Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
Proc Natl Acad Sci U S A ; 120(4): e2208924120, 2023 01 24.
Article in English | MEDLINE | ID: mdl-36652486

ABSTRACT

Nitro-fatty acids (NO2-FAs) are unsaturated fatty acid nitration products that exhibit anti-inflammatory actions in experimental mouse models of autoimmune and allergic diseases. These electrophilic molecules interfere with intracellular signaling pathways by reversible post-translational modification of nucleophilic amino-acid residues. Several regulatory proteins have been identified as targets of NO2-FAs, modifying their activity and promoting gene expression changes that result in anti-inflammatory effects. Herein, we report the effects of nitro-oleic acid (NO2-OA) on pro-inflammatory T cell functions, showing that 9- and 10-NOA, but not their oleic acid precursor, decrease T cell proliferation, expression of activation markers CD25 and CD71 on the plasma membrane, and IL-2, IL-4, and IFN-γ cytokine gene expressions. Moreover, we have found that NO2-OA inhibits the transcriptional activity of nuclear factor of activated T cells (NFAT) and that this inhibition takes place through the regulation of the phosphatase activity of calcineurin (CaN), hindering NFAT dephosphorylation, and nuclear translocation in activated T cells. Finally, using mass spectrometry-based approaches, we have found that NO2-OA nitroalkylates CaNA on four Cys (Cys129, 228, 266, and 372), of which only nitroalkylation on Cys372 was of importance for the regulation of CaN phosphatase activity in cells, disturbing functional CaNA/CaNB heterodimer formation. These results provide evidence for an additional mechanism by which NO2-FAs exert their anti-inflammatory actions, pointing to their potential as therapeutic bioactive lipids for the modulation of harmful T cell-mediated immune responses.


Subject(s)
Calcineurin , Nitrogen Dioxide , Mice , Animals , Calcineurin/metabolism , Oleic Acid , Protein Processing, Post-Translational , Fatty Acids/metabolism
2.
Int J Mol Sci ; 23(21)2022 Oct 31.
Article in English | MEDLINE | ID: mdl-36362060

ABSTRACT

Cyclooxygenase (COX) is the key enzyme in prostanoid synthesis from arachidonic acid (AA). Two isoforms, named COX-1 and COX-2, are expressed in mammalian tissues. The expression of COX-2 isoform is induced by several stimuli including cytokines and mitogens, and this induction is inhibited by glucocorticoids (GCs). We have previously shown that the transcriptional induction of COX-2 occurs early after T cell receptor (TCR) triggering, suggesting functional implications of this enzyme in T cell activation. Here, we show that dexamethasone (Dex) inhibits nuclear factor of activated T cells (NFAT)-mediated COX-2 transcriptional induction upon T cell activation. This effect is dependent on the presence of the GC receptor (GR), but independent of a functional DNA binding domain, as the activation-deficient GRLS7 mutant was as effective as the wild-type GR in the repression of NFAT-dependent transcription. Dex treatment did not disturb NFAT dephosphorylation, but interfered with activation mediated by the N-terminal transactivation domain (TAD) of NFAT, thus pointing to a negative cross-talk between GR and NFAT at the nuclear level. These results unveil the ability of GCs to interfere with NFAT activation and the induction of pro-inflammatory genes such as COX-2, and explain some of their immunomodulatory properties in activated human T cells.


Subject(s)
Cyclooxygenase 2 , Receptors, Glucocorticoid , T-Lymphocytes , Humans , Cyclooxygenase 2/genetics , Cyclooxygenase 2/metabolism , Dexamethasone/pharmacology , Glucocorticoids/pharmacology , Glucocorticoids/metabolism , Lymphocyte Activation , Mammals/metabolism , Receptors, Glucocorticoid/metabolism , T-Lymphocytes/metabolism , Transcriptional Activation , NFATC Transcription Factors/genetics , NFATC Transcription Factors/metabolism
3.
Front Cell Dev Biol ; 9: 673973, 2021.
Article in English | MEDLINE | ID: mdl-34026763

ABSTRACT

Nitric oxide (NO) and electrophilic cyclopentenone prostaglandins (CyPG) are local mediators that modulate cellular response to oxidative stress in different pathophysiological processes. In particular, there is increasing evidence about their functional role during inflammation and immune responses. Although the mechanistic details about their relationship and functional interactions are still far from resolved, NO and CyPG share the ability to promote redox-based post-translational modification (PTM) of proteins that play key roles in cellular homeostasis, signal transduction and transcription. NO-induced S-nitrosylation and S-glutathionylation as well as cyclopentenone-mediated adduct formation, are a few of the main PTMs by which intra- and inter-cellular signaling are regulated. There is a growing body of evidence indicating that actin and actin-binding proteins are susceptible to covalent PTM by these agents. It is well known that the actin cytoskeleton is key for the establishment of interactions among leukocytes, endothelial and muscle cells, enabling cellular activation and migration. In this review we analyze the current knowledge about the actions exerted by NO and CyPG electrophilic lipids on the regulation of actin dynamics and cytoskeleton organization, and discuss some open questions regarding their functional relevance in the regulation of intercellular communication.

4.
J Acoust Soc Am ; 138(4): 2046-53, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26520288

ABSTRACT

Commerson's dolphins (Cephalorhynchus commersonii) inhabit coastal waters of Southern South America and Kerguelen Islands. Limited information exists about the acoustic repertoire of this species in the wild. Here, echolocation signals from free-ranging Commerson's dolphins were recorded in Bahía San Julián, Argentina. Signal parameters were calculated and a cluster analysis was made on 3180 regular clicks. Three clusters were obtained based on peak frequency (129, 137, and 173 kHz) and 3 dB bandwidth (8, 6, and 5 kHz). The 428 buzz clicks were analyzed separately. They consisted of clicks emitted with a median inter-click interval of 3.5 ms, peak frequency at 131 kHz, 3 dB bandwidth of 9 kHz, 10 dB bandwidth of 18 kHz, and duration of 56 µs. Buzz clicks were significantly shorter and with a lower peak frequency and a broader bandwidth than most of the regular clicks. This study provided the first description of different echolocation signals, including on- and off-axis signals, recorded from Commerson's dolphins in the wild, most likely as a result of animals at several distances and orientations to the recording device. This information could be useful while doing passive acoustic monitoring.


Subject(s)
Dolphins/physiology , Echolocation , Age Factors , Algorithms , Animals , Bays , Cluster Analysis , Meteorological Concepts , Seasons , Sound Spectrography
6.
PLoS Negl Trop Dis ; 9(8): e0004025, 2015.
Article in English | MEDLINE | ID: mdl-26305786

ABSTRACT

Inflammation plays an important role in the pathophysiology of Chagas disease, caused by Trypanosoma cruzi. Prostanoids are regulators of homeostasis and inflammation and are produced mainly by myeloid cells, being cyclooxygenases, COX-1 and COX-2, the key enzymes in their biosynthesis from arachidonic acid (AA). Here, we have investigated the expression of enzymes involved in AA metabolism during T. cruzi infection. Our results show an increase in the expression of several of these enzymes in acute T. cruzi infected heart. Interestingly, COX-2 was expressed by CD68+ myeloid heart-infiltrating cells. In addition, infiltrating myeloid CD11b+Ly6G- cells purified from infected heart tissue express COX-2 and produce prostaglandin E2 (PGE2) ex vivo. T. cruzi infections in COX-2 or PGE2-dependent prostaglandin receptor EP-2 deficient mice indicate that both, COX-2 and EP-2 signaling contribute significantly to the heart leukocyte infiltration and to the release of chemokines and inflammatory cytokines in the heart of T. cruzi infected mice. In conclusion, COX-2 plays a detrimental role in acute Chagas disease myocarditis and points to COX-2 as a potential target for immune intervention.


Subject(s)
Chagas Disease/immunology , Cyclooxygenase 2/immunology , Dinoprostone/immunology , Myocarditis/immunology , Receptors, Prostaglandin E, EP2 Subtype/immunology , Trypanosoma cruzi/physiology , Animals , Chagas Disease/complications , Chagas Disease/enzymology , Chagas Disease/genetics , Cyclooxygenase 2/genetics , Cytokines/genetics , Cytokines/immunology , Humans , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Myocarditis/etiology , Myocarditis/genetics , Myocardium/enzymology , Myocardium/immunology , Receptors, Prostaglandin E, EP2 Subtype/genetics , Trypanosoma cruzi/immunology
7.
Article in English | MEDLINE | ID: mdl-25736222

ABSTRACT

Liver X receptors (LXRs) are nuclear receptors that act as ligand-dependent transcription factors forming permissive heterodimers with retinoid X receptors (RXRs). In this study we aimed to assess the effect of LXR/RXR activation on the transcriptional induction of pro-inflammatory genes including cyclooxygenase-2 (COX-2) and microsomal prostaglandin E2 synthase-1 (mPGES-1) in activated macrophages. Our study shows that LXR ligands such as oxysterols, GW3965 or TO901317, as well as RXR ligands like 9cis retinoic acid or SR11237, decreased LPS-induced expression of COX-2 and mPGES-1. Consequently, LPS-dependent PGE2 production was substantially reduced in macrophages treated with LXR/RXR ligands. The inhibitory effects of LXR/RXR activation on LPS-induced expression of COX-2 and mPGES-1 in macrophages, occurred by a mechanism involving interference with transcriptional activation of these genes. LXR/RXR activation interfered with the activity of transcription factors essential in the up-regulation of the expression of pro-inflammatory genes in these cells, such as NFκB, but also Egr-1, which had not been previously associated with LXR-mediated gene repression. As this transcription factor is involved in the regulation of a variety of genes involved in inflammatory processes, LXR and RXR-mediated interference with Egr-1 signaling could represent an important event mediating the anti-inflammatory effects of these receptors in macrophages.


Subject(s)
Early Growth Response Protein 1/metabolism , Gene Expression/drug effects , Lipopolysaccharides/metabolism , Macrophages/metabolism , Orphan Nuclear Receptors/pharmacology , Animals , Cell Line , Humans , Ligands , Liver X Receptors , Mice , Monocytes/metabolism , Orphan Nuclear Receptors/antagonists & inhibitors , Orphan Nuclear Receptors/metabolism
8.
J Immunol ; 191(1): 395-406, 2013 Jul 01.
Article in English | MEDLINE | ID: mdl-23733875

ABSTRACT

Cyclooxygenase (Cox)-2 dependent PGs modulate several functions in many pathophysiological processes, including migration of immune cells. In this study, we addressed the role of Cox-2 in macrophage migration by using in vivo and in vitro models. Upon thioglycolate challenge, CD11b(+) F4/80(+) macrophages showed a diminished ability to migrate to the peritoneal cavity in cox-2(-/-) mice. In vivo migration of cox-2(-/-) macrophages from the peritoneal cavity to lymph nodes, as well as cell adhesion to the mesothelium, was reduced in response to LPS. In vitro migration of cox-2(-/-) macrophages toward MCP-1, RANTES, MIP-1α, or MIP-1ß, as well as cell adhesion to ICAM-1 or fibronectin, was impaired. Defects in cell migration were not due to changes in chemokine receptor expression. Remarkably, cox-2(-/-) macrophages showed a deficiency in focal adhesion formation, with reduced phosphorylation of paxillin (Tyr(188)). Interestingly, expression of the p110γ catalytic subunit of PI3K was severely reduced in the absence of Cox-2, leading to defective Akt phosphorylation, as well as cdc42 and Rac-1 activation. Our results indicate that the paxillin/p110γ-PI3K/Cdc42/Rac1 axis is defective in cox-2(-/-) macrophages, which results in impaired cell adhesion and migration.


Subject(s)
Cell Migration Inhibition/immunology , Class Ib Phosphatidylinositol 3-Kinase/deficiency , Cyclooxygenase 2/deficiency , Macrophages, Peritoneal/enzymology , Macrophages, Peritoneal/immunology , Phosphatidylinositol 3-Kinases/deficiency , Signal Transduction/immunology , Animals , Cell Adhesion/genetics , Cell Adhesion/immunology , Cell Migration Inhibition/genetics , Cells, Cultured , Class Ib Phosphatidylinositol 3-Kinase/physiology , Cyclooxygenase 2/genetics , Cyclooxygenase 2/physiology , Macrophages, Peritoneal/pathology , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Models, Immunological , Phosphatidylinositol 3-Kinases/physiology , Signal Transduction/genetics , cdc42 GTP-Binding Protein/deficiency , rac1 GTP-Binding Protein/deficiency
9.
Front Pharmacol ; 4: 23, 2013.
Article in English | MEDLINE | ID: mdl-23494519

ABSTRACT

Sigma (σ) receptor ligands are essentially known for their effects on the nervous system although recent studies have shown their potential effects modulating some other pathophysiological processes as cell proliferation, cancer, and the immune response. Here, we have analyzed the actions of σ-1 and σ-2 receptors ligands on T cell activation. Our results show that treatment of Jurkat T cells with σ-2 agonists decreased the induction of the expression of Interleukin (IL)-2, Tumor necrosis factor (TNF)-α, and Cyclooxygenase (COX)-2 by activated T cells in a dose-dependent manner. These effects take place at the transcriptional level since σ-2 agonists BD-737 and CB-184 diminished the activity of the promoters of those genes. Those immunosuppressive effects could be attributable to interference with transcription factor activation. Induced transcription mediated by Nuclear factor (NF)-κB or Nuclear Factor of Activated T cells (NFAT) was inhibited by σ-2 agonists. These effects seem to be specific for σ-2 agonists as no significant effects on T cell activation by σ-1 ligands PRE-084 and BD-1063 were found. Our results provide new insights into the immunomodulatory actions of σ ligands and describe a new property of σ-2 agonists, through inhibition of activation of transcription factors as NFAT by which these compounds are regulating gene expression. This may have important consequences on the possible therapeutic use of those compounds.

10.
J Acoust Soc Am ; 131(6): EL448-53, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22713020

ABSTRACT

Franciscana dolphins are small odontocetes hard to study in the field. In particular, little is known on their echolocation behavior in the wild. In this study we recorded 357 min and analyzed 1019 echolocation signals in the Rio Negro Estuary, Argentina. The clicks had a peak frequency at 139 kHz, and a bandwidth of 19 kHz, ranging from 130 to 149 kHz. This is the first study describing echolocation signals of franciscana dolphins in the wild, showing the presence of narrow-band high frequency signals in these dolphins. Whether they use other vocalizations to communicate or not remains uncertain.


Subject(s)
Dolphins/physiology , Echolocation/physiology , Animals , Sound Spectrography
11.
Biochem J ; 443(2): 451-61, 2012 Apr 15.
Article in English | MEDLINE | ID: mdl-22268508

ABSTRACT

PG (prostaglandin) E2 plays an important role in the modulation of the immune response and the inflammatory process. In the present study, we describe a PGE2 positive feedback for COX (cyclo-oxygenase)-2 and mPGES-1 [microsomal PGES (PGE synthase)-1] expression in the macrophage cell line RAW 264.7. Our results show that PGE2 induces COX-2 and mPGES-1 expression, an effect mimicked by dbcAMP (dibutyryl-cAMP) or forskolin. Furthermore, the cAMP signalling pathway co-operates with LPS (lipopolysaccharide) in the induction of COX-2 and mPGES-1 transcriptional activation. Analysis of the involvement of PGE receptors [EPs (E-prostanoids)] showed that incubation with EP2 agonists up-regulated both COX2 and mPGES-1 mRNA levels. Moreover, EP2 receptor overexpression enhanced the transcriptional activation of COX2 and mPGES-1 promoters. This induction was repressed by the PKA (protein kinase A) inhibitor H89. Activation of the PGE2/EP2/PKA signalling pathway induced the phosphorylation of CREB [CRE (cAMP-response element)-binding protein] in macrophages and stimulated the specific binding of this transcription factor to COX2 and mPGES-1 promoters. Deletion or mutation of potential CRE sites in both promoters diminished their transcriptional activity. In summary, the results of the present study demonstrate that activation of PKA/CREB signalling through the EP2 receptor by PGE2 plays a key role in the expression of COX-2 and mPGES-1 in activated macrophages.


Subject(s)
Cyclooxygenase 2/metabolism , Dinoprostone/metabolism , Intramolecular Oxidoreductases/metabolism , Lipopolysaccharides/immunology , Macrophages/metabolism , Signal Transduction , Animals , Cell Line , Cyclic AMP/metabolism , Cyclooxygenase 2/genetics , Intramolecular Oxidoreductases/genetics , Macrophages/immunology , Mice , Promoter Regions, Genetic , Prostaglandin-E Synthases , Transcription, Genetic , Transcriptional Activation , Up-Regulation
12.
Int Immunopharmacol ; 10(10): 1295-304, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20709632

ABSTRACT

Anti-inflammatory efficacy of non-steroidal anti-inflammatory drugs (NSAIDs) has been related to their properties as inhibitors of cyclooxygenase (COX)-mediated prostaglandin (PG) synthesis. However, recent studies have suggested that variations of the in vivo anti-inflammatory actions among different NSAIDs could not be solely explained by COX inhibition. Here, we have analyzed the effects on T cell activation of novel 4,5-dihydro-3 trifluoromethyl pyrazole anti-inflammatory drugs with different potencies as COX-2 inhibitors, namely E-6087, E-6232, E-6231, E-6036 and E-6259 as well as the chemically related COX-2 inhibitor Celecoxib. These drugs inhibited mitogen-mediated T cell proliferation as well as Interleukin (IL)-2, tumor necrosis factor (TNF)-α and Interferon (IFN)-γ synthesis by activated T cells, independently of their ability to inhibit COX-2 enzymatic activity. Immunosuppressive effects of these drugs seem to be due to their interference on transcription factor activation as induced transcription from Nuclear Factor (NF)-κB and Nuclear Factor of Activated T cells (NFAT)-dependent enhancers was inhibited in a dose-dependent manner, being the latter effect the most sensitive to the action of those compounds. Both NFAT dephosphorylation, required for its nuclear translocation, as well as transcriptional activity of a GAL4-NFAT chimera were diminished in the presence of these compounds. These findings provide new insights into the molecular mechanisms involved in the immunomodulatory and anti-inflammatory actions of NSAIDs, which may have important implications in anti-inflammatory therapy, through inhibition of NFAT.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Cyclooxygenase 2 Inhibitors/pharmacology , Lymphocyte Activation/drug effects , Pyrazoles/pharmacology , T-Lymphocytes/drug effects , T-Lymphocytes/physiology , Animals , Anti-Inflammatory Agents, Non-Steroidal/chemistry , Exudates and Transudates/chemistry , Gastric Mucosa/drug effects , Gene Expression Regulation/physiology , Humans , Inflammation/metabolism , Jurkat Cells , Molecular Structure , NF-kappa B/metabolism , NFATC Transcription Factors/metabolism , Pyrazoles/chemistry , Rats
13.
Clin Cancer Res ; 16(16): 4207-16, 2010 Aug 15.
Article in English | MEDLINE | ID: mdl-20682710

ABSTRACT

PURPOSE: 2-(3,4-dihydroxyphenil)-ethanol (DPE), a polyphenol present in olive oil, has been found to attenuate the growth of colon cancer cells, an effect presumably related to its anti-inflammatory activity. EXPERIMENTAL DESIGN: To further explore the effects of DPE on angiogenesis and tumor growth we investigated the in vivo efficacy of DPE in a HT-29 xenograft model and in vitro activities in colon cancer cells exposed to interleukin-1beta (IL-1beta) and prostaglandin E-2 (PGE-2). RESULTS: DPE (10 mg/kg/day for 14 days) inhibited tumor growth, reducing vessel lumina and blood perfusion to tumor, and diminished expression of hypoxia inducible factor-1alpha (HIF-1alpha), vascular endothelial growth factor (VEGF), and microsomal prostaglandin-E synthase-1 (mPGEs-1). In vitro, DPE (100 mumol/L) neither affected cell proliferation nor induced apoptosis in HT-29 and WiDr cells. DPE prevented the IL-1beta-mediated increase of mPGEs-1 expression and PGE-2 generation, as it did the silencing of HIF-1alpha. Moreover, DPE blocked mPGEs-1-dependent expression of VEGF and inhibited endothelial sprouting induced by tumor cells in a coculture system. PGE-2 triggers a feed-forward loop involving HIF-1alpha, which impinges on mPGEs-1 and VEGF expression, events prevented by DPE via extracellular signal-related kinase 1/2. The reduction of PGE-2 and VEGF levels, caused by DPE, was invariably associated with a marked decrease in HIF-1alpha expression and activity, independent of proteasome activity, indicating that the DPE effects on tumor growth and angiogenesis are dependent on the inhibition of HIF-1alpha translation. CONCLUSIONS: We show that the in vivo DPE antitumor effect is associated with anti-inflammatory and antiangiogenic activities resulting from the downregulation of the HIF-1alpha/mPGEs-1/VEGF axis.


Subject(s)
Antineoplastic Agents/pharmacology , Hypoxia-Inducible Factor 1, alpha Subunit/drug effects , Neoplasms, Experimental/drug therapy , Neovascularization, Pathologic/drug therapy , Plant Oils/pharmacology , Signal Transduction/drug effects , Animals , Blotting, Western , Cell Line, Tumor , Cell Proliferation/drug effects , Coculture Techniques , Enzyme-Linked Immunosorbent Assay , Flavonoids/pharmacology , Gene Expression/drug effects , HT29 Cells , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/antagonists & inhibitors , Intramolecular Oxidoreductases/biosynthesis , Intramolecular Oxidoreductases/drug effects , Mice , Mice, Nude , Microsomes/drug effects , Neoplasms, Experimental/metabolism , Olive Oil , Phenols/pharmacology , Plant Oils/chemistry , Polyphenols , Prostaglandin-E Synthases , RNA, Small Interfering , Receptors, Vascular Endothelial Growth Factor/antagonists & inhibitors , Receptors, Vascular Endothelial Growth Factor/drug effects , Reverse Transcriptase Polymerase Chain Reaction , Transfection , Xenograft Model Antitumor Assays
14.
Cell Signal ; 22(10): 1427-36, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20546888

ABSTRACT

Prostaglandin (PG) E(2) is a potent lipid mediator that plays an essential role in inflammation, fever and pain. It is produced from arachidonic acid (AA) by a cascade of enzymatic reactions involving cyclooxygenases (COX-1 and -2) and prostaglandin E synthases (cPGES, mPGES-1 and -2). Functional coupling of the inducible enzymes COX-2 and mPGES-1 has been proposed for increased production of PGE(2) in different cell types. PGE(2) produced by macrophages plays an essential role in the pathogenesis of inflammatory diseases. Here, we have investigated the mechanisms involved in the regulation of COX-2 and mPGES-1 expressions in murine macrophages upon bacterial lipopolysaccharide (LPS) treatment. LPS stimulation induced the coordinated synthesis of COX-2 and mPGES-1 that resulted in an enhanced production of PGE(2) in RAW 264.7 macrophages. Furthermore, we show the involvement of NF-kappaB and Egr-1 transcription factors in the transcriptional induction of these enzymes. LPS treatment promoted specific binding of NF-kappaB to both COX-2 and mPGES-1 promoters. Site-directed mutagenesis, electrophoretic mobility shift assays and ChIP assays allowed the identification of a sequence acting as a NF-kappaB recognition site in the murine mPGES-1 promoter. Furthermore, LPS induced the expression of Egr-1 that cooperated with NF-kappaB in the up-regulation of COX-2 and mPGES-1. Inhibition of Egr-1 expression reduced substantially LPS-mediated induction of COX-2 and mPGES-1 expression, resulting in a decrease in PGE(2) production. Our findings point out to Egr-1 and NF-kappaB cooperation as determinant for PGE2 synthesis by macrophages in inflammatory processes through the coordinated regulation of COX-2 and mPGES-1.


Subject(s)
Cyclooxygenase 2/genetics , Early Growth Response Protein 1/metabolism , Intramolecular Oxidoreductases/genetics , Macrophages/enzymology , NF-kappa B/metabolism , Animals , Cyclooxygenase 2/biosynthesis , Early Growth Response Protein 1/antagonists & inhibitors , Intramolecular Oxidoreductases/biosynthesis , Lipopolysaccharides/pharmacology , Macrophages/drug effects , Mice , Promoter Regions, Genetic , Prostaglandin-E Synthases , Transcription, Genetic , Up-Regulation
15.
PPAR Res ; 2008: 352437, 2008.
Article in English | MEDLINE | ID: mdl-18670614

ABSTRACT

A growing body of evidence indicates that PPAR (peroxisome proliferator-activated receptor) alpha agonists might have therapeutic usefulness in antitumoral therapy by decreasing abnormal cell growth, and reducing tumoral angiogenesis. Most of the anti-inflammatory and antineoplastic properties of PPAR ligands are due to their inhibitory effects on transcription of a variety of genes involved in inflammation, cell growth and angiogenesis. Cyclooxygenase (COX)-2 and vascular endothelial growth factor (VEGF) are crucial agents in inflammatory and angiogenic processes. They also have been significantly associated to cell proliferation, tumor growth, and metastasis, promoting tumor-associated angiogenesis. Aberrant expression of VEGF and COX-2 has been observed in a variety of tumors, pointing to these proteins as important therapeutic targets in the treatment of pathological angiogenesis and tumor growth. This review summarizes the current understanding of the role of PPARalpha and its ligands in the regulation of COX-2 and VEGF gene expression in the context of tumor progression.

16.
Arch Physiol Biochem ; 114(3): 201-9, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18629685

ABSTRACT

Prostanoids, including prostaglandins (PGs) and thromboxanes (TXs) are synthesized from arachidonic acid by the combined action of cyclooxygenases (COXs) and PG and TX synthases. Finally after their synthesis, prostanoids are quickly released to the extracellular medium exerting their effects upon interaction with prostanoid receptors present in the neighbouring cells. These agents exert important actions in the cardiovascular system, modulating vascular homeostasis and participating in the pathogenesis of vascular diseases as thrombosis and atherosclerosis. Among prostanoids, Tromboxane (TX)A(2), a potent platelet activator and vasoconstrictor and prostacyclin (PGI2), a platelet inhibitor and vasodilator, are the most important in controlling vascular homeostasis. Although multiple studies using pharmacological inhibitors and genetically deficient mice have demonstrated the importance of prostanoid-mediated actions on cardiovascular physiology, further analysis on the prostanoid mediated actions in the vascular system are required to better understand the benefits and risks for the use of COX inhibitors in cardiovascular diseases.


Subject(s)
Cardiovascular Diseases/physiopathology , Prostaglandins/physiology , Homeostasis , Humans
17.
Cell Signal ; 19(8): 1652-61, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17398070

ABSTRACT

Nuclear factor of activated T cells (NFAT) plays a prominent role in gene transcription during the immune response. Growing evidence demonstrates the implication of inducible phosphorylation of the transactivation domain (TAD) of NFAT in transcriptional activation of genes. We have analyzed the regulation of NFATc2 activation by Cot/Tpl2 and protein kinase C zeta (PKCzeta) in T cells. Our results show that PKCzeta and Cot/Tpl2 cooperate in regulating the transactivation activity mediated by the amino-terminal domain of NFATc2. Neither Cot/Tpl2 kinase nor PKCzeta-mediated induction of the transactivation activity of NFATc2 was affected by cyclosporin-A treatment, supporting a calcineurin independent route in the signaling pathways mediating NFATc2 activation. Co-precipitation experiments showed physical interaction among Cot/Tpl2, PKCzeta and NFATc2. Analysis of the transactivation activity of deletions in the N-terminal region of NFATc2, suggested the involvement of amino acids 52-64 of NFATc2 in the induction of its transactivating function by PKCzeta. This kinase in vitro phosphorylates NFATc2 and deletion and mutational studies identified Ser53 and Ser56 (of the SPPS motif) as substrates for PKCzeta. Thus, our results suggest that PKCzeta phosphorylation of Ser53 and Ser56 in the N-terminal TAD from NFATc2 potentiates its transactivating function in human T cells.


Subject(s)
Gene Expression Regulation, Enzymologic , MAP Kinase Kinase Kinases/metabolism , NFATC Transcription Factors/chemistry , NFATC Transcription Factors/metabolism , Protein Kinase C/metabolism , Proto-Oncogene Proteins/metabolism , Genes, Reporter , Humans , Jurkat Cells , Luciferases/metabolism , NFATC Transcription Factors/genetics , Phosphorylation , Protein Kinase C/analysis , Protein Structure, Tertiary , Transcription, Genetic , Transfection
18.
Cardiovasc Res ; 70(3): 446-56, 2006 Jun 01.
Article in English | MEDLINE | ID: mdl-16458868

ABSTRACT

Endothelial cells play an active role in the maintenance of homeostasis. Endothelial injury can give rise to endothelial dysfunction in which the profile of mediators released by endothelial cells is altered. Among these mediators are factors that participate in the development of many cardiovascular disorders. Some of the most important are the prostanoids, which can modulate the progression of atherosclerosis, arterial hypertension, and angiogenesis. Prostanoids are produced by the sequential actions of cyclooxygenases and specific synthases and exert their actions through diverse cell-surface and nuclear receptors. The profile of prostanoids produced depends on cell type and the changing pathophysiological status, and these factors similarly affect the great array of biological responses elicited by these molecules. The resulting complexity enables extremely subtle and highly complex responses, and this provides opportunities for the development of targeted therapeutic approaches.


Subject(s)
Cardiovascular Diseases/metabolism , Endothelium, Vascular/metabolism , Prostaglandins/physiology , Signal Transduction/physiology , Animals , Cardiovascular Diseases/pathology , Endothelial Cells/metabolism , Gene Expression , Humans , Neovascularization, Pathologic , Prostaglandins/genetics
19.
Cell Signal ; 18(8): 1262-9, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16326073

ABSTRACT

Growing evidence shows that Interleukin (IL)-1beta and Cyclooxygenase 2 (COX-2) play a crucial role in the pathogenesis of inflammatory diseases and tumor growth, particularly in the gastrointestinal tract. Here, we have analyzed the regulation of COX-2 by IL-1beta in the human colon carcinoma cell line Caco-2, showing that COX-2 induction by this cytokine is due to both nuclear factor (NF)-kappaB-dependent transcriptional and p38 mitogen-activated protein kinase (MAPK)-mediated post-transcriptional mechanisms. Treatment of these cells with IL-1beta increased the levels of COX-2 mRNA and protein and hence the production of PGE2. IL-1beta induced NF-kappaB activation in Caco-2 cells, promoting the binding of this transcription factor to DNA and increasing NF-kappaB-dependent transcription. Inhibition of NF-kappaB activation diminished IL-1beta-mediated transcriptional activation of COX-2. Furthermore, mutation or deletion of a putative NF-kappaB binding site in the human COX-2 promoter greatly diminished its induction by IL-1beta. In addition, this cytokine induced a rapid increase in p38 MAPK activation. Interestingly, inhibition of p38 MAPK by SB203580 severely decreased induction of COX-2 expression by IL-1beta. p38 MAPK signalling was required for IL-1beta-dependent stabilization of COX-2 transcript. Given the importance of COX-2 expression in intestinal inflammation and colon carcinogenesis, these findings contribute to determine the key signalling pathways involved in the regulation of COX-2 expression in colorectal cells by inflammatory stimuli, such as IL-1beta.


Subject(s)
Colonic Neoplasms/enzymology , Cyclooxygenase 2/genetics , Interleukin-1/pharmacology , Up-Regulation/drug effects , Up-Regulation/genetics , Caco-2 Cells , Gene Expression Regulation, Enzymologic/drug effects , Humans , NF-kappa B/metabolism , RNA Stability/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Transcription, Genetic , p38 Mitogen-Activated Protein Kinases/metabolism
20.
Biochem J ; 395(1): 81-8, 2006 Apr 01.
Article in English | MEDLINE | ID: mdl-16343055

ABSTRACT

Recent evidence indicates that PPAR (peroxisome-proliferator-activated receptor) alpha ligands possess anti-inflammatory and antitumoural properties owing to their inhibitory effects on the expression of genes that are involved in the inflammatory response. However, the precise molecular mechanisms underlying these effects are poorly understood. In the present study, we show that tumour promoter PMA-mediated induction of genes that are significantly associated with inflammation, tumour growth and metastasis, such as COX-2 (cyclo-oxygenase 2) and VEGF (vascular endothelial growth factor), is inhibited by PPARalpha ligands in the human colorectal carcinoma cell line SW620. PPARalpha activators LY-171883 and WY-14,643 were able to diminish transcriptional induction of COX-2 and VEGF by inhibiting AP-1 (activator protein-1)-mediated transcriptional activation induced by PMA or by c-Jun overexpression. The actions of these ligands on AP-1 activation and COX-2 and VEGF transcriptional induction were found to be dependent on PPARalpha expression. Our studies demonstrate the existence of a negative cross-talk between the PPARalpha- and AP-1-dependent signalling pathways in these cells. PPARalpha interfered with at least two steps within the pathway leading to AP-1 activation. First, PPARalpha activation impaired AP-1 binding to a consensus DNA sequence. Secondly, PPARalpha ligands inhibited c-Jun transactivating activity. Taken together, these findings provide new insight into the anti-inflammatory and anti-tumoural properties of PPARalpha activation, through the inhibition of the induction of AP-1-dependent genes that are involved in inflammation and tumour progression.


Subject(s)
Colorectal Neoplasms/metabolism , Cyclooxygenase 2 Inhibitors/pharmacology , Cyclooxygenase 2/metabolism , Gene Expression Regulation, Neoplastic/drug effects , PPAR alpha/agonists , Transcription Factor AP-1/antagonists & inhibitors , Vascular Endothelial Growth Factor A/genetics , Caco-2 Cells , Colorectal Neoplasms/pathology , Cyclooxygenase 2/genetics , Humans , Ligands , PPAR alpha/metabolism , Promoter Regions, Genetic/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction/drug effects , Tetradecanoylphorbol Acetate/pharmacology , Transcription Factor AP-1/metabolism , Transcriptional Activation/drug effects , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...