adenosine-5--(n-ethylcarboxamide) and Inflammation

adenosine-5--(n-ethylcarboxamide) has been researched along with Inflammation* in 17 studies

Other Studies

17 other study(ies) available for adenosine-5--(n-ethylcarboxamide) and Inflammation

ArticleYear
Testicular adenosine acts as a pro-inflammatory molecule: role of testicular peritubular cells.
    Molecular human reproduction, 2021, 07-01, Volume: 27, Issue:7

    Extracellular ATP has been described to be involved in inflammatory cytokine production by human testicular peritubular cells (HTPCs). The ectonucleotidases ENTPD1 and NT5E degrade ATP and have been reported in rodent testicular peritubular cells. We hypothesized that if a similar situation exists in human testis, ATP metabolites may contribute to cytokine production. Indeed, ENTPD1 and NT5E were found in situ and in vitro in HTPCs. Malachite green assays confirmed enzyme activities in HTPCs. Pharmacological inhibition of ENTPD1 (by POM-1) significantly reduced pro-inflammatory cytokines evoked by ATP treatment, suggesting that metabolites of ATP, including adenosine, are likely involved. We focused on adenosine and detected three of the four known adenosine receptors in HTPCs. One, A2B, was also found in situ in peritubular cells of human testicular sections. The A2B agonist BAY60-6583 significantly elevated levels of IL6 and CXCL8, a result also obtained with adenosine and its analogue NECA. Results of siRNA-mediated A2B down-regulation support a role of this receptor. In mouse peritubular cells, in contrast to HTPCs, all four of the known adenosine receptors were detected; when challenged with adenosine, cytokine expression levels significantly increased. Organotypic short-term testis cultures yielded comparable results and indicate an overall pro-inflammatory action of adenosine in the mouse testis. If transferable to the in vivo situation, our results may implicate that interference with the generation of ATP metabolites or interference with adenosine receptors could reduce inflammatory events in the testis. These novel insights may provide new avenues for treatment of sterile inflammation in male subfertility and infertility.

    Topics: 5'-Nucleotidase; Adenosine; Adenosine Triphosphate; Adenosine-5'-(N-ethylcarboxamide); Adult; Aminopyridines; Animals; Apyrase; Cells, Cultured; Cytokines; GPI-Linked Proteins; Humans; Infertility, Male; Inflammation; Male; Mice; Mice, Inbred C57BL; Middle Aged; Receptor, Adenosine A2B; Receptors, Purinergic P1; RNA Interference; RNA, Small Interfering; Testis

2021
Impact of Adenosine Analogue, Adenosine-5'-N-Ethyluronamide (NECA), on Insulin Signaling in Skeletal Muscle Cells.
    BioMed research international, 2021, Volume: 2021

    Rat L6 skeletal muscle cells were cultured in 25 cm. Adenosine-5'-N-ethyluronamide (NECA), a stable adenosine analogue, significantly stimulate inflammatory mediator (IL-6) (. A novel crosstalk between adenosine analogue and insulin has been demonstrated for the first time; evidence has been gathered in vitro for the effects of NECA and insulin treatment on intracellular signaling pathways, in particular glycolysis and insulin sensitivity in skeletal muscle cells.

    Topics: Acetyl-CoA Carboxylase; Acyl-CoA Dehydrogenase, Long-Chain; Adenosine; Adenosine-5'-(N-ethylcarboxamide); Animals; Carnitine O-Palmitoyltransferase; Hexokinase; Inflammation; Insulin; Interleukin-6; Muscle Fibers, Skeletal; Muscle, Skeletal; Myoblasts; Nuclear Receptor Subfamily 4, Group A, Member 1; Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha; Phosphofructokinases; Pyruvate Dehydrogenase Acetyl-Transferring Kinase; Rats; Receptors, Cytoplasmic and Nuclear; Reverse Transcriptase Polymerase Chain Reaction; Signal Transduction; Transcription Factors

2021
Ecto-5'-nucleotidase (CD73) attenuates inflammation after spinal cord injury by promoting macrophages/microglia M2 polarization in mice.
    Journal of neuroinflammation, 2018, May-22, Volume: 15, Issue:1

    Immune activation, specifically activation of macrophages and resident microglia, leading to inflammation is a key component in the progression of spinal cord injury (SCI). Macrophages/microglia exist in two states-the classically activated M1 phenotype that confers pro-inflammatory effects or the alternatively activated M2 phenotype that confers anti-inflammatory effects. Ecto-5'-nucleotidase (CD73) is an immunosuppressive molecule intricately involved in adaptive and innate immune responses and is able to dephosphorylate AMP to adenosine. However, it is not known if CD73 is able to modulate the macrophages/microglia transformation between the M1 and M2 phenotypes.. We used gene-deficient mice to determine the role of CD73 in macrophages/microglia polarization post-SCI in vivo. We used small interference RNA (siRNA) or pcDNA3.1 to inhibit or overexpress CD73 in BV2 cells to verify anterior discovery in vitro. A combination of molecular and histological methods was used to detect the macrophages/microglia polarization and explore the mechanism both in vivo and in vitro.. We found that SCI induced the upregulation of CD73 expression. CD73 deficient mice were noted to demonstrate overwhelming immune responses, few anti-inflammatory phenotype macrophages/microglia, and had a poorer locomotor recovery in comparison to wild-type mice that were also inflicted with SCI. In vitro studies found that CD73 suppression inhibited the expression of characteristic microglial anti-inflammatory polarization markers in BV2 cells, while the converse was noted in CD73 overexpression. Subsequent experiments confirmed that CD73 promoted microglia alternative activation by stimulating p38 MAPK.. We were able to conclude that CD73 imparts neuroprotective effects by mediating macrophages/microglia polarization. These findings allow for better understanding of the modulatory factors involved in triggering the change in macrophages/microglia phenotypes, therefore uncovering additional molecules and pathways that may be targeted in the innovation of novel SCI therapies.

    Topics: 5'-Nucleotidase; Adenosine-5'-(N-ethylcarboxamide); Animals; Antineoplastic Agents; Apoptosis; Arginase; Cell Polarity; Disease Models, Animal; Inflammation; Lipopolysaccharides; Macrophages; Male; Mice; Mice, Inbred C57BL; Mice, Knockout; Microglia; Motor Disorders; Nitric Oxide Synthase Type II; Spinal Cord Injuries; Up-Regulation

2018
CD73-generated adenosine is critical for immune regulation during Toxoplasma gondii infection.
    Infection and immunity, 2015, Volume: 83, Issue:2

    As an obligate intracellular pathogen, the apicomplexan parasite Toxoplasma gondii evades immune system-mediated clearance by undergoing stage differentiation to persist indefinitely in susceptible hosts. Previously, we found that mice deficient in the ectoenzyme CD73, which generates adenosine in the extracellular matrix, were resistant to chronic toxoplasmosis after oral infection with T. gondii. Resistance in CD73 knockout mice was due to a delay in parasite differentiation in the central nervous system (CNS). To further clarify the role of CD73 and extracellular adenosine in T. gondii pathogenesis, we infected wild-type (WT) and CD73(-/-) mice with T. gondii cysts systemically by the intraperitoneal (i.p.) route. In contrast to oral infection, i.p. infected CD73(-/-) mice were highly susceptible to immune-mediated pathology, with significantly increased infiltration of neutrophils and T cells into the peritoneal cavity. Administration of the broad-spectrum adenosine receptor agonist 5'-N-ethylcarboxamidoadenosine (NECA) protected CD73(-/-) mice against T. gondii-induced immunopathology, suggesting that the absence of CD73-generated adenosine led to the increased susceptibility in these mice. Peritoneal exudate cells from infected CD73(-/-) mice produced higher levels of the inflammatory mediators nitric oxide, tumor necrosis factor alpha (TNF-α), and interleukin-1β (IL-1β), without enhanced parasite killing or clearance. Bone marrow chimeras established that CD73 expression in both hematopoietic and nonhematopoietic compartments contributes to limiting T. gondii-induced immunopathology. In addition, mice deficient in the adenosine receptor A(2A) were more susceptible to immunopathology during intraperitoneal infection with T. gondii than WT mice. Thus, extracellular adenosine is a key immune regulator that limits collateral tissue damage due to an intracellular pathogen and promotes host survival.

    Topics: 5'-Nucleotidase; Adenosine; Adenosine-5'-(N-ethylcarboxamide); Animals; Antineoplastic Agents; CD4-Positive T-Lymphocytes; CD8-Positive T-Lymphocytes; Cells, Cultured; Inflammation; Inflammation Mediators; Interleukin-1beta; Lymphocyte Activation; Mice; Mice, Inbred C57BL; Mice, Knockout; Neutrophil Infiltration; Neutrophils; Nitric Oxide; Purinergic P1 Receptor Agonists; Toxoplasma; Toxoplasmosis; Tumor Necrosis Factor-alpha

2015
Protective effect of adenosine receptors against lipopolysaccharide-induced acute lung injury.
    American journal of physiology. Lung cellular and molecular physiology, 2014, Mar-15, Volume: 306, Issue:6

    Acute lung injury and acute respiratory distress syndrome (ALI/ARDS) affect 200,000 people a year in the USA. Pulmonary vascular and specifically endothelial cell (EC) barrier compromise is a hallmark of these diseases. We have recently shown that extracellular adenosine enhances human pulmonary (EC) barrier via activation of adenosine receptors (ARs) in cell cultures. On the basis of these data, we hypothesized that activation of ARs might exert barrier-protective effects in a model of ALI/ARDS in mice. To test this hypothesis, we examined the effects of pre- and posttreatment of adenosine and 5'-N-ethylcarboxamidoadenosine (NECA), a nonselective stable AR agonist, on LPS-induced lung injury. Mice were given vehicle or LPS intratracheally followed by adenosine, NECA, or vehicle instilled via the internal jugular vein. Postexperiment cell counts, Evans Blue Dye albumin (EBDA) extravasation, levels of proteins, and inflammatory cytokines were analyzed. Harvested lungs were used for histology and myeloperoxidase studies. Mice challenged with LPS alone demonstrated an inflammatory response typical of ALI. Cell counts, EBDA extravasation, as well as levels of proteins and inflammatory cytokines were decreased in adenosine-treated mice. Histology displayed reduced infiltration of neutrophils. NECA had a similar effect on LPS-induced vascular barrier compromise. Importantly, posttreatment with adenosine or NECA recovers lung vascular barrier and reduces inflammation induced by LPS challenge. Furthermore, adenosine significantly attenuated protein degradation of A2A and A3 receptors induced by LPS. Collectively, our results demonstrate that activation of ARs protects and restores vascular barrier functions and reduces inflammation in LPS-induced ALI.

    Topics: Acute Lung Injury; Adenosine; Adenosine-5'-(N-ethylcarboxamide); Animals; Bronchoalveolar Lavage Fluid; Capillary Permeability; Cell Count; Cytokines; Endothelial Cells; Endothelium; Inflammation; Interleukin-6; Lipopolysaccharides; Lung; Mice; Mice, Inbred C57BL; Purinergic P1 Receptor Agonists; Receptors, Purinergic P1; Respiratory Distress Syndrome; Tumor Necrosis Factor-alpha

2014
Adenosine promotes alternative macrophage activation via A2A and A2B receptors.
    FASEB journal : official publication of the Federation of American Societies for Experimental Biology, 2012, Volume: 26, Issue:1

    Adenosine has been implicated in suppressing the proinflammatory responses of classically activated macrophages induced by Th1 cytokines. Alternative macrophage activation is induced by the Th2 cytokines interleukin (IL)-4 and IL-13; however, the role of adenosine in governing alternative macrophage activation is unknown. We show here that adenosine treatment of IL-4- or IL-13-activated macrophages augments the expression of alternative macrophage markers arginase-1, tissue inhibitor of matrix metalloproteinase-1 (TIMP-1), and macrophage galactose-type C-type lectin-1. The stimulatory effect of adenosine required primarily A(2B) receptors because the nonselective adenosine receptor agonist 5'-N-ethylcarboxamidoadenosine (NECA) increased both arginase activity (EC(50)=261.8 nM) and TIMP-1 production (EC(50)=80.67 nM), and both pharmacologic and genetic blockade of A(2B) receptors prevented the effect of NECA. A(2A) receptors also contributed to the adenosine augmentation of IL-4-induced TIMP-1 release, as both adenosine and NECA were less efficacious in augmenting TIMP-1 release by A(2A) receptor-deficient than control macrophages. Of the transcription factors known to drive alternative macrophage activation, CCAAT-enhancer-binding protein β was required, while cAMP response element-binding protein and signal transducer and activator of transcription 6 were dispensable in mediating the effect of adenosine. We propose that adenosine receptor activation suppresses inflammation and promotes tissue restitution, in part, by promoting alternative macrophage activation.

    Topics: Adenosine; Adenosine-5'-(N-ethylcarboxamide); Animals; Arginase; CCAAT-Enhancer-Binding Protein-beta; Cell Line; Cyclic AMP Response Element-Binding Protein; Extracellular Space; Inflammation; Interleukin-13; Interleukin-4; Macrophages; Male; Mice; Mice, Inbred C57BL; Mice, Knockout; Receptor, Adenosine A2A; Receptor, Adenosine A2B; STAT6 Transcription Factor; Tissue Inhibitor of Metalloproteinase-1; Toll-Like Receptor 4; Vasodilator Agents

2012
The effect of adenosine on pro-inflammatory cytokine production by porcine T cells.
    Veterinary immunology and immunopathology, 2012, Jan-15, Volume: 145, Issue:1-2

    Adenosine is a well described anti-inflammatory modulator of immune responses. The aim of the present study was to describe the role of common adenosine agonist 5'-N-ethylcarboxamidoadenosine (NECA) in cytokine production by main porcine T cell subpopulations. TNF-α, IFN-γ, IL-2 and IL-10 were detected by multicolor flow cytometry together with cell surface markers CD3, CD4 and CD8. It was found that NECA inhibits (in a dose-dependent manner) production of pro-inflammatory TNF-α and Th1-associated cytokines IFN-γ, IL-2 in all concanavalin A-stimulated T cell subpopulations. Moreover, production of IL-10 was potentiated in all T cell subpopulations tested. These corresponded well with the fact that all T cell subsets expressed mRNA for adenosine receptor (AR) subtypes to comparable extents. Contrary to concanavalin A-stimulated cells, NECA had a moderate effect on PMA-stimulated T cells, suggesting that AR in pigs acts via signaling pathways not associated with protein-kinase C. Non-selective antagonist CGS15943 as well as allosteric modulator SCH202676 failed to reverse the effect of NECA in pigs. In conclusion, NECA has an anti-inflammatory effect on porcine T cell subpopulations.

    Topics: Adenosine; Adenosine-5'-(N-ethylcarboxamide); Animals; CD3 Complex; CD4-Positive T-Lymphocytes; CD8-Positive T-Lymphocytes; Concanavalin A; Dose-Response Relationship, Drug; Flow Cytometry; Inflammation; Interferon-gamma; Interleukin-10; Interleukin-2; Quinazolines; Swine; T-Lymphocyte Subsets; T-Lymphocytes; Triazoles; Tumor Necrosis Factor-alpha

2012
Links between insulin resistance, adenosine A2B receptors, and inflammatory markers in mice and humans.
    Diabetes, 2011, Volume: 60, Issue:2

    To determine the mechanisms by which blockade of adenosine A(2B) receptors (A(2B)Rs) reduces insulin resistance.. We investigated the effects of deleting or blocking the A(2B)R on insulin sensitivity using glucose tolerance tests (GTTs) and hyperinsulinemic-euglycemic clamps in mouse models of type 2 diabetes. The effects of diabetes on A(2B)R transcription and signaling were measured in human and mouse macrophages and mouse endothelial cells. In addition, tag single nucleotide polymorphisms (SNPs) in ~42 kb encompassing the A(2B)R gene, ADORA2B, were evaluated for associations with markers of diabetes and inflammation.. Treatment of mice with the nonselective adenosine receptor agonist 5'-N-ethylcarboxamidoadensoine (NECA) increased fasting blood glucose and slowed glucose disposal during GTTs. These responses were inhibited by A(2B)R deletion or blockade and minimally affected by deletion of A(1)Rs or A(2A)Rs. During hyperinsulinemic-euglycemic clamp of diabetic KKA(Y) mice, A(2B)R antagonism increased glucose infusion rate, reduced hepatic glucose production, and increased glucose uptake into skeletal muscle and brown adipose tissue. Diabetes caused a four- to sixfold increase in A(2B)R mRNA in endothelial cells and macrophages and resulted in enhanced interleukin (IL)-6 production in response to NECA due to activation of protein kinases A and C. Five consecutive tag SNPs in ADORA2B were highly correlated with IL-6 and C-reactive protein (CRP). Diabetes had a highly significant independent effect on variation in inflammatory markers. The strength of associations between several ADORA2B SNPs and inflammatory markers was increased when accounting for diabetes status.. Diabetes affects the production of adenosine and the expression of A(2B)Rs that stimulate IL-6 and CRP production, insulin resistance, and the association between ADORA2B SNPs and inflammatory markers. We hypothesize that increased A(2B)R signaling in diabetes increases insulin resistance in part by elevating proinflammatory mediators. Selective A(2B)R blockers may be useful to treat insulin resistance.

    Topics: Adenosine-5'-(N-ethylcarboxamide); Animals; Biomarkers; Blood Glucose; C-Reactive Protein; Cells, Cultured; Diabetes Mellitus, Type 2; Glucose Clamp Technique; Glucose Tolerance Test; Humans; Inflammation; Insulin; Insulin Resistance; Interleukin-6; Liver; Macrophages; Mice; Mice, Transgenic; Polymorphism, Single Nucleotide; Receptor, Adenosine A2B; Reverse Transcriptase Polymerase Chain Reaction

2011
Microsphere-based flow cytometry protease assays for use in protease activity detection and high-throughput screening.
    Current protocols in cytometry, 2010, Volume: Chapter 13

    This protocol describes microsphere-based protease assays for use in flow cytometry and high-throughput screening. This platform measures a loss of fluorescence from the surface of a microsphere due to the cleavage of an attached fluorescent protease substrate by a suitable protease enzyme. The assay format can be adapted to any site or protein-specific protease of interest and results can be measured in both real time and as endpoint fluorescence assays on a flow cytometer. Endpoint assays are easily adapted to microplate format for flow cytometry high-throughput analysis and inhibitor screening.

    Topics: Animals; Biotinylation; Flow Cytometry; Fluorescence Resonance Energy Transfer; Green Fluorescent Proteins; High-Throughput Screening Assays; Humans; Inflammation; Kinetics; Microspheres; Peptide Hydrolases; Peptides; Reproducibility of Results; Temperature

2010
A new role for the A2b adenosine receptor in regulating platelet function.
    Journal of thrombosis and haemostasis : JTH, 2010, Volume: 8, Issue:4

    Activation of platelets is a critical component of atherothrombosis and plays a central role in the progression of unstable cardiovascular syndromes. Adenosine, acting through adenosine receptors, increases intracellular cAMP levels and inhibits platelet aggregation. The A2a adenosine receptor has already been recognized as a mediator of adenosine-dependent effects on platelet aggregation, and here we present a new role for the A2b adenosine receptor (A2bAR) in this process.. As compared with platelets from wild-type controls, platelets derived from A2bAR knockout mice have significantly greater ADP receptor activation-induced aggregation. Although mouse megakaryocytes and platelets express low levels of the A2bAR transcript, this gene is highly upregulated following injury and systemic inflammation in vivo. Under these conditions, A2bAR-mediated inhibition of platelet aggregation significantly increases. Our studies also identify a novel mechanism by which the A2bAR could regulate platelet aggregation; namely, ablation of the A2bAR leads to upregulated expression of the P2Y1 ADP receptor, whereas A2bAR-mediated or direct elevation of cAMP has the opposite effect. Thus, the A2bAR regulates platelet function beyond mediating the immediate effect of adenosine on aggregation.. Taken together, these investigations show for the first time that the platelet A2bAR is upregulated under stress in vivo, plays a significant role in regulating ADP receptor expression, and inhibits agonist-induced platelet aggregation.

    Topics: Adenosine A2 Receptor Agonists; Adenosine Diphosphate; Adenosine-5'-(N-ethylcarboxamide); Animals; Blood Platelets; Cells, Cultured; Cyclic AMP; Disease Models, Animal; Femoral Artery; Genotype; Inflammation; Lipopolysaccharides; Megakaryocytes; Mice; Mice, Inbred C57BL; Mice, Knockout; Phenotype; Platelet Aggregation; Receptor, Adenosine A2B; Receptors, Purinergic P2; Receptors, Purinergic P2Y1; RNA, Messenger; Time Factors; Up-Regulation

2010
Involvement of A1 adenosine receptors in altered vascular responses and inflammation in an allergic mouse model of asthma.
    American journal of physiology. Heart and circulatory physiology, 2010, Volume: 299, Issue:1

    Poor lung function and respiratory disorders like asthma have a positive correlation with the development of adverse cardiovascular events. Increased adenosine levels are associated with lung inflammation that could lead to altered vascular responses and systemic inflammation. We hypothesized that asthmatic lung inflammation has systemic effects through A(1) adenosine receptors (A(1)AR) and investigated the effects of aerosolized adenosine on vascular reactivity and inflammation, using A(1)AR knockout (A(1)KO) and corresponding wild-type (A(1)WT) mice that were divided into three experimental groups each: control (CON), allergen sensitized and challenged (SEN), and SEN + aerosolized adenosine (SEN + AD). Animals were sensitized with ragweed (200 microg ip; days 1 and 6), followed by 1% ragweed aerosol challenges (days 11 to 13). On day 14, the SEN + AD groups received one adenosine aerosol challenge (6 mg/ml) for 2 min, and aortae were collected on day 15. 5'-N-ethylcarboxamidoadenosine (NECA; nonselective adenosine analog) induced concentration-dependent aortic relaxation in the A(1)WT CON group, which was impaired in the A(1)WT SEN and SEN + AD groups. All groups of A(1)KO mice showed similar (no significant difference) concentration-dependent relaxation to NECA. The A(1)WT SEN and SEN + AD groups had a significantly higher contraction to selective A(1) agonist 2-chloro-N(6)-cyclopentyladenosine (CCPA) compared with the CON group. Western blot data showed that aortic A(1)AR expression was significantly increased in WT SEN and SEN + AD mice compared with CON mice. Gene expression of ICAM-1 and IL-5 was significantly increased in allergic A(1)WT aorta and were undetected in the A(1)KO groups. A(1)WT allergic mice had significantly higher airway hyperresponsiveness (enhanced pause) to NECA, with adenosine aerosol further enhancing it. In conclusion, allergic A(1)WT mice showed altered vascular reactivity, increased airway hyperresponsiveness, and systemic inflammation. These data suggest that A(1)AR is proinflammatory systemically in this model of allergic asthma.

    Topics: Adenosine; Adenosine-5'-(N-ethylcarboxamide); Administration, Inhalation; Aerosols; Ambrosia; Animals; Antigens, Plant; Aorta; Asthma; Blotting, Western; Bronchial Hyperreactivity; Bronchoconstriction; Disease Models, Animal; Dose-Response Relationship, Drug; Inflammation; Inflammation Mediators; Intercellular Adhesion Molecule-1; Interleukin-5; Male; Mice; Mice, Inbred BALB C; Mice, Knockout; Plethysmography, Whole Body; Polymerase Chain Reaction; Receptor, Adenosine A1; RNA, Messenger; Vasodilation

2010
Intranasal administration of NECA can induce both anti-inflammatory and pro-inflammatory effects in BALB/c mice: evidence for A 2A receptor sub-type mediation of NECA-induced anti-inflammatory effects.
    Pulmonary pharmacology & therapeutics, 2009, Volume: 22, Issue:3

    The role of adenosine in allergic inflammation is unclear. This study investigated the effects of the non-selective adenosine receptor agonist, 5-N-ethylcarboxamidoadenosine (NECA), on immunized only and immunized and airway challenged mice. The adenosine receptor sub-type(s) mediating the NECA effects and the A(2A) receptor mRNA expression were also investigated. In mice that were only immunized, intranasal NECA (1 mM) administration caused a significant increase in bronchoalveolar lavage total cell count (TCC), neutrophils and eosinophils (>1.5-, >6 and >60-fold, respectively). Two and four intranasal ovalbumin (OVA) challenges induced a significant (P < 0.05) increase in TCC (>2.1- and >4-fold, respectively) and eosinophils (>350- and >1700-fold, respectively). Real-time PCR analysis showed that the A(2A) receptor sub-type mRNA was significantly increased (P < 0.05) in the lung tissue of immunized mice following both two and four OVA challenges. NECA (0.3 mM) treatment caused a significant reduction in the increase induced by the two and four OVA challenges in the TCC by 46.1% and 56.6%, respectively, eosinophils by 70.1% and 75.6%, respectively, and in the A(2A) receptor sub-type mRNA by 43.2% and 41.0%, respectively. Treatment with the A(2A) receptor antagonist, 7-(2-phenylethyl)-5-amino-2-(2-furyl)-pyrazolo-[4,3-e]-1,2,4-triazolo[1,5-c]pyrimidine), SCH-58261, completely reversed both the NECA-mediated reduction in TCC and eosinophilia. Moreover, OVA challenge of immunized mice, over 2 consecutive days, resulted in a significant (P < 0.05) increase in TCC (4.5-fold) and eosinophils (>2000-fold) that was detected 72 h later. NECA (0.3 mM) treatment, at 24 and 48 h post OVA challenge, significantly reduced the increase in both TCC and eosinophils by 45.0% and 74.8%, respectively. Our data show that in immunized, but not OVA-challenged mice, high dose of NECA (1 mM) induces an inflammatory airway response. In contrast, in models of inflammation, NECA, at mainly 0.3 mM, induces a significant anti-inflammatory effect when administered prior to the induction of airway inflammation or therapeutically following its establishment. The data also indicate that the anti-inflammatory action of NECA seems to be mediated via the A(2A) receptor sub-type and hence the use of selective A(2A) receptor agonists as potential therapeutic agents in the treatment of inflammatory diseases such as asthma should be investigated further.

    Topics: Adenosine A2 Receptor Agonists; Adenosine A2 Receptor Antagonists; Adenosine-5'-(N-ethylcarboxamide); Administration, Intranasal; Animals; Anti-Inflammatory Agents; Cell Count; Dose-Response Relationship, Drug; Immunization; Inflammation; Lung; Male; Mice; Mice, Inbred BALB C; Ovalbumin; Purinergic P1 Receptor Agonists; Purinergic P1 Receptor Antagonists; Receptor, Adenosine A2A; Respiratory System; Reverse Transcriptase Polymerase Chain Reaction; RNA, Messenger; Vasodilator Agents

2009
Adenosine signaling mediates SUMO-1 modification of IkappaBalpha during hypoxia and reoxygenation.
    The Journal of biological chemistry, 2009, May-15, Volume: 284, Issue:20

    Small ubiquitin-like modifier 1 (SUMO-1) modification of IkappaBalpha has been described to actively participate in NFkappaB regulation. Following proteosomal degradation of IkappaBalpha, an auto-regulatory loop consisting of transcriptional activation of IkappaBalpha gene and SUMO-1 modification of newly synthesized IkappaBalpha proceeds. The SUMOylated IkappaBalpha form is resistant to signal-induced degradation, consequently halting NFkappaB activation. We describe a mechanistic model by which adenosine (Ado) signaling results in significant accumulation of SUMO-1 modified IkappaBalpha with subsequent attenuation of NFkappaB activation. Using models of hypoxia followed by reoxygenation (H/R), we have documented an H/R cycle-dependent increase in extracellular Ado correlating with increases in the cytoplasmic pool of IkappaBalpha/SUMO-1. We demonstrate a dose-dependent increase in IkappaBalpha/SUMO in cells treated with the general Ado receptor agonist NECA and abolished by Ado receptor antagonists. Experiments in cells exposed to cycles of H/R followed by hypoxia demonstrated differential patterns of SUMOylation and phosphorylation of IkappaBalpha, greatly impacting its proteosomal degradation by the 26 S proteasome. Assays targeting knockdown and overexpression of SUMO-1 demonstrated significant regulation of NFkappaB activation and NFkappaB-mediated gene transcription (interleukin-6). These results were confirmed in vivo using wild type and cd73 null mouse lung tissue. In summary, we present an endogenous mechanism by which cells and tissues acquire anti-inflammatory properties by recruiting a nondegradable form of IkappaBalpha, a major control point for NFkappaB activation via Ado signaling.

    Topics: 5'-Nucleotidase; Adenosine; Adenosine-5'-(N-ethylcarboxamide); Animals; Cell Hypoxia; Gene Knockdown Techniques; HeLa Cells; Humans; Hypoxia; I-kappa B Proteins; Inflammation; Interleukin-6; Lung; Mice; Mice, Mutant Strains; NF-kappa B; NF-KappaB Inhibitor alpha; Pneumonia; Proteasome Endopeptidase Complex; Protein Processing, Post-Translational; Purinergic P1 Receptor Agonists; Receptors, Purinergic P1; SUMO-1 Protein; Transcription, Genetic; Vasodilator Agents

2009
Involvement of mast cells in adenosine-mediated bronchoconstriction and inflammation in an allergic mouse model.
    The Journal of pharmacology and experimental therapeutics, 2005, Volume: 313, Issue:1

    In allergen-induced asthma, activation of lung mast cells leads to bronchial constriction, increased mucus secretion, and an increase in the localization of inflammatory cells to the airways. The purpose of this study was to explore the role of mast cells in adenosine-mediated airway reactivity and inflammation using the mast cell degranulating agent, compound 48/80 (C48/80). Mice were sensitized and challenged with ragweed (or 0.9% saline) followed by C48/80 administration twice a day in increasing doses for 5 days. Dose-responsiveness to the nonspecific adenosine receptor agonist 5'-N-ethylcarboxamidoadenosine (NECA) was established, and lung lavage was performed 24 h later for cell differential analysis to evaluate inflammation. At a dose of 375 microg/ml (aerosolized NECA), C48/80 pretreatment resulted in a significant attenuation in airway reactivity when compared with sensitized control mice (330.07 versus 581.57%, respectively). Lung lavage from the C48/80 treated mice showed a decrease in eosinophils (17.7 versus 60.9%, respectively) and an increase in macrophages when compared with the sensitized control group (76.4 versus 30.8%, respectively). These results support the conclusion that mast cell degranulation plays an important role in adenosine receptor-mediated airway hyperresponsiveness and inflammation.

    Topics: Adenosine; Adenosine-5'-(N-ethylcarboxamide); Ambrosia; Animals; Bronchoalveolar Lavage Fluid; Bronchoconstriction; Cell Count; Cell Degranulation; Dose-Response Relationship, Drug; Inflammation; Leukocyte Count; Mast Cells; Methacholine Chloride; Mice; Muscarinic Agonists; p-Methoxy-N-methylphenethylamine; Respiratory Hypersensitivity

2005
Effect of buflomedil on the neutrophil-endothelial cell interaction under inflammatory and hypoxia conditions.
    Haemostasis, 1996, Volume: 26 Suppl 4

    In hypoxia/ischaemia and ischaemia/reperfusion, human neutrophils are likely to play an important role in the development of endothelial cell damage in the microcirculation. Buflomedil hypochloride improves the capillary perfusion in such related situations, evoking a possible effect upon neutrophils. Using in vitro models of cell adhesion, buflomedil decreased 100% of histamine related neutrophil adhesion (flow system) and partially inhibited adhesion after IL-1-4 hours (flow and stable systems). Hypoxia induced neutrophil adhesion (4 hours) was also reduced by buflomedil, which decreased the expression of P-selectin at the surface of endothelial cells. As adenosine (NECA) exhibited the same results in hypoxia and theophylline inhibited them, such results support an action of buflomedil presumably via the A2 receptor.

    Topics: Adenosine; Adenosine-5'-(N-ethylcarboxamide); Adrenergic alpha-2 Receptor Antagonists; Adrenergic alpha-Antagonists; Cell Adhesion; Cell Hypoxia; Cells, Cultured; Cytokines; Endothelium, Vascular; Humans; Inflammation; Intercellular Adhesion Molecule-1; Interleukin-1; Neutrophils; P-Selectin; Pyrrolidines; Theophylline; Umbilical Veins

1996
Spinal cord adenosine receptor stimulation in rats inhibits peripheral neutrophil accumulation. The role of N-methyl-D-aspartate receptors.
    The Journal of clinical investigation, 1996, Dec-15, Volume: 98, Issue:12

    The effect of spinal adenosine receptor ligation on peripheral leukocyte accumulation was studied in two rat models of inflammation. Neutrophil infiltration into dermal inflammatory sites was signficantly reduced by adenosine A1 receptor agonists injected through intrathecal catheters. These effects were reversed by N-methyl-D-aspartate (NMDA), and were mimicked by (+/-)-2-amino-5-phosphonopentanoic acid (AP-5), a glutamate NMDA receptor antagonist. Peripheral adenosine levels, as measured in air pouch exudates, decreased markedly in inflamed pouches but remained near normal after intrathecal treatment with AP-5. Moreover, the antiinflammatory effects of intrathecal A1 receptor agonists and AP-5 were reversed by an adenosine A2 receptor antagonist administered intraperitoneally. Hence, central NMDA receptor activity can regulate neutrophil accumulation in peripheral inflammatory sites by reducing local levels of adenosine, an antiinflammatory autacoid which inhibits neutrophil function through A2 receptor activation. This represents a previously unknown pathway by which the central nervous system influences inflammatory responses.

    Topics: 6-Cyano-7-nitroquinoxaline-2,3-dione; Adenosine; Adenosine-5'-(N-ethylcarboxamide); Animals; Anti-Inflammatory Agents; Carrageenan; Catheterization; Central Nervous System; Dexamethasone; Excitatory Amino Acid Antagonists; Inflammation; N-Methylaspartate; Neutrophils; Peroxidase; Phenethylamines; Propionates; Purinergic P1 Receptor Antagonists; Rats; Receptors, Glutamate; Receptors, N-Methyl-D-Aspartate; Receptors, Purinergic P1; Signal Transduction; Skin; Spinal Cord; Theobromine

1996
Immune-related intestinal chloride secretion. II. Effect of adenosine on T84 cell line.
    The American journal of physiology, 1990, Volume: 258, Issue:5 Pt 1

    The inflammatory mediator adenosine caused sustained Cl- secretion across monolayers of T84 cells. The effect was promptly reversed by the adenosine receptor antagonist 8-phenyltheophylline and appeared to be mediated through an adenosine A2-receptor [rank order of potency: 5'-(N-ethyl)-carboxamido-adenosine (NECA) greater than adenosine greater than (-)-N6-(phenylisopropyl)adenosine (PIA) greater than or equal to (+)-PIA]. High doses of adenosine and its analogues increased cellular adenosine 3',5'-cyclic monophosphate (cAMP) but not guanosine 3',5'-cyclic monophosphate (cGMP) or free cytosolic Ca2+. However, lower concentrations of adenosine had maximal effects on Cl- secretion with little or no effect on cAMP. In other respects, Cl- secretion resembled that induced by cAMP-mediated secretagogues such as vasoactive intestinal peptide (VIP). Addition of both low and high doses of NECA activated basolateral K+ and apical Cl- channels, exhibited synergism with Ca2(+)-mediated secretagogues, did not produce additive effects with VIP or Escherichia coli heat-stable enterotoxin, and was associated with cAMP-dependent protein kinase-mediated protein phosphorylation. The results suggest that either adenosine mobilizes an intracellular pool of cAMP that is extremely efficiently coupled to the cAMP-dependent protein kinase and is thereafter rapidly destroyed or that second messenger(s) other than cAMP, cGMP, or Ca2+ are able to activate Cl- secretion in the T84 cell line. In the latter case, such messenger(s), as yet unidentified, might represent a final common pathway for cyclic nucleotide-activated Cl- secretion.

    Topics: 2-Chloroadenosine; Adenosine; Adenosine-5'-(N-ethylcarboxamide); Calcium; Cell Line; Chlorides; Cyclic AMP; Cyclic GMP; Cytosol; Electrolytes; Epithelium; Histamine; Humans; Inflammation; Kinetics; Phenylisopropyladenosine; Phosphorylation; Protein Kinases; Theophylline

1990