Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
1.
Int J Mol Sci ; 23(15)2022 Jul 29.
Article in English | MEDLINE | ID: mdl-35955542

ABSTRACT

Systemic lupus erythematosus is a complex autoimmune disorder mostly mediated by B-cells in which costimulatory signals are involved. This immune dysregulation can cause tissue damage and inflammation of the kidney, resulting in lupus nephritis and chronic renal failure. Given the previous experience reported with CTLA4-Ig as well as recent understanding of the PD-1 pathway in this setting, our group was encouraged to evaluate, in the NZBWF1 model, a human fusion recombinant protein (Hybri) with two domains: CTLA4, blocking the CD28-CD80 costimulatory pathway, and PD-L2, exacerbating the PD-1-PD-L2 coinhibitory pathway. After achieving good results in this model, we decided to validate the therapeutic effect of Hybri in the more severe MRL/lpr model of lupus nephritis. The intraperitoneal administration of Hybri prevented the progression of proteinuria and anti-dsDNA antibodies to levels like those of cyclophosphamide and reduced the histological score, infiltration of B-cells, T-cells, and macrophages and immune deposition in both lupus-prone models. Additionally, Hybri treatment produced changes in both inflammatory-related circulating cytokines and kidney gene expression. To summarize, both in vivo studies revealed that the Hybri effect on costimulatory-coinhibitory pathways may effectively mitigate lupus nephritis, with potential for use as a maintenance therapy.


Subject(s)
Lupus Erythematosus, Systemic , Lupus Nephritis , Animals , Antibodies, Antinuclear , Disease Models, Animal , Humans , Immunomodulation , Kidney/metabolism , Mice , Mice, Inbred MRL lpr , Programmed Cell Death 1 Receptor/metabolism , Recombinant Proteins/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 J Mol Sci ; 21(24)2020 Dec 18.
Article in English | MEDLINE | ID: mdl-33353159

ABSTRACT

Cardiovascular mortality increases with decreasing renal function although the cause is yet unknown. Here, we have investigated whether low chronic inflammation in chronic kidney diseases (CKD) could contribute to increased risk for coronary artery diseases (CAD). Thus, a prospective case-control study was conducted in patients with CAD and CKD undergoing coronary artery bypass graft surgery with the aim of detecting differences in cardiovascular outcomes, epicardial adipose tissue volume, and inflammatory marker activity associated with renal dysfunction. Expression of membrane CD14 and CD16, inflammatory cytokines and chemokines, mitogen-activated protein (MAP) kinases and hsa-miR-30a-5p were analyzed in peripheral blood mononuclear cells (PBMCs). Epicardial fat volume and tissue inflammation in perivascular adipose tissue and in the aorta were also studied. In the present study, 151 patients were included, 110 with CAD (51 with CKD) and 41 nonCAD controls (15 with CKD). CKD increased the risk of cardiac surgery-associated acute kidney injury (CSA-AKI) as well as the 30-day mortality after cardiac surgery. Higher counts of CD14++CD16+ monocytes were associated with vascular inflammation, with an increased expression of IL1ß, and with CKD in CAD patients. Expression of hsa-miR-30a-5p was correlated with hypertension. We conclude that CKD patients show an increased risk of CSA-AKI and mortality after cardiovascular surgery, associated with the expansion of the CD14++CD16+ subset of proinflammatory monocytes and with IL1ß expression. We propose that inflammation associated with CKD may contribute to atherosclerosis (ATH) pathogenesis.


Subject(s)
Acute Kidney Injury/etiology , Cardiac Surgical Procedures/mortality , Cardiovascular Diseases/mortality , Inflammation/complications , Renal Insufficiency, Chronic/physiopathology , Acute Kidney Injury/pathology , Aged , Cardiovascular Diseases/etiology , Cardiovascular Diseases/pathology , Case-Control Studies , Female , Humans , Male , Middle Aged , Prognosis , Prospective Studies , Survival Rate
5.
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.

6.
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
7.
Sci Rep ; 8(1): 6786, 2018 Apr 25.
Article in English | MEDLINE | ID: mdl-29691448

ABSTRACT

A correction to this article has been published and is linked from the HTML and PDF versions of this paper. The error has not been fixed in the paper.

8.
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
9.
Sci Rep ; 7(1): 17515, 2017 12 13.
Article in English | MEDLINE | ID: mdl-29235543

ABSTRACT

ß-cell proliferation is a rare event in adult pancreatic islets. To study the replication-related ß-cell biology we designed a replicating ß-cells sorting system for gene expression experiments. Replicating ß-cells were identified by EdU incorporation and purified by flow cytometry. For ß-cell separation islet cells were sorted by size, granularity and Newport Green fluorescence emission that was combined with emitted fluorescence for EdU-labelled replicating cells sorting. The purity of the resulting sorted populations was evaluated by insulin staining and EdU for ß-cell identification and for replicating cells, respectively. Total RNA was isolated from purified cell-sorted populations for gene expression analysis. Cell sorting of dispersed islet cells resulted in 96.2% purity for insulin positivity in the collected ß-cell fraction and 100% efficiency of the EdU-based cell separation. RNA integrity was similar between FACS-sorted replicating and quiescent ß-cells. Global transcriptome analysis of replicating vs quiescent ß-cells showed the expected enrichment of categories related to cell division and DNA replication. Indeed, key genes in the spindle check-point were the most upregulated genes in replicating ß-cells. This work provides a method that allows for the isolation of replicating ß-cells, a very scarce population in adult pancreatic islets.


Subject(s)
Cell Separation/methods , Insulin-Secreting Cells , Alternative Splicing , Animals , Cell Proliferation/physiology , Cells, Cultured , Fluorescent Antibody Technique , Gene Expression , Gene Expression Profiling , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/metabolism , Male , Rats, Wistar , Transcriptome
10.
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
11.
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
13.
Arch Dermatol ; 148(10): 1159-64, 2012 Oct.
Article in English | MEDLINE | ID: mdl-23069952

ABSTRACT

OBJECTIVE: To validate dermoscopy as a real-time noninvasive diagnostic imaging technique for actinic keratosis (AK). DESIGN: Prospective study to validate a diagnostic test. SETTING: Dermatology department of a tertiary university hospital in Fuenlabrada, Madrid, Spain. PATIENTS: A total of 178 patients with a clinical diagnosis of AK participated in the study. MAIN OUTCOME MEASURES: An independent blinded comparison was performed between dermoscopy results and histopathological findings, the gold standard for the diagnosis of AK. All the patients underwent both diagnostic tests. RESULTS: One hundred seventy-eight lesions were evaluated. The concordance between dermoscopy results and histopathological findings was 0.917. The sensitivity of dermoscopy for the diagnosis of AK was 98.7%, with a specificity of 95.0%, a positive likelihood ratio of 19.74, and a negative likelihood ratio of 0.01. A diagnostic algorithm that combined follicular openings and erythematous pseudonetwork demonstrated a sensitivity of 95.6% and a specificity of 95.0% for the diagnosis of AK. CONCLUSIONS: The sensitivity and specificity of dermoscopy for the diagnosis of AK were high, as was the concordance between dermoscopy results and histopathological findings. As a real-time noninvasive diagnostic imaging technique for AK, dermoscopy may be incorporated in the management of patients with these lesions.


Subject(s)
Dermoscopy , Keratosis, Actinic/pathology , Adult , Aged , Aged, 80 and over , Algorithms , Chi-Square Distribution , Double-Blind Method , Female , Humans , Keratosis, Actinic/diagnosis , Likelihood Functions , Male , Middle Aged , Prospective Studies , Sensitivity and Specificity
14.
J Pharm Pharm Sci ; 15(3): 407-19, 2012.
Article in English | MEDLINE | ID: mdl-22974789

ABSTRACT

PURPOSE: P-glycoprotein (Pgp) is a member of the ABC-transporter family that transports substances across cellular membranes acting as an efflux pump extruding drugs out of the cells. Pgp plays a key role on the pharmacokinetics of several drugs. Herein, we have studied the effects of immunosuppressants on Pgp function, assessing rhodamine-123 (Rho123) uptake and efflux in different T-cell subsets. METHODS: Different immunosuppressants such as Cyclosporine (CsA), Rapamycin (Rapa) and Tacrolimus (Tac) were used to assess the in vitro effect on Pgp function of main T-cell subsets among healthy volunteers. We measured Rho123 uptake, efflux and kinetic of extrusion in CD4+ and CD8+ subsets by flow cytometry. Antigen-specific memory T-cell responses were assessed by measuring T-cell proliferation and cytokine secretion using an allogeneic mixed lymphocyte reaction. RESULTS: Rho123 uptake in groups treated with CsA and CsA+Rapa was significantly decreased compared to non-treated group and the other immunosupressants in both T cells subsets. Pgp activity was also reduced in CsA and CsA+Rapa compared to the other immunosupressants but it was only significant in the CsA group for CD8+ subset. Kinetic extrusion of Rho123 by Pgp in all groups was faster in CD8+ T cells. All immunosuppressants and the specific Pgp inhibitor PSC833 diminished antigen-primed T-cell proliferation, especially CD8+ T-cell subset. CONCLUSIONS: Our data indicate that small molecules immunosuppressants, especially CsA, inhibit Pgp activity and T-cell function being the CD8+ T cells more susceptible to this effect. These findings support the importance of Pgp when designing combined immunosuppressive regimens.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , CD4-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/drug effects , Immunosuppressive Agents/pharmacology , T-Lymphocyte Subsets/drug effects , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/metabolism , Cell Proliferation/drug effects , Cells, Cultured , Cyclosporine/pharmacology , Cytokines/metabolism , Humans , Rhodamine 123/metabolism , Sirolimus/pharmacology , T-Lymphocyte Subsets/metabolism , Tacrolimus/pharmacology
15.
PLoS One ; 6(4): e18588, 2011 Apr 05.
Article in English | MEDLINE | ID: mdl-21483692

ABSTRACT

Glioblastoma multiforme (GBM) is the most common and aggressive primary brain tumor in adults. Despite concerted efforts to improve current therapies and develop novel clinical approaches, patient survival remains poor. As such, increasing attention has focused on developing new therapeutic strategies that specifically target the apoptotic pathway in order to improve treatment responses. Recently, nutlins, small-molecule antagonists of MDM2, have been developed to inhibit p53-MDM2 interaction and activate p53 signaling in cancer cells. Glioma cell lines and primary cultured glioblastoma cells were treated with nutlin-3a. Nutlin-3a induced p53-dependent G1- and G2-M cell cycle arrest and apoptosis in glioma cell lines with normal TP53 status. In addition, nutlin-arrested glioma cells show morphological features of senescence and persistent induction of p21 protein. Furthermore, senescence induced by nutlin-3a might be depending on mTOR pathway activity. In wild-type TP53 primary cultured cells, exposure to nutlin-3a resulted in variable degrees of apoptosis as well as cellular features of senescence. Nutlin-3a-induced apoptosis and senescence were firmly dependent on the presence of functional p53, as revealed by the fact that glioblastoma cells with knockdown p53 with specific siRNA, or cells with mutated or functionally impaired p53 pathway, were completely insensitive to the drug. Finally, we also found that nutlin-3a increased response of glioma cells to radiation therapy. The results provide a basis for the rational use of MDM2 antagonists as a novel treatment option for glioblastoma patients.


Subject(s)
Apoptosis/drug effects , Cellular Senescence/drug effects , Glioblastoma/pathology , Imidazoles/pharmacology , Piperazines/pharmacology , Tumor Suppressor Protein p53/metabolism , Apoptosis/radiation effects , Cell Cycle/drug effects , Cell Cycle/radiation effects , Cell Line, Tumor , Cellular Senescence/radiation effects , Gene Silencing , Glioblastoma/genetics , Glioblastoma/metabolism , Humans , Mutation , Proto-Oncogene Proteins c-mdm2/antagonists & inhibitors , Tumor Suppressor Protein p53/deficiency , Tumor Suppressor Protein p53/genetics
16.
J Pharm Pharm Sci ; 12(3): 357-66, 2009.
Article in English | MEDLINE | ID: mdl-20067710

ABSTRACT

P-glycoprotein (Pgp), a protein codified by Multi Drug Resistance (MDR1) gene, has a detoxifying function and might influence the toxicity and pharmacokinetics and pharmacodynamics of drugs. Sampling strategies to improve Pgp studies could be useful to optimize the sensitivity and the reproducibility of efflux assays. This study aimed to compare Pgp expression and efflux activity by measuring Rhodamine123 (Rh123) retention in lymphocytes stored under different conditions, in order to evaluate the potential utility of any of the storing conditions in Pgp functionality. Our results show no change in protein expression of Pgp by confocal studies and Western blotting, nor changes at the mRNA level (qRT-PCR). No differences in Rh123 efflux by Pgp activity assays were found between fresh and frozen lymphocytes after 24 hours of blood extraction, using either of the two Pgp specific inhibitors (VP and PSC833). Different working conditions in the 24 hours post blood extraction do not affect Rh123 efflux. These results allow standardization of Pgp activity measurement in different individuals with different timing of blood sampling and in different geographic areas.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Fluorescent Dyes/metabolism , Lymphocytes/physiology , Rhodamine 123/metabolism , ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors , ATP Binding Cassette Transporter, Subfamily B, Member 1/biosynthesis , Adult , Annexin A5/genetics , Apoptosis , CD3 Complex , Cell Line, Tumor , Cell Survival , Cryopreservation , Dactinomycin/analogs & derivatives , Female , Flow Cytometry , Humans , Male , Middle Aged , RNA, Messenger/metabolism , Time Factors , Young Adult
18.
J Leukoc Biol ; 80(6): 1473-9, 2006 Dec.
Article in English | MEDLINE | ID: mdl-16940331

ABSTRACT

Apoptosis of B cell chronic lymphocytic leukemia (B-CLL) cells is regulated by the PI-3K-Akt pathway. In the present work, we have analyzed the mechanisms of Akt phosphorylation in B-CLL cells. Freshly isolated cells present basal Akt phosphorylation, which is PI-3K-dependent, as incubation with the PI-3K inhibitor LY294002 decreased Ser-473 and Thr-308 phosphorylation in most samples analyzed (seven out of 10). In three out of 10 cases, inhibition of protein kinase C (PKC) inhibited basal Akt phosphorylation. Stromal cell-derived factor-1alpha, IL-4, and B cell receptor activation induced PI-3K-dependent Akt phosphorylation. PMA induced the phosphorylation of Akt at Ser-473 and Thr-308 and the phosphorylation of Akt substrates, independently of PI-3K in B-CLL cells. In contrast, PKC-mediated phosphorylation of Akt was PI-3K-dependent in normal B cells. Finally, a specific inhibitor of PKCbeta blocked the phosphorylation and activation of Akt by PMA in B-CLL cells. Taken together, these results suggest a model in which Akt could be activated by two different pathways (PI-3K and PKCbeta) in B-CLL cells.


Subject(s)
Apoptosis , Leukemia, Lymphocytic, Chronic, B-Cell/enzymology , Phosphatidylinositol 3-Kinases/metabolism , Protein Kinase C/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Apoptosis/drug effects , Carcinogens/pharmacology , Chromones/pharmacology , Enzyme Inhibitors/pharmacology , Humans , Models, Biological , Morpholines/pharmacology , Phosphoinositide-3 Kinase Inhibitors , Phosphorylation/drug effects , Protein Kinase C/antagonists & inhibitors , Protein Kinase C beta , Protein Processing, Post-Translational/drug effects , Receptors, Cytokine/metabolism , Signal Transduction/drug effects , Tetradecanoylphorbol Acetate/pharmacology , Tumor Cells, Cultured
20.
Neurochem Int ; 49(3): 276-84, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16522342

ABSTRACT

The brain tissue obtained after death is subjected to several circumstances that can affect RNA integrity. The present study has been directed to reveal possible pitfalls and to control RNA normalization in post-mortem samples in order to recognize the limitations and minimize errors when using TaqMan PCR technology. This has been carried out in samples of the frontal cortex in a series of control and diseased cases covering Parkinson's disease, dementia with Lewy bodies pure form and common form, and Alzheimer's disease. Special attention has been paid to the value of the agonal state, post-mortem delay and pH of the nervous tissue as approximate predictors of the quality of RNA, as well as to the use of the Bioanalyzer to confirm RNA preservation. In addition, since possible disease-modified mRNAs have to be normalized with ideal unaltered RNAs, TaqMan human endogenous control plates have been used to determine the endogenous control most appropriate for the study. beta-glucuronidase (GUS) and beta-actin were good endogenous controls because their expression levels showed a small variation across a representative number of control and pathological cases. RNA stability was also analysed in a paradigm mimicking cumulative delay in tissue processing. GUS mRNA levels were not modified although beta-actin mRNA levels showed degradation at 22 h. Finally, the control of RNA degradation for the normalization of genes of interest was also tested. mRNA expression levels for superoxide dismutase 1 (SOD1) and metalloproteinase domain 22 (ADAM22) were examined at several artificial post-mortem times, and their expression levels compared with those for putative controls beta-actin and GUS. In our paradigm, the expressions of SOD1 and ADAM22 were apparently not modified when normalized with beta-actin. Yet their expression levels were reduced with post-mortem delay when values were normalized with GUS. Taken together, these observations point to practical consequences in TaqMan PCR studies. Short post-mortem delays and acceptable pH of the brain are not sufficient to rule out RNA degradation. The selection of adequate endogenous controls is pivotal in the study. beta-actin and GUS are found to be good endogenous controls in these pathologies, although GUS but not beta-actin expression levels are preserved in samples with long post-mortem delay.


Subject(s)
Brain/metabolism , Brain/pathology , Polymerase Chain Reaction/methods , RNA Stability/physiology , RNA, Messenger/biosynthesis , Aged , Aged, 80 and over , Female , Gene Expression Regulation/physiology , Humans , Male , Middle Aged , RNA, Messenger/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...