Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 24(13)2023 Jun 29.
Article in English | MEDLINE | ID: mdl-37446007

ABSTRACT

Some chemoattractants and leukocytes such as M1 and M2 macrophages are known to be involved in the development of glomerulosclerosis during diabetic nephropathy (DN). In the course of diabetes, an altered and defective cellular metabolism leads to the increase in adenosine levels, and thus to changes in the polarity (M1/M2) of macrophages. MRS1754, a selective antagonist of the A2B adenosine receptor (A2BAR), attenuated glomerulosclerosis and decreased macrophage-myofibroblast transition in DN rats. Therefore, we aimed to investigate the effect of MRS1754 on the glomerular expression/secretion of chemoattractants, the intraglomerular infiltration of leukocytes, and macrophage polarity in DN rats. Kidneys/glomeruli of non-diabetic, DN, and MRS1754-treated DN rats were processed for transcriptomic analysis, immunohistopathology, ELISA, and in vitro macrophage migration assays. The transcriptomic analysis identified an upregulation of transcripts and pathways related to the immune system in the glomeruli of DN rats, which was attenuated using MRS1754. The antagonism of the A2BAR decreased glomerular expression/secretion of chemoattractants (CCL2, CCL3, CCL6, and CCL21), the infiltration of macrophages, and their polarization to M2 in DN rats. The in vitro macrophages migration induced by conditioned-medium of DN glomeruli was significantly decreased using neutralizing antibodies against CCL2, CCL3, and CCL21. We concluded that the pharmacological blockade of the A2BAR decreases the transcriptional expression of genes/pathways related to the immune response, protein expression/secretion of chemoattractants, as well as the infiltration of macrophages and their polarization toward the M2 phenotype in the glomeruli of DN rats, suggesting a new mechanism implicated in the antifibrotic effect of MRS1754.


Subject(s)
Acetamides , Adenosine A2 Receptor Antagonists , Cell Polarity , Chemotactic Factors , Diabetic Nephropathies , Kidney Glomerulus , Macrophages , Purines , Diabetic Nephropathies/genetics , Diabetic Nephropathies/immunology , Kidney Glomerulus/drug effects , Kidney Glomerulus/metabolism , Chemotactic Factors/antagonists & inhibitors , Chemotactic Factors/genetics , Chemotactic Factors/metabolism , Cell Polarity/drug effects , Cell Polarity/immunology , Macrophages/drug effects , Macrophages/immunology , Adenosine A2 Receptor Antagonists/pharmacology , Receptor, Adenosine A2B , Acetamides/pharmacology , Purines/pharmacology , Animals , Rats , Cell Movement/drug effects , Male , Rats, Sprague-Dawley , Transcription, Genetic/drug effects , Protein Biosynthesis/drug effects , Immunity/drug effects , Immunity/genetics
2.
Clin Epigenetics ; 13(1): 56, 2021 03 16.
Article in English | MEDLINE | ID: mdl-33726839

ABSTRACT

BACKGROUND: An intronic GGGGCC (G4C2) hexanucleotide repeat expansion (HRE) in the C9ORF72 gene is the most common cause of amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD), referred to as C9ALS/FTD. No cure or effective treatment exist for C9ALS/FTD. Three major molecular mechanisms have emerged to explain C9ALS/FTD disease mechanisms: (1) C9ORF72 loss-of-function through haploinsufficiency, (2) dipeptide repeat (DPR) proteins mediated toxicity by the translation of the repeat RNAs, and more controversial, (3) RNA-mediated toxicity by bidirectional transcription of the repeats that form intranuclear RNA foci. Recent studies indicate a double-hit pathogenic mechanism in C9ALS/FTD, where reduced C9ORF72 protein levels lead to impaired clearance of toxic DPRs. Here we explored whether pharmacological compounds can revert these pathological hallmarks in vitro and cognitive impairment in a C9ALS/FTD mouse model (C9BAC). We specifically focused our study on small molecule inhibitors targeting chromatin-regulating proteins (epidrugs) with the goal of increasing C9ORF72 gene expression and reduce toxic DPRs. RESULTS: We generated luciferase reporter cell lines containing 10 (control) or ≥ 90 (mutant) G4C2 HRE located between exon 1a and 1b of the human C9ORF72 gene. In a screen of 14 different epidrugs targeting bromodomains, chromodomains and histone-modifying enzymes, we found that several bromodomain and extra-terminal domain (BET) inhibitors (BETi), including PFI-1 and JQ1, increased luciferase reporter activity. Using primary cortical cultures from C9BAC mice, we further found that PFI-1 treatment increased the expression of V1-V3 transcripts of the human mutant C9ORF72 gene, reduced poly(GP)-DPR inclusions but enhanced intranuclear RNA foci. We also tested whether JQ1, an BETi previously shown to reach the mouse brain by intraperitoneal (i.p.) injection, can revert behavioral abnormalities in C9BAC mice. Interestingly, it was found that JQ1 administration (daily i.p. administration for 7 days) rescued hippocampal-dependent cognitive deficits in C9BAC mice. CONCLUSIONS: Our findings place BET bromodomain inhibitors as a potential therapy for C9ALS/FTD by ameliorating C9ORF72-associated pathological and behavioral abnormalities. Our finding that PFI-1 increases accumulation of intranuclear RNA foci is in agreement with recent data in flies suggesting that nuclear RNA foci can be neuroprotective by sequestering repeat transcripts that result in toxic DPRs.


Subject(s)
C9orf72 Protein/genetics , Enzyme Inhibitors/metabolism , Epigenesis, Genetic , Frontotemporal Dementia/genetics , Frontotemporal Dementia/physiopathology , Histone Acetyltransferases/genetics , Transcription Factors/genetics , Animals , Cell Line , Disease Models, Animal , Gene Expression Regulation , Humans , Mice
3.
Immunotherapy ; 13(4): 309-326, 2021 03.
Article in English | MEDLINE | ID: mdl-33397152

ABSTRACT

Aim: Whole dead tumor cells can be used as antigen source and the induction of protective immune response could be enhanced by damage-associated molecular patterns. Materials & methods: We generated whole dead tumor cells called B16-immunogenic cell bodies (ICBs) from B16 melanoma cells by nutrient starvation and evaluated the in vivo antitumor effect of B16-ICBs plus ATP and polymyxin B (PMB). Results: The subcutaneous immunization with B16-ICBs + PMB + ATP a 50% of tumor-free animals and induced a significant delay in tumor growth in a prophylactic approach. These results correlated with maturation of bone marrow-derived dendritic cells and activation of T CD8+ lymphocytes in vitro. Conclusion: Altogether, ICB + ATP + PMB is efficient in inducing the antitumor efficacy of the whole dead tumor cells vaccine.


Subject(s)
Adenosine Triphosphate/immunology , Cancer Vaccines/immunology , Melanoma, Experimental/immunology , Polymyxin B/immunology , Adenosine Triphosphate/administration & dosage , Alarmins/administration & dosage , Alarmins/immunology , Animals , Antigen Presentation , Antigens, Neoplasm/immunology , CD40 Antigens/metabolism , Cancer Vaccines/administration & dosage , Cytokines/metabolism , Dendritic Cells/immunology , Dendritic Cells/metabolism , Immunization , Melanoma, Experimental/pathology , Melanoma, Experimental/prevention & control , Mice , Mice, Inbred C57BL , Phagocytosis , Polymyxin B/administration & dosage , Spleen/immunology , Tumor Cells, Cultured
5.
Diabetol Metab Syndr ; 12: 66, 2020.
Article in English | MEDLINE | ID: mdl-32774470

ABSTRACT

BACKGROUND: In type I diabetes mellitus (T1DM) pancreatic ß cells are destroyed. Treatment entails exogenous insulin administration and strict diet control, yet optimal glycemic control is hardly attainable. Islet transplant could be an alternative in patients with poor glycemic control, but inefficient islet purification and autoimmune response of patients is still a challenge. For these reasons, it is necessary to explore new cellular sources and immunological isolation methods oriented to develop T1DM cell-based therapies. AIMS: We postulate human adipose-derived stem cell (hASC) as an adequate source to generate pancreatic islet cells in vitro, and to produce islet-like structures. Furthermore, we propose microencapsulation of these aggregates as an immunological isolation strategy. METHODS: hASC obtained from lipoaspirated fat tissue from human donors were differentiated in vitro to insulin (Ins) and glucagon (Gcg) producing cells. Then, insulin producing cells (IPC) and glucagon producing cells (GPC) were cocultured in low adhesion conditions to form cellular aggregates, and later encapsulated in a sodium alginate polymer. Expression of pancreatic lineage markers and secretion of insulin or glucagon in vitro were analyzed. RESULTS: The results show that multipotent hASC efficiently differentiate to IPC and GPC, and express pancreatic markers, including insulin or glucagon hormones which they secrete upon stimulation (fivefold for insulin in IPC, and fourfold for glucagon, compared to undifferentiated cells). In turn, calculation of the Feret diameter and area of cellular aggregates revealed mean diameters of ~ 80 µm, and 65% of the aggregates reached 4000 µm2 at 72 h of formation. IPC/GPC aggregates were then microencapsulated in sodium-alginate polymer microgels, which were found to be more stable when stabilized with Ba2+, yielding average diameters of ~ 300 µm. Interestingly, Ba2+-microencapsulated aggregates respond to high external glucose with insulin secretion. CONCLUSIONS: The IPC/GPC differentiation process from hASC, followed by the generation of cellular aggregates that are later microencapsulated, could represent a possible treatment for T1DM.

6.
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
7.
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
8.
J Mol Biol ; 432(10): 3222-3238, 2020 05 01.
Article in English | MEDLINE | ID: mdl-32198114

ABSTRACT

Several mechanisms directing a rapid transcriptional reactivation of genes immediately after mitosis have been described. However, little is known about the maintenance of repressive signals during mitosis. In this work, we address the role of Ski in the repression of gene expression during M/G1 transition in mouse embryonic fibroblasts (MEFs). We found that Ski localises as a distinct pair of dots at the pericentromeric region of mitotic chromosomes, and the absence of the protein is related to high acetylation and low tri-methylation of H3K9 in pericentromeric major satellite. Moreover, differential expression assays in early G1 cells showed that the presence of Ski is significantly associated with repression of genes localised nearby to pericentromeric DNA. In mitotic cells, chromatin immunoprecipitation assays confirmed the association of Ski to major satellite and the promoters of the most repressed genes: Mmp3, Mmp10 and Mmp13. These genes are at pericentromeric region of chromosome 9. In these promoters, the presence of Ski resulted in increased H3K9 tri-methylation levels. This Ski-dependent regulation is also observed during interphase. Consequently, Mmp activity is augmented in Ski-/- MEFs. Altogether, these data indicate that association of Ski with the pericentromeric region of chromosomes during mitosis is required to maintain the silencing bookmarks of underlying chromatin.


Subject(s)
Centromere/genetics , DNA-Binding Proteins/metabolism , Fibroblasts/cytology , Histones/metabolism , Matrix Metalloproteinases, Secreted/genetics , Proto-Oncogene Proteins/metabolism , Acetylation , Animals , Cells, Cultured , Centromere/metabolism , Down-Regulation , Fibroblasts/metabolism , Matrix Metalloproteinase 10/genetics , Matrix Metalloproteinase 13/genetics , Matrix Metalloproteinase 3/genetics , Methylation , Mice , Mitosis , Promoter Regions, Genetic , Transcriptional Activation
10.
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
11.
Mol Oncol ; 12(2): 151-165, 2018 02.
Article in English | MEDLINE | ID: mdl-28614631

ABSTRACT

Increased expression of the TRPM4 channel has been reported to be associated with the progression of prostate cancer. However, the molecular mechanism underlying its effect remains unknown. This work found that decreasing TRPM4 levels leads to the reduced proliferation of PC3 cells. This effect was associated with a decrease in total ß-catenin protein levels and its nuclear localization, and a significant reduction in Tcf/Lef transcriptional activity. Moreover, TRPM4 silencing increases the Ser33/Ser37/Thr41 ß-catenin phosphorylated population and reduces the phosphorylation of GSK-3ß at Ser9, suggesting an increase in ß-catenin degradation as the underlying mechanism. Conversely, TRPM4 overexpression in LNCaP cells increases the Ser9 inhibitory phosphorylation of GSK-3ß and the total levels of ß-catenin and its nonphosphorylated form. Finally, PC3 cells with reduced levels of TRPM4 showed a decrease in basal and stimulated phosphoactivation of Akt1, which is likely responsible for the decrease in GSK-3ß activity in these cells. Our results also suggest that the effect of TRPM4 on Akt1 is probably mediated by an alteration in the calcium/calmodulin-EGFR axis, linking TRPM4 activity with the observed effects in ß-catenin-related signaling pathways. These results suggest a role for TRPM4 channels in ß-catenin oncogene signaling and underlying mechanisms, highlighting this ion channel as a new potential target for future therapies in prostate cancer.


Subject(s)
Cell Proliferation/genetics , Glycogen Synthase Kinase 3 beta/metabolism , Prostatic Neoplasms/metabolism , Proto-Oncogene Proteins c-akt/metabolism , TRPM Cation Channels/metabolism , beta Catenin/metabolism , Calcium/metabolism , Calmodulin/metabolism , Cell Line, Tumor , Disease Progression , Glycogen Synthase Kinase 3 beta/genetics , HEK293 Cells , Humans , Male , PC-3 Cells , Phosphorylation/genetics , Prostatic Neoplasms/genetics , Proto-Oncogene Proteins c-akt/genetics , TRPM Cation Channels/genetics , beta Catenin/genetics
12.
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
13.
Immunobiology ; 217(3): 307-15, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22118797

ABSTRACT

Regulatory T cells (Treg) are important in the development of immune tolerance under normal physiological conditions. However, in pathological situations such as cancer, Treg increases have been correlated with bad prognoses. Treg depletion can be achieved in vitro under several stimuli, including the activation of the purinergic P2X7 receptor. Our aim was to determine whether polymyxin B (PMB), which is a positive modulator of this receptor, could affect mice Treg depletion by ATP and related compounds. For that purpose, we evaluated by flow cytometry changes in Treg populations under several treatments with PMB and/or purinergic agonists and antagonists. We found that both ATP and NAD induce a dose-dependent decrease on the Treg CD4+ CD25+ population. PMB not only potentiated the effect of exogenous ATP and NAD, but also decreased the CD4+ CD25+ population when it was applied alone. While ATP mediated effects are related to the P2X7 receptor, PMB effects appear to be related to another mechanism. We conclude that PMB positively modulates the depletion of the CD4+ CD25+ population of Treg. Therefore PMB could constitute a non-canonical drug with potential use on Treg depletion and cancer treatment.


Subject(s)
Anti-Bacterial Agents/pharmacology , Lymphocyte Depletion , Polymyxin B/pharmacology , Purinergic Agonists/pharmacology , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , Adenosine Triphosphate/pharmacology , Animals , Cells, Cultured , Drug Synergism , Female , Immunophenotyping , Male , Mice , Mice, Inbred C57BL
SELECTION OF CITATIONS
SEARCH DETAIL
...