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










Publication year range
1.
Int. microbiol ; 22(1): 131-141, mar. 2019. graf, tab
Article in English | IBECS | ID: ibc-184821

ABSTRACT

The relationships between specific type IV pili (TFP) groups and antibiotic resistance, biofilm formation, and bacterial motility were determined in 190 Pseudomonas aeruginosa clinical isolates. While motility and biofilm formation were determined by phenotypic assays, the presence of TFP was determined by PCR assay and antibiotic susceptibility by disk diffusion. The results showed a high ability to form biofilm (97.4%), multidrug resistance (44.7%), and the presence of a high number of motile isolates. We also found an association between strong biofilm production and multidrug resistance. Furthermore, TFP group III was associated with strong biofilm production. In contrast, the isolates with TFP group II and those without any TFP were associated with non-strong biofilm production. Regarding motility, TFP group II was associated with higher percentages of swarming, swimming, and twitching, while TFP group I showed lower percentages of swarming and twitching, and TFP group III showed lower levels of swarming and swimming. In conclusion, these findings highlight the differences in P. aeruginosa phenotypes related to the presence of specific TFP groups and their potential implications in clinical settings


No disponible


Subject(s)
Humans , Biofilms/growth & development , Fimbriae, Bacterial/physiology , Pseudomonas Infections/microbiology , Pseudomonas aeruginosa/isolation & purification , Anti-Bacterial Agents/pharmacology , Drug Resistance, Bacterial , Locomotion , Pseudomonas aeruginosa/physiology , Disk Diffusion Antimicrobial Tests , Polymerase Chain Reaction , Pseudomonas aeruginosa/drug effects , Pseudomonas aeruginosa/genetics
2.
Int Microbiol ; 22(1): 131-141, 2019 Mar.
Article in English | MEDLINE | ID: mdl-30810940

ABSTRACT

The relationships between specific type IV pili (TFP) groups and antibiotic resistance, biofilm formation, and bacterial motility were determined in 190 Pseudomonas aeruginosa clinical isolates. While motility and biofilm formation were determined by phenotypic assays, the presence of TFP was determined by PCR assay and antibiotic susceptibility by disk diffusion. The results showed a high ability to form biofilm (97.4%), multidrug resistance (44.7%), and the presence of a high number of motile isolates. We also found an association between strong biofilm production and multidrug resistance. Furthermore, TFP group III was associated with strong biofilm production. In contrast, the isolates with TFP group II and those without any TFP were associated with non-strong biofilm production. Regarding motility, TFP group II was associated with higher percentages of swarming, swimming, and twitching, while TFP group I showed lower percentages of swarming and twitching, and TFP group III showed lower levels of swarming and swimming. In conclusion, these findings highlight the differences in P. aeruginosa phenotypes related to the presence of specific TFP groups and their potential implications in clinical settings.


Subject(s)
Biofilms/growth & development , Fimbriae, Bacterial/physiology , Locomotion , Pseudomonas Infections/microbiology , Pseudomonas aeruginosa/isolation & purification , Pseudomonas aeruginosa/physiology , Anti-Bacterial Agents/pharmacology , Disk Diffusion Antimicrobial Tests , Drug Resistance, Bacterial , Humans , Polymerase Chain Reaction , Pseudomonas aeruginosa/drug effects , Pseudomonas aeruginosa/genetics
3.
J Immunol Methods ; 375(1-2): 100-10, 2012 Jan 31.
Article in English | MEDLINE | ID: mdl-21996427

ABSTRACT

We developed a flow cytometry-based assay to simultaneously quantify multiple leukocyte populations in the marginated vascular, interstitial, and alveolar compartments of the mouse lung. An intravenous injection of a fluorescently labeled anti-CD45 antibody was used to label circulating and marginated vascular leukocytes. Following vascular flushing to remove non-adherent cells and collection of broncho-alveolar lavage (BAL) fluid, lungs were digested and a second fluorescent anti-CD45 antibody was added ex vivo to identify cells not located in the vascular space. In the naïve mouse lung, we found about 11 million CD45+ leukocytes, of which 87% (9.5 million) were in the vascular marginated compartment, consisting of 17% NK cells, 17% neutrophils, 57% mononuclear myeloid cells (monocytes, macrophage precursors and dendritic cells), and 10% T cells (CD4+, CD8+, and invariant NKT cells). Non-vascular compartments including the interstitial compartment contained 7.7×10(5)cells, consisting of 49% NK cells, 25% dendritic cells, and 16% other mononuclear myeloid cells. The alveolar compartment was overwhelmingly populated by macrophages (5.63×10(5)cells, or 93%). We next studied leukocyte margination and extravasation into the lung following acid injury, a model of gastric aspiration. At 1 h after injury, neutrophils were markedly elevated in the blood while all other circulating leukocytes declined by an average of 79%. At 4 h after injury, there was a peak in the numbers of marginated neutrophils, NK cells, CD4+ and CD8+ T cells and a peak in the number of alveolar NK cells. Most interstitial cells consisted of DCs, neutrophils, and CD4+ T cells, and most alveolar compartment cells consisted of macrophages, neutrophils, and NK cells. At 24 h after injury, there was a decline in the number of all marginated and interstitial leukocytes and a peak in alveolar neutrophils. In sum, we have developed a novel assay to study leukocyte margination and trafficking following pulmonary inflammation and show that marginated cells comprise a large fraction of lung leukocytes that increases shortly after lung injury. This assay may be of interest in future studies to determine if leukocytes become activated upon adherence to the endothelium, and have properties that distinguish them from interstitial and circulating cells.


Subject(s)
Cell Movement/physiology , Leukocytes/immunology , Lung Injury/immunology , Lung Injury/pathology , Animals , Bronchoalveolar Lavage Fluid/immunology , Cell Movement/immunology , Dendritic Cells/immunology , Dendritic Cells/metabolism , Dendritic Cells/pathology , Flow Cytometry/methods , Leukocyte Common Antigens/immunology , Leukocyte Count/methods , Leukocytes/metabolism , Leukocytes/pathology , Lung Injury/metabolism , Macrophages/immunology , Macrophages/metabolism , Macrophages/pathology , Male , Mice , Mice, Inbred C57BL , Pneumonia/immunology , Pneumonia/metabolism , Pneumonia/pathology , Reproducibility of Results
4.
J Thorac Cardiovasc Surg ; 139(2): 474-82, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19909990

ABSTRACT

OBJECTIVE: Adenosine A(2A) receptor activation potently attenuates lung ischemia-reperfusion injury. This study tests the hypothesis that adenosine A(2A) receptor activation attenuates ischemia-reperfusion injury by inhibiting CD4+ T cell activation and subsequent neutrophil infiltration. METHODS: An in vivo model of lung ischemia-reperfusion injury was used. C57BL/6 mice were assigned to either sham group (left thoracotomy) or 7 study groups that underwent ischemia-reperfusion (1 hour of left hilar occlusion plus 2 hours of reperfusion). ATL313, a selective adenosine A(2A) receptor agonist, was administered 5 minutes before reperfusion with or without antibody depletion of neutrophils or CD4+ T cells. After reperfusion, the following was measured: pulmonary function using an isolated, buffer-perfused lung system, T cell infiltration by immunohistochemistry, myeloperoxidase and proinflammatory cytokine/chemokine levels in bronchoalveolar lavage fluid, lung wet/dry weight, and microvascular permeability. RESULTS: ATL313 significantly improved pulmonary function and reduced edema and microvascular permeability after ischemia-reperfusion compared with control. Immunohistochemistry and myeloperoxidase content demonstrated significantly reduced infiltration of neutrophils and CD4+ T cells after ischemia-reperfusion in ATL313-treated mice. Although CD4+ T cell-depleted and neutrophil-depleted mice displayed significantly reduced lung injury, no additional protection occurred when ATL313 was administered to these mice. Expression of tumor necrosis factor-alpha, interleukin 17, KC, monocyte chemotactic protein-1, macrophage inflammatory protein-1, and RANTES were significantly reduced in neutrophil- and CD4+ T cell-depleted mice and reduced further by ATL313 only in neutrophil-depleted mice. CONCLUSIONS: These results demonstrate that CD4+ T cells play a key role in mediating lung inflammation after ischemia-reperfusion. ATL313 likely exerts its protective effect largely through activation of adenosine A(2A) receptors on CD4+ T cells and neutrophils.


Subject(s)
Adenosine A2 Receptor Agonists , Lung Diseases/prevention & control , Piperidines/pharmacology , Reperfusion Injury/prevention & control , Animals , CD4-Positive T-Lymphocytes , Capillary Permeability/drug effects , Capillary Permeability/physiology , Chemokines/biosynthesis , Cytokines/biosynthesis , Immunohistochemistry , Male , Mice , Mice, Inbred C57BL , Neutrophil Infiltration/physiology , Respiratory Function Tests
5.
Am J Physiol Lung Cell Mol Physiol ; 297(3): L467-74, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19574419

ABSTRACT

The adenosine A(2B) receptor (A(2B)R) has a wide tissue distribution that includes fibroblasts and endothelial and epithelial cells. The recent generation of an A(2B)R(-/-) mouse constructed with a beta-galactosidase (beta-gal) reporter gene under control of the endogenous promoter has provided a valuable tool to quantify A(2B)R promoter activity (29). To determine the sites of expression of the A(2B) receptor in the mouse lung, histological and flow cytometric analysis of beta-gal reporter gene expression in various lung cell populations was performed. The major site of A(2B)R promoter activity was found to be the type II alveolar epithelial cells (AECs), identified by coexpression of prosurfactant protein C, with relatively less expression in alveolar macrophages, bronchial epithelial cells, and cells of the vasculature. Highly purified type II AECs were prepared by fluorescence-activated sorting of enhanced green fluorescent protein (eGFP)-positive cells from transgenic mice expressing eGFP under control of the surfactant protein C promoter (21). The type II cells expressed 89-fold higher A(2B)R mRNA than pulmonary leukocytes, and the A(2B)R was shown to be functional, as treatment of purified type II AECs with the nonspecific adenosine receptor agonist 5'-N-ethylcarboxamidoadenosine (NECA) induced an increase in intracellular cAMP greater that the beta-adrenergic agonist isoproterenol that was inhibited completely following treatment by ATL-802, a novel, highly potent (K(i) = 8.6 nM), and selective (>900 fold over other adenosine receptor subtypes) antagonist of the mouse A(2B)R.


Subject(s)
Epithelial Cells/metabolism , Pulmonary Alveoli/cytology , Receptor, Adenosine A2B/metabolism , Adenosine A2 Receptor Antagonists , Animals , Bronchi/cytology , Cyclic AMP/metabolism , Epithelial Cells/cytology , Gene Expression Regulation , Genes, Reporter , Macrophages/cytology , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/metabolism , Promoter Regions, Genetic/genetics , Pulmonary Surfactant-Associated Protein C/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptor, Adenosine A2B/genetics , Recombinant Proteins/metabolism , beta-Galactosidase/metabolism
6.
Blood ; 114(3): 667-76, 2009 Jul 16.
Article in English | MEDLINE | ID: mdl-19433855

ABSTRACT

Ischemia-reperfusion injury (IRI) triggers an inflammatory cascade that is initiated by the activation of CD1d-restricted iNKT cells. In sickle cell disease (SCD), misshapen erythrocytes evoke repeated transient bouts of microvascular IRI. Compared with C57BL/6 controls, NY1DD mice have more numerous and activated (CD69(+), interferon-gamma(+) [IFN-gamma(+)]) lung, liver, and spleen iNKT cells that are hyperresponsive to hypoxia/reoxygenation. NY1DD mice have increased pulmonary levels of IFN-gamma, IFN-gamma-inducible chemokines (CXCL9, CXCL10), and elevated numbers of lymphocytes expressing the chemokine receptor CXCR3. Treating NY1DD mice with anti-CD1d antibody to inhibit iNKT cell activation reverses baseline pulmonary dysfunction manifested as elevated vascular permeability, decreased arterial oxygen saturation, and increased numbers of activated leukocytes. Anti-CD1d antibodies decrease pulmonary levels of IFN-gamma and CXCR3 chemokines. Neutralization of CXCR3 receptors ameliorates pulmonary dysfunction. Crossing NY1DD to lymphocyte-deficient Rag1(-/-) mice decreases pulmonary dysfunction. This is counteracted by the adoptive transfer of 1 million NKT cells. Like mice, people with SCD have increased numbers of activated circulating iNKT cells expressing CXCR3. Together, these data indicate that iNKT cells play a pivotal role in sustaining inflammation in SCD mice by a pathway involving IFN-gamma and production of chemotactic CXCR3 chemokines and that this mechanism may translate to human disease.


Subject(s)
Anemia, Sickle Cell/pathology , Inflammation/etiology , Interferon-gamma/biosynthesis , Killer Cells, Natural/immunology , Lung/pathology , Receptors, CXCR3/biosynthesis , Adoptive Transfer , Anemia, Sickle Cell/immunology , Animals , Chemokines/biosynthesis , Humans , Interferon-gamma/metabolism , Killer Cells, Natural/transplantation , Mice
7.
Am J Physiol Heart Circ Physiol ; 293(3): H1371-83, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17513486

ABSTRACT

In the microcirculation, longitudinal conduction of vasomotor responses provides an essential means of coordinating flow distribution among vessels in a complex network. Spread of current along the vessel axis can display a regenerative component, which leads to propagation of vasomotor signals over many millimeters; the ionic basis for the regenerative response is unknown. We examined the responses to 10 s of focal electrical stimulation (30 Hz, 2 ms, 30 V) of mouse cremaster arterioles to test the hypothesis that voltage-dependent Na(+) (Na(v)) and Ca(2+) channels might be activated in long-distance signaling in microvessels. Electrical stimulation evoked a vasoconstriction at the site of stimulation and a spreading, nondecremental conducted dilation. Endothelial damage (air bubble) blocked conduction of the vasodilation, indicating an involvement of the endothelium. The Na(v) channel blocker bupivacaine also blocked conduction, and TTX attenuated it. The Na(v) channel activator veratridine induced an endothelium-dependent dilation. The Na(v) channel isoforms Na(v)1.2, Na(v)1.6, and Na(v)1.9 were detected in the endothelial cells of cremaster arterioles by immunocytochemistry. These findings are consistent with the involvement of Na(v) channels in the conducted response. BAPTA buffering of endothelial cell Ca(2+) delayed and reduced the conducted dilation, which was almost eliminated by Ni(2+), amiloride, or deletion of alpha(1H) T-type Ca(2+) (Ca(v)3.2) channels. Blockade of endothelial nitric oxide synthase or Ca(2+)-activated K(+) channels also inhibited the conducted vasodilation. Our findings indicate that an electrically induced signal can propagate along the vessel axis via the endothelium and can induce sequential activation of Na(v) and Ca(v)3.2 channels. The resultant Ca(2+) influx activates endothelial nitric oxide synthase and Ca(2+)-activated K(+) channels, triggering vasodilation.


Subject(s)
Calcium Channels, T-Type/physiology , Calcium Channels/physiology , Endothelium, Vascular/physiology , Sodium Channels/physiology , Vasomotor System/physiology , Animals , Bupivacaine/pharmacology , Calcium Signaling/physiology , Electric Stimulation , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microcirculation/physiology , Nitric Oxide/metabolism , Potassium Channels, Calcium-Activated/physiology , Sodium Channel Blockers/pharmacology , Vasodilation/drug effects , Vasodilation/physiology , Veratridine/pharmacology
8.
Circ Res ; 100(2): 246-54, 2007 Feb 02.
Article in English | MEDLINE | ID: mdl-17218602

ABSTRACT

Second messenger signaling between endothelial cells (ECs) and vascular smooth muscle cells (VSMCs) is poorly understood, but intracellular Ca2+ concentrations ([Ca2+]i) in the 2 cells are coordinated, possibly through gap junctions at the myoendothelial junction. To study heterocellular calcium signaling, we used a vascular cell coculture model composed of monolayers of ECs and VSMCs. Stimulation of either cell type leads to an increase in [Ca2+]i in the stimulated cell and a secondary increase in [Ca2+]i in the other cell type that was blocked by gap junction inhibitors. To determine which second messengers are involved, we initially depleted Ca2+ stores in the endoplasmic reticulum Ca2+ with thapsigargin in ECs or VSMCs, but this had no effect on heterocellular calcium signaling. Alternatively, we loaded ECs or VSMCs with 1,2-bis(2-aminophenoxy)ethane-N,N,N',N'-tetraacetic acid (BAPTA) to buffer changes in [Ca2+]i. BAPTA loading of ECs inhibited agonist-induced increases in intracellular calcium concentration ([Ca2+]i), in both ECs and VSMCs. In contrast, BAPTA loading of the VSMCs blunted the VSMC response but did not alter the secondary increase in EC [Ca2+]i. Xestospongin C (an inositol 1,4,5-trisphosphate receptor inhibitor) had no effect on the secondary Ca2+ response, but when xestospongin C or thapsigargin was loaded into ECs and BAPTA into VSMCs, intercellular Ca2+ signaling was completely blocked. We conclude that 1,4,5-trisphosphate and Ca2+ originating in the VSMCs induces the secondary increase in EC [Ca2+]i but stimulation of the ECs generates a Ca2+ dependent response in the VSMCs.


Subject(s)
Calcium Signaling/physiology , Calcium/physiology , Endothelium, Vascular/physiology , Gap Junctions/physiology , Inositol 1,4,5-Trisphosphate/physiology , Muscle, Smooth, Vascular/physiology , Animals , Cell Communication/physiology , Cells, Cultured , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Mice , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/metabolism
9.
Circulation ; 114(19): 2056-64, 2006 Nov 07.
Article in English | MEDLINE | ID: mdl-17060376

ABSTRACT

BACKGROUND: We previously used adenosine A2A receptor (A2AR) knockout (KO) mice and bone marrow transplantation to show that the infarct-sparing effect of A2AR activation at reperfusion is primarily due to effects on bone marrow-derived cells. In this study we show that CD4+ but not CD8+ T lymphocytes contribute to myocardial ischemia/reperfusion injury. METHOD AND RESULTS: After a 45-minute occlusion of the left anterior descending coronary artery and reperfusion, T cells accumulate in the infarct zone within 2 minutes. Addition of 10 microg/kg of the A2AR agonist ATL146e 5 minutes before reperfusion produces a significant reduction in T-cell accumulation and a significant reduction in infarct size (percentage of risk area) measured at 24 hours. In Rag1 KO mice lacking mature lymphocytes, infarct size is significantly smaller than in C57BL/6 mice. Infarct size in Rag1 KO mice is increased to the level of B6 mice by adoptive transfer of 50 million CD4+ T lymphocytes derived from C57BL/6 or A2AR KO but not interferon-gamma KO mice. ATL146e completely blocked the increase in infarct size in Rag1 KO mice reconstituted with B6 but not A2AR KO CD4+ T cells. The number of neutrophils in the reperfused heart at 24 hours after infarction correlated well with the number of lymphocytes and infarct size. CONCLUSIONS: These results strongly suggest that the infarct-sparing effect of A2AR activation is primarily due to inhibition of CD4+ T-cell accumulation and activation in the reperfused heart.


Subject(s)
CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/pathology , Myocardial Infarction/metabolism , Myocardial Infarction/prevention & control , Receptor, Adenosine A2A/physiology , Animals , Bone Marrow Cells/immunology , Bone Marrow Cells/metabolism , Bone Marrow Cells/pathology , CD4-Positive T-Lymphocytes/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocardial Infarction/genetics , Myocardial Infarction/pathology , Myocardial Reperfusion/methods , Neutrophil Activation/genetics , Neutrophil Activation/immunology , Receptor, Adenosine A2A/deficiency , Receptor, Adenosine A2A/genetics
10.
Am J Physiol Heart Circ Physiol ; 290(2): H692-9, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16199480

ABSTRACT

Platelets become activated during myocardial infarction (MI), but the direct contribution of activated platelets to myocardial reperfusion injury in vivo has yet to be reported. We tested the hypothesis that activated platelets contribute importantly to reperfusion injury during MI in mice. After 30 min of ischemia and 60 min of reperfusion, P-selectin knockout mice had a significantly smaller infarct size than that of wild-type mice (P < 0.05). Platelets were detected by P-selectin antibody in the previously ischemic region of wild-type mice as early as 2 min postreperfusion after 45 min, but not 20 min, of ischemia. The appearance of neutrophils in the heart was delayed when compared with platelets. Flow cytometry showed that the number of activated platelets more than doubled after 45 min of ischemia when compared with 20 min of ischemia or sham treatment (P < 0.05). Platelet-rich or platelet-poor plasma was then transfused from either sham-operated or infarcted mice after 45 and 10 min of ischemia-reperfusion to mice undergoing 20 and 60 min of ischemia-reperfusion. Infarct size was increased by threefold and platelet accumulation was remarkably enhanced in mice treated with wild-type, MI-activated platelet-rich plasma but not in mice receiving either platelet-poor plasma from wild types or MI-activated platelet-rich plasma from P-selectin knockout mice. In conclusion, circulating platelets become activated early during reperfusion and their activation depends on the duration of the preceding coronary occlusion and is proportional to the extent of myocardial injury. Activated platelets play an important role in the process of myocardial ischemia-reperfusion injury, and platelet-derived P-selectin is a critical mediator.


Subject(s)
Myocardial Reperfusion Injury/blood , Myocardial Reperfusion Injury/pathology , Platelet Activation , Animals , Blood Platelets/pathology , Coronary Disease/complications , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocardial Infarction/etiology , Myocardial Infarction/pathology , Myocardial Reperfusion Injury/immunology , Myocardial Reperfusion Injury/metabolism , Myocardium/pathology , Neutrophil Infiltration , P-Selectin/metabolism , Time Factors
11.
Am J Physiol Heart Circ Physiol ; 290(4): H1410-8, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16284233

ABSTRACT

Following myocardial infarction (MI), contractile dysfunction develops not only in the infarct zone but also in noninfarcted regions of the left ventricle remote from the infarct zone. Inflammatory activation secondary to MI stimulates inducible nitric oxide synthase (iNOS) induction with excess production of nitric oxide. We hypothesized that the anti-inflammatory effects of selective A(2A)-adenosine receptor (A(2A)AR) stimulation would suppress inflammation and preserve cardiac function in the remote zone early after MI. A total of 53 mice underwent 60 min of coronary occlusion followed by 24 h of reperfusion. The A(2A)AR agonist (ATL146e, 2.4 microg/kg) was administered intraperitoneally 1, 3, and 6 h postreperfusion. Because of the 1-h delay in treatment after MI, ATL146e had no effect on infarct size, as demonstrated by contrast-enhanced cardiac MRI (n = 18) performed 24 h post-MI. ATL146e did however preserve global cardiac function at that time by limiting contractile dysfunction in remote regions [left ventricle wall thickening: 51 +/- 4% in treated (n = 9) vs. 29 +/- 3% in nontreated groups (n = 9), P < 0.01]. RT-PCR, immunohistochemistry, and Western blot analysis indicated that iNOS mRNA and protein expression were significantly reduced by ATL146e treatment in both infarcted and noninfarcted zones. Similarly, elevations in plasma nitrate-nitrite after MI were substantially blunted by ATL146e (P < 0.01). Finally, treatment with ATL146e reduced NF-kappaB activation in the myocardium by over 50%, not only in the infarct zone but also in noninfarcted regions (P < 0.05). In conclusion, A(2A)AR stimulation after MI suppresses inflammatory activation and preserves cardiac function, suggesting the potential utility of A(2A)AR agonists against acute heart failure in the immediate post-MI period.


Subject(s)
Cytokines/immunology , Myocardial Contraction/immunology , Myocardial Reperfusion Injury/immunology , Nitric Oxide Synthase Type II/immunology , Receptor, Adenosine A2A/immunology , Ventricular Dysfunction, Left/immunology , Animals , Mice , Myocardial Reperfusion Injury/complications , Tissue Distribution , Ventricular Dysfunction, Left/etiology
12.
J Immunol ; 174(8): 5040-6, 2005 Apr 15.
Article in English | MEDLINE | ID: mdl-15814735

ABSTRACT

Activation of the A2A adenosine receptor (A(2A)R) during reperfusion of various tissues has been found to markedly reduce ischemia-reperfusion injury. In this study, we used bone marrow transplantation (BMT) to create chimeric mice that either selectively lack or selectively express the A(2A)R on bone marrow-derived cells. Bolus i.p. injection of the selective A2A agonist, 4-[3-[6-amino-9-(5-cyclopropylcarbamoyl-3,4-dihydroxy-tetrahydro-furan-2-yl)-9H-purin-2-yl]-prop-2-ynyl]-piperidine-1-carboxylic acid methyl ester (ATL313; 3 microg/kg), at the time of reperfusion protects wild-type (wt) mice from liver ischemia-reperfusion injury. ATL313 also protects wt/wt (donor/recipient BMT mouse chimera) and wt/knockout chimera but produces modest protection of knockout/wt chimera as assessed by alanine aminotransferase activity, induction of cytokine transcripts (RANTES, IFN-gamma-inducible protein-10, IL-1alpha, IL-1-beta, IL-1Ralpha, IL-18, IL-6, and IFN-gamma), or histological criteria. ATL313, which is highly selective for the A(2A)R, produces more liver protection of chimeric BMT mice than 4-[3-[6-amino-9-(5-ethylcarbamoyl-3,4-dihydroxy-tetrahydro-furan-2-yl)-9H-purin-2-yl]-prop-2-ynyl]-cyclohexanecarboxylic acid methyl ester, which is rapidly metabolized in mice to produce 4-[3-[6-amino-9-(5-ethylcarbamoyl-3,4-dihydroxy-tetrahydro-furan-2-yl)-9H-purin-2-yl]-prop-2-ynyl]-cyclohexanecarboxylic acid, which has similar affinity for the A(2A)R and the proinflammatory A3 adenosine receptor. GFP chimera mice were created to show that vascular endothelial cells in the injured liver do not account for liver protection because they are not derived by transdifferentiation of bone marrow precursors. The data suggest that activation of the A(2A)R on bone marrow-derived cells is primarily responsible for protecting the liver from reperfusion injury.


Subject(s)
Bone Marrow Cells/metabolism , Liver/injuries , Liver/metabolism , Receptor, Adenosine A2A/metabolism , Reperfusion Injury/prevention & control , Adenosine A2 Receptor Agonists , Alanine Transaminase/blood , Animals , Bone Marrow Cells/cytology , Bone Marrow Transplantation , Cell Differentiation , Chimera , Cyclohexanecarboxylic Acids/pharmacology , Cytokines/genetics , Endothelium, Vascular/cytology , Liver/blood supply , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Purines/pharmacology , Receptor, Adenosine A2A/deficiency , Receptor, Adenosine A2A/genetics , Reperfusion Injury/metabolism
13.
Circulation ; 111(17): 2190-7, 2005 May 03.
Article in English | MEDLINE | ID: mdl-15851591

ABSTRACT

BACKGROUND: A2A-adenosine receptor (A2AAR) activation on reperfusion after ischemia reduces the size of myocardial infarction, but the mechanism of action has not been fully defined. METHODS AND RESULTS: We created chimeric mice by bone marrow transplantation from A2AAR-knockout or green fluorescent donor mice to irradiated congenic C57BL/6 (B6) recipients. In the GFP chimeras, we were unable to detect green fluorescent-producing cells in the vascular endothelium, indicating that bone marrow-derived cells were not recruited to endothelium at appreciable levels after bone marrow transplantation and/or acute myocardial infarction. Injection of 5 or 10 microg/kg of a potent and selective agonist of A2AAR, ATL146e, had no effect on hemodynamic parameters but reduced infarct size in B6 mice after 45 minutes of left anterior descending artery occlusion followed by 24 hours of reperfusion to 42.5+/-3.0% and 39.3+/-4.7% of risk region, respectively, compared with 61.0+/-2.3% in vehicle-treated B6 mice (P<0.05). Myocardial myeloperoxidase activity in the risk region measured at 4 hours after reperfusion was significantly reduced by ATL146e. The salutary effects of ATL146e were absent in A2AAR-knockout mice or in mice treated with a selective A2AAR antagonist, ZM241385. ATL146e also reduced infarct size and myeloperoxidase in B6/B6 (donor/recipient) chimeras (P<0.05) but not in A2AAR-knockout/B6 chimeras. In immunocompromised Rag-1-KO mice, infarct size was significantly reduced compared with B6 mice but was not further reduced by ATL146e. CONCLUSIONS: The results indicate that A2AAR activation on bone marrow-derived cells, specifically T or B lymphocytes, is responsible for the infarct-sparing and antiinflammatory effects of ATL146e administered at the time of reperfusion after coronary occlusion.


Subject(s)
Lymphocytes/metabolism , Myocardial Infarction/prevention & control , Myocardial Reperfusion/methods , Receptor, Adenosine A2A/metabolism , Adenosine A2 Receptor Agonists , Adenosine A2 Receptor Antagonists , Animals , Bone Marrow Cells , Bone Marrow Transplantation , Inflammation/prevention & control , Mice , Mice, Knockout , Myocardial Infarction/drug therapy , Peroxidase/metabolism , Transplantation Chimera
SELECTION OF CITATIONS
SEARCH DETAIL
...