Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
1.
Pharmaceuticals (Basel) ; 17(5)2024 Apr 30.
Article in English | MEDLINE | ID: mdl-38794149

ABSTRACT

Glioblastoma (GB) is the most aggressive and common primary malignant tumor of the brain and central nervous system. Without treatment, the average patient survival time is about six months, which can be extended to fifteen months with multimodal therapies. The chemoresistance observed in GB is, in part, attributed to the presence of a subpopulation of glioblastoma-like stem cells (GSCs) that are characterized by heightened tumorigenic capacity and chemoresistance. GSCs are situated in hypoxic tumor niches, where they sustain and promote the stem-like phenotype and have also been correlated with high chemoresistance. GSCs have the particularity of generating high levels of extracellular adenosine (ADO), which causes the activation of the A3 adenosine receptor (A3AR) with a consequent increase in the expression and activity of genes related to chemoresistance. Therefore, targeting its components is a promising alternative for treating GB. This analysis determined genes that were up- and downregulated due to A3AR blockades under both normoxic and hypoxic conditions. In addition, possible candidates associated with chemoresistance that were positively regulated by hypoxia and negatively regulated by A3AR blockades in the same condition were analyzed. We detected three potential candidate genes that were regulated by the A3AR antagonist MRS1220 under hypoxic conditions: LIMD1, TRIB2, and TGFB1. Finally, the selected markers were correlated with hypoxia-inducible genes and with the expression of adenosine-producing ectonucleotidases. In conclusion, we detected that hypoxic conditions generate extensive differential gene expression in GSCs, increasing the expression of genes associated with chemoresistance. Furthermore, we observed that MRS1220 could regulate the expression of LIMD1, TRIB2, and TGFB1, which are involved in chemoresistance and correlate with a poor prognosis, hypoxia, and purinergic signaling.

2.
Cells ; 13(10)2024 May 16.
Article in English | MEDLINE | ID: mdl-38786068

ABSTRACT

Induction of the adenosine receptor A2B (A2BAR) expression in diabetic glomeruli correlates with an increased abundance of its endogenous ligand adenosine and the progression of kidney dysfunction. Remarkably, A2BAR antagonism protects from proteinuria in experimental diabetic nephropathy. We found that A2BAR antagonism preserves the arrangement of podocytes on the glomerular filtration barrier, reduces diabetes-induced focal adhesion kinase (FAK) activation, and attenuates podocyte foot processes effacement. In spreading assays using human podocytes in vitro, adenosine enhanced the rate of cell body expansion on laminin-coated glass and promoted peripheral pY397-FAK subcellular distribution, while selective A2BAR antagonism impeded these effects and attenuated the migratory capability of podocytes. Increased phosphorylation of the Myosin2A light chain accompanied the effects of adenosine. Furthermore, when the A2BAR was stimulated, the cells expanded more broadly and more staining of pS19 myosin was detected which co-localized with actin cables, suggesting increased contractility potential in cells planted onto a matrix with a stiffness similar to of the glomerular basement membrane. We conclude that A2BAR is involved in adhesion dynamics and contractile actin bundle formation, leading to podocyte foot processes effacement. The antagonism of this receptor may be an alternative to the intervention of glomerular barrier deterioration and proteinuria in the diabetic kidney disease.


Subject(s)
Cell Adhesion , Diabetes Mellitus, Experimental , Focal Adhesion Protein-Tyrosine Kinases , Podocytes , Proteinuria , Receptor, Adenosine A2B , Podocytes/metabolism , Podocytes/drug effects , Podocytes/pathology , Animals , Humans , Proteinuria/metabolism , Rats , Receptor, Adenosine A2B/metabolism , Cell Adhesion/drug effects , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Male , Diabetic Nephropathies/metabolism , Diabetic Nephropathies/pathology , Diabetic Nephropathies/drug therapy , Adenosine A2 Receptor Antagonists/pharmacology , Adenosine/metabolism , Adenosine/pharmacology , Cell Movement/drug effects , Phosphorylation/drug effects , Myosin Light Chains/metabolism
3.
Int J Mol Sci ; 24(8)2023 Apr 11.
Article in English | MEDLINE | ID: mdl-37108208

ABSTRACT

Glioblastoma (GBM) is the most common and malignant primary brain cancer in adults. Without treatment the mean patient survival is approximately 6 months, which can be extended to 15 months with the use of multimodal therapies. The low effectiveness of GBM therapies is mainly due to the tumor infiltration into the healthy brain tissue, which depends on GBM cells' interaction with the tumor microenvironment (TME). The interaction of GBM cells with the TME involves cellular components such as stem-like cells, glia, endothelial cells, and non-cellular components such as the extracellular matrix, enhanced hypoxia, and soluble factors such as adenosine, which promote GBM's invasiveness. However, here we highlight the role of 3D patient-derived glioblastoma organoids cultures as a new platform for study of the modeling of TME and invasiveness. In this review, the mechanisms involved in GBM-microenvironment interaction are described and discussed, proposing potential prognosis biomarkers and new therapeutic targets.


Subject(s)
Brain Neoplasms , Glioblastoma , Adult , Humans , Glioblastoma/therapy , Glioblastoma/pathology , Brain Neoplasms/therapy , Brain Neoplasms/pathology , Endothelial Cells/pathology , Brain/pathology , Extracellular Matrix/pathology , Tumor Microenvironment , Cell Line, Tumor
4.
Int J Mol Sci ; 23(16)2022 Aug 12.
Article in English | MEDLINE | ID: mdl-36012307

ABSTRACT

Glioblastoma is the most common and aggressive primary brain tumor, characterized by its high chemoresistance and the presence of a cell subpopulation that persists under hypoxic niches, called glioblastoma stem-like cells (GSCs). The chemoresistance of GSCs is mediated in part by adenosine signaling and ABC transporters, which extrude drugs outside the cell, such as the multidrug resistance-associated proteins (MRPs) subfamily. Adenosine promotes MRP1-dependent chemoresistance under normoxia. However, adenosine/MRPs-dependent chemoresistance under hypoxia has not been studied until now. Transcript and protein levels were determined by RT-qPCR and Western blot, respectively. MRP extrusion capacity was determined by intracellular 5 (6)-Carboxyfluorescein diacetate (CFDA) accumulation. Cell viability was measured by MTS assays. Cell cycle and apoptosis were determined by flow cytometry. Here, we show for the first time that MRP3 expression is induced under hypoxia through the A2B adenosine receptor. Hypoxia enhances MRP-dependent extrusion capacity and the chemoresistance of GSCs. Meanwhile, MRP3 knockdown decreases GSC viability under hypoxia. Downregulation of the A2B receptor decreases MRP3 expression and chemosensibilizes GSCs treated with teniposide under hypoxia. These data suggest that hypoxia-dependent activation of A2B adenosine receptor promotes survival of GSCs through MRP3 induction.


Subject(s)
Brain Neoplasms , Glioblastoma , Multidrug Resistance-Associated Proteins , Adenosine/metabolism , Brain Neoplasms/metabolism , Drug Resistance, Neoplasm , Glioblastoma/metabolism , Humans , Hypoxia/metabolism , Multidrug Resistance-Associated Proteins/metabolism , Neoplastic Stem Cells/metabolism , Receptor, Adenosine A2B/metabolism , Receptors, Purinergic P1/metabolism
5.
Biology (Basel) ; 11(2)2022 Feb 16.
Article in English | MEDLINE | ID: mdl-35205179

ABSTRACT

Glioblastoma (GBM) is the most frequent and aggressive brain tumor, characterized by great resistance to treatments, as well as inter- and intra-tumoral heterogeneity. GBM exhibits infiltration, vascularization and hypoxia-associated necrosis, characteristics that shape a unique microenvironment in which diverse cell types are integrated. A subpopulation of cells denominated GBM stem-like cells (GSCs) exhibits multipotency and self-renewal capacity. GSCs are considered the conductors of tumor progression due to their high tumorigenic capacity, enhanced proliferation, invasion and therapeutic resistance compared to non-GSCs cells. GSCs have been classified into two molecular subtypes: proneural and mesenchymal, the latter showing a more aggressive phenotype. Tumor microenvironment and therapy can induce a proneural-to-mesenchymal transition, as a mechanism of adaptation and resistance to treatments. In addition, GSCs can transition between quiescent and proliferative substates, allowing them to persist in different niches and adapt to different stages of tumor progression. Three niches have been described for GSCs: hypoxic/necrotic, invasive and perivascular, enhancing metabolic changes and cellular interactions shaping GSCs phenotype through metabolic changes and cellular interactions that favor their stemness. The phenotypic flexibility of GSCs to adapt to each niche is modulated by dynamic epigenetic modifications. Methylases, demethylases and histone deacetylase are deregulated in GSCs, allowing them to unlock transcriptional programs that are necessary for cell survival and plasticity. In this review, we described the effects of GSCs plasticity on GBM progression, discussing the role of GSCs niches on modulating their phenotype. Finally, we described epigenetic alterations in GSCs that are important for stemness, cell fate and therapeutic resistance.

6.
Int J Mol Sci ; 21(22)2020 Nov 21.
Article in English | MEDLINE | ID: mdl-33233484

ABSTRACT

Pretransplant graft inflammation could be involved in the worse prognosis of deceased donor (DD) kidney transplants. A2A adenosine receptor (A2AR) can stimulate anti-inflammatory M2 macrophages, leading to fibrosis if injury and inflammation persist. Pre-implantation biopsies of kidney donors (47 DD and 21 living donors (LD)) were used to analyze expression levels and activated intracellular pathways related to inflammatory and pro-fibrotic processes. A2AR expression and PKA pathway were enhanced in DD kidneys. A2AR gene expression correlated with TGF-ß1 and other profibrotic markers, as well as CD163, C/EBPß, and Col1A1, which are highly expressed in DD kidneys. TNF-α mRNA levels correlated with profibrotic and anti-inflammatory factors such as TGF-ß1 and A2AR. Experiments with THP-1 cells point to the involvement of the TNF-α/NF-κB pathway in the up-regulation of A2AR, which induces the M2 phenotype increasing CD163 and TGF-ß1 expression. In DD kidneys, the TNF-α/NF-κB pathway could be involved in the increase of A2AR expression, which would activate the PKA-CREB axis, inducing the macrophage M2 phenotype, TGF-ß1 production, and ultimately, fibrosis. Thus, in inflamed DD kidneys, an increase in A2AR expression is associated with the onset of fibrosis, which may contribute to graft dysfunction and prognostic differences between DD and LD transplants.


Subject(s)
Cyclic AMP Response Element-Binding Protein/genetics , Cyclic AMP-Dependent Protein Kinases/genetics , Kidney Transplantation , Receptor, Adenosine A2A/genetics , Fibrosis/genetics , Fibrosis/pathology , Fibrosis/therapy , Gene Expression Regulation/genetics , Humans , Inflammation/genetics , Inflammation/pathology , Inflammation/therapy , Kidney/metabolism , Kidney/pathology , Macrophages/metabolism , Macrophages/pathology , NF-kappa B/genetics , Tissue Donors , Transforming Growth Factor beta1/genetics , Tumor Necrosis Factor-alpha/genetics
7.
Cells ; 9(8)2020 08 18.
Article in English | MEDLINE | ID: mdl-32824670

ABSTRACT

Glioblastoma multiforme is one of the most malignant types of cancer. This is mainly due to a cell subpopulation with an extremely aggressive potential, called glioblastoma stem-like cells (GSCs). These cells produce high levels of extracellular adenosine which has been associated with increased chemoresistance, migration, and invasion in glioblastoma. In this study, we attempted to elucidate the mechanisms that control extracellular adenosine levels in GSC subtypes. By using primary and U87MG-derived GSCs, we associated increased extracellular adenosine with the mesenchymal phenotype. [3H]-adenosine uptake occurred mainly through the equilibrative nucleoside transporters (ENTs) in GSCs, but mesenchymal GSCs have lower expression and ENT1-mediated uptake activity than proneural GSCs. By analyzing expression and enzymatic activity, we determined that ecto-5'-nucleotidase (CD73) is predominantly expressed in proneural GSCs, driving AMPase activity. While in mesenchymal GSCs, both CD73 and Prostatic Acid Phosphatase (PAP) contribute to the AMP (adenosine monophosphate) hydrolysis. We did not observe significant differences between the expression of proteins involved in the metabolization of adenosine among the GCSs subtypes. In conclusion, the lower expression and activity of the ENT1 transporter in mesenchymal GSCs contributes to the high level of extracellular adenosine that these GSCs present.


Subject(s)
Adenosine/metabolism , Brain Neoplasms/metabolism , Equilibrative Nucleoside Transporter 1/metabolism , Extracellular Space/metabolism , Glioblastoma/metabolism , Neoplastic Stem Cells/metabolism , 5'-Nucleotidase/metabolism , Acid Phosphatase/metabolism , Biological Transport , Brain Neoplasms/pathology , Cell Line, Tumor , GPI-Linked Proteins/metabolism , Glioblastoma/pathology , Humans
8.
Biochim Biophys Acta Mol Basis Dis ; 1866(7): 165796, 2020 07 01.
Article in English | MEDLINE | ID: mdl-32289379

ABSTRACT

Progressive diabetic nephropathy (DN) and loss of renal function correlate with kidney fibrosis. Crosstalk between TGF-ß and adenosinergic signaling contributes to the phenotypic transition of cells and to renal fibrosis in DN models. We evaluated the role of TGF-ß on NT5E gene expression coding for the ecto-5`-nucleotidase CD73, the limiting enzyme in extracellular adenosine production. We showed that high d-glucose may predispose HK-2 cells towards active transcription of the proximal promoter region of the NT5E gene while additional TGF-ß results in full activation. The epigenetic landscape of the NT5E gene promoter was modified by concurrent TGF-ß with occupancy by the p300 co-activator and the phosphorylated forms of the Smad2/3 complex and RNA Pol II. Transcriptional induction at NT5E in response to TGF-ß was earlier compared to the classic responsiveness genes PAI-1 and Fn1. CD73 levels and AMPase activity were concomitantly increased by TGF-ß in HK-2 cells. Interestingly, we found increased CD73 content in urinary extracellular vesicles only in diabetic patients with renal repercussions. Further, CD73-mediated AMPase activity was increased in the urinary sediment of DN patients. We conclude that the NT5E gene is a target of the profibrotic TGF-ß cascade and is a traceable marker of progressive DN.


Subject(s)
5'-Nucleotidase/genetics , Diabetic Nephropathies/genetics , Fibrosis/genetics , Transforming Growth Factor beta/genetics , Adenosine/biosynthesis , Biomarkers/metabolism , Cell Line , Diabetic Nephropathies/pathology , E1A-Associated p300 Protein/genetics , Epigenesis, Genetic/genetics , Epithelial Cells/metabolism , Epithelial Cells/pathology , Fibrosis/pathology , GPI-Linked Proteins/genetics , Gene Expression Regulation , Humans , Kidney/metabolism , Kidney/pathology , Kidney Tubules, Proximal/metabolism , Kidney Tubules, Proximal/pathology , Nucleotidases/genetics , Promoter Regions, Genetic/genetics , RNA Polymerase II/genetics
9.
Cells ; 9(4)2020 04 23.
Article in English | MEDLINE | ID: mdl-32340145

ABSTRACT

Diabetic nephropathy (DN) is considered the main cause of kidney disease in which myofibroblasts lead to renal fibrosis. Macrophages were recently identified as the major source of myofibroblasts in a process known as macrophage-myofibroblast transition (MMT). Adenosine levels increase during DN and in vivo administration of MRS1754, an antagonist of the A2B adenosine receptor (A2BAR), attenuated glomerular fibrosis (glomerulosclerosis). We aimed to investigate the association between A2BAR and MMT in glomerulosclerosis during DN. Kidneys/glomeruli of non-diabetic, diabetic, and MRS1754-treated diabetic (DM+MRS1754) rats were processed for histopathologic, transcriptomic, flow cytometry, and cellular in vitro analyses. Macrophages were used for in vitro cell migration/transmigration assays and MMT studies. In vivo MRS1754 treatment attenuated the clinical and histopathological signs of glomerulosclerosis in DN rats. Transcriptomic analysis demonstrated a decrease in chemokine-chemoattractants/cell-adhesion genes of monocytes/macrophages in DM+MRS1754 glomeruli. The number of intraglomerular infiltrated macrophages and MMT cells increased in diabetic rats. This was reverted by MRS1754 treatment. In vitro cell migration/transmigration decreased in macrophages treated with MRS1754. Human macrophages cultured with adenosine and/or TGF-ß induced MMT, a process which was reduced by MRS1754. We concluded that pharmacologic blockade of A2BAR attenuated some clinical signs of renal dysfunction and glomerulosclerosis, and decreased intraglomerular macrophage infiltration and MMT in DN rats.


Subject(s)
Diabetic Nephropathies/metabolism , Diabetic Nephropathies/pathology , Macrophages/pathology , Monocytes/pathology , Myofibroblasts/pathology , Receptor, Adenosine A2B/metabolism , Acetamides/pharmacology , Adenosine A2 Receptor Antagonists/pharmacology , Animals , Biomarkers/metabolism , Cell Adhesion Molecules/metabolism , Chemokines/metabolism , Chemotactic Factors/pharmacology , Fibrosis , Humans , Kidney Glomerulus/drug effects , Kidney Glomerulus/pathology , Macrophages/drug effects , Macrophages/metabolism , Male , Monocytes/drug effects , Monocytes/metabolism , Myofibroblasts/drug effects , Myofibroblasts/metabolism , Purines/pharmacology , Rats, Sprague-Dawley , Transcription, Genetic/drug effects
10.
Cells ; 8(11)2019 10 30.
Article in English | MEDLINE | ID: mdl-31671624

ABSTRACT

Glioblastoma is the brain tumor with the worst prognosis. This is mainly due to a cell subpopulation with an extremely aggressive potential, called glioblastoma stem-like cells (GSCs). These cells produce high levels of extracellular adenosine, which are increased even more under hypoxic conditions. Under hypoxia, adenosine signaling is related to HIF-2α expression, enhancing cell aggressiveness. Adenosine can be degraded using recombinant adenosine deaminase (ADA) to revert its pathological effects. The aim of this study was to degrade adenosine using ADA in order to decrease malignancy of GSCs. Adenosine depletion was performed using recombinant ADA. Migration and invasion were measured by transwell and matrigel-coated transwell assay, respectively. HIF-2α-dependent cell migration/invasion decreased in GSCs treated with ADA under hypoxia. MRPs-mediated chemoresistance and colony formation decreased in treatment with ADA. In conclusion, adenosine depletion using adenosine deaminase decreases GSCs aggressiveness.


Subject(s)
Adenosine/deficiency , Brain Neoplasms/pathology , Cell Movement , Cell Proliferation , Drug Resistance, Neoplasm , Glioblastoma/pathology , Neoplastic Stem Cells/pathology , Adenosine/metabolism , Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis , Basic Helix-Loop-Helix Transcription Factors/metabolism , Brain Neoplasms/drug therapy , Brain Neoplasms/metabolism , Cell Adhesion , Glioblastoma/drug therapy , Glioblastoma/metabolism , Humans , Hypoxia , Neoplasm Invasiveness , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Tumor Cells, Cultured , Vincristine/pharmacology
11.
Int J Mol Sci ; 20(18)2019 Sep 13.
Article in English | MEDLINE | ID: mdl-31540220

ABSTRACT

Diabetic nephropathy (DN) is the main cause of end-stage renal disease, which remains incurable. The progression of DN is associated with progressive and irreversible renal fibrosis and also high levels of adenosine. Our aim was to evaluate the effects of ADORA3 antagonism on renal injury in streptozotocin-induced diabetic rats. An ADORA3 antagonist that was administered in diabetic rats greatly inhibited the levels of inflammatory interleukins IL-1ß and IL-18, meanwhile when adenosine deaminase was administered, there was a non-selective attenuation of the inflammatory mediators IL-1ß, IL-18, IL-6, and induction of IL-10. The ADORA3 antagonist attenuated the high glucose-induced activation of caspase 1 in HK2 cells in vitro. Additionally, ADORA3 antagonisms blocked the increase in caspase 1 and the nuclear localization of NFκB in the renal tubular epithelium of diabetic rats, both events that are involved in regulating the production and activation of IL-1ß and IL-18. The effects of the A3 receptor antagonist resulted in the attenuation of kidney injury, as evidenced by decreased levels of the pro-fibrotic marker α-SMA at histological levels and the restoration of proteinuria in diabetic rats. We conclude that ADORA3 antagonism represents a potential therapeutic target that mechanistically works through the selective blockade of the NLRP3 inflammasome.


Subject(s)
Adenosine A3 Receptor Antagonists/administration & dosage , Caspase 1/metabolism , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/drug therapy , Diabetic Nephropathies/prevention & control , Adenosine A3 Receptor Antagonists/pharmacology , Adenosine Deaminase/adverse effects , Animals , Cell Line , Diabetes Mellitus, Experimental/enzymology , Diabetic Nephropathies/chemically induced , Disease Models, Animal , Gene Expression Regulation/drug effects , Humans , Injections, Intraperitoneal , Interleukin-18/metabolism , Interleukin-1beta/metabolism , Kidney Tubules/drug effects , Kidney Tubules/enzymology , Male , Rats , Streptozocin
12.
Cancer Lett ; 446: 112-122, 2019 04 01.
Article in English | MEDLINE | ID: mdl-30660649

ABSTRACT

Glioblastoma (GBM) is the brain tumor with the worst prognosis composed of a cell subpopulation called Glioblastoma Stem-like Cells (GSCs) responsible for tumor recurrence mediated by cell invasion. GSCs persist in a hypoxic microenvironment which promotes extracellular adenosine production and activation of the A3 Adenosine Receptor (A3AR), therefore, the aim of this study was to determine the role of extracellular adenosine and A3AR on GSCs invasion under hypoxia. GSCs were obtained from a U87MG cell line and primary cultures of GBM patients, and then incubated under normoxia or hypoxia. Gene expression was evaluated by RNAseq, RT-qPCR, and western blot. Cell migration was measured by spreading and transwell boyden chamber assays; cell invasion was evaluated by Matrigel-coated transwell, ex vivo brain slice, and in vivo xenograft assays. The contribution of A3AR on cell migration/invasion was evaluated using the A3AR antagonist, MRS1220. Extracellular adenosine production was higher under hypoxia than normoxia, mainly by the catalytic action of the prostatic acid phosphatase (PAP), promoting cell migration/invasion in a HIF-2-dependent process. A3AR blockade decreased cell migration/invasion and the expression of Epithelial-Mesenchymal Transition markers. In conclusion, high levels of extracellular adenosine production enhance cell migration/invasion of GSCs, through HIF-2/PAP-dependent activation of A3AR under hypoxia.


Subject(s)
Adenosine/metabolism , Brain Neoplasms/metabolism , Cell Movement , Glioblastoma/metabolism , Neoplastic Stem Cells/metabolism , Receptor, Adenosine A3/metabolism , Acid Phosphatase/genetics , Acid Phosphatase/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Cell Line, Tumor , Glioblastoma/genetics , Glioblastoma/pathology , Humans , Mice, Inbred NOD , Mice, SCID , Neoplasm Invasiveness , Neoplastic Stem Cells/pathology , Receptor, Adenosine A3/genetics , Signal Transduction , Tumor Cells, Cultured , Tumor Hypoxia , Tumor Microenvironment
13.
Int J Mol Sci ; 19(4)2018 Apr 18.
Article in English | MEDLINE | ID: mdl-29670017

ABSTRACT

Glioblastoma (GBM) is a neoplasm characterized by an extensive blood vessel network. Hypoxic niches of GBM can induce tumorigenic properties of a small cell subpopulation called Glioblastoma stem-like cells (GSCs) and can also increase extracellular adenosine generation which activates the A3 adenosine receptor (A3AR). Moreover, GSCs potentiates the persistent neovascularization in GBM. The aim of this study was to determine if A3AR blockade can reduce the vasculogenesis mediated by the differentiation of GSCs to Endothelial Cells (ECs) under hypoxia. We evaluated the expression of endothelial cell markers (CD31, CD34, CD144, and vWF) by fluorescence-activated cell sorting (FACS), and vascular endothelial growth factor (VEGF) secretion by ELISA using MRS1220 (A3AR antagonist) under hypoxia. We validate our results using U87MG-GSCs A3AR knockout (GSCsA3-KO). The effect of MRS1220 on blood vessel formation was evaluated in vivo using a subcutaneous GSCs-tumor model. GSCs increased extracellular adenosine production and A3AR expression under hypoxia. Hypoxia also increased the percentage of GSCs positive for endothelial cell markers and VEGF secretion, which was in turn prevented when using MRS1220 and in GSCsA3-KO. Finally, in vivo treatment with MRS1220 reduced tumor size and blood vessel formation. Blockade of A3AR decreases the differentiation of GSCs to ECs under hypoxia and in vivo blood vessel formation.


Subject(s)
Cell Differentiation , Endothelial Cells/metabolism , Endothelial Cells/pathology , Glioblastoma/metabolism , Glioblastoma/pathology , Receptor, Adenosine A3/metabolism , Adenosine/pharmacology , Adenosine A3 Receptor Antagonists/pharmacology , Animals , Biomarkers, Tumor/metabolism , Cell Differentiation/drug effects , Cell Hypoxia/drug effects , Cell Line, Tumor , Endothelial Cells/drug effects , Humans , Male , Models, Biological , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Neovascularization, Physiologic/drug effects , Rats, Sprague-Dawley
14.
Mol Aspects Med ; 60: 38-51, 2018 04.
Article in English | MEDLINE | ID: mdl-29222067

ABSTRACT

The role of extracellular vesicles in cancer biology has emerged as a focus of the study of great importance and has been shown to directly influence tumour development in several cancers including brain tumours, such as gliomas. Gliomas are the most aggressive brain tumours, and in the last time, a considerable effort has been made to understand their biology. Studies focus in the signalling pathways involved in the processes of angiogenesis, viability, drug resistance and immune response evasion, as well as gliomas ability to infiltrate healthy tissue, a phenomenon regulated by the migratory and invasive capacity of the cells within a tumour. In this review, we summarize the different types and classifications of extracellular vesicles, their intravesicular content, and their role in the regulation of tumour progression processes in glioma.


Subject(s)
Brain Neoplasms/diagnosis , Brain Neoplasms/metabolism , Extracellular Vesicles/metabolism , Glioblastoma/diagnosis , Glioblastoma/metabolism , Animals , Biomarkers, Tumor , Cell-Derived Microparticles/metabolism , Disease Progression , Exosomes/metabolism , Humans , Tumor Microenvironment
15.
Sci Rep ; 7(1): 9439, 2017 08 25.
Article in English | MEDLINE | ID: mdl-28842605

ABSTRACT

Deficient insulin signaling is a key event mediating diabetic glomerulopathy. Additionally, diabetic kidney disease has been related to increased levels of adenosine. Therefore, we tested a link between insulin deficiency and dysregulated activity of the equilibrative nucleoside transporters (ENTs) responsible for controlling extracellular levels of adenosine. In ex vivo glomeruli, high D-glucose decreased nucleoside uptake mediated by ENT1 and ENT2 transporters, resulting in augmented extracellular levels of adenosine. This condition was reversed by exposure to insulin. Particularly, insulin through insulin receptor/PI3K pathway markedly upregulated ENT2 uptake activity to restores the extracellular basal level of adenosine. Using primary cultured rat podocytes as a cellular model, we found insulin was able to increase ENT2 maximal velocity of transport. Also, PI3K activity was necessary to maintain ENT2 protein levels in the long term. In glomeruli of streptozotocin-induced diabetic rats, insulin deficiency leads to decreased activity of ENT2 and chronically increased extracellular levels of adenosine. Treatment of diabetic rats with adenosine deaminase attenuated both the glomerular loss of nephrin and proteinuria. In conclusion, we evidenced ENT2 as a target of insulin signaling and sensitive to dysregulation in diabetes, leading to chronically increased extracellular adenosine levels and thereby setting conditions conducive to kidney injury.


Subject(s)
Adenosine/metabolism , Diabetic Nephropathies/genetics , Diabetic Nephropathies/metabolism , Equilibrative-Nucleoside Transporter 2/genetics , Insulin/metabolism , Animals , Biopsy , Diabetic Nephropathies/pathology , Equilibrative-Nucleoside Transporter 2/metabolism , Extracellular Space/metabolism , Gene Expression Regulation , Kinetics , Male , Phosphatidylinositol 3-Kinases/metabolism , Rats , Signal Transduction
16.
Purinergic Signal ; 13(4): 479-488, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28721552

ABSTRACT

We aim to investigate whether overweight/obese pregnant women have elevated plasma levels of adenosine associated with increased consumption of high-calorie food. Sixty women were included. They were divided into lean (n = 23 and n = 12) or overweight/obese (n = 7 and n = 18) non-pregnant and pregnant women, respectively. Clinical records and maternal blood samples were collected after informed consent. A self-reported dietary questionnaire was also completed. Plasma adenosine levels were determined with high-performance liquid chromatography. Biochemical parameters, including glucose, total protein, and lipid profile, were determined using standard colorimetric assays. Adenosine levels were higher in pregnant women than in non-pregnant women (18.7 ± 1.6 vs 10.8 ± 1.3 nM/µg protein, respectively, p < 0.0001). Overweight/obese pregnant women (21.9 ± 2.5 nM/µg protein) exhibited higher adenosine levels than lean pregnant (14.5 ± 1.0 nM/µg protein, p = 0.04) or non-pregnant women (11.7 ± 1.5 nM/µg protein, p = 0.0005). Also, pregnant women with elevated weight gain exhibited higher (26.2 ± 3.7 nM/µg protein) adenosine levels than those with adequate weight gain (14.9 ± 1.4 nM/µg protein, p = 0.03). These differences were not statistically significant compared with those of pregnant women with reduced weight gain (17.4 ± 2.1 nM/µg protein, p = 0.053). Body mass index and adenosine only in pregnant women were positively correlated (r = 0.39, p = 0.02). While, polyunsaturated fatty acid (PUFA) consumption was negatively correlated with plasma adenosine levels only in non-pregnant women (r = -0.33, p = 0.03). Pregnancy is associated with high plasma adenosine levels, which are further elevated in pregnant women who are overweight/obese. High PUFA intake might reduce plasma adenosine levels in non-pregnant women.


Subject(s)
Adenosine/blood , Obesity/blood , Overweight/blood , Pregnancy Complications/blood , Adult , Body Mass Index , Cross-Sectional Studies , Diet , Female , Humans , Pregnancy , Weight Gain
17.
Mol Aspects Med ; 55: 140-151, 2017 06.
Article in English | MEDLINE | ID: mdl-28223127

ABSTRACT

Glioblastoma multiforme (GBM) is considered the most common and aggressive tumour of the central nervous system and is characterized for being highly chemoresistant. This property is mainly due to the activation of Multiple Drug Resistance (MDR) mechanisms that protect cancer cells from structurally and morphologically different drugs. Overexpression and increased ABC transporters activity is one of the most important MDR mechanisms at the clinical level, and both its expression and activity are elevated in GBM cells. Within the tumour, there is a subpopulation called glioblastoma stem-like cells (GSCs), which due to its high tumourigenic capacity and chemoresistance, have been postulated as the main responsible for tumour recurrence. The GSCs inhabit hypoxic tumour zones, niches that apart from maintaining and promoting stem phenotype have also been correlated with high chemoresistance. Of the signalling pathways activated during hypoxia, purinergic signalling has been highly associated to the induction of MDR mechanisms. Through its receptors, the nucleoside adenosine has been shown to promotes the chemoresistance mediated by ABC transporters. Therefore, targeting its components is a promising alternative for GBM treatment. In this review, we will discuss chemoresistance in GSCs and the effect of the hypoxic microenvironment and adenosine on MDR mechanisms.


Subject(s)
Adenosine/genetics , Drug Resistance, Multiple/genetics , Drug Resistance, Neoplasm/genetics , Glioblastoma/drug therapy , ATP-Binding Cassette Transporters/genetics , Adenosine/metabolism , Cell Hypoxia/genetics , Glioblastoma/genetics , Glioblastoma/pathology , Humans , Neoplastic Stem Cells/drug effects , Signal Transduction , Tumor Microenvironment/genetics
18.
Mol Aspects Med ; 55: 75-89, 2017 06.
Article in English | MEDLINE | ID: mdl-28109856

ABSTRACT

Adenosine is a nucleoside that is particularly interesting to many scientific and clinical communities as it has important physiological and pathophysiological roles in the kidney. The distribution of adenosine receptors has only recently been elucidated; therefore it is likely that more biological roles of this nucleoside will be unveiled in the near future. Since the discovery of the involvement of adenosine in renal vasoconstriction and regulation of local renin production, further evidence has shown that adenosine signaling is also involved in the tubuloglomerular feedback mechanism, sodium reabsorption and the adaptive response to acute insults, such as ischemia. However, the most interesting finding was the increased adenosine levels in chronic kidney diseases such as diabetic nephropathy and also in non-diabetic animal models of renal fibrosis. When adenosine is chronically increased its signaling via the adenosine receptors may change, switching to a state that induces renal damage and produces phenotypic changes in resident cells. This review discusses the physiological and pathophysiological roles of adenosine and pays special attention to the mechanisms associated with switching homeostatic nucleoside levels to increased adenosine production in kidneys affected by CKD.


Subject(s)
Adenosine/genetics , Kidney/physiology , Renal Insufficiency, Chronic/physiopathology , Humans , Kidney/metabolism , Kidney Tubules/metabolism , Kidney Tubules/physiopathology , Renal Insufficiency, Chronic/genetics , Signal Transduction , Vasoconstriction/genetics , Vasoconstriction/physiology
19.
Mol Aspects Med ; 55: 45-61, 2017 06.
Article in English | MEDLINE | ID: mdl-27871900

ABSTRACT

Regulation of blood flow depends on systemic and local release of vasoactive molecules such as insulin and adenosine. These molecules cause vasodilation by activation of plasma membrane receptors at the vascular endothelium. Adenosine activates at least four subtypes of adenosine receptors (A1AR, A2AAR, A2BAR, A3AR), of which A2AAR and A2BAR activation leads to increased cAMP level, generation of nitric oxide, and relaxation of the underlying smooth muscle cell layer. Vasodilation caused by adenosine also depends on plasma membrane hyperpolarization due to either activation of intermediate-conductance Ca2+-activated K+ channels in vascular smooth muscle or activation of ATP-activated K+ channels in the endothelium. Adenosine also causes vasoconstriction via a mechanism involving A1AR activation resulting in lower cAMP level and increased thromboxane release. Insulin has also a dual effect causing NO-dependent vasodilation, but also sympathetic activity- and increased endothelin 1 release-dependent vasoconstriction. Interestingly, insulin effects require or are increased by activation or inactivation of adenosine receptors. This is phenomenon described for d-glucose and l-arginine transport where A2AAR and A2BAR play a major role. Other studies show that A1AR activation could reduce insulin release from pancreatic ß-cells. Whether adenosine modulation of insulin biological effect is a phenomenon that depends on co-localization of adenosine receptors and insulin receptors, and adenosine plasma membrane transporters is something still unclear. This review summarizes findings addressing potential involvement of adenosine receptors to modulate insulin effect via insulin receptors with emphasis in the human vasculature.


Subject(s)
Adenosine/metabolism , Endothelium, Vascular/metabolism , Insulin/metabolism , Adenosine/genetics , Endothelium, Vascular/pathology , Glucose/metabolism , Humans , Insulin/genetics , Muscle, Smooth, Vascular/metabolism , Nitric Oxide/metabolism , Receptor, Adenosine A2A/genetics , Receptor, Adenosine A2A/metabolism , Receptor, Adenosine A2B/genetics , Receptor, Adenosine A2B/metabolism , Receptor, Adenosine A3/genetics , Receptor, Adenosine A3/metabolism , Signal Transduction , Vasoconstriction/genetics
20.
Oncotarget ; 7(41): 67373-67386, 2016 Oct 11.
Article in English | MEDLINE | ID: mdl-27634913

ABSTRACT

MRP1 transporter correlates positively with glioma malignancy and the Multiple Drug Resistance (MDR) phenotype in Glioblastoma Multiforme (GBM). Evidence shows that the MRP1 transporter is controlled by the adenosine signalling axis. The aim of this study was to identify the role of adenosine on the MDR phenotype in Glioblastoma Stem-like Cells (GSCs), the cell population responsible for the tumorigenic and chemoresistance capabilities of this tumour. We found that GSCs have increased intrinsic capacity to generate extracellular adenosine, thus controlling MRP1 transporter expression and activity via activation of the adenosine A3 receptor (A3AR). We showed PI3K/Akt and MEK/ERK1/2 signaling pathways downstream A3AR to control MRP1 in GSCs. In vitro pharmacological blockade of A3AR had a chemosensitizing effect, enhancing the actions of antitumour drugs and decreasing cell viability and proliferation of GSCs. In addition, we produced an in vivo xenograft model by subcutaneous inoculation of human GSCs in NOD/SCID-IL2Rg null mice. Pharmacological blockade of A3AR generated a chemosensitizing effect, enhancing the effectiveness of the MRP1 transporter substrate, vincristine, reducing tumour size and the levels of CD44 and Nestin stem cell markers as well as the Ki-67 proliferation indicator. In conclusion, we demonstrated the chemosensitizing effect of A3AR blockade on GSCs.


Subject(s)
Brain Neoplasms/pathology , Drug Resistance, Neoplasm/physiology , Glioblastoma/pathology , Multidrug Resistance-Associated Proteins/metabolism , Neoplastic Stem Cells/pathology , Receptor, Adenosine A3/metabolism , Animals , Brain Neoplasms/metabolism , Glioblastoma/metabolism , Heterografts , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Neoplastic Stem Cells/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...