Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters











Database
Publication year range
1.
Heart Vessels ; 26(6): 637-45, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21221603

ABSTRACT

The importance of the interaction between natural killer (NK) cells and dendritic cells (DCs) in the expansion of antiviral and antitumor immune responses is well documented; however, limited information on NK/DC interaction during atherosclerosis is available. Inflammation plays an important role in the development of atherosclerosis, and oxidized low-density lipoprotein (ox-LDL) is believed to play a critical role in the development and progression of atherosclerosis. In this study, we developed a NK/DC coculture system to examine the role of ox-LDL in modulating the interaction of mice NK cells and DCs. Fresh NK cells were cocultured with DCs in the absence or presence of ox-LDL. We examined the cytokines released during the interaction. This report provides the first evidence of an enhancement effect by ox-LDL on the NK/DC crosstalk. Notably, we found that ox-LDL significantly promoted the interaction of NK cells and DCs via CD48-2B4 contact-dependent mechanisms. These findings highlight the importance of NK/DCs crosstalk in atherosclerosis and provide new information about the possible mechanisms of atherosclerosis.


Subject(s)
Antigens, CD/metabolism , Cell Communication , Dendritic Cells/metabolism , Killer Cells, Natural/metabolism , Lipoproteins, LDL/metabolism , Receptors, Immunologic/metabolism , Animals , Atherosclerosis/immunology , Atherosclerosis/metabolism , CD48 Antigen , Coculture Techniques , Cytotoxicity, Immunologic , Dendritic Cells/immunology , Female , Humans , Inflammation/immunology , Inflammation/metabolism , Inflammation Mediators/metabolism , Interferon-gamma/metabolism , Interleukin-12/metabolism , K562 Cells , Killer Cells, Natural/immunology , Mice , Mice, Inbred C57BL , Signaling Lymphocytic Activation Molecule Family
2.
Zhonghua Yi Xue Za Zhi ; 88(12): 835-9, 2008 Mar 25.
Article in Chinese | MEDLINE | ID: mdl-18756989

ABSTRACT

OBJECTIVE: To investigate the effect of pioglitazone (Pio) on dendritic cell-(DC) specific intercellular adhesion molecule-3 grabbing nonintegrin (DC-SIGN) expression in DCs and explore the possible mechanism of Pio inhibiting DC adhesion and transmigration. METHODS: DCs derived from human peripheral blood mononuclear cells were cultured and divided into 6 groups: blank control group, Pio 0.1 micromol/L group, Pio 1.0 micromol/L group, Pio 10 micromol/L group, GW9662, a peroxisome proliferator activated receptor (PPAR)-gamma antagonist, 10 micromol/L group, and GW9662 10 micromol/L + Pio 10 micromol/L group. Western blotting was used to detect the protein expression of DC-SIGN 24 h later. Human umbilical vein endothelial cells (HUVECs) were obtained and co-cultured with the DCs undergoing different treatments. Immunofluorescence test was used to detect the protein expression of DC-SIGN. DCs labeled with 5-chloromethylfluorescein diacetate (CMFDA) were added into the monocellular layer of fused ECs. Blank DCs and DCs pretreated with anti-DC-SIGN antibody were used as blank and experimental groups. Laser confocal microscopy was used to observe the adhesion ability of the DCs. HUVECs were inoculated into the upper chamber of Transwell plate and CMFDA-labeled DCs of above mentioned groups were added to the mono-cellular layer of these ECs. Serum-free culture medium with monocyte chemoattractant protein-1 was added into the lower chamber. Eight hours later the transmigration ability was observed. RESULTS: Western blotting showed that the DC-SIGN protein expression levels of the DCs of the Pio 1.0 micromol/L and Pio 10 micromol/L group were 0.96 +/- 0.09 and 0.80 +/- 0.08 respectively, both significantly lower than that of the blank control group (1.25 +/- 0.23, P < 0.05 and P < 0.01); and the DC-SIGN protein expression level of the GW9662 10 micromol/L + Pio 10 micromol/L group was 1.10 +/- 0.12, significantly higher than that of the Pio 10 micromol/L group (P < 0.05). Immunofluorescence test showed that the DC-SIGN protein expression levels of the DCs of the Pio 1.0 micromol/L and Pio 10 micromol/L group were 22.3 +/- 5.4 and 14.4 +/- 2.3 respectively, both significantly lower than that of the control group (29.5 +/- 5.1, P < 0.05 and P < 0.01), and the DC-SIGN protein expression level of the GW9662 10 micromol/L + Pio 10 micromol/L group was 24.9 +/- 4.3, significantly higher than that of the Pio 10 micromol/L group (P < 0.01), and not significantly different from that of the blank control group (P > 0.05). The adhesion rates of the Pio 1.0 micromol/L and Pio 10 micromol/L groups were 10.8% +/- 2.0% and 7.6% +/- 1.5% respectively, both significantly lower than that of the control group (13.4% +/- 2.1%, P < 0.05 and P < 0.01); and the adhesion rate of the GW9662 + Pio 10 micromol/L group was 12.1% +/- 1.9%, significantly higher than that of the Pio 10 micromol/L group (P < 0.01), and not significantly different from that of the blank control group (P > 0.05). The transmigration inhibition rate of DCs of the Pio 0.1, 1.0, and 10 micromol/L groups were 4.1%, 12.9%, and 17.2% compared with the transmigration rate of the blank control group (P < 0.05, P < 0.05, and P < 0.01). The transmigration rate of the GW9662 + Pio 10 micromol/L group was significantly higher than that of the Pio 10 micromol/L group (P < 0.05), and not significantly different from that of the blank control group (P > 0.05). The transmigration rate of the anti-DC-SIGN intervention group was lower by 17.8% than that of the control group (P < 0.01). CONCLUSION: Pio down-regulates the DC-SIGN protein expression and inhibits DC adhesion and transmigration through the pathway of PPAR-gamma.


Subject(s)
Cell Adhesion Molecules/metabolism , Cell Movement/drug effects , Dendritic Cells/drug effects , Lectins, C-Type/metabolism , Receptors, Cell Surface/metabolism , Thiazolidinediones/pharmacology , Anilides/pharmacology , Blotting, Western , Cell Adhesion/drug effects , Cell Line , Cells, Cultured , Coculture Techniques , Dendritic Cells/cytology , Dendritic Cells/metabolism , Down-Regulation , Endothelial Cells/cytology , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Humans , Microscopy, Confocal , Pioglitazone
3.
Int Immunopharmacol ; 7(9): 1241-50, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17630203

ABSTRACT

Elevated plasma homocysteine (Hcy) is an independent risk factor for atherosclerosis, which is recognized as inflammatory and immune responses. The purpose of this study was to investigate the effect of Hcy on the interaction between dendritic cells (DCs) and endothelial cells (ECs) by upregulating the expression of DC-specific ICAM-3-grabbing nonintegrin (DC-SIGN) in cultured DCs. The immunophenotype of Hcy-treated DCs was monitored by flow cytometry. Then, they were coincubated with cultured human umbilical vein endothelial cells, and adhesion of DCs to ECs, and migration of DCs through an endothelial monolayer growing on the insert of a transwell plate, were assessed using a confocal microscope and a multi-detection microplate reader. The expression of DC-SIGN on Hcy-stimulated DCs was assessed by Western blot and immunofluorescence staining. The presence of Hcy did not change the phenotype of immature and mature DCs. Hcy promoted adhesion of DCs to ECs and migration in a concentration-dependent fashion. This effect was inhibited by an anti-DC-SIGN monoclonal antibody. The expression of DC-SIGN on DCs was significantly upregulated by Hcy in a concentration-dependent manner. Taken together, our results show for the first time that Hcy can potentiate the adhesion of DCs to ECs and migration by upregulating the expression of DC-SIGN on DCs, suggesting a novel role of Hcy in the pathogenesis of human vascular disease.


Subject(s)
Cell Adhesion Molecules/physiology , Cell Movement/physiology , Dendritic Cells/metabolism , Endothelial Cells/metabolism , Homocysteine/physiology , Lectins, C-Type/physiology , Receptors, Cell Surface/physiology , Cell Adhesion/physiology , Cells, Cultured , Dendritic Cells/pathology , Endothelial Cells/pathology , Homocysteine/toxicity , Humans
4.
Article in Chinese | MEDLINE | ID: mdl-21186583

ABSTRACT

AIM: To determine whether activation of kappa-opioid receptor with U50,488H, a selective kappa-opioid receptor agonist, produces any changes in electrical uncoupling during prolonged ischemia and whether these changes in electrical uncoupling is associated with the cardioprotection induced by kappa-opioid receptor activation, and to explore the possible mechanism. METHODS: (1) To observe the effect of U50,488H (10(-7), 10(-6), 3 x10(-6) and 10(-5) mol/L), a selective kappa-opioid receptor agonist, or with a selective kappa-opioid receptor antagonist nor-BNI (5 x 10(-6) mol/L), or with a mitochondrial K(ATP) channel inhibitor 5-HD on myocardium during ischemia/reperfusion in isolated perfused rat heart. Parameters of measurements include hemodynamic data, formazan content, heart rate, coronary flow, and lactate dehydrogenase (LDH). (2) To examine the effect of U50,488H of different concentration on electrical coupling parameters (including onset of uncoupling, plateau time, slope, and fold increase in r1) during 70 min myocardial ischemia in isolated perfused rat heart. RESULTS: (1) Pretreatment with U50,488H concentration dependently increased formazan content and reduced LDH release induced by 30 min of ischemia and 120 min of reperfusion. (2) The onset of electrical uncoupling and plateau time during prolonged ischemia was delayed by kappa-opioid receptor activation with U50,488H. (3) Linear regression analysis shown that the increase in formazan content and decrease in LDH release produced by kappa-opioid receptor activation was associated with delayed electrical uncoupling during prolonged ischemia. (4) The effects of U50,488H on formazan content, LDH release and on electrical coupling were abolished by nor-BNI, or 5-HD. CONCLUSION: This results demonstrate that the onset of electrical uncoupling during prolonged ischemia is delayed by kappa-opioid receptor activation with a selective kappa-opioid receptor agonist U50,488H, and that delayed electrical uncoupling is associated with the cardioprotection induced by kappa-opioid receptor activation with U50,488H. These effects of kappa-opioid receptor activation with U50,488H are mediated by mitochondrial K(ATP) channels.


Subject(s)
Heart/physiopathology , Myocardial Ischemia/physiopathology , Receptors, Opioid, kappa , 3,4-Dichloro-N-methyl-N-(2-(1-pyrrolidinyl)-cyclohexyl)-benzeneacetamide, (trans)-Isomer/pharmacology , Animals , Antihypertensive Agents , Heart/drug effects , In Vitro Techniques , Male , Myocardium/metabolism , Naltrexone/analogs & derivatives , Naltrexone/pharmacology , Potassium Channels/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Opioid, kappa/agonists
5.
Scand Cardiovasc J ; 39(6): 375-82, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16352491

ABSTRACT

OBJECTIVES: To determine whether the kappa-opioid receptor agonist U50,488H affects electrical uncoupling during prolonged ischemia and, if so, whether the changes are associated with its cardioprotective action. DESIGN: The isolated rat heart was perfused in a Langendorff apparatus. Formazan content, lactate dehydrogenase (LDH) and hemodynamic parameters were measured to confirm the cardioprotective effect of U50,488H. The effects of U50,488H on electrical coupling during prolonged ischemia were also measured. RESULTS: U50,488H concentration-dependently increased formazan content and reduced LDH release, and the ameliorating effect of 10(-5) mol/L U50,488H was abolished by 5 x 10(-6) mol/L nor-binaltorphimine (nor-BNI), a selective kappa-opioid receptor antagonist, or 10(-4) mol/L 5-hydroxydecanoate (5-HD), a selective mitochondrial ATP-sensitive K(+) (K(ATP)) channel blocker. The onset of electrical uncoupling during prolonged ischemia was delayed by U50,488H, and the delay was not only abolished, but also advanced by nor-BNI or 5-HD relative to the control group. CONCLUSIONS: These results demonstrate that delayed uncoupling during prolonged ischemia is associated with the cardioprotection of U50,488H, and these effects of U50,488H are mediated by mitochondrial K(ATP) channels.


Subject(s)
3,4-Dichloro-N-methyl-N-(2-(1-pyrrolidinyl)-cyclohexyl)-benzeneacetamide, (trans)-Isomer/pharmacology , Cardiotonic Agents/pharmacology , Heart/drug effects , Myocardial Ischemia/prevention & control , Potassium Channels/drug effects , Receptors, Opioid, kappa/agonists , Animals , Cell Communication , Electrophysiologic Techniques, Cardiac , Formazans , Gap Junctions/drug effects , HSC70 Heat-Shock Proteins/drug effects , Heart/physiopathology , Male , Myocardial Ischemia/physiopathology , Rats , Rats, Sprague-Dawley , Time Factors
6.
Clin Exp Pharmacol Physiol ; 32(8): 655-62, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16120193

ABSTRACT

1. It is known that infusion of the gap junction uncoupler heptanol, before ischaemia or during reperfusion, limits myocardial infarct size. However, whether this cardiac effect is linked to the effect of heptanol on communication across gap junctions has not been elucidated. The aims of the present study were to examine the effect of heptanol on infarct size, arrhythmias and myocardial tissue resistance and to assess whether changes in electrical coupling correlate with cardiac protection. 2. Rat isolated, perfused hearts were subjected to a 24 min infusion of heptanol (0.05, 0.1, 0.5 or 1.0 mmol/L) followed by 20 min regional ischaemia and 60 min reperfusion, or by 70 min global no-flow ischaemia. The effective refractory period, action potential and conduction velocity were measured in papillary muscles from the right ventricle. Heptanol markedly decreased arrhythmia scores during ischaemia and reperfusion, as well as reducing infarct size to a degree similar to that induced by ischaemic preconditioning. In the prolonged ischaemia model, heptanol delayed the onset of uncoupling, increased time to plateau and decreased the maximal rate of uncoupling during ischaemia. Ischaemic preconditioning had similar effects on these parameters. In papillary muscle, heptanol reduced the conduction velocity of the action potential in a dose-dependent manner, but had no significant effect on resting potential, action potential amplitude, action potential duration, maximal upstroke of depolarization or effective refractory period. 3. These results demonstrate that treatment with the gap junction uncoupler heptanol confers cardioprotection against ischaemia and this effect is related to delayed electrical uncoupling during prolonged ischaemia.


Subject(s)
Heart/drug effects , Heptanol/pharmacology , Myocardial Ischemia/prevention & control , Myocardial Ischemia/physiopathology , Animals , Dose-Response Relationship, Drug , Electrophysiology , Gap Junctions/drug effects , Male , Myocardial Reperfusion Injury/prevention & control , Organ Culture Techniques , Papillary Muscles/drug effects , Rats , Rats, Sprague-Dawley
7.
Article in English | MEDLINE | ID: mdl-17282127

ABSTRACT

Two series of experiments were performed in the perfused isolated rat heart to determine whether stimulation of κ-opioid receptor with U50,488H, a selective κ-opioid receptor agonist, produces any changes in electrical coupling during prolonged ischemia and whether these changes in electrical coupling is associated with the cardioprotection induced by U50,488H. It was found that U50,488H concentration dependently increased formazan content and reduced lactate dehydrogenase (LDH) release induced by 30 min of ischemia and 120 min of reperfusion, and the ameliorating effect of 10-5mol/L U50,488H was abolished by 5x10-6mol/L nor-binaltorphimine (nor-BNI), a selective Κ-opioid receptor antagonist, or 10-4mol/L 5-hydroxydecanoate (5-HD), a selective mitochondrial ATP-sensitive K+(KATP) channels blocker. The onset of electrical uncoupling during prolonged ischemia was delayed by U50,488H, and delaying effect was not only abolished, but also advanced by nor-BNI or 5-HD compared with control group. These results demonstrate that delayed electrical uncoupling is associated with the cardioprotection induced by U50,488H. These effects of U50,488H are mediated by mitochondrial KATPchannels.

8.
Article in English | MEDLINE | ID: mdl-17282126

ABSTRACT

The aims of the present study were to examine the effect of heptanol on electrical coupling during ischemia, and to assess whether changes in electrical coupling by heptanol is associated with its cardiac protection. Perfused isolated rat hearts were subjected to a 24 min infusion of heptanol (0.05, 0.1, 0.5 or 1.0 mmol/L) followed by 70 min of global no-flow ischemia or by 20 min of regional ischemia and 60 min of reperfusion. Heptanol markedly decreased arrhythmia scores during ischemia and reperfusion as well as reduced infarct size to a degree similar to that induced by ischemic preconditioning. In the prolonged ischemia model, heptanol delayed the onset of uncoupling, increased time to plateau, and decreased the maximal rate of uncoupling during ischemia. Ischemic preconditioning had similar effects on these parameters. These results demonstrate that treatment with the gap junction uncoupler heptanol confers cardioprotection against ischemia, and this effect is related to delayed electrical uncoupling during prolonged ischemia.

SELECTION OF CITATIONS
SEARCH DETAIL