing for production, uptake and utilization of lactate. In sum, aggressive breast cancer is dependent on metabolic coupling.


Asunto(s)
Neoplasias de la Mama , Carcinoma , Humanos , Femenino , Neoplasias de la Mama/patología , Proteínas Reguladoras de la Apoptosis/metabolismo , Glucólisis , Ácido Láctico/metabolismo , FN-kappa B/metabolismo , Apoptosis , Línea Celular Tumoral , Microambiente Tumoral , Proteína p53 Supresora de Tumor/metabolismo
3.
Int J Mol Sci ; 23(3)2022 Feb 08.
Artículo en Inglés | MEDLINE | ID: mdl-35163828

RESUMEN

Hyperactivation of the KEAP1-NRF2 axis is a common molecular trait in carcinomas from different origin. The transcriptional program induced by NRF2 involves antioxidant and metabolic genes that render cancer cells more capable of dealing with oxidative stress. The TP53-Induced Glycolysis and Apoptosis Regulator (TIGAR) is an important regulator of glycolysis and the pentose phosphate pathway that was described as a p53 response gene, yet TIGAR expression is detected in p53-null tumors. In this study we investigated the role of NRF2 in the regulation of TIGAR in human carcinoma cell lines. Exposure of carcinoma cells to electrophilic molecules or overexpression of NRF2 significantly increased expression of TIGAR, in parallel to the known NRF2 target genes NQO1 and G6PD. The same was observed in TP53KO cells, indicating that NRF2-mediated regulation of TIGAR is p53-independent. Accordingly, downregulation of NRF2 decreased the expression of TIGAR in carcinoma cell lines from different origin. As NRF2 is essential in the bone, we used mouse primary osteoblasts to corroborate our findings. The antioxidant response elements for NRF2 binding to the promoter of human and mouse TIGAR were described. This study provides the first evidence that NRF2 controls the expression of TIGAR at the transcriptional level.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/genética , Factor 2 Relacionado con NF-E2/metabolismo , Neoplasias/genética , Osteoblastos/citología , Monoéster Fosfórico Hidrolasas/genética , Proteína p53 Supresora de Tumor/genética , Células A549 , Animales , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica , Técnicas de Silenciamiento del Gen , Glucosafosfato Deshidrogenasa/genética , Células HCT116 , Células HeLa , Humanos , Ratones , NAD(P)H Deshidrogenasa (Quinona)/genética , Neoplasias/metabolismo , Osteoblastos/metabolismo , Cultivo Primario de Células , Regiones Promotoras Genéticas
4.
Gastroenterology ; 159(1): 273-288, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32169429

RESUMEN

BACKGROUND & AIMS: We investigated mechanisms of hepatic stellate cell (HSC) activation, which contributes to liver fibrogenesis. We aimed to determine whether activated HSCs increase glycolysis, which is regulated by 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase-3 (PFKFB3), and whether this pathway might serve as a therapeutic target. METHODS: We performed studies with primary mouse HSCs, human LX2 HSCs, human cirrhotic liver tissues, rats and mice with liver fibrosis (due to bile duct ligation [BDL] or administration of carbon tetrachloride), and CPEB4-knockout mice. Glycolysis was inhibited in cells and mice by administration of a small molecule antagonist of PFKFB3 (3-[3-pyridinyl]-1-[4-pyridinyl]-2-propen-1-one [3PO]). Cells were transfected with small interfering RNAs that knock down PFKFB3 or CPEB4. RESULTS: Up-regulation of PFKFB3 protein and increased glycolysis were early and sustained events during HSC activation and accompanied by increased expression of markers of fibrogenesis; incubation of HSCs with 3PO or knockdown of PFKFB3 reduced their activation and proliferation. Mice with liver fibrosis after BDL had increased hepatic PFKFB3; injection of 3PO immediately after the surgery prevented HSC activation and reduced the severity of liver fibrosis compared with mice given vehicle. Levels of PFKFB3 protein were increased in fibrotic liver tissues from patients compared with non-fibrotic liver. Up-regulation of PFKFB3 in activated HSCs did not occur via increased transcription, but instead via binding of CPEB4 to cytoplasmic polyadenylation elements within the 3'-untranslated regions of PFKFB3 messenger RNA. Knockdown of CPEB4 in LX2 HSCs prevented PFKFB3 overexpression and cell activation. Livers from CPEB4-knockout had decreased PFKFB3 and fibrosis after BDL or administration of carbon tetrachloride compared with wild-type mice. CONCLUSIONS: Fibrotic liver tissues from patients and rodents (mice and rats) have increased levels of PFKFB3 and glycolysis, which are essential for activation of HSCs. Increased expression of PFKFB3 is mediated by binding of CPEB4 to its untranslated messenger RNA. Inhibition or knockdown of CPEB4 or PFKFB3 prevents HSC activation and fibrogenesis in livers of mice.


Asunto(s)
Células Estrelladas Hepáticas/patología , Cirrosis Hepática Experimental/patología , Cirrosis Hepática/patología , Fosfofructoquinasa-2/metabolismo , Proteínas de Unión al ARN/metabolismo , Animales , Tetracloruro de Carbono/toxicidad , Línea Celular , Regulación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Glucólisis , Humanos , Hígado/citología , Hígado/patología , Cirrosis Hepática Experimental/inducido químicamente , Cirrosis Hepática Experimental/genética , Masculino , Ratones , Ratones Noqueados , Fosfofructoquinasa-2/genética , Cultivo Primario de Células , Proteínas de Unión al ARN/genética , Ratas , Regulación hacia Arriba
5.
FASEB J ; 34(9): 11816-11837, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32666604

RESUMEN

The prevalence of nonalcoholic fatty liver disease (NAFLD) has increased drastically due to the global obesity pandemic but at present there are no approved therapies. Here, we aimed to revert high-fat diet (HFD)-induced obesity and NAFLD in mice by enhancing liver fatty acid oxidation (FAO). Moreover, we searched for potential new lipid biomarkers for monitoring liver steatosis in humans. We used adeno-associated virus (AAV) to deliver a permanently active mutant form of human carnitine palmitoyltransferase 1A (hCPT1AM), the key enzyme in FAO, in the liver of a mouse model of HFD-induced obesity and NAFLD. Expression of hCPT1AM enhanced hepatic FAO and autophagy, reduced liver steatosis, and improved glucose homeostasis. Lipidomic analysis in mice and humans before and after therapeutic interventions, such as hepatic AAV9-hCPT1AM administration and RYGB surgery, respectively, led to the identification of specific triacylglyceride (TAG) specie (C50:1) as a potential biomarker to monitor NAFFLD disease. To sum up, here we show for the first time that liver hCPT1AM gene therapy in a mouse model of established obesity, diabetes, and NAFLD can reduce HFD-induced derangements. Moreover, our study highlights TAG (C50:1) as a potential noninvasive biomarker that might be useful to monitor NAFLD in mice and humans.


Asunto(s)
Biomarcadores/metabolismo , Carnitina O-Palmitoiltransferasa/metabolismo , Ácidos Grasos/metabolismo , Terapia Genética/métodos , Metabolismo de los Lípidos , Hígado/metabolismo , Enfermedad del Hígado Graso no Alcohólico/terapia , Animales , Carnitina O-Palmitoiltransferasa/genética , Diabetes Mellitus/etiología , Diabetes Mellitus/metabolismo , Dieta Alta en Grasa/efectos adversos , Modelos Animales de Enfermedad , Humanos , Hígado/patología , Masculino , Ratones Endogámicos C57BL , Enfermedad del Hígado Graso no Alcohólico/etiología , Enfermedad del Hígado Graso no Alcohólico/genética , Obesidad/etiología , Obesidad/metabolismo , Oxidación-Reducción , Triglicéridos/metabolismo
6.
Int J Mol Sci ; 22(14)2021 Jul 11.
Artículo en Inglés | MEDLINE | ID: mdl-34299056

RESUMEN

The glycolytic modulator TP53-Inducible Glycolysis and Apoptosis Regulator (TIGAR) is overexpressed in several types of cancer and has a role in metabolic rewiring during tumor development. However, little is known about the role of this enzyme in proliferative tissues under physiological conditions. In the current work, we analysed the role of TIGAR in primary human lymphocytes stimulated with the mitotic agent Concanavalin A (ConA). We found that TIGAR expression was induced in stimulated lymphocytes through the PI3K/AKT pathway, since Akti-1/2 and LY294002 inhibitors prevented the upregulation of TIGAR in response to ConA. In addition, suppression of TIGAR expression by siRNA decreased the levels of the proliferative marker PCNA and increased cellular ROS levels. In this model, TIGAR was found to support the activity of glucose 6-phosphate dehydrogenase (G6PDH), the first enzyme of the pentose phosphate pathway (PPP), since the inhibition of TIGAR reduced G6PDH activity and increased autophagy. In conclusion, we demonstrate here that TIGAR is upregulated in stimulated human lymphocytes through the PI3K/AKT signaling pathway, which contributes to the redirection of the carbon flux to the PPP.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/metabolismo , Concanavalina A/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Linfocitos/metabolismo , Mitógenos/farmacología , Fosfatidilinositol 3-Quinasas/química , Monoéster Fosfórico Hidrolasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Apoptosis , Proteínas Reguladoras de la Apoptosis/genética , Autofagia , Glucólisis , Humanos , Linfocitos/efectos de los fármacos , Vía de Pentosa Fosfato , Monoéster Fosfórico Hidrolasas/genética , Transducción de Señal
7.
Mol Cell Biochem ; 448(1-2): 187-197, 2018 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-29435871

RESUMEN

Lymphocyte activation is associated with rapid increase of both the glycolytic activator fructose 2,6-bisphosphate (Fru-2,6-P2) and the enzyme responsible for its synthesis, 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase (PFK-2/FBPase-2). PFKFB3 gene, which encodes for the most abundant PFK-2 isoenzyme in proliferating tissues, has been found overexpressed during cell activation in several models, including immune cells. However, there is limited knowledge on the pathways underlying PFKFB3 regulation in human T-lymphocytes, and the role of this gene in human immune response. The aim of this work is to elucidate the molecular mechanisms of PFKFB3 induction during human T-lymphocyte activation by mitotic agents. The results obtained showed PFKFB3 induction during human T-lymphocyte activation by mitogens such as phytohemagglutinin (PHA). PFKFB3 increase occurred concomitantly with GLUT-1, HK-II, and PCNA upregulation, showing that mitotic agents induce a metabolic reprograming process that is required for T-cell proliferation. PI3K-Akt pathway inhibitors, Akti-1/2 and LY294002, reduced PFKFB3 gene induction by PHA, as well as Fru-2,6-P2 and lactate production. Moreover, both inhibitors blocked activation and proliferation in response to PHA, showing the importance of PI3K/Akt signaling pathway in the antigen response of T-lymphocytes. These results provide a link between metabolism and T-cell antigen receptor signaling in human lymphocyte biology that can help to better understand the importance of modulating both pathways to target complex diseases involving the activation of the immune system.


Asunto(s)
Regulación de la Expresión Génica/inmunología , Activación de Linfocitos , Fosfatidilinositol 3-Quinasas/inmunología , Fosfofructoquinasa-2/inmunología , Proteínas Proto-Oncogénicas c-akt/inmunología , Transducción de Señal/inmunología , Linfocitos T/inmunología , Proliferación Celular/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Fitohemaglutininas/farmacología , Transducción de Señal/efectos de los fármacos , Linfocitos T/citología
8.
J Biol Chem ; 291(51): 26291-26303, 2016 Dec 16.
Artículo en Inglés | MEDLINE | ID: mdl-27803158

RESUMEN

A subgroup of breast cancers has several metabolic compartments. The mechanisms by which metabolic compartmentalization develop in tumors are poorly characterized. TP53 inducible glycolysis and apoptosis regulator (TIGAR) is a bisphosphatase that reduces glycolysis and is highly expressed in carcinoma cells in the majority of human breast cancers. Hence we set out to determine the effects of TIGAR expression on breast carcinoma and fibroblast glycolytic phenotype and tumor growth. The overexpression of this bisphosphatase in carcinoma cells induces expression of enzymes and transporters involved in the catabolism of lactate and glutamine. Carcinoma cells overexpressing TIGAR have higher oxygen consumption rates and ATP levels when exposed to glutamine, lactate, or the combination of glutamine and lactate. Coculture of TIGAR overexpressing carcinoma cells and fibroblasts compared with control cocultures induce more pronounced glycolytic differences between carcinoma and fibroblast cells. Carcinoma cells overexpressing TIGAR have reduced glucose uptake and lactate production. Conversely, fibroblasts in coculture with TIGAR overexpressing carcinoma cells induce HIF (hypoxia-inducible factor) activation with increased glucose uptake, increased 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase-3 (PFKFB3), and lactate dehydrogenase-A expression. We also studied the effect of this enzyme on tumor growth. TIGAR overexpression in carcinoma cells increases tumor growth in vivo with increased proliferation rates. However, a catalytically inactive variant of TIGAR did not induce tumor growth. Therefore, TIGAR expression in breast carcinoma cells promotes metabolic compartmentalization and tumor growth with a mitochondrial metabolic phenotype with lactate and glutamine catabolism. Targeting TIGAR warrants consideration as a potential therapy for breast cancer.


Asunto(s)
Neoplasias de la Mama/metabolismo , Ácido Glutámico/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Ácido Láctico/metabolismo , Apoptosis/genética , Proteínas Reguladoras de la Apoptosis , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Técnicas de Cocultivo , Femenino , Fibroblastos/metabolismo , Fibroblastos/patología , Ácido Glutámico/genética , Glucólisis/genética , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Isoenzimas/genética , Isoenzimas/metabolismo , L-Lactato Deshidrogenasa/genética , L-Lactato Deshidrogenasa/metabolismo , Lactato Deshidrogenasa 5 , Células MCF-7 , Fosfofructoquinasa-2/genética , Fosfofructoquinasa-2/metabolismo , Monoéster Fosfórico Hidrolasas , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo
9.
Am J Physiol Endocrinol Metab ; 310(6): E440-51, 2016 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-26714846

RESUMEN

Neuregulin (NRG) is an EGF-related growth factor that binds to the tyrosine kinase receptors ErbB3 and ErbB4, thus inducing tissue development and muscle glucose utilization during contraction. Here, we analyzed whether NRG has systemic effects regulating glycemia in control and type 2 diabetic rats. To this end, recombinant NRG (rNRG) was injected into Zucker diabetic fatty (ZDF) rats and their respective lean littermates 15 min before a glucose tolerance test (GTT) was performed. rNRG enhanced glucose tolerance without promoting the activation of the insulin receptor (IR) or insulin receptor substrates (IRS) in muscle and liver. However, in control rats, rNRG induced the phosphorylation of protein kinase B (PKB) and glycogen synthase kinase-3 (GSK-3) in liver but not in muscle. In liver, rNRG increased ErbB3 tyrosine phosphorylation and its binding to phosphatidylinositol 3-kinase (PI3K), thus indicating that rNRG activates the ErbB3/PI3K/PKB signaling pathway. rNRG increased glycogen content in liver but not in muscle. rNRG also increased the content of fructose-2,6-bisphosphate (Fru-2,6-P2), an activator of hepatic glycolysis, and lactate in liver but not in muscle. Increases in lactate were abrogated by wortmannin, a PI3K inhibitor, in incubated hepatocytes. The liver of ZDF rats showed a reduced content of ErbB3 receptors, entailing a minor stimulation of the rNRG-induced PKB/GSK-3 cascade and resulting in unaltered hepatic glycogen content. Nonetheless, rNRG increased hepatic Fru-2,6-P2 and augmented lactate both in liver and in plasma of diabetic rats. As a whole, rNRG improved response to the GTT in both control and diabetic rats by enhancing hepatic glucose utilization.


Asunto(s)
Glucemia/efectos de los fármacos , Diabetes Mellitus Tipo 2/metabolismo , Hígado/efectos de los fármacos , Músculo Esquelético/efectos de los fármacos , Neurregulinas/farmacología , Animales , Glucemia/metabolismo , Estudios de Casos y Controles , Fructosadifosfatos/metabolismo , Glucosa/metabolismo , Prueba de Tolerancia a la Glucosa , Glucógeno Sintasa Quinasa 3/efectos de los fármacos , Glucógeno Sintasa Quinasa 3/metabolismo , Insulina , Proteínas Sustrato del Receptor de Insulina/efectos de los fármacos , Proteínas Sustrato del Receptor de Insulina/metabolismo , Ácido Láctico/metabolismo , Hígado/metabolismo , Glucógeno Hepático/metabolismo , Músculo Esquelético/metabolismo , Fosfatidilinositol 3-Quinasa/efectos de los fármacos , Fosfatidilinositol 3-Quinasa/metabolismo , Fosforilación/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas , Ratas Zucker , Receptor ErbB-3/efectos de los fármacos , Receptor ErbB-3/metabolismo , Receptor de Insulina/efectos de los fármacos , Receptor de Insulina/metabolismo
10.
FASEB J ; 29(4): 1414-25, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25550462

RESUMEN

The skeleton acts as an endocrine organ that regulates energy metabolism and calcium and phosphorous homeostasis through the secretion of osteocalcin (Oc) and fibroblast growth factor 23 (FGF23). However, evidence suggests that osteoblasts secrete additional unknown factors that contribute to the endocrine function of bone. To search for these additional factors, we generated mice with a conditional osteoblast-specific deletion of p38α MAPK known to display profound defects in bone homeostasis. Herein, we show that impaired osteoblast function is associated with a strong decrease in body weight and adiposity (P < 0.01). The differences in adiposity were not associated with diminished caloric intake, but rather reflected 20% increased energy expenditure and the up-regulation of uncoupling protein-1 (Ucp1) in white adipose tissue (WAT) and brown adipose tissue (BAT) (P < 0.05). These alterations in lipid metabolism and energy expenditure were correlated with a decrease in the blood levels of neuropeptide Y (NPY) (40% lower) rather than changes in the serum levels of insulin, Oc, or FGF23. Among all Npy-expressing tissues, only bone and primary osteoblasts showed a decline in Npy expression (P < 0.01). Moreover, the intraperitoneal administration of recombinant NPY partially restored the WAT weight and adipocyte size of p38α-deficient mice (P < 0.05). Altogether, these results further suggest that, in addition to Oc, other bone-derived signals affect WAT and energy expenditure contributing to the regulation of energy metabolism.


Asunto(s)
Tejido Adiposo/metabolismo , Proteína Quinasa 14 Activada por Mitógenos/metabolismo , Osteoblastos/enzimología , Adipocitos/citología , Adipocitos/metabolismo , Tejido Adiposo Pardo/metabolismo , Tejido Adiposo Blanco/metabolismo , Adiposidad , Animales , Peso Corporal , Desarrollo Óseo , Tamaño de la Célula , Metabolismo Energético , Femenino , Factor-23 de Crecimiento de Fibroblastos , Regulación de la Expresión Génica , Homeostasis , Canales Iónicos/genética , Metabolismo de los Lípidos , Masculino , Ratones , Ratones Noqueados , Proteínas Mitocondriales/genética , Proteína Quinasa 14 Activada por Mitógenos/deficiencia , Proteína Quinasa 14 Activada por Mitógenos/genética , Neuropéptido Y/sangre , Neuropéptido Y/genética , Obesidad/enzimología , Obesidad/prevención & control , Osteocalcina/metabolismo , Embarazo , Transducción de Señal , Proteína Desacopladora 1 , Regulación hacia Arriba
11.
Arterioscler Thromb Vasc Biol ; 35(6): 1463-71, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25882065

RESUMEN

OBJECTIVE: Although it is accepted that macrophage glycolysis is upregulated under hypoxic conditions, it is not known whether this is linked to a similar increase in macrophage proinflammatory activation and whether specific energy demands regulate cell viability in the atheromatous plaque. APPROACH AND RESULTS: We studied the interplay between macrophage energy metabolism, polarization, and viability in the context of atherosclerosis. Cultured human and murine macrophages and an in vivo murine model of atherosclerosis were used to evaluate the mechanisms underlying metabolic and inflammatory activity of macrophages in the different atherosclerotic conditions analyzed. We observed that macrophage energetics and inflammatory activation are closely and linearly related, resulting in dynamic calibration of glycolysis to keep pace with inflammatory activity. In addition, we show that macrophage glycolysis and proinflammatory activation mainly depend on hypoxia-inducible factor and on its impact on glucose uptake, and on the expression of hexokinase II and ubiquitous 6-phosphofructo-2-kinase. As a consequence, hypoxia potentiates inflammation and glycolysis mainly via these pathways. Moreover, when macrophages' ability to increase glycolysis through 6-phosphofructo-2-kinase is experimentally attenuated, cell viability is reduced if subjected to proinflammatory or hypoxic conditions, but unaffected under control conditions. In addition to this, granulocyte-macrophage colony-stimulating factor enhances anerobic glycolysis while exerting a mild proinflammatory activation. CONCLUSIONS: These findings, in human and murine cells and in an animal model, show that hypoxia potentiates macrophage glycolytic flux in concert with a proportional upregulation of proinflammatory activity, in a manner that is dependent on both hypoxia-inducible factor -1α and 6-phosphofructo-2-kinase.


Asunto(s)
Aterosclerosis/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Macrófagos/metabolismo , Fosfofructoquinasa-2/metabolismo , Animales , Hipoxia de la Célula , Modelos Animales de Enfermedad , Glucólisis , Humanos , Inflamación/metabolismo , Ratones , Factor de Necrosis Tumoral alfa/metabolismo
12.
Anal Bioanal Chem ; 408(6): 1715-9, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26753978

RESUMEN

Here we report a new approach for studying protein oligomerization in cells using a single electrophoresis gel. We combined the use of a crosslinking reagent for sample preparation, such as glutaraldehyde, with the analysis of oligomers by Tris-acetate polyacrylamide gel electrophoresis. The use of a 3-15% Tris-acetate polyacrylamide gradient gel allows for the simultaneous analysis of proteins of masses ranging from 10 to 500 kDa. We showed the usefulness of this method for analyzing endogenous p53 oligomerization with high resolution and sensitivity in human cells. Oligomerization analysis was dependent on the crosslinker concentration used. We also showed that this method could be used to study the regulation of oligomerization. In all experiments, Tris-acetate polyacrylamide gel electrophoresis proved to be a robust, manageable, and cost- and time-efficient method that provided excellent results using a single gel. This approach can be easily extrapolated to the study of other oligomers. All of these features make this method a highly useful tool for the analysis of protein oligomerization.


Asunto(s)
Electroforesis en Gel de Poliacrilamida/métodos , Proteínas/análisis , Acetatos/química , Bleomicina/farmacología , Línea Celular , Reactivos de Enlaces Cruzados/química , Doxorrubicina/farmacología , Glutaral/química , Humanos , Peso Molecular , Multimerización de Proteína/efectos de los fármacos , Proteínas/metabolismo , Proteína p53 Supresora de Tumor/análisis , Proteína p53 Supresora de Tumor/metabolismo
13.
J Biol Chem ; 289(21): 14782-95, 2014 May 23.
Artículo en Inglés | MEDLINE | ID: mdl-24722987

RESUMEN

The tumor suppressor p53 is a transcription factor that coordinates the cellular response to several kinds of stress. p53 inactivation is an important step in tumor progression. Oligomerization of p53 is critical for its posttranslational modification and its ability to regulate the transcription of target genes necessary to inhibit tumor growth. Here we report that the HECT E3 ubiquitin ligase HERC2 interacts with p53. This interaction involves the CPH domain of HERC2 (a conserved domain within Cul7, PARC, and HERC2 proteins) and the last 43 amino acid residues of p53. Through this interaction, HERC2 regulates p53 activity. RNA interference experiments showed how HERC2 depletion reduces the transcriptional activity of p53 without affecting its stability. This regulation of p53 activity by HERC2 is independent of proteasome or MDM2 activity. Under these conditions, up-regulation of cell growth and increased focus formation were observed, showing the functional relevance of the HERC2-p53 interaction. This interaction was maintained after DNA damage caused by the chemotherapeutic drug bleomycin. In these stressed cells, p53 phosphorylation was not impaired by HERC2 knockdown. Interestingly, p53 mutations that affect its tetramerization domain disrupted the HERC2-p53 interaction, suggesting a role for HERC2 in p53 oligomerization. This regulatory role was shown using cross-linking assays. Thus, the inhibition of p53 activity after HERC2 depletion can be attributed to a reduction in p53 oligomerization. Ectopic expression of HERC2 (residues 2292-2923) confirmed these observations. Together, these results identify HERC2 as a novel regulator of p53 signaling.


Asunto(s)
Multimerización de Proteína , Proteína p53 Supresora de Tumor/química , Proteína p53 Supresora de Tumor/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Línea Celular Tumoral , Daño del ADN , Expresión Génica , Células HEK293 , Células HeLa , Humanos , Immunoblotting , Mutación , Fosforilación , Unión Proteica , Interferencia de ARN , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteína p53 Supresora de Tumor/genética , Ubiquitina-Proteína Ligasas/genética
14.
Lasers Surg Med ; 47(9): 765-72, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26414998

RESUMEN

BACKGROUND AND OBJECTIVES: As the population ages, osteometabolic diseases and osteoporotic fractures emerge, resulting in substantial healthcare resource utilization and impaired quality of life. Many types of mechanical stimulation have the potential of being recognized by bone cells after a mechanical sign is transformed into a biological one (a process called mechanotransduction). The therapeutic ultrasound (TU) is one of several resources capable of promoting bone cell mechanical stimulation. Therefore, the main purpose of present study was to evaluate the effect of TU on the proliferation of pre-osteoblasts using in vitro bioassays. STUDY DESIGN/MATERIALS AND METHODS: We used MC3T3-E1 pre-osteoblast lineage cells kept in Alpha medium. Cells were treated using pulsed mode therapeutic ultrasound, with frequency of 1 MHz, intensity of 0.2 W/cm(2) (SATA), duty cycle of 20%, for 30 minutes. Nifedipine and rapamycin were used to further investigate the role of L-type Ca(2+) channels and mTOR pathway. Intracellular calcium, TGF-ß1, magnesium, and the mRNA levels of osteopontin, osteonectin, NF-κB1, p38α were evaluated. RESULTS: The results show that TU stimulates the growth of MC3T3-E1 cells and decreases the supernatant calcium and magnesium content. Also, it increases intracellular calcium, activates NF-κB1 and mTOR complex via p38α. Moreover, TU promoted a decrease in the TGF-ß1 synthesis, which is a cell growth inhibitor. CONCLUSIONS: Therapeutic ultrasound, with frequency of 1 MHz, intensity of 0.2 W/cm(2) (SATA) and pulsed mode, for 30 minutes, was able to increase the proliferation of preosteoblast-like bone cells. This effect was mediated by a calcium influx, with a consequent activation of the mTOR pathway, through increased NF-κB1 and p38α.


Asunto(s)
Proliferación Celular/efectos de la radiación , Proteína Quinasa 14 Activada por Mitógenos/fisiología , FN-kappa B/fisiología , Osteoblastos/efectos de la radiación , Serina-Treonina Quinasas TOR/fisiología , Terapia por Ultrasonido , Células 3T3 , Animales , Técnicas de Cultivo de Célula , Diferenciación Celular , Ratones , Osteoblastos/metabolismo , Osteoblastos/patología
15.
J Biol Chem ; 288(20): 14264-14275, 2013 May 17.
Artículo en Inglés | MEDLINE | ID: mdl-23564456

RESUMEN

Osteogenesis depends on a coordinated network of signals and transcription factors such as Runx2 and Osterix. Recent evidence indicates that microRNAs (miRNAs) act as important post-transcriptional regulators in a large number of processes, including osteoblast differentiation. In this study, we performed miRNA expression profiling and identified miR-322, a BMP-2-down-regulated miRNA, as a regulator of osteoblast differentiation. We report miR-322 gain- and loss-of-function experiments in C2C12 and MC3T3-E1 cells and primary cultures of murine bone marrow-derived mesenchymal stem cells. We demonstrate that overexpression of miR-322 enhances BMP-2 response, increasing the expression of Osx and other osteogenic genes. Furthermore, we identify Tob2 as a target of miR-322, and we characterize the specific Tob2 3'-UTR sequence bound by miR-322 by reporter assays. We demonstrate that Tob2 is a negative regulator of osteogenesis that binds and mediates degradation of Osx mRNA. Our results demonstrate a new molecular mechanism controlling osteogenesis through the specific miR-322/Tob2 regulation of specific target mRNAs. This regulatory circuit provides a clear example of a complex miRNA-transcription factor network for fine-tuning the osteoblast differentiation program.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Regulación de la Expresión Génica , MicroARNs/metabolismo , Osteoblastos/citología , Osteogénesis/fisiología , Factores de Transcripción/metabolismo , Células 3T3 , Animales , Secuencia de Bases , Diferenciación Celular , Línea Celular , Proliferación Celular , Perfilación de la Expresión Génica , Células HeLa , Humanos , Ratones , MicroARNs/genética , MicroARNs/fisiología , Datos de Secuencia Molecular , Estabilidad del ARN , ARN Mensajero/metabolismo , Homología de Secuencia de Ácido Nucleico , Factor de Transcripción Sp7
16.
J Biol Chem ; 288(15): 10640-51, 2013 Apr 12.
Artículo en Inglés | MEDLINE | ID: mdl-23457334

RESUMEN

Reciprocal regulation of metabolism and signaling allows cells to modulate their activity in accordance with their metabolic resources. Thus, amino acids could activate signal transduction pathways that control cell metabolism. To test this hypothesis, we analyzed the effect of amino acids on fructose-2,6-bisphosphate (Fru-2,6-P2) metabolism. We demonstrate that amino acids increase Fru-2,6-P2 concentration in HeLa and in MCF7 human cells. In conjunction with this, 6-phosphofructo-2-kinase activity, glucose uptake, and lactate concentration were increased. These data correlate with the specific phosphorylation of heart 6-phosphofructo-2-kinase/fructose-2,6-biphosphatase (PFKFB2) isoenzyme at Ser-483. This activation was mediated by the PI3K and p38 signaling pathways. Furthermore, Akt inactivation blocked PFKFB2 phosphorylation and Fru-2,6-P2 production, thereby suggesting that the above signaling pathways converge at Akt kinase. In accordance with these results, kinase assays showed that amino acid-activated Akt phosphorylated PFKFB2 at Ser-483 and that knockdown experiments confirmed that the increase in Fru-2,6-P2 concentration induced by amino acids was due to PFKFB2. In addition, similar effects on Fru-2,6-P2 metabolism were observed in freshly isolated rat cardiomyocytes treated with amino acids, which indicates that these effects are not restricted to human cancer cells. In these cardiomyocytes, the glucose consumption and the production of lactate and ATP suggest an increase of glycolytic flux. Taken together, these results demonstrate that amino acids stimulate Fru-2,6-P2 synthesis by Akt-dependent PFKFB2 phosphorylation and activation and show how signaling and metabolism are inextricably linked.


Asunto(s)
Aminoácidos/metabolismo , Glucólisis/fisiología , Miocitos Cardíacos/enzimología , Fosfofructoquinasa-2/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal/fisiología , Adenosina Trifosfato/genética , Adenosina Trifosfato/metabolismo , Aminoácidos/genética , Animales , Activación Enzimática/fisiología , Fructosadifosfatos/genética , Fructosadifosfatos/metabolismo , Glucosa/genética , Glucosa/metabolismo , Células HEK293 , Células HeLa , Humanos , Ácido Láctico/metabolismo , Masculino , Miocitos Cardíacos/citología , Fosfofructoquinasa-2/genética , Fosforilación/fisiología , Proteínas Proto-Oncogénicas c-akt/genética , Ratas , Ratas Sprague-Dawley , Serina/genética , Serina/metabolismo
17.
Biochem J ; 452(3): 531-43, 2013 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-23548149

RESUMEN

PFK-2/FBPase-2 (6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase) catalyses the synthesis and degradation of Fru-2,6-P2 (fructose 2,6-bisphosphate), a key modulator of glycolysis and gluconeogenesis. The PFKFB3 gene is involved in cell proliferation owing to its role in carbohydrate metabolism. In the present study we analysed the mechanism of regulation of PFKFB3 as an immediate early gene controlled by stress stimuli that activates the p38/MK2 [MAPK (mitogen-activated protein kinase)-activated protein kinase 2] pathway. We report that exposure of HeLa and T98G cells to different stress stimuli (NaCl, H2O2, UV radiation and anisomycin) leads to a rapid increase (15-30 min) in PFKFB3 mRNA levels. The use of specific inhibitors in combination with MK2-deficient cells implicate control by the protein kinase MK2. Transient transfection of HeLa cells with deleted gene promoter constructs allowed us to identify an SRE (serum-response element) to which SRF (serum-response factor) binds and thus transactivates PFKFB3 gene transcription. Direct binding of phospho-SRF to the SRE sequence (-918 nt) was confirmed by ChIP (chromatin immunoprecipiation) assays. Moreover, PFKFB3 isoenzyme phosphorylation at Ser461 by MK2 increases PFK-2 activity. Taken together, the results of the present study suggest a multimodal mechanism of stress stimuli affecting PFKFB3 transcriptional regulation and kinase activation by protein phosphorylation, resulting in an increase in Fru-2,6-P2 concentration and stimulation of glycolysis in cancer cells.


Asunto(s)
Sistema de Señalización de MAP Quinasas/fisiología , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Estrés Oxidativo , Fosfofructoquinasa-2/química , Fosforilación , Proteínas Quinasas p38 Activadas por Mitógenos/química , Proteínas Quinasas p38 Activadas por Mitógenos/fisiología , Secuencia de Aminoácidos , Activación Enzimática/fisiología , Glucólisis/genética , Células HeLa , Humanos , Proteínas Quinasas Activadas por Mitógenos/genética , Datos de Secuencia Molecular , Neoplasias/química , Neoplasias/genética , Neoplasias/patología , Estrés Oxidativo/genética , Fosfofructoquinasa-2/genética , Fosfofructoquinasa-2/metabolismo , Fosforilación/genética , Unión Proteica/genética , Proteínas Quinasas p38 Activadas por Mitógenos/genética
18.
Biochem J ; 442(2): 345-56, 2012 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-22115192

RESUMEN

PFKFB (6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase) catalyses the synthesis and degradation of Fru-2,6-P2 (fructose-2,6-bisphosphate), a key modulator of glycolysis and gluconeogenesis. The PFKFB3 gene is extensively involved in cell proliferation owing to its key role in carbohydrate metabolism. In the present study we analyse its mechanism of regulation by progestins in breast cancer cells. We report that exposure of T47D cells to synthetic progestins (ORG2058 or norgestrel) leads to a rapid increase in Fru-2,6-P2 concentration. Our Western blot results are compatible with a short-term activation due to PFKFB3 isoenzyme phosphorylation and a long-term sustained action due to increased PFKFB3 protein levels. Transient transfection of T47D cells with deleted gene promoter constructs allowed us to identify a PRE (progesterone-response element) to which PR (progesterone receptor) binds and thus transactivates PFKFB3 gene transcription. PR expression in the PR-negative cell line MDA-MB-231 induces endogenous PFKFB3 expression in response to norgestrel. Direct binding of PR to the PRE box (-3490 nt) was confirmed by ChIP (chromatin immunoprecipiation) experiments. A dual mechanism affecting PFKFB3 protein and gene regulation operates in order to assure glycolysis in breast cancer cells. An immediate early response through the ERK (extracellular-signal-regulated kinase)/RSK (ribosomal S6 kinase) pathway leading to phosphorylation of PFKFB3 on Ser461 is followed by activation of mRNA transcription via cis-acting sequences on the PFKFB3 promoter.


Asunto(s)
Neoplasias de la Mama/metabolismo , Fosfofructoquinasa-2/metabolismo , Congéneres de la Progesterona/farmacología , Secuencia de Bases , Neoplasias de la Mama/genética , Línea Celular Tumoral , Activación Enzimática/efectos de los fármacos , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Sistema de Señalización de MAP Quinasas , Norgestrel/farmacología , Fosfofructoquinasa-2/genética , Pregnenodionas/farmacología , Regiones Promotoras Genéticas , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Neoplásico/genética , ARN Neoplásico/metabolismo , Receptores de Progesterona/metabolismo
19.
J Biol Chem ; 286(8): 6128-42, 2011 Feb 25.
Artículo en Inglés | MEDLINE | ID: mdl-21131356

RESUMEN

The activity of mammalian target of rapamycin (mTOR) complexes regulates essential cellular processes, such as growth, proliferation, or survival. Nutrients such as amino acids are important regulators of mTOR complex 1 (mTORC1) activation, thus affecting cell growth, protein synthesis, and autophagy. Here, we show that amino acids may also activate mTOR complex 2 (mTORC2). This activation is mediated by the activity of class I PI3K and of Akt. Amino acids induced a rapid phosphorylation of Akt at Thr-308 and Ser-473. Whereas both phosphorylations were dependent on the presence of mTOR, only Akt phosphorylation at Ser-473 was dependent on the presence of rictor, a specific component of mTORC2. Kinase assays confirmed mTORC2 activation by amino acids. This signaling was functional, as demonstrated by the phosphorylation of Akt substrate FOXO3a. Interestingly, using different starvation conditions, amino acids can selectively activate mTORC1 or mTORC2. These findings identify a new signaling pathway used by amino acids underscoring the crucial importance of these nutrients in cell metabolism and offering new mechanistic insights.


Asunto(s)
Aminoácidos/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal/fisiología , Factores de Transcripción/metabolismo , Aminoácidos/farmacología , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Proteína Forkhead Box O3 , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/metabolismo , Células HEK293 , Células HeLa , Humanos , Fosfatidilinositol 3-Quinasas/genética , Fosforilación/efectos de los fármacos , Fosforilación/fisiología , Proteínas Proto-Oncogénicas c-akt/genética , Proteína Asociada al mTOR Insensible a la Rapamicina , Transducción de Señal/efectos de los fármacos , Factores de Transcripción/genética
20.
J Biol Chem ; 286(22): 19247-58, 2011 Jun 03.
Artículo en Inglés | MEDLINE | ID: mdl-21464136

RESUMEN

Macrophages activated through Toll receptor triggering increase the expression of the A(2A) and A(2B) adenosine receptors. In this study, we show that adenosine receptor activation enhances LPS-induced pfkfb3 expression, resulting in an increase of the key glycolytic allosteric regulator fructose 2,6-bisphosphate and the glycolytic flux. Using shRNA and differential expression of A(2A) and A(2B) receptors, we demonstrate that the A(2A) receptor mediates, in part, the induction of pfkfb3 by LPS, whereas the A(2B) receptor, with lower adenosine affinity, cooperates when high adenosine levels are present. pfkfb3 promoter sequence deletion analysis, site-directed mutagenesis, and inhibition by shRNAs demonstrated that HIF1α is a key transcription factor driving pfkfb3 expression following macrophage activation by LPS, whereas synergic induction of pfkfb3 expression observed with the A(2) receptor agonists seems to depend on Sp1 activity. Furthermore, levels of phospho-AMP kinase also increase, arguing for increased PFKFB3 activity by phosphorylation in long term LPS-activated macrophages. Taken together, our results show that, in macrophages, endogenously generated adenosine cooperates with bacterial components to increase PFKFB3 isozyme activity, resulting in greater fructose 2,6-bisphosphate accumulation. This process enhances the glycolytic flux and favors ATP generation helping to develop and maintain the long term defensive and reparative functions of the macrophages.


Asunto(s)
Adenosina/metabolismo , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Glucólisis/efectos de los fármacos , Lipopolisacáridos/farmacología , Macrófagos Peritoneales/enzimología , Fosfofructoquinasa-2/biosíntesis , Receptor Toll-Like 4/agonistas , Adenosina/genética , Adenosina Trifosfato/biosíntesis , Adenosina Trifosfato/genética , Secuencia de Aminoácidos , Animales , Línea Celular , Fructosadifosfatos/genética , Fructosadifosfatos/metabolismo , Regulación Enzimológica de la Expresión Génica/fisiología , Glucólisis/fisiología , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Isoenzimas/biosíntesis , Isoenzimas/genética , Activación de Macrófagos/efectos de los fármacos , Activación de Macrófagos/fisiología , Macrófagos Peritoneales/citología , Ratones , Ratones Noqueados , Mutagénesis Sitio-Dirigida , Fosfofructoquinasa-2/genética , Receptor de Adenosina A2A/genética , Receptor de Adenosina A2A/metabolismo , Receptor de Adenosina A2B , Eliminación de Secuencia , Factor de Transcripción Sp1/genética , Factor de Transcripción Sp1/metabolismo , Receptor Toll-Like 4/genética , Receptor Toll-Like 4/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA