Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
1.
Nat Commun ; 13(1): 7113, 2022 11 19.
Article in English | MEDLINE | ID: mdl-36402789

ABSTRACT

NRAS-mutated melanoma lacks a specific line of treatment. Metabolic reprogramming is considered a novel target to control cancer; however, NRAS-oncogene contribution to this cancer hallmark is mostly unknown. Here, we show that NRASQ61-mutated melanomas specific metabolic settings mediate cell sensitivity to sorafenib upon metabolic stress. Mechanistically, these cells are dependent on glucose metabolism, in which glucose deprivation promotes a switch from CRAF to BRAF signaling. This scenario contributes to cell survival and sustains glucose metabolism through BRAF-mediated phosphorylation of 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase-2/3 (PFKFB2/PFKFB3). In turn, this favors the allosteric activation of phosphofructokinase-1 (PFK1), generating a feedback loop that couples glycolytic flux and the RAS signaling pathway. An in vivo treatment of NRASQ61 mutant melanomas, including patient-derived xenografts, with 2-deoxy-D-glucose (2-DG) and sorafenib effectively inhibits tumor growth. Thus, we provide evidence for NRAS-oncogene contributions to metabolic rewiring and a proof-of-principle for the treatment of NRASQ61-mutated melanoma combining metabolic stress (glycolysis inhibitors) and previously approved drugs, such as sorafenib.


Subject(s)
Melanoma , Proto-Oncogene Proteins B-raf , Humans , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins B-raf/metabolism , Sorafenib/pharmacology , Cell Line, Tumor , Mutation , Melanoma/drug therapy , Melanoma/genetics , Melanoma/metabolism , Glycolysis/genetics , Glucose/metabolism , Stress, Physiological , Phosphofructokinase-2/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , GTP Phosphohydrolases/genetics , GTP Phosphohydrolases/metabolism
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.
Int Immunopharmacol ; 91: 107025, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33360369

ABSTRACT

BACKGROUND: The ability of dendritic cells (DCs) to regulate adaptive immunity makes them interesting cells to be used as therapeutic targets modulating alloimmune responses. Mycophenolic acid (MPA) is an immunosuppressor commonly used in transplantation, and its effect on DCs has not been fully investigated. METHODS: Monocyte-derived DCs were obtained from healthy volunteers and cultured for 7 days. Cells were treated with MPA on day 2 and matured by lipopolysaccharide (LPS) stimulation. Functionality of mature DC (mDCs) was evaluated by allogeneic mixed lymphocytes reaction. Surface expression of maturation markers (CD40, CD83, CD86, and ICAM-1) was analyzed in both immature DCs (iDCs) and mDCs by flow cytometry. To assess transcriptional regulation and protein subcellular location, RT-PCR and confocal microscopy were used, respectively. RESULTS: MPA decreased surface expression of all maturation markers in mDCs and significantly abrogated DCs-induced allogeneic T-cell proliferation after MPA pre-treatment. In iDCs, the reduced surface protein expression after MPA paralleled with mRNA downregulation of their genes. In mDCs, the mRNA levels of ICAM-1, CD40 and CD83 were enhanced in MPA-treated mDCs with an increase in the expression of CD83 and ICAM-1 near the Golgi compared to non-treated mDCs. In contrast, mRNA levels of CD86 were diminished after MPA treatment. CONCLUSIONS: The reduced surface markers expression in mDCs exerted by MPA produced a decline in their capacity to activate immune responses. Moreover, the inhibition of guanosine-derived nucleotide biosynthesis by MPA treatment leads to DC maturation interference by two mechanisms depending on the marker, transcriptional downregulation or disrupted intracellular protein trafficking.


Subject(s)
Antigens, CD/metabolism , Dendritic Cells/drug effects , Immunosuppressive Agents/pharmacology , Intercellular Adhesion Molecule-1/metabolism , Mycophenolic Acid/pharmacology , Transcription, Genetic/drug effects , Antigens, CD/genetics , B7-2 Antigen/genetics , B7-2 Antigen/metabolism , CD40 Antigens/genetics , CD40 Antigens/metabolism , Cell Proliferation , Cells, Cultured , Coculture Techniques , Dendritic Cells/immunology , Dendritic Cells/metabolism , Humans , IMP Dehydrogenase/antagonists & inhibitors , IMP Dehydrogenase/metabolism , Immunoglobulins/genetics , Immunoglobulins/metabolism , Intercellular Adhesion Molecule-1/genetics , Lymphocyte Activation , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Phenotype , Protein Transport , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , CD83 Antigen
5.
Gastroenterology ; 159(1): 273-288, 2020 07.
Article in English | MEDLINE | ID: mdl-32169429

ABSTRACT

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.


Subject(s)
Hepatic Stellate Cells/pathology , Liver Cirrhosis, Experimental/pathology , Liver Cirrhosis/pathology , Phosphofructokinase-2/metabolism , RNA-Binding Proteins/metabolism , Animals , Carbon Tetrachloride/toxicity , Cell Line , Gene Expression Regulation , Gene Knockdown Techniques , Glycolysis , Humans , Liver/cytology , Liver/pathology , Liver Cirrhosis, Experimental/chemically induced , Liver Cirrhosis, Experimental/genetics , Male , Mice , Mice, Knockout , Phosphofructokinase-2/genetics , Primary Cell Culture , RNA-Binding Proteins/genetics , Rats , Up-Regulation
6.
iScience ; 20: 434-448, 2019 Oct 25.
Article in English | MEDLINE | ID: mdl-31627130

ABSTRACT

Cancer cells rely on mTORC1 activity to coordinate mitogenic signaling with nutrients availability for growth. Based on the metabolic function of E2F1, we hypothesize that glucose catabolism driven by E2F1 could participate on mTORC1 activation. Here, we demonstrate that glucose potentiates E2F1-induced mTORC1 activation by promoting mTORC1 translocation to lysosomes, a process that occurs independently of AMPK activation. We showed that E2F1 regulates glucose metabolism by increasing aerobic glycolysis and identified the PFKFB3 regulatory enzyme as an E2F1-regulated gene important for mTORC1 activation. Furthermore, PFKFB3 and PFK1 were found associated to lysosomes and we demonstrated that modulation of PFKFB3 activity, either by substrate accessibility or expression, regulates the translocation of mTORC1 to lysosomes by direct interaction with Rag B and subsequent mTORC1 activity. Our results support a model whereby a glycolytic metabolon containing phosphofructokinases transiently interacts with the lysosome acting as a sensor platform for glucose catabolism toward mTORC1 activity.

7.
Alzheimers Dement ; 15(7): 917-926, 2019 07.
Article in English | MEDLINE | ID: mdl-31175028

ABSTRACT

INTRODUCTION: Age- and sex-stratified incidence rates of uncommon dementia subtypes are imprecise and scarce. METHODS: We used data from 7357 newly diagnosed individuals aged between 30.6 and 101.0 years from the Registry of Dementia of Girona during 2007-2016 to determine the incidence rates of uncommon dementia subtypes stratified by sex and age groups and to describe their clinical characteristics. RESULTS: Uncommon dementia subtypes were classified according to their etiology. The incidence rate of uncommon dementia subtypes was 27.8 cases per 100,000 person-years for those aged 30 years and older, 3.7 cases per 100,000 person-years for people aged less than 65 years, and 110.9 per 100,000 person-years for those aged 65 years and older. Age, sex, dementia severity, and medical comorbidities were different depending on the dementia subtype. DISCUSSION: There are differences in the incidence rates and the demographic and clinical characteristics among uncommon dementia subtypes for age and sex groups.


Subject(s)
Dementia , Demography , Registries , Adult , Age Factors , Aged , Comorbidity , Dementia/classification , Dementia/epidemiology , Female , Humans , Incidence , Male , Middle Aged , Sex Factors , Spain/epidemiology
8.
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.

9.
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
10.
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
11.
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
12.
Drugs Aging ; 34(4): 303-310, 2017 04.
Article in English | MEDLINE | ID: mdl-28258538

ABSTRACT

BACKGROUND: Acetylcholinesterase inhibitors (AChEIs) and the N-methyl D-aspartate-antagonist memantine are indicated for the symptomatic treatment of Alzheimer's disease (AD). OBJECTIVES: Our aims were to describe the baseline characteristics of patients with AD according to prescription of these treatments after the diagnostic work-up to describe long-term trends in the use of these medications and to identify baseline characteristics associated with the frequency of use of each treatment. METHODS: This was a cohort study with a sample of 2992 patients with AD recorded in the Registry of Dementias of Girona (ReDeGi) between 2007 and 2014. Consumption of AChEIs and memantine was assessed using the Pharmacy Unit database from the Public Catalan Healthcare Service. We used generalized estimating equation analyses to identify the baseline characteristics associated with the consumption of AChEIs and memantine over time. RESULTS: Most of the patients (70.4%; 95% confidence interval [CI] 68.7-72.0) were prescribed antidementia medication at the time of diagnosis. Of these, 75.0% (95% CI 73.1-76.8) were prescribed AChEIs, 14.7% (95% CI 13.2-16.3) were prescribed an AChEI plus memantine, and 10.3% (95% CI 9.0-11.6) were prescribed memantine. Advanced age reduced the likelihood of AChEI consumption. Mild dementia severity increased the use of AChEIs, and moderate-advanced dementia increased the likelihood of memantine consumption. After diagnosis, the likelihood of AChEI consumption decreased from the first year until the fifth, whereas the likelihood of memantine consumption, either alone or in combination with AChEIs, increased. CONCLUSIONS: Antidementia drug use in this study showed the initial use of AChEIs alone with later use of AChEIs in combination with memantine and memantine alone in older patients with severe AD. Our findings are in agreement with current clinical practice guidelines for the pharmacological treatment of AD.


Subject(s)
Alzheimer Disease/drug therapy , Cholinesterase Inhibitors/therapeutic use , Drug Utilization/trends , Memantine/therapeutic use , Aged , Aged, 80 and over , Cohort Studies , Female , Humans , Male , Registries , Spain
13.
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
14.
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
15.
Am J Physiol Endocrinol Metab ; 310(6): E440-51, 2016 Mar 15.
Article in English | MEDLINE | ID: mdl-26714846

ABSTRACT

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.


Subject(s)
Blood Glucose/drug effects , Diabetes Mellitus, Type 2/metabolism , Liver/drug effects , Muscle, Skeletal/drug effects , Neuregulins/pharmacology , Animals , Blood Glucose/metabolism , Case-Control Studies , Fructosediphosphates/metabolism , Glucose/metabolism , Glucose Tolerance Test , Glycogen Synthase Kinase 3/drug effects , Glycogen Synthase Kinase 3/metabolism , Insulin , Insulin Receptor Substrate Proteins/drug effects , Insulin Receptor Substrate Proteins/metabolism , Lactic Acid/metabolism , Liver/metabolism , Liver Glycogen/metabolism , Muscle, Skeletal/metabolism , Phosphatidylinositol 3-Kinase/drug effects , Phosphatidylinositol 3-Kinase/metabolism , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Zucker , Receptor, ErbB-3/drug effects , Receptor, ErbB-3/metabolism , Receptor, Insulin/drug effects , Receptor, Insulin/metabolism
16.
Diabetes ; 64(9): 3093-103, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26015548

ABSTRACT

Patients with nonalcoholic fatty liver disease (NAFLD) are reported to have low growth hormone (GH) production and/or hepatic GH resistance. GH replacement can resolve the fatty liver condition in diet-induced obese rodents and in GH-deficient patients. However, it remains to be determined whether this inhibitory action of GH is due to direct regulation of hepatic lipid metabolism. Therefore, an adult-onset, hepatocyte-specific, GH receptor (GHR) knockdown (aLivGHRkd) mouse was developed to model hepatic GH resistance in humans that may occur after sexual maturation. Just 7 days after aLivGHRkd, hepatic de novo lipogenesis (DNL) was increased in male and female chow-fed mice, compared with GHR-intact littermate controls. However, hepatosteatosis developed only in male and ovariectomized female aLivGHRkd mice. The increase in DNL observed in aLivGHRkd mice was not associated with hyperactivation of the pathway by which insulin is classically considered to regulate DNL. However, glucokinase mRNA and protein levels as well as fructose-2,6-bisphosphate levels were increased in aLivGHRkd mice, suggesting that enhanced glycolysis drives DNL in the GH-resistant liver. These results demonstrate that hepatic GH actions normally serve to inhibit DNL, where loss of this inhibitory signal may explain, in part, the inappropriate increase in hepatic DNL observed in NAFLD patients.


Subject(s)
Glucokinase/genetics , Growth Hormone/metabolism , Lipogenesis/genetics , Liver/metabolism , Non-alcoholic Fatty Liver Disease/genetics , RNA, Messenger/metabolism , Receptors, Somatotropin/genetics , Animals , Disease Models, Animal , Female , Fructosediphosphates , Gene Knockdown Techniques , Glucokinase/metabolism , Glycolysis , Insulin/metabolism , Lipogenesis/physiology , Male , Mice , Non-alcoholic Fatty Liver Disease/metabolism , Ovariectomy , Receptors, Somatotropin/metabolism
17.
J Am Med Dir Assoc ; 15(7): 497-503, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24637090

ABSTRACT

OBJECTIVES: Psychotropic drugs are usually prescribed to deal with behavioral and psychological symptoms of dementia, especially when nonpharmacologic approaches are not available or have limited efficacy. Poor outcomes and serious adverse events of the drugs used must be addressed, and risk-benefit ratios need to be considered. The aim of this longitudinal study was to describe the evolution of dispensation of psychotropic drugs in patients with Alzheimer's disease (AD) and to identify the associated demographic and clinical variables. METHODS: Longitudinal study using 698 cases with AD included in the Registry of Dementias of Girona in 2007 and 2008 and followed up during 3 years. Drugs were categorized according to the Anatomical Therapeutic Chemical classification. Binary logistic regression analyses were used to detect the variables associated with the use of antipsychotics, selective serotonin reuptake inhibitors (SSRIs), anxiolytics, and hypnotics. RESULTS: Of the patients, 51.2% consumed antipsychotics at least once during the three years of the study, whereas 73.3% and 58.2% consumed SSRIs and anxiolytics, respectively; 32.8% used hypnotics. Antipsychotic use was associated with a diagnosis of AD with delusions) [odds ratio (OR) = 5.7] and with increased behavior disorders (OR = 1.2). Patients with AD with depressed mood were more likely to be treated with SSRIs (OR = 3.1), while being a woman was associated with increased dispensation of anxiolytics (OR = 1.9) and SSRIs (OR = 2.2). CONCLUSIONS: Consumption of psychotropic drugs by the patients with AD registered in the Registry of Dementias of Girona is very high. Despite all the described adverse effects and recommendations of caution in their use, antipsychotics still are extensively used.


Subject(s)
Alzheimer Disease/drug therapy , Drug Utilization , Psychotropic Drugs/therapeutic use , Aged , Aged, 80 and over , Female , Humans , Logistic Models , Longitudinal Studies , Male , Middle Aged , Odds Ratio , Registries , Spain
18.
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
19.
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
20.
Am J Alzheimers Dis Other Demen ; 27(4): 260-6, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22605780

ABSTRACT

INTRODUCTION: Clinical evidence indicates that acetylcholinesterase inhibitors (AChEIs) are not efficacious to treat frontotemporal lobar degeneration (FTLD). The British Association for Psychopharmacology recommends avoiding the use of AChEI and memantine in patients with FTLD. METHODS: Cross-sectional design using 1092 cases with Alzheimer's disease (AD) and 64 cases with FTLD registered by the Registry of Dementias of Girona. Bivariate analyses were performed, and binary logistic regressions were used to detect variables associated with antidementia drugs consumption. RESULTS: The AChEIs were consumed by 57.6% and 42.2% of the patients with AD and FTLD, respectively. Memantine was used by 17.2% and 10.9% of patients with AD and FTLD, respectively. Binary logistic regressions yielded no associations with antidementia drugs consumption. CONCLUSIONS: There is a discrepancy regarding clinical practice and the recommendations based upon clinical evidence. The increased central nervous system drug use detected in FTLD requires multicentric studies aiming at finding the best means to treat these patients.


Subject(s)
Alzheimer Disease/drug therapy , Cholinesterase Inhibitors/therapeutic use , Frontotemporal Lobar Degeneration/drug therapy , Memantine/therapeutic use , Aged , Aged, 80 and over , Cross-Sectional Studies , Female , Humans , Male , Registries , Regression Analysis
SELECTION OF CITATIONS
SEARCH DETAIL
...