Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
Cancers (Basel) ; 16(2)2024 Jan 19.
Article in English | MEDLINE | ID: mdl-38275867

ABSTRACT

Purpose. To assess the impact of the COVID-19 pandemic on adherence to oral endocrine therapy in patients diagnosed with breast cancer in the public healthcare system in Catalonia (Spain). Methods. Retrospective cohort study in patients starting endocrine therapy from 2017 to 2021. Adherence was measured during the first year of treatment, and the impact of the pandemic was calculated according to the calendar year and whether the first year of treatment included the peak period of the pandemic in our setting (March-September 2020). Analyses were performed using a chi-square test and multivariable logistic regression, with results stratified by year, age group, and drug type. Results. Mean overall adherence during the first year of treatment was 89.6% from 2017 to 2021. In contrast, the patients who started treatment in 2019 and 2020 and whose treatment included the peak pandemic period presented an adherence of 87.0% and 86.5%, respectively. Young age and tamoxifen or combination therapy were predictors of low adherence. An increase in neoadjuvant therapy was also observed in 2020. Conclusions. The COVID-19 pandemic had only a modest impact on adherence to endocrine therapy (≈3%), despite the enormous disruptions for patients, the healthcare system in general, and cancer care in particular that were occurring in that period.

2.
Int J Mol Sci ; 23(3)2022 Feb 08.
Article in English | MEDLINE | ID: mdl-35163828

ABSTRACT

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.


Subject(s)
Apoptosis Regulatory Proteins/genetics , NF-E2-Related Factor 2/metabolism , Neoplasms/genetics , Osteoblasts/cytology , Phosphoric Monoester Hydrolases/genetics , Tumor Suppressor Protein p53/genetics , A549 Cells , Animals , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Glucosephosphate Dehydrogenase/genetics , HCT116 Cells , HeLa Cells , Humans , Mice , NAD(P)H Dehydrogenase (Quinone)/genetics , Neoplasms/metabolism , Osteoblasts/metabolism , Primary Cell Culture , Promoter Regions, Genetic
3.
Int J Mol Sci ; 22(14)2021 Jul 11.
Article in English | MEDLINE | ID: mdl-34299056

ABSTRACT

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.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Concanavalin A/pharmacology , Gene Expression Regulation/drug effects , Lymphocytes/metabolism , Mitogens/pharmacology , Phosphatidylinositol 3-Kinases/chemistry , Phosphoric Monoester Hydrolases/metabolism , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Apoptosis , Apoptosis Regulatory Proteins/genetics , Autophagy , Glycolysis , Humans , Lymphocytes/drug effects , Pentose Phosphate Pathway , Phosphoric Monoester Hydrolases/genetics , Signal Transduction
4.
Front Oncol ; 8: 331, 2018.
Article in English | MEDLINE | ID: mdl-30234009

ABSTRACT

For a long time, pioneers in the field of cancer cell metabolism, such as Otto Warburg, have focused on the idea that tumor cells maintain high glycolytic rates even with adequate oxygen supply, in what is known as aerobic glycolysis or the Warburg effect. Recent studies have reported a more complex situation, where the tumor ecosystem plays a more critical role in cancer progression. Cancer cells display extraordinary plasticity in adapting to changes in their tumor microenvironment, developing strategies to survive and proliferate. The proliferation of cancer cells needs a high rate of energy and metabolic substrates for biosynthesis of biomolecules. These requirements are met by the metabolic reprogramming of cancer cells and others present in the tumor microenvironment, which is essential for tumor survival and spread. Metabolic reprogramming involves a complex interplay between oncogenes, tumor suppressors, growth factors and local factors in the tumor microenvironment. These factors can induce overexpression and increased activity of glycolytic isoenzymes and proteins in stromal and cancer cells which are different from those expressed in normal cells. The fructose-6-phosphate/fructose-1,6-bisphosphate cycle, catalyzed by 6-phosphofructo-1-kinase/fructose 1,6-bisphosphatase (PFK1/FBPase1) isoenzymes, plays a key role in controlling glycolytic rates. PFK1/FBpase1 activities are allosterically regulated by fructose-2,6-bisphosphate, the product of the enzymatic activity of the dual kinase/phosphatase family of enzymes: 6-phosphofructo-2-kinase/fructose 2,6-bisphosphatase (PFKFB1-4) and TP53-induced glycolysis and apoptosis regulator (TIGAR), which show increased expression in a significant number of tumor types. In this review, the function of these isoenzymes in the regulation of metabolism, as well as the regulatory factors modulating their expression and activity in the tumor ecosystem are discussed. Targeting these isoenzymes, either directly or by inhibiting their activating factors, could be a promising approach for treating cancers.

5.
Expert Opin Ther Targets ; 22(8): 659-674, 2018 08.
Article in English | MEDLINE | ID: mdl-29985086

ABSTRACT

INTRODUCTION: It has been known for over half a century that tumors exhibit an increased demand for nutrients to fuel their rapid proliferation. Interest in targeting cancer metabolism to treat the disease has been renewed in recent years with the discovery that many cancer-related pathways have a profound effect on metabolism. Considering the recent increase in our understanding of cancer metabolism and the enzymes and pathways involved, the question arises as to whether metabolism is cancer's Achilles heel. Areas covered: This review summarizes the role of 6-Phosphofructo-2-kinase/fructose-2,6-bisphosphatase 3 (PFKFB3) in glycolysis, cell proliferation, and tumor growth, discussing PFKFB3 gene and isoenzyme regulation and the changes that occur in cancer and inflammatory diseases. Pharmacological options currently available for selective PFKFB3 inhibition are also reviewed. Expert opinion: PFKFB3 plays an important role in sustaining the development and progression of cancer and might represent an attractive target for therapeutic strategies. Nevertheless, clinical trials are needed to follow up on the promising results from preclinical studies with PFKFB3 inhibitors. Combination therapies with PFKFB3 inhibitors, chemotherapeutic drugs, or radiotherapy might improve the efficacy of cancer treatments targeting PFKFB3.


Subject(s)
Molecular Targeted Therapy , Neoplasms/drug therapy , Phosphofructokinase-2/metabolism , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacology , Cell Proliferation/physiology , Disease Progression , Drug Development/methods , Gene Expression Regulation, Neoplastic , Glycolysis/physiology , Humans , Neoplasms/genetics , Neoplasms/pathology , Phosphofructokinase-2/genetics
6.
Mol Cell Biochem ; 448(1-2): 187-197, 2018 Nov.
Article in English | MEDLINE | ID: mdl-29435871

ABSTRACT

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.


Subject(s)
Gene Expression Regulation/immunology , Lymphocyte Activation , Phosphatidylinositol 3-Kinases/immunology , Phosphofructokinase-2/immunology , Proto-Oncogene Proteins c-akt/immunology , Signal Transduction/immunology , T-Lymphocytes/immunology , Cell Proliferation/drug effects , Gene Expression Regulation/drug effects , Humans , Phytohemagglutinins/pharmacology , Signal Transduction/drug effects , T-Lymphocytes/cytology
7.
FEBS J ; 284(20): 3437-3454, 2017 10.
Article in English | MEDLINE | ID: mdl-28834297

ABSTRACT

In human cancers, transforming growth factor-ß1 (TGF-ß1) plays a dual role by acting as both a tumor suppressor and a promoter of tumor metastasis. Although TGF-ß1 contributes to the metabolic reprogramming of cancer cells and tumor-associated stromal cells, little is known of the molecular mechanisms connecting this cytokine with enhanced glycolysis. PFKFB3 is a homodymeric bifunctional enzyme, belonging to the family of 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatases, that controls the conversion of fructose-6-phosphate (Fru-6-P) to fructose-2,6-bisphosphate (Fru-2,6-P2 ). This metabolite is important for the dynamic regulation of glycolytic flux by allosterically activating phosphofructokinase-1, a rate-limiting enzyme in glycolysis. The PFKFB3 gene is involved in cell proliferation via its role in carbohydrate metabolism. Here, we studied the mechanisms connecting TGF-ß1, glucose metabolism, and PFKFB3 in glioblastoma cell lines. We demonstrate that TGF-ß1 upregulates PFKFB3 mRNA and protein expression resulting in an increase in fructose 2,6-bisphosphate concentration, glucose uptake, glycolytic flux and lactate production. Moreover, these increases in PFKFB3 mRNA and protein expression and Fru-2,6-P2 concentration were reduced when the Smad3, p38 mitogen-activated protein kinase (MAPK), and phosphoinositide 3-kinase (PI3K)/Akt signaling pathways were inhibited. We demonstrate that inhibition of PFKFB3 activity with 3PO or siRNA-mediated knockdown of PFKFB3 significantly eliminated the capacity of the T98G cells to form colonies by TGF-ß1, one of the hallmarks of transformation. Taken together, these results show that TGF-ß1 induces PFKFB3 expression through activation of the p38 MAPK and PI3K/Akt signaling pathways that complement and converge with early activation of Smad signaling. This suggests that PFKFB3 induction by TGF-ß1 can be one of the main mechanisms mediating the reprogramming of glioma cells.


Subject(s)
Glioblastoma/metabolism , Glycolysis/drug effects , Phosphofructokinase-2/metabolism , Phosphoinositide-3 Kinase Inhibitors , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Smad Proteins/antagonists & inhibitors , Transforming Growth Factor beta1/pharmacology , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , Cell Proliferation/drug effects , Fructosediphosphates/metabolism , Glioblastoma/drug therapy , Glioblastoma/pathology , Glucose/metabolism , Humans , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , Smad Proteins/metabolism , Tumor Cells, Cultured , Tumor Stem Cell Assay , p38 Mitogen-Activated Protein Kinases/metabolism
8.
J Biol Chem ; 291(51): 26291-26303, 2016 Dec 16.
Article in English | MEDLINE | ID: mdl-27803158

ABSTRACT

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.


Subject(s)
Breast Neoplasms/metabolism , Glutamic Acid/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Lactic Acid/metabolism , Apoptosis/genetics , Apoptosis Regulatory Proteins , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Coculture Techniques , Female , Fibroblasts/metabolism , Fibroblasts/pathology , Glutamic Acid/genetics , Glycolysis/genetics , Humans , Intracellular Signaling Peptides and Proteins/genetics , Isoenzymes/genetics , Isoenzymes/metabolism , L-Lactate Dehydrogenase/genetics , L-Lactate Dehydrogenase/metabolism , Lactate Dehydrogenase 5 , MCF-7 Cells , Phosphofructokinase-2/genetics , Phosphofructokinase-2/metabolism , Phosphoric Monoester Hydrolases , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
9.
FEBS Lett ; 590(17): 2915-26, 2016 09.
Article in English | MEDLINE | ID: mdl-27491040

ABSTRACT

Neoplastic cells metabolize higher amounts of glucose relative to normal cells in order to cover increased energetic and anabolic needs. Inhibition of the glycolytic enzyme 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase 3 (PFKFB3) diminishes cancer cell proliferation and tumour growth in animals. In this work, we investigate the crosstalk between PFKFB3 and TIGAR (TP53-Induced Glycolysis and Apoptosis Regulator), a protein known to protect cells from oxidative stress. Our results show consistent TIGAR induction in HeLa cells in response to PFKFB3 knockdown. Upon PFKFB3 silencing, cells undergo oxidative stress and trigger Akt phosphorylation. This leads to induction of a TIGAR-mediated prosurvival pathway that reduces both oxidative stress and cell death. As TIGAR is known to have a role in DNA repair, it could serve as a potential target for the development of effective antineoplastic therapies.


Subject(s)
Intracellular Signaling Peptides and Proteins/biosynthesis , Neoplasms/genetics , Oxidative Stress/genetics , Phosphofructokinase-2/biosynthesis , Apoptosis Regulatory Proteins , Cell Proliferation/genetics , DNA Repair/genetics , Gene Expression Regulation, Neoplastic , HeLa Cells , Humans , Intracellular Signaling Peptides and Proteins/genetics , Neoplasms/pathology , Phosphofructokinase-2/genetics , Phosphoric Monoester Hydrolases , Phosphorylation , Proto-Oncogene Proteins c-akt/genetics
10.
Biochem J ; 452(3): 531-43, 2013 Jun 15.
Article in English | MEDLINE | ID: mdl-23548149

ABSTRACT

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.


Subject(s)
MAP Kinase Signaling System/physiology , Mitogen-Activated Protein Kinases/metabolism , Oxidative Stress , Phosphofructokinase-2/chemistry , Phosphorylation , p38 Mitogen-Activated Protein Kinases/chemistry , p38 Mitogen-Activated Protein Kinases/physiology , Amino Acid Sequence , Enzyme Activation/physiology , Glycolysis/genetics , HeLa Cells , Humans , Mitogen-Activated Protein Kinases/genetics , Molecular Sequence Data , Neoplasms/chemistry , Neoplasms/genetics , Neoplasms/pathology , Oxidative Stress/genetics , Phosphofructokinase-2/genetics , Phosphofructokinase-2/metabolism , Phosphorylation/genetics , Protein Binding/genetics , p38 Mitogen-Activated Protein Kinases/genetics
11.
J Biol Chem ; 288(15): 10640-51, 2013 Apr 12.
Article in English | MEDLINE | ID: mdl-23457334

ABSTRACT

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.


Subject(s)
Amino Acids/metabolism , Glycolysis/physiology , Myocytes, Cardiac/enzymology , Phosphofructokinase-2/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/physiology , Adenosine Triphosphate/genetics , Adenosine Triphosphate/metabolism , Amino Acids/genetics , Animals , Enzyme Activation/physiology , Fructosediphosphates/genetics , Fructosediphosphates/metabolism , Glucose/genetics , Glucose/metabolism , HEK293 Cells , HeLa Cells , Humans , Lactic Acid/metabolism , Male , Myocytes, Cardiac/cytology , Phosphofructokinase-2/genetics , Phosphorylation/physiology , Proto-Oncogene Proteins c-akt/genetics , Rats , Rats, Sprague-Dawley , Serine/genetics , Serine/metabolism
12.
Am J Physiol Endocrinol Metab ; 303(6): E695-707, 2012 Sep 15.
Article in English | MEDLINE | ID: mdl-22811469

ABSTRACT

Sertoli cells play a central role in the control and maintenance of spermatogenesis by secreting growth factors, in response to hormonal stimulation, that participate in the paracrine regulation of this process. In this study, we investigated how the hormonal regulation of spermatogenesis modulates 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase (PFKFB) isozyme expression in two mouse spermatogenic cell lines, GC-1 spg and GC-2 spd (ts). For this purpose, TM4 Sertoli cells were used to obtain conditioned medium that was treated or not with dihydrotestosterone for 2 days [dihydrotestosterone conditioned medium (TCM) and basal conditioned medium (BCM), respectively]. We observed an increase in the expression of PFKFB4 along with a decrease in PFKFB3 in spermatogenic cell lines treated with TCM. These effects were inhibited by the antiandrogen drug flutamide and by heat-inactivated TCM, indicating the protein nature of the TCM mediator and its dependence on Sertoli cell stimulation by dihydrotestosterone. In addition, adult rat testes treated with the GnRH antagonist Degarelix exhibited a reduction in the expression of PFKFB4 in germ cells. Addition of exogenous FGF-2 mimicked the changes in the Pfkfb gene expression, whereas neutralizing antibodies against FGF-2 abolished them. Interestingly, similar effects on Pfkfb gene expression were observed using different MAPK inhibitors (U-0126, PD-98059, and H-89). Luciferase analysis of Pfkfb4 promoter constructs demonstrated that a putative CRE-binding sequence located at -1,463 relative to the transcription start site is required to control Pfkfb4 gene expression after TCM treatment. Pulldown assays showed the binding of the CREB transcription factor to this site. Altogether, these results show how the paracrine regulation orchestrated by Sertoli cells in response to testosterone controls glycolysis in germ cells.


Subject(s)
Enzyme Induction , Fibroblast Growth Factor 2/metabolism , MAP Kinase Signaling System , Paracrine Communication , Phosphofructokinase-2/biosynthesis , Sertoli Cells/metabolism , Spermatogonia/metabolism , Androgen Antagonists/pharmacology , Animals , Antibodies, Neutralizing/pharmacology , Cell Line , Dihydrotestosterone/antagonists & inhibitors , Dihydrotestosterone/metabolism , Enzyme Induction/drug effects , Enzyme Repression/drug effects , Fibroblast Growth Factor 2/antagonists & inhibitors , Humans , MAP Kinase Signaling System/drug effects , Male , Mice , Mice, Inbred BALB C , Paracrine Communication/drug effects , Phosphofructokinase-2/genetics , Phosphofructokinase-2/metabolism , Protein Kinase Inhibitors/pharmacology , Rats , Rats, Sprague-Dawley , Recombinant Proteins/biosynthesis , Recombinant Proteins/metabolism , Sertoli Cells/cytology , Sertoli Cells/drug effects , Spermatogonia/cytology , Spermatogonia/drug effects
13.
Biochem J ; 442(2): 345-56, 2012 Mar 01.
Article in English | MEDLINE | ID: mdl-22115192

ABSTRACT

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.


Subject(s)
Breast Neoplasms/metabolism , Phosphofructokinase-2/metabolism , Progesterone Congeners/pharmacology , Base Sequence , Breast Neoplasms/genetics , Cell Line, Tumor , Enzyme Activation/drug effects , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , MAP Kinase Signaling System , Norgestrel/pharmacology , Phosphofructokinase-2/genetics , Pregnenediones/pharmacology , Promoter Regions, Genetic , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Neoplasm/genetics , RNA, Neoplasm/metabolism , Receptors, Progesterone/metabolism
14.
Radiother Oncol ; 101(1): 132-9, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21864926

ABSTRACT

BACKGROUND AND PURPOSE: The TP53 induced glycolysis and apoptosis regulator (TIGAR) functions to lower fructose-2,6-bisphosphate (Fru-2,6-P(2)) levels in cells, consequently decreasing glycolysis and leading to the scavenging of reactive oxygen species (ROS), which correlate with a higher resistance to cell death. The decrease in intracellular ROS levels in response to TIGAR may also play a role in the ability of p53 to protect from the accumulation of genomic lesions. Given these good prospects of TIGAR for metabolic regulation and p53-response modulation, we analyzed the effects of TIGAR knockdown in U87MG and T98G glioblastoma-derived cell lines. METHODS/RESULTS: After TIGAR-knockdown in glioblastoma cell lines, different metabolic parameters were assayed, showing an increase in Fru-2,6-P(2), lactate and ROS levels, with a concomitant decrease in reduced glutathione (GSH) levels. In addition, cell growth was inhibited without evidence of apoptotic or autophagic cell death. In contrast, a clear senescent phenotype was observed. We also found that TIGAR protein levels were increased shortly after irradiation. In addition, avoiding radiotherapy-triggered TIGAR induction by gene silencing resulted in the loss of capacity of glioblastoma cells to form colonies in culture and the delay of DNA repair mechanisms, based in γ-H2AX foci, leading cells to undergo morphological changes compatible with a senescent phenotype. Thus, the results obtained raised the possibility to consider TIGAR as a therapeutic target to increase radiotherapy effects. CONCLUSION: TIGAR abrogation provides a novel adjunctive therapeutic strategy against glial tumors by increasing radiation-induced cell impairment, thus allowing the use of lower radiotherapeutic doses.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Apoptosis/genetics , Glioblastoma/radiotherapy , Intracellular Signaling Peptides and Proteins/metabolism , Radiation Tolerance/genetics , Reactive Oxygen Species/metabolism , Tumor Suppressor Protein p53/metabolism , Blotting, Western , Down-Regulation , Fluorescent Antibody Technique , Glioblastoma/pathology , Glycolysis/genetics , Humans , Intracellular Signaling Peptides and Proteins/genetics , Phosphoric Monoester Hydrolases , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Sensitivity and Specificity , Tumor Cells, Cultured/metabolism , Tumor Cells, Cultured/radiation effects , Tumor Suppressor Protein p53/genetics
15.
J Biol Chem ; 286(22): 19247-58, 2011 Jun 03.
Article in English | MEDLINE | ID: mdl-21464136

ABSTRACT

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.


Subject(s)
Adenosine/metabolism , Gene Expression Regulation, Enzymologic/drug effects , Glycolysis/drug effects , Lipopolysaccharides/pharmacology , Macrophages, Peritoneal/enzymology , Phosphofructokinase-2/biosynthesis , Toll-Like Receptor 4/agonists , Adenosine/genetics , Adenosine Triphosphate/biosynthesis , Adenosine Triphosphate/genetics , Amino Acid Sequence , Animals , Cell Line , Fructosediphosphates/genetics , Fructosediphosphates/metabolism , Gene Expression Regulation, Enzymologic/physiology , Glycolysis/physiology , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Isoenzymes/biosynthesis , Isoenzymes/genetics , Macrophage Activation/drug effects , Macrophage Activation/physiology , Macrophages, Peritoneal/cytology , Mice , Mice, Knockout , Mutagenesis, Site-Directed , Phosphofructokinase-2/genetics , Receptor, Adenosine A2A/genetics , Receptor, Adenosine A2A/metabolism , Receptor, Adenosine A2B , Sequence Deletion , Sp1 Transcription Factor/genetics , Sp1 Transcription Factor/metabolism , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/metabolism
16.
FEBS J ; 276(16): 4555-68, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19645723

ABSTRACT

The ubiquitous isoform of 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase (uPFK-2), a product of the Pfkfb3 gene, plays a crucial role in the control of glycolytic flux. In this study, we demonstrate that Pfkfb3 gene expression is increased in streptozotocin-induced diabetic mouse liver. The Pfkfb3/-3566 promoter construct linked to the luciferase reporter gene was delivered to the liver via hydrodynamic gene transfer. This promoter was upregulated in streptozotocin-induced diabetic mouse liver compared with transfected healthy cohorts. In addition, increases were observed in Pfkfb3 mRNA and uPFK-2 protein levels, and intrahepatic fructose-2,6-bisphosphate concentration. During streptozotocin-induced diabetes, phosphorylation of both p38 mitogen-activated protein kinase and Akt was detected, together with the overexpression of the proliferative markers cyclin D and E2F. These findings indicate that uPFK-2 induction is coupled to enhanced hepatocyte proliferation in streptozotocin-induced diabetic mouse liver. Expression decreased when hepatocytes were treated with either rapamycin or LY 294002. This shows that uPFK-2 regulation is phosphoinositide 3-kinase-Akt-mammalian target of rapamycin dependent. These results indicate that fructose-2,6-bisphosphate is essential to the maintenance of the glycolytic flux necessary for providing energy and biosynthetic precursors to dividing cells.


Subject(s)
Cell Proliferation , Liver/enzymology , Phosphofructokinase-2/genetics , Signal Transduction , Transcription, Genetic , Animals , Diabetes Mellitus, Experimental , Fructosediphosphates/analysis , Glycolysis , Mice , RNA, Messenger/analysis , Up-Regulation
17.
Biochem Biophys Res Commun ; 367(4): 748-54, 2008 Mar 21.
Article in English | MEDLINE | ID: mdl-18191036

ABSTRACT

The bifunctional enzyme 6-phosphofructo-2-kinase/fructose 2,6-bisphosphatase (PFK-2) catalyzes the synthesis and degradation of fructose 2,6-bisphosphate (Fru-2,6-P(2)), a signalling molecule that controls the balance between glycolysis and gluconeogenesis in several cell types. Four genes, designated Pfkfb1-4, code several PFK-2 isozymes that differ in their kinetic properties, molecular masses, and regulation by protein kinases. In rat tissues, Pfkfb3 gene accounts for eight splice variants and two of them, ubiquitous and inducible PFK-2 isozymes, have been extensively studied and related to cell proliferation and tumour metabolism. Here, we characterize a new kidney- and liver-specific Pfkfb3 isozyme, a product of the RB2K3 splice variant, and demonstrate that its expression, in primary cultured hepatocytes, depends on hepatic cell proliferation and dedifferentiation. In parallel, our results provide further evidence that ubiquitous PFK-2 is a crucial isozyme in supporting growing and proliferant cell metabolism.


Subject(s)
Hepatocytes/cytology , Hepatocytes/enzymology , Kidney/cytology , Kidney/enzymology , Phosphofructokinase-2/metabolism , Animals , Cell Differentiation , Cell Proliferation , Cells, Cultured , Down-Regulation , Isoenzymes/metabolism , Male , Organ Specificity , Rats , Rats, Sprague-Dawley , Tissue Distribution
18.
Biochem Biophys Res Commun ; 365(2): 291-7, 2008 Jan 11.
Article in English | MEDLINE | ID: mdl-17991424

ABSTRACT

Fructose 2,6-bisphosphate (Fru-2,6-P2) is an important metabolite that controls glycolytic and gluconeogenic pathways in several cell types. Its synthesis and degradation are catalyzed by the bifunctional enzyme 6-phosphofructo-2-kinase/fructose 2,6-bisphosphatase (PFK-2). Four genes, designated Pfkfb1-4, codify the different PFK-2 isozymes. The Pfkfb3 gene product, ubiquitous PFK-2 (uPFK-2), has the highest kinase/bisphosphatase activity ratio and is associated with proliferation and tumor metabolism. A transgenic mouse model that overexpresses uPFK-2 under the control of the phosphoenolpyruvate carboxykinase promoter was designed to promote sustained and elevated Fru-2,6-P2 levels in the liver. Our results demonstrate that in diet-induced obesity, high Fru-2,6-P2 levels in transgenic livers caused changes in hepatic gene expression profiles for key gluconeogenic and lipogenic enzymes, as well as an accumulation of lipids in periportal cells, and weight gain.


Subject(s)
Liver/physiology , Phosphofructokinase-2/metabolism , Weight Gain/physiology , Animals , Mice , Mice, Transgenic , Phosphofructokinase-2/genetics , Up-Regulation
19.
Liver Transpl ; 12(11): 1615-25, 2006 Nov.
Article in English | MEDLINE | ID: mdl-17058268

ABSTRACT

Hepatic ischemia-reperfusion injury associated with liver transplantation is an as yet unresolved problem in clinical practice. Preconditioning protects the liver against the deleterious effects of ischemia, although the mechanism underlying this preconditioning is still unclear. To profile gene expression patterns involved in hepatic ischemic preconditioning, we analyzed the changes in gene expression in rat livers by DNA microarray analysis. Approximately 116 genes were found to have altered gene expression after 8 hours of cold ischemia. Moreover, the expression of 218 genes was modified by classic preconditioning followed by the same ischemia process. Given the importance of the effects of ischemic preconditioning (IP) in minimizing the liver damage induced by sustained ischemia before reperfusion, this study analyzed the putative genes involved in the beneficial role of IP in liver grafts undergoing cold ischemia before its implantation in the recipient (IP+I). Great differences were found in the gene expression pattern of ischemic preconditioning + long cold ischemia (IP+I) group when compared with the long cold ischemia alone condition (I), which could explain the protective regulatory mechanisms that take place after preconditioning. Twenty-six genes that were downregulated in cold ischemia were found upregulated after preconditioning preceding a long cold ischemia period. These would be genes activated or maintained by preconditioning. Heat shock protein genes and 3-hydroxy-3-methylglutaryl-coenzyme A reductase are among the most markedly induced transcripts.


Subject(s)
Cold Ischemia , Gene Expression , Ischemic Preconditioning , Liver/blood supply , Liver/metabolism , Oligonucleotide Array Sequence Analysis , Animals , Computer Systems , Down-Regulation , Gene Expression Profiling , Gene Expression Regulation , Heat-Shock Proteins/genetics , Hydroxymethylglutaryl CoA Reductases/genetics , Male , Polymerase Chain Reaction , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Time Factors , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL
...