Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Cell Physiol Biochem ; 52(6): 1339-1360, 2019.
Article in English | MEDLINE | ID: mdl-31050282

ABSTRACT

BACKGROUND/AIMS: Melanocortin receptors (MCRs) belong to a hormonal signalling pathway with multiple homeostatic and protective actions. Microvascular and umbilical vein endothelial cells (ECs) express components of the melanocortin system, including the type 1 receptor (MC1R), playing a role in modulating inflammation and vascular tone. Since ECs exhibit a remarkable heterogeneity, we investigated whether human artery ECs express any functional MCR and whether its activation affects cell migration. METHODS: We used reverse transcription real-time PCR to examine the expression of melanocortin system components in primary human artery ECs. We assessed MC1R protein expression and activity by western blot, immunohistochemistry, cAMP production, and intracellular Ca²âº mobilization assays. We performed gap closure and scratch tests to examine cell migration after stimulation with alpha-melanocyte-stimulating hormone (α-MSH), the receptor highest-affinity natural ligand. We assessed differential time-dependent transcriptional changes in migrating cells by microarray analysis. RESULTS: We showed that human aortic ECs (HAoECs) express a functionally active MC1R. Unlike microvascular ECs, arterial cells did not express the α-MSH precursor proopiomelanocortin, nor produced the hormone. MC1R engagement with a single pulse of α-MSH accelerated HAoEC migration both in the directional migration assay and in the scratch wound healing test. This was associated with an enhancement in Ca²âº signalling and inhibition of cAMP elevation. Time-course genome-wide expression analysis in HAoECs undergoing directional migration allowed identifying dynamic co-regulation of genes involved in extracellular matrix-receptor interaction, vesicle-mediated trafficking, and metal sensing - which have all well-established influences on EC motility -, without affecting the balance between pro- and anticoagulant genes. CONCLUSION: Our work broadens the knowledge on peripherally expressed MC1R. These results indicate that the receptor is constitutively expressed by arterial ECs and provide evidence of a novel homeostatic function for MC1R, whose activation may participate in preventing/healing endothelial dysfunction or denudation in macrovascular arteries.


Subject(s)
Receptor, Melanocortin, Type 1/metabolism , Aorta/cytology , Calcium Signaling/drug effects , Cell Movement/drug effects , Cell Proliferation/drug effects , Cyclic AMP/metabolism , Egtazic Acid/analogs & derivatives , Egtazic Acid/pharmacology , Endothelial Cells/cytology , Endothelial Cells/metabolism , Gene Expression Regulation/drug effects , Humans , Ki-67 Antigen/metabolism , Oligopeptides/pharmacology , Receptor, Melanocortin, Type 1/genetics , alpha-MSH/pharmacology
2.
Data Brief ; 8: 836-50, 2016 Sep.
Article in English | MEDLINE | ID: mdl-27508230

ABSTRACT

We previously showed that Aminaphtone, a drug used in the treatment of chronic venous insufficiency, modulates several vasoactive factors, such as endothelin-1 and adhesion molecules. Here, we provide data of time-course experiments about the effects of Aminaphtone on gene expression at the genome-wide level in human endothelial cells undergoing cytokine stimulation in vitro. ECV-304 endothelial cells were incubated with interleukin-1ß (IL-1ß) in the presence or absence of Aminaphtone for 1, 3, and 6 h. Gene expression profiles were analyzed by microarray. This article contains complete data on the genes significantly modulated by the drug over time. The data are supplemental to our original research article reporting detailed analysis of the actions of Aminaphtone on IL-1ß stimulated endothelial cells at the molecular level, "Gene expression profiling reveals novel protective effects of Aminaphtone on ECV304 endothelial cells" (Salazar et al., 2016) [1].

3.
Eur J Pharmacol ; 782: 59-69, 2016 Jul 05.
Article in English | MEDLINE | ID: mdl-27083548

ABSTRACT

Aminaphtone, a drug used in the treatment of chronic venous insufficiency (CVI), showed a remarkable role in the modulation of several vasoactive factors, like endothelin-1 and adhesion molecules. We analysed in vitro the effects of Aminaphtone on whole-genome gene expression and production of different inflammatory proteins. ECV-304 endothelial cells were stimulated with IL-1ß 100U/ml in the presence or absence of Aminaphtone 6µg/ml. Gene expression profiles were compared at 1, 3, and 6h after stimulation by microarray. Supernatants of ECV-304 cultures were analysed at 3, 6, 12, and 24h by multiplex ELISA for production of several cytokine and chemokines. Microarrays showed a significant down-regulation at all times of a wide range of inflammatory genes. Aminaphtone appeared also able to modulate the regulation of immune response process (down-regulating cytokine biosynthesis, transcripts involved in lymphocyte differentiation and cell proliferation, and cytokine-cytokine receptor interaction) and to regulate genes engaged in homeostasis, secretion, body fluid levels, response to hypoxia, cell division, and cell-to-cell communication and signalling. Results were confirmed and extended analysing the secretome, which showed significant reduction of the release of 14 cytokines and chemokines. These effects are predicted to be mediated by interaction with different transcription factors. Aminaphtone was able to modulate the expression of inflammatory molecules relevant to the pathogenesis of several conditions in which the endothelial dysfunction is the main player and early event, like scleroderma, lung fibrosis, or atherosclerosis.


Subject(s)
Endothelial Cells/drug effects , Endothelial Cells/metabolism , Gene Expression Profiling , para-Aminobenzoates/pharmacology , Cell Line , Cytokines/genetics , Cytokines/metabolism , Humans , Immunomodulation/drug effects , Interleukin-1beta/pharmacology
4.
J Leukoc Biol ; 98(3): 409-21, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25990243

ABSTRACT

The function of human circulating PACs has been described extensively. However, little focus has been placed on understanding how these cells differ in their functions in the presence of microenvironments mimicking vascular inflammation. We hypothesized that exposure to proinflammatory cytokines or the oxLDL, an autoantigen abundant in advanced atherosclerotic plaques, converts PACs into immune-modulating/proinflammatory cells. Hence, we examined the effect of oxLDL and inflammatory stimuli on their phenotype by use of a functional genomics model based on secretome and whole genome transcriptome profiling. PACs obtained from culturing a PBMC fraction in angiogenic medium were primed with DC differentiation cytokines and then exposed to proinflammatory cytokines or oxLDL. Under these conditions, PACs converted into APCs, expressed maturation markers CD80 and CD83, and showed an increased up-regulation of CD86. APCcy and APCox induced a robust T cell BrdU incorporation. Despite a similar ability to induce lymphocyte proliferation, APCcy and APCox differed for the secretory pathway and mRNA expression. Analysis of the differentially expressed genes identified 4 gene "clusters," showing reciprocal modulation in APCcy vs. APCox, justifying, according to functional genomics analyses, a different putative function of the cells in antigen processing. Together, these data show that treatment with inflammatory cytokines or oxLDL converts human PAC phenotypes and functions into that of APCs with similar lymphocyte-activating ability but distinct maturation degree and paracrine functions.


Subject(s)
Antigen-Presenting Cells/pathology , Cell Movement/drug effects , Inflammation/pathology , Lipoproteins, LDL/pharmacology , Neovascularization, Physiologic/drug effects , Antigen Presentation/drug effects , Antigen-Presenting Cells/drug effects , Antigen-Presenting Cells/metabolism , Cell Differentiation/drug effects , Cluster Analysis , Cytokines/metabolism , Cytokines/pharmacology , Gene Expression Profiling , Gene Expression Regulation/drug effects , Genomics , Humans , Immunophenotyping , Mitogens/pharmacology , Oligonucleotide Array Sequence Analysis , Phenotype , T-Lymphocytes/drug effects , T-Lymphocytes/immunology
5.
Differentiation ; 82(1): 9-17, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21492995

ABSTRACT

High levels of DNA methyltransferase 1 (DNMT1), hypermethylation, and downregulation of GAD(67) and reelin have been described in GABAergic interneurons of patients with schizophrenia (SZ) and bipolar (BP) disorders. However, overexpression of DNMT1 is lethal, making it difficult to assess the direct effect of high levels of DNMT1 on neuronal development in vivo. We therefore used Dnmt1(tet/tet) mouse ES cells that overexpress DNMT1 as an in vitro model to investigate the impact of high levels of DNMT1 on neuronal differentiation. Although there is down-regulation of DNMT1 during early stages of differentiation in wild type and Dnmt1(tet/tet) ES cell lines, neurons derived from Dnmt1(tet/tet) cells showed abnormal dendritic arborization and branching. The Dnmt1(tet/tet) neuronal cells also showed elevated levels of functional N-methyl d-aspartate receptor (NMDAR), a feature also reported in some neurological and neurodegenerative disorders. Considering the roles of reelin and GAD(67) in neuronal networking and excitatory/inhibitory balance, respectively, we studied methylation of these genes' promoters in Dnmt1(tet/tet) ES cells and neurons. Both reelin and GAD(67) promoters were not hypermethylated in the Dnmt1(tet/tet) ES cells and neurons, suggesting that overexpression of DNMT1 may not directly result in methylation-mediated repression of these two genes. Taken together, our results suggest that overexpression of DNMT1 in ES cells results in an epigenetic change prior to the onset of differentiation. This epigenetic change in turn results in abnormal neuronal differentiation and upregulation of functional NMDA receptor.


Subject(s)
Cell Differentiation , DNA (Cytosine-5-)-Methyltransferases/metabolism , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Neurons/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Up-Regulation , Animals , Cells, Cultured , DNA (Cytosine-5-)-Methyltransferase 1 , DNA (Cytosine-5-)-Methyltransferases/genetics , Immunoblotting , Mice , Neurons/cytology , Neurons/pathology , Reelin Protein , Reverse Transcriptase Polymerase Chain Reaction
6.
Sleep Breath ; 15(1): 3-11, 2011 Jan.
Article in English | MEDLINE | ID: mdl-19924457

ABSTRACT

PURPOSE: The relationship between obstructive sleep apnea (OSA) and atherosclerosis-related inflammation has been poorly investigated, particularly focusing on functional responses of immune cells playing a key role in atherogenesis and in comparison with control groups with similar cardiovascular risk factors which are known to be themselves associated with inflammation. We sought to determine cellular tumor necrosis factor-alpha (TNF-α) production from peripheral blood mononuclear cells (PBMCs) and interleukin (IL)-8 release from neutrophils (PMNs) in patients studied for suspected OSA. METHODS: Thirty-six consecutive patients who underwent a nocturnal complete cardiorespiratory evaluation for suspected OSA were initially evaluated. Serum, PBMCs, and PMNs were isolated (at baseline and after 12 weeks) from patients with apnea-ipopnea index (AHI) >20 (OSA group, n = 16) and from control patients with AHI <5 (nonOSA group, n = 11). All patients continued the same pharmacological therapy for 12 weeks; the OSA group was additionally treated with nocturnal continuous positive-airway-pressure ventilation (cPAP). RESULTS: The two groups had similar clinical characteristics (prevalence of hypertension, dyslipidemia, diabetes, and cardio-metabolic therapies) except for obesity. Resting and stimulated TNF-α production from PBMCs and IL-8 release from PMNs were similar in the two groups. Serum cytokines resulted within the normal range. In the OSA group, cPAP was not associated with changes in cellular responses. CONCLUSIONS: In patients showing similar prevalence of major cardiovascular risk factors and cardio-metabolic therapies, differing for the presence or absence of OSA, cytokine productions from PBMC and PMN were similar and were not modified during cPAP therapy. Studies designed to investigate OSA-associated inflammation should carefully match the control group subjects.


Subject(s)
Cytokines/blood , Interleukin-8/blood , Monocytes/immunology , Neutrophils/immunology , Sleep Apnea, Obstructive/immunology , Tumor Necrosis Factor-alpha/blood , Adult , Aged , Continuous Positive Airway Pressure , Female , Humans , Male , Middle Aged , Reference Values , Sleep Apnea, Obstructive/therapy
7.
PLoS One ; 5(3): e9831, 2010 Mar 23.
Article in English | MEDLINE | ID: mdl-20352123

ABSTRACT

Deletion analysis of mouse DNMT1, the primary maintenance methyltransferase in mammals, showed that most of the N-terminal regulatory domain (amino acid residues 412-1112) is required for its enzymatic activity. Although analysis of deletion mutants helps to identify regions of a protein sequence required for a particular activity, amino acid deletions can have drastic effects on protein structure and/or stability. Alternative approaches represented by rational design and directed evolution are resource demanding, and require high-throughput selection or screening systems. We developed Regional Frame-shift Mutagenesis (RFM) as a new approach to identify portions required for the methyltransferase activity of DNMT1 within the N-terminal 89-905 amino acids. In this method, a short stretch of amino acids in the wild-type protein is converted to a different amino acid sequence. The resultant mutant protein retains the same amino acid length as the wild type, thereby reducing physical constrains on normal folding of the mutant protein. Using RFM, we identified three small regions in the amino-terminal one-third of the protein that are essential for DNMT1 function. Two of these regions (amino acids 124-160 and 341-368) border a large disordered region that regulates maintenance methylation activity. This organization of DNMT1's amino terminus suggests that the borders define the position of the disordered region within the DNMT1 protein, which in turn allows for its proper function.


Subject(s)
DNA (Cytosine-5-)-Methyltransferases/metabolism , Animals , DNA (Cytosine-5-)-Methyltransferase 1 , DNA, Complementary/metabolism , Embryonic Stem Cells/cytology , Frameshift Mutation , Gene Deletion , Immunohistochemistry/methods , Mice , Mutagenesis , Mutagenesis, Site-Directed , Mutation , Plasmids/metabolism , Protein Folding , Protein Structure, Tertiary , Transcription, Genetic
8.
Clin Neuropharmacol ; 32(3): 133-9, 2009.
Article in English | MEDLINE | ID: mdl-18978486

ABSTRACT

Long-term treatment of Parkinson disease (PD) is frequently associated with l-3,4-dihydroxyphenylalanine (L-DOPA)-induced dyskinesias (LIDs). L-DOPA-induced dyskinesias are likely due to changes in the signal transduction pathways, at the striatal level, related to pulsatile stimulation of dopamine receptors. We investigated whether markers of this phenomenon can also be detected peripherally. We analyzed mRNA expression for D5 (D1-like) and D3 (D2-like) receptors and levels of second messengers, such as cAMP and free intracellular Ca2+ ([Ca2+]i), in peripheral blood lymphocytes of PD patients with (LID+) or without LIDs (LID-). Patients with PD showed depressed [Ca2+]i rise in response to mitogen-induced activation. The defect was more pronounced in LID+ (-33% with respect to healthy controls) than in LID- patients (-20%). Peripheral blood lymphocyte levels of cAMP were decreased in both LID+ (3.8 +/- 2.9 pmol/10 cells) and LID- patients (4.2 +/- 2.4 pmol/10(6) cells), with respect to controls (6 +/- 2.6 pmol/10(6) cells). No differences were found in dopamine receptor mRNA expression. Our results demonstrate that second messenger levels are altered in the peripheral blood lymphocytes of PD patients treated with dopaminergic agents and that patients with LIDs show further alterations in the regulation of [Ca2+]i homeostasis. This may represent a distinctive trait of patients prone to develop dyskinetic movements.


Subject(s)
Akathisia, Drug-Induced/etiology , Antiparkinson Agents/adverse effects , Calcium/metabolism , Homeostasis/drug effects , Levodopa/adverse effects , Parkinson Disease/metabolism , Aged , Carbonyl Cyanide p-Trifluoromethoxyphenylhydrazone/pharmacology , Case-Control Studies , Cyclic AMP/metabolism , Female , Humans , Ionophores/pharmacology , Lactones/pharmacology , Lymphocytes/drug effects , Lymphocytes/metabolism , Male , Middle Aged , Mitogens/pharmacology , Parkinson Disease/drug therapy , Phytohemagglutinins/pharmacology , RNA, Messenger/metabolism , Receptors, Dopamine D3/genetics , Receptors, Dopamine D3/metabolism , Receptors, Dopamine D5/genetics , Receptors, Dopamine D5/metabolism , Sesquiterpenes/pharmacology
9.
Blood ; 109(2): 632-42, 2007 Jan 15.
Article in English | MEDLINE | ID: mdl-16985181

ABSTRACT

CD4+CD25+ regulatory T lymphocytes (Tregs) are specialized T cells playing a key role in the control of immune homeostasis. Here, we show that human Tregs constitutively express tyrosine hydroxylase (TH, EC 1.14.16.2), the rate-limiting enzyme in the synthesis of catecholamines, and contain substantial amounts of dopamine, norepinephrine, and epinephrine, which are released upon treatment with reserpine. Catecholamine release results in reduced production of interleukin-10 and transforming growth factor-beta by Tregs, and in down-regulation of Treg-dependent inhibition of effector T-lymphocyte (Teff) proliferation, which occurs without affecting the production of tumor necrosis factor-alpha or interferon-gamma. Tregs and Teffs express on the cell membrane both D1-like and D2-like dopaminergic receptors to a similar extent (12%-29% of the cells). Catecholamine-dependent down-regulation of Tregs is, however, selectively reversed by pharmacological blockade of dopaminergic D1-like receptors, which in Tregs only (and not in Teffs) are also expressed at the level of mRNA and are functionally coupled to intracellular production of cAMP. These findings indicate that in human Tregs endogenous catecholamines subserve an autocrine/paracrine loop involving dopaminergic pathways and resulting in down-regulation of Treg function.


Subject(s)
Autocrine Communication/drug effects , Catecholamines/metabolism , Paracrine Communication/drug effects , Phytohemagglutinins/pharmacology , T-Lymphocytes, Regulatory/immunology , Tyrosine 3-Monooxygenase/biosynthesis , Autocrine Communication/immunology , CD4 Antigens/immunology , Cell Proliferation , Down-Regulation , Forkhead Transcription Factors/drug effects , Forkhead Transcription Factors/immunology , Humans , Interleukin-10/biosynthesis , Interleukin-2 Receptor alpha Subunit/immunology , Paracrine Communication/immunology , Phytohemagglutinins/immunology , RNA, Messenger/drug effects , RNA, Messenger/immunology , Reserpine/pharmacology , T-Lymphocytes/immunology , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/enzymology , Transforming Growth Factor beta/biosynthesis , Tyrosine 3-Monooxygenase/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...