Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Clin Exp Pharmacol Physiol ; 49(8): 805-812, 2022 08.
Article in English | MEDLINE | ID: mdl-35577580

ABSTRACT

Atherosclerosis is associated with a haemostatic imbalance characterized by excessive activation of pro-inflammatory and pro-coagulant pathways. Non-vitamin K antagonists oral anticoagulant (NOACs) may reduce the incidence of cardiovascular events, cerebral ischemia, thromboembolic events and atherosclerosis. Chronic inflammation, vascular proliferation and the development of atherosclerosis is also influenced by 25-hydroxycholesterol (25-OHC). The aim of the study was to assess the effect of rivaroxaban and dabigatran on the messenger RNA (mRNA) expression of anti-inflammatory cytokines transforming growth factor ß (TGF-ß), interleukin (IL)-37, IL-35 as well as of pro-inflammatory cytokines IL-18 and IL-23, in endothelial cells damaged by 25-OHC. Human umbilical vascular endothelial cells (HUVECs) were treated with 25-OHC (10 µg/mL), rivaroxaban (100, 500 ng/mL), dabigatran (100, 500 ng/mL), 25-OHC + rivaroxaban, and 25-OHC + dabigatran. The mRNA expression of TGF-ß, IL-37, IL-35 subunits EBI3 and p35, IL-18, and IL-23 was analysed using real-time polymerase chain reaction (PCR). The results showed that 25-OHC decreased TGF-ß and IL-37 mRNA expression and increased EBI3, p35, IL-18, IL-23 mRNA expression in endothelial cell as compared to an untreated control (P < .05). Messenger RNA expression of TGF-ß and IL-37 significantly increased following stimulation with rivaroxaban and dabigatran as compared to an untreated control (P < .01). In HUVECs pre-treated with oxysterol, rivaroxaban and dabigatran increased mRNA expression of TGF-ß, IL-37 and decreased mRNA expression of EBI3, p35, IL-23 and IL-18 as compared to 25-OHC (P < .01). Our finding suggests that both rivaroxaban and dabigatran inhibit the inflammatory activation caused by oxysterol in vitro.


Subject(s)
Atherosclerosis , Cytokines , Dabigatran , Human Umbilical Vein Endothelial Cells , Hydroxycholesterols , Rivaroxaban , Administration, Oral , Anticoagulants/pharmacology , Anticoagulants/therapeutic use , Atherosclerosis/drug therapy , Atherosclerosis/genetics , Atherosclerosis/immunology , Atrial Fibrillation/drug therapy , Cytokines/genetics , Cytokines/immunology , Dabigatran/pharmacology , Dabigatran/therapeutic use , Endothelial Cells/drug effects , Endothelial Cells/immunology , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/immunology , Humans , Hydroxycholesterols/administration & dosage , Hydroxycholesterols/adverse effects , Hydroxycholesterols/pharmacology , Interleukin-18/genetics , Interleukin-18/immunology , Interleukin-23/genetics , Interleukin-23/immunology , Oxysterols/administration & dosage , Oxysterols/adverse effects , Oxysterols/pharmacology , RNA, Messenger/genetics , RNA, Messenger/immunology , Rivaroxaban/pharmacology , Rivaroxaban/therapeutic use , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/immunology
2.
Biomed Pharmacother ; 147: 112679, 2022 Mar.
Article in English | MEDLINE | ID: mdl-35121342

ABSTRACT

Recent data showed that dabigatran can reduce not only procoagulatory effects but also block proinflammatory stimuli by inhibiting the expression of cytokines and chemokines and reducing thrombin-induced endothelial permeability. The aim of our study was to assess the effect of dabigatran on the integrity and inflammatory properties of endothelial cells stimulated by 25-hydroxycholesterol (25-OHC, oxysterol). HUVECs (Human Umbilical Vein Endothelial Cells) were stimulated with 25-hydroxycholesterol 10 µg/ml, dabigatran 100 ng/ml or 500 ng/ml and 25-hydroxycholesterol + dabigatran (100 ng/ml, 500 ng/ml). HUVEC integrity and permeability was measured in the RTCA-DP xCELLigence system and by the paracellular flux system. The mRNA expression of ICAM-1, VEGF, IL-33, MCP-1 and TNF-α was analyzed by Real-time PCR. Cell apoptosis and viability was measured by flow cytometry. VEGF protein concentration was assessed in supernatants by ELISA. VE-cadherin expression in endothelial cells was evaluated by confocal microscopy. Pre-stimulation of HUVECs with 25-OHC decreased endothelial cell integrity (p < 0.001) and increased the expression of IL-33, ICAM-1, MCP-1, VEGF, TNF-α mRNA (p < 0.01) compared to unstimulated controls. Following stimulation of HUVECs with dabigatran 100 ng/ml or 500 ng/ml restored HUVEC integrity interrupted by 25-OHC (p < 0.001). In HUVECs pre-stimulated with oxysterol, dabigatran stimulation decreased mRNA expression of the proinflammatory cytokines IL-33 and TNF-α, chemokines MCP-1 ICAM-1 and VEGF (p < 0.01). Dabigatran 500 mg/ml+ 25-OHC increased the endothelial expression of VE-cadherin as compared to 25-OHC (p < 0.01). Our findings suggest that dabigatran stabilizes the endothelial barrier and inhibits the inflammation caused by oxysterol.


Subject(s)
Chemokines/drug effects , Cytokines/drug effects , Dabigatran/pharmacology , Endothelial Cells/drug effects , Oxysterols/pharmacology , Dose-Response Relationship, Drug , Human Umbilical Vein Endothelial Cells , Humans , Inflammation Mediators/metabolism , RNA, Messenger
4.
Tissue Barriers ; 9(4): 1956284, 2021 Oct 02.
Article in English | MEDLINE | ID: mdl-34323663

ABSTRACT

BACKGROUND: Rivaroxaban is one of the direct factor Xa inhibitors. Its function in the inactivated coagulation cascade is unclear. The aim of the study was to assess the effect of rivaroxaban on the endothelial integrity and inflammatory properties of endothelial cells stimulated by 25-hydroxycholesterol (25-OHC). METHODS: HUVECs were stimulated with 25-OHC, rivaroxaban and 25-OHC+ rivaroxaban. HUVEC integrity and permeability were measured using the xCELLigence system and paracellular flux assay. The mRNA expression of tissue factor, ICAM-1, VEGF, IL-33, MCP-1, TNF-α was analyzed in the real-time PCR. Apoptosis and viability were measured by flow cytometry. The VEGF protein concentration was assessed by ELISA. The confocal microscope was used to evaluate the expression of VE-cadherin in endothelial cells. RESULTS: 25-OHC decreased endothelial cell integrity and increased the mRNA expression of IL-33, tissue factor, ICAM-1, MCP-1, VEGF, TNF-α as compared to unstimulated controls. Following the stimulation with rivaroxaban, HUVEC restored integrity disrupted by 25-OHC (p < .01). In HUVECs pre-stimulated with oxysterol, rivaroxaban decreased mRNA expression of IL-33, TNF-α, chemokines MCP-1, ICAM-1, VEGF and tissue factor (p < .01). Rivaroxaban 100 mg/ml+25-OHC increased the VE-cadherin expression in endothelium as compared to 25-OHC (p < .05). CONCLUSION: Our finding suggests that rivaroxaban may restore the endothelial barrier and inhibit the inflammatory activation caused by oxysterol in vitro.


Subject(s)
Oxysterols , Rivaroxaban , Endothelium, Vascular , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Oxysterols/metabolism , Oxysterols/pharmacology , Rivaroxaban/metabolism , Rivaroxaban/pharmacology , Rivaroxaban/therapeutic use
5.
J Interferon Cytokine Res ; 40(1): 43-53, 2020 01.
Article in English | MEDLINE | ID: mdl-31460824

ABSTRACT

Postprandial hypertriglyceridemia is an independent risk factor for cardiovascular disease. The aim of this study was to assess the effects of a single fat-rich meal on barrier functions and inflammatory status on human umbilical vascular endothelial cells (HUVECs), furthermore we assess the effects of mixture of palmitic acid and 25-hydroxycholesterol (PA +25OHCH) on integrity of endothelial cells and their inflammatory properties. HUVECs were induced with serum of healthy volunteers taken before, and 3 h after, the consumption of a meal with a standardized daily required dose of fats. In addition, endothelial cells were induced with PA +25OHCH (800 µM/L+10 µg/mL). Total cholesterol, triglycerides (TGs), high-density lipoprotein cholesterol, low-density lipoprotein cholesterol, high sensitivity c-reactive protein, and glucose were measured at fasting and postprandially. HUVEC integrity was measured in the RTCA-DP xCELLigence system. mRNA expression of interleukin (IL)-33, IL-32, intercellular adhesion molecule-1 (ICAM-1), monocyte chemoattractant protein-1 (MCP-1), CX3C-chemokine, vascular endothelial growth factor (VEGF) occludin, and VE-cadherin was analyzed by real-time polymerase chain reaction. Viability and apoptosis were assessed in flow cytometry. The level of VEGF and IL-33 in fasting and postprandial serum was assessed by enzyme-linked immunosorbent assay (ELISA). Three hours after consumption of a fatty meal, all patients displayed increased levels of TGs and Toll-like receptors (110 ± 37 mg/dL versus 182 ± 64 mg/dL P < 0.05) (24 ± 11 mg/dL versus 42 ± 14 mg/dL P < 0.05). Postprandial serum and PA +25OHCH caused >20% decrease of HUVEC integrity than fasting serum (P < 0.001). HUVEC disintegration was accompanied by a decrease of occludin mRNA expression as compared with fasting serum (P < 0.05). The fatty meal affected neither VE-cadherin mRNA expression nor its apoptosis (P > 0.05). Mixture of PA +25OHCH caused decrease of VE-cadherin mRNA expression as compared with fasting serum (P < 0.01). PA +25OHCH did not affect HUVEC apoptosis (P > 0.05). Postprandial serum and PA +25OHCH caused increase of IL-33, MCP-1, ICAM-1, IL-32, VEGF, and CX3C-chemokine mRNA expression as compared with fasting serum (P < 0.05). Moreover, level of VEGF in fatty serum was significantly higher (P < 0.001). Postprandial lipemia after a single fatty meal may destabilize the endothelial barrier and initiate inflammatory processes.


Subject(s)
Human Umbilical Vein Endothelial Cells/drug effects , Inflammation/chemically induced , Triglycerides/administration & dosage , Triglycerides/adverse effects , Adult , Cells, Cultured , Cytokines/genetics , Cytokines/metabolism , Female , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Hydroxycholesterols/pharmacology , Inflammation/metabolism , Male , Meals , Palmitic Acid/pharmacology , Postprandial Period , RNA, Messenger/genetics , RNA, Messenger/metabolism , Triglycerides/blood
6.
APMIS ; 128(1): 10-19, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31642122

ABSTRACT

Atherogenesis is associated with chronic gut infections; however, the mechanisms are not clear. The aim of the study was to determine whether lipopolysaccharide of E. coli (E. coli LPS) may affect endothelial barrier and modify IL-10 expression in dendritic cells (DCs). Human umbilical vein endothelial cells (HUVECs) and monocyte-derived DCs were treated with E. coli LPS, apolipoprotein B100 (ApoB100) and 7-ketocholesterol (7-kCH) - harmful oxidized form of cholesterol. The effect of E. coli LPS, 7-kCH and ApoB100 on the barrier functions of HUVECs in real-time cell electric impedance sensing system (RTCA-DP) was assessed. Furthermore, the effect of 7-kCH and ApoB100 on barrier functions of HUVECs co-cultured with DCs previously treated with LPS was analyzed. Both E. coli LPS and 7-kCH decreased barrier functions of HUVECs and reduced tight junction protein mRNA expression, whereas ApoB100 increased endothelial barrier. In DCs, ApoB100 and E. coli LPS decreased IL-10 mRNA expression. In HUVECs co-cultured with DCs treated with LPS and subsequently pulsed with ApoB100 or 7-kCH, IL-10 mRNA expression was lower. E. coli LPS-exposed DCs diminished the protective effect of ApoB100 on endothelial integrity and led to the decrease in occludin mRNA expression. LPS potentially derived from gut microflora may destabilize endothelial barrier together with oxidized cholesterol and intensify the immunogenicity of ApoB100.


Subject(s)
Dendritic Cells/drug effects , Escherichia coli/immunology , Human Umbilical Vein Endothelial Cells/drug effects , Interleukin-10/genetics , Lipopolysaccharides/immunology , Tight Junctions/drug effects , Apolipoprotein B-100/pharmacology , Cells, Cultured , Coculture Techniques , Dendritic Cells/immunology , Escherichia coli/chemistry , Humans , Ketocholesterols/pharmacology , Lipopolysaccharides/pharmacology , Occludin/genetics , Signal Transduction/drug effects , Tight Junction Proteins/genetics
7.
Adv Clin Exp Med ; 28(9): 1243-1248, 2019 Sep.
Article in English | MEDLINE | ID: mdl-31430072

ABSTRACT

BACKGROUND: FoxP3 is a marker of human T regulatory cells (Tregs), which are supposed to play an important role in the pathophysiology of atherosclerosis. Interleukin 10 (IL-10) is a cytokine with pleiotropic, immunoregulatory properties, produced mostly by Tregs and B regulatory cells. Due to their anti-inflammatory action, both Tregs and IL-10 are believed to inhibit plaque development and decrease atherosclerosis progression. The effect of hypolipidemic drugs - statins or ezetimibe - on FoxP3-positive Tregs and anti-inflammatory cytokines, such as IL-10, is still unclear. OBJECTIVES: The objective of the study was to investigate the effects of 3 different therapies of equivalent hypolipidemic activity: atorvastatin, rosuvastatin, and combination therapy of atorvastatin and ezetimibe on FoxP3-Tregs transcription factor and IL-10 mRNA expression in peripheral blood mononuclear cells (PBMCs) from patients with stable coronary artery disease (CAD). MATERIAL AND METHODS: Sixty-five patients with diagnosed CAD participated in the study. They were randomly assigned to 3 therapeutic groups: atorvastatin at a dose of 40 mg/day (A40 group); rosuvastatin 20 mg/day (R20 group); and atorvastatin 10 mg/day combined with ezetimibe 10 mg/day (A10+E10 group). After 1 month and 6 months of therapy, the mRNA expression for FoxP3 and IL-10 in PBMCs was evaluated using real-time polymerase chain reaction (RT-PCR) and lipid parameters. RESULTS: An improvement in lipid parameters was observed in each of the groups studied; however, hypolipidemic treatment did not induce any change in FoxP3 and IL-10 mRNA expression. After 6 months, an increase in FoxP3 mRNA expression was noted in A40 group as compared to R20 group. CONCLUSIONS: None of the therapies of equal hypolipidemic efficacy affected FoxP3 and IL-10 mRNA expression in patients with stable CAD.


Subject(s)
Anticholesteremic Agents , Coronary Artery Disease , Hydroxymethylglutaryl-CoA Reductase Inhibitors , Anticholesteremic Agents/therapeutic use , Atorvastatin/therapeutic use , Coronary Artery Disease/genetics , Coronary Artery Disease/metabolism , Ezetimibe/therapeutic use , Forkhead Transcription Factors/metabolism , Gene Expression Regulation , Humans , Interleukin-10/metabolism , Leukocytes, Mononuclear , RNA, Messenger
8.
Adv Clin Exp Med ; 26(3): 461-466, 2017.
Article in English | MEDLINE | ID: mdl-28791821

ABSTRACT

BACKGROUND: IL-22 is expressed at barrier surfaces, which suggests its critical role in the maintenance of normal barrier homeostasis and tissue repair. IL-22 can both promote pathological inflammation and prevent the destruction of tissues. The functional outcomes of IL-22 on vascular smooth muscle cells, which are shown to regulate immune processes within the vascular wall and which are involved in certain pathologies, have not been analyzed. OBJECTIVES: The effect of IL-22 on the expression of novel antiand pro-inflammatory and barrier disrupting cytokines, apoptosis and the expression of adhesive molecules in human primary aortic smooth muscle cells (AoSMC) was investigated. MATERIAL AND METHODS: Human AoSMC were induced with IL-22 for 24 h in vitro. The influence of IL-22 on IL-35 subunits EBI3 and p35, IL-33, IFN-γ and VEGF mRNA expression in Ao-SMC were assessed using real-time PCR. Additionally, the surface expression of ICAM-1 and apoptosis of AoSMC were analyzed in the flow cytometer. RESULTS: IL-22 caused a 2- and 3-fold increase of mRNA expression of the EBI3 and p35 IL-35 subunits, and a 40% decrease of IL-33 mRNA expression in AoSMC. Additionally, IL-22 decreased ICAM-1 expression on the surface of AoSMC by 30%. However, IL-22 affected neither IFN-γ and VEGF mRNA expression in AoSMC nor their apoptosis and viability. CONCLUSIONS: Our data suggest that IL-22, which is released by Th22 and NK cells, may be an agent affecting the inflammatory response of AoSMC, and thus it may regulate immune homeostasis of the vascular wall.


Subject(s)
Aorta/metabolism , Inflammation/metabolism , Interleukins/metabolism , Myocytes, Smooth Muscle/metabolism , Apoptosis/physiology , Cells, Cultured , Humans , Intercellular Adhesion Molecule-1/metabolism , Interferon-gamma/metabolism , Interleukin-33/metabolism , RNA, Messenger/metabolism , Vascular Endothelial Growth Factor A/metabolism , Interleukin-22
9.
Arch Med Sci ; 13(4): 778-784, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28721145

ABSTRACT

INTRODUCTION: Atherosclerosis leading to coronary artery disease (CAD) is a chronic inflammatory condition. Interleukin 35 (IL-35) released by regulatory T cells (Tregs) has been found to be associated with CAD in the Chinese population. However, nothing is known about the relation between IL-35 concentrations and cholesterol levels. The aim of the study was to assess the levels of IL-35 in CAD patients and healthy subjects from a Caucasian population, and to analyze the relationship between IL-35 and the levels of total cholesterol, low-density lipoprotein (LDL) and high-density lipoprotein (HDL) cholesterol, left ventricular ejection fraction (LVEF), sex and postmenopausal status. MATERIAL AND METHODS: Thirty-one patients with CAD and 30 healthy controls were included in the study. Levels of plasma IL-35 were analyzed by ELISA. The LVEF was assessed by transthoracic echocardiographic examination. Plasma levels of cholesterol fractions and C-reactive protein (CRP) were assessed by immunoenzymatic methods. RESULTS: The CAD patients had higher levels of IL-35 as compared to healthy controls (58.1 ±16.6 pg/ml vs. 5.35 ±3.35 pg/ml; p < 0.001). IL-35 levels negatively correlated with total and LDL cholesterol concentrations (R = -0.31, p < 0.01) and positively correlated with HDL cholesterol in men (R = 0.53, p < 0.01). In women, IL-35 levels negatively correlated with LVEF (R = -0.29, p < 0.05) and positively with the duration of postmenopausal status (R = 0.55, p < 0.01). CONCLUSIONS: These results suggest a possible association between high levels of IL-35 and CAD.

10.
Postepy Hig Med Dosw (Online) ; 71(0): 162-169, 2017 Mar 02.
Article in English | MEDLINE | ID: mdl-28258676

ABSTRACT

BACKGROUND: Human vascular endothelial function and integrity may be regulated by many non-specific factors. However, the potential influence of specific antigens via an IgE-mediated mechanism remains unknown. The aim of the study was to determine the expression of the IgE receptors FcεRI and FcεRII in the human vascular endothelium and to assess their relevance in the IgE-mediated regulation of endothelial integrity. MATERIAL/METHODS: FcεRI and FcεRII expression in human umbilical vein endothelial cells (HUVEC) was genetically assessed by PCR with respective primers and sequencing. HUVEC were cultured with IL-4, and changes in FcεRI and FcεRII mRNA expression were analyzed by real-time PCR. Changes in the integrity of endothelium pre-treated with anti-BSA-DNP IgE following exposure to the specific BSA-DNP antigen was assessed using the Real-time Cell Electric Impedance Sensing system (RTCA-DP). RESULTS: PCR and sequencing revealed the expression of FcεRI and FcεRII receptors in the human vascular endothelium. IL-4 caused respective 2- and 3-fold increases in FcεRI and FcεRII mRNA expression. Exposure of endothelium pre-treated with anti-BSA-DNP IgE to specific BSA-DNP antigen led to a 20% increase of endothelial integrity (p<0.05) after 24 hours, but only in cells pre-incubated with IL-4. CONCLUSIONS: The presence of FcεRI and FcεRII may allow the human vascular endothelium to respond to a specific antigen by increasing its integrity via an IgE-mediated mechanism.


Subject(s)
Antibodies, Anti-Idiotypic/immunology , Immunoglobulin E/immunology , Receptors, IgE/immunology , Cells, Cultured , Humans , Interleukin-4/immunology
11.
Immunol Lett ; 179: 43-46, 2016 11.
Article in English | MEDLINE | ID: mdl-27646628

ABSTRACT

The low-grade inflammation present in obese visceral adipose tissue impairs glucose metabolism, and contributes to the development of insulin resistance and weight gain. Immune processes occurring in response to the deposition of cholesterol within the vascular walls support atherosclerotic plaque growth and contribute to the cardiovascular complications. In both the obese adipose tissue and the atherosclerotic plaque, the Th1-type immune environment dominates over the Th2/Treg-type due to the overproduction of pro-inflammatory cytokines (IFN-γ, IL-6, TNF-α) and the deficiency of Th2-type processes and interleukins (IL-4, IL-5, IL-10, IL-13). So far, Th2 cells and eosinophils have been considered as the main providers of Th2-type mediators and the basis of Th2-type immunity in tissues. However, recently discovered innate lymphoid type 2 cells (ILC2s), which infiltrate lean visceral adipose tissue and the vascular wall, are believed to orchestrate local Th2-like immune responses. Upon activation by tissue-derived IL-33 and IL-25, ILCs2 secrete mostly IL-4, IL-5, IL-9 and IL-13: cytokines responsible for the accumulation of eosinophils and polarization of alternatively-activated macrophages, which altogether create the beneficial anti-inflammatory and metabo-regulatory environment in the adipose tissue and the vascular wall. Consequently, ILC2s-orchestrated immune environment seems to prevent obesity and atherosclerosis. Thus, ILCs2 appear to be the emerging immune regulators of immune and metabolic homeostasis in both adipose tissue and the vascular wall.


Subject(s)
Atherosclerosis/etiology , Atherosclerosis/metabolism , Immunity, Innate , Immunomodulation , Lymphocyte Subsets/immunology , Lymphocyte Subsets/metabolism , Obesity/etiology , Obesity/metabolism , Adipose Tissue/immunology , Adipose Tissue/metabolism , Adipose Tissue/pathology , Animals , Cytokines/metabolism , Energy Metabolism , Humans , Lymphocyte Subsets/cytology , Metabolic Diseases/etiology , Metabolic Diseases/metabolism
12.
Vascul Pharmacol ; 73: 57-63, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26231284

ABSTRACT

The vascular endothelium forms a barrier that controls flow of solutes and proteins and the entry of leukocytes into tissue. Injured tissue releases IL-33, which then alarms the immune system and attracts Th2 cells, thus increasing local concentration of IL-4. The aim of the study was to assess the influence of IL-33 and IL-4 on barrier functions of the human endothelium, expression of tight and adherent junction proteins, apoptosis and adhesive molecule surface expression in human endothelium in order to describe the mechanism of this effect. IL-33 and IL-4 decreased endothelial integrity and increased permeability. When added together, both cytokines lowered the endothelial integrity twice as much as used alone. This effect was accompanied by the down-regulation of occludin and VE-cadherin mRNA expression. Additionally, IL-4, but not IL-33, induced cell apoptosis. Both IL-33 and IL-4 showed the additive potency to down-regulate VE-cadherin mRNA expression. IL-33, unlike IL-4, increased the surface expression of ICAM-1, but not PECAM-1 in endothelial cells. Our results indicate that IL-33 may reversibly destabilize the endothelial barrier, thus accelerating the supply with immunomodulators and assisting leukocytes to reach wounded tissue. However, extended and less-controlled down-regulation of endothelial barrier, which may be a consequence of IL-33-initiated, but in fact IL-4-induced apoptosis of endothelial cells, may be deleterious and may eventually lead to the aggravation of inflammatory processes and the prolongation of tissue dysfunction.


Subject(s)
Capillary Permeability/drug effects , Human Umbilical Vein Endothelial Cells/drug effects , Interleukin-33/pharmacology , Interleukin-4/pharmacology , Antigens, CD/genetics , Antigens, CD/metabolism , Apoptosis/drug effects , Cadherins/genetics , Cadherins/metabolism , Cells, Cultured , Gene Expression Regulation , Human Umbilical Vein Endothelial Cells/metabolism , Human Umbilical Vein Endothelial Cells/pathology , Humans , Intercellular Adhesion Molecule-1/metabolism , Occludin/genetics , Occludin/metabolism , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , RNA, Messenger/metabolism , Recombinant Proteins/pharmacology , Time Factors
13.
Pharmacol Rep ; 67(2): 376-81, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25712667

ABSTRACT

BACKGROUND: Interleukin-35 (IL-35) is a novel immunomodulatory cytokine produced by CD4+ 25+ foxp3+ regulatory T-cells (T regs). Vascular smooth muscle cells (VSMCs) are involved in local immune homeostasis and certain chronic inflammatory pathologies. The effect of IL-35 on electrical impedance reflecting tissue integrity, the surface expression of ICAM-1 and mRNA expression of IL-32, as well as apoptosis in human primary aortic smooth muscle cells (Ao-SMCs) was investigated. METHODS: The influence of IL-35 on Ao-SMC integrity was assessed with the real-time cell electric impedance sensing system (RTCA-DP) based on normalized Cell Index (nCI). Additionally, Ao-SMCs were stimulated with IL-35 in order to assess ICAM-1 surface expression and apoptosis in flow cytometer. IL-32 mRNA expression was measured using real-time PCR. RESULTS: We found that the nCI of Ao-SMCs induced with IL-35 was lower after 12, 24 and 48h of incubation than the nCI of unstimulated cells. IL-35 slightly enhanced ICAM-1 surface expression and increased IL-32 mRNA expression in Ao-SMCs after 24h of induction. However, IL-35 did not affect Ao-SMC apoptosis, necrosis or viability. CONCLUSION: Our data suggest that IL-35 may be an agent affecting the inflammatory properties of AoSMCs and thus it may regulate immune homeostasis of the vascular wall. Hence, IL-35 may be a novel player affecting Ao-SMC-controlled arterial wall immune homeostasis.


Subject(s)
Aorta/cytology , Apoptosis/drug effects , Cell Survival/drug effects , Intercellular Adhesion Molecule-1/biosynthesis , Interleukins/pharmacology , Myocytes, Smooth Muscle/drug effects , Aorta/drug effects , Electric Impedance , Humans , Interleukins/biosynthesis , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/metabolism , Primary Cell Culture
14.
Arch Med Sci ; 10(4): 669-75, 2014 Aug 29.
Article in English | MEDLINE | ID: mdl-25276149

ABSTRACT

INTRODUCTION: The aim of this study was to evaluate the effect of melatonin on blood pressure in patients with essential hypertension receiving medical treatment and with type 2 diabetes in good metabolic control. MATERIAL AND METHODS: The study lasted 8 weeks. Patients were equipped with a 24-hour ambulatory blood pressure monitor and took melatonin (3 mg a day in the evening) for 4 weeks. The patients were divided into four groups: group 1 (n = 32) including dippers, group 2 (n = 34) non-dippers treated with melatonin; and two control groups: group 3 (n = 28) including dippers and group 4 (n = 30) non-dippers treated without melatonin. After 4 weeks patients took melatonin for the next 4 weeks (5 mg a day). In each visit were analyzed: systolic, diastolic and mean blood pressure in both day and night time. RESULTS: We observed that 29.5% non-dippers (n = 10) treated with melatonin in a dose of 3 mg/day achieved features of dippers compared to control group (p < 0.05). Five mg of melatonin per day restored normal diurnal blood pressure rhythm in 32.4% non-dippers (n = 11, p < 0.05). In non-dippers treated with melatonin significant decreases of diastolic, systolic and mean night blood pressure values (p < 0.05) were observed. CONCLUSIONS: More than 30% of non-dippers with type 2 diabetes treated with melatonin were restored to the normal circadian rhythm of blood pressure. The effect of melatonin in both doses (3 mg and 5 mg) was significant for non-dippers only and included nocturnal systolic, diastolic and mean arterial pressure.

15.
Food Chem Toxicol ; 69: 289-93, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24727230

ABSTRACT

The intestinal epithelium is exposed to oxygenated cholesterol products present in foodstuffs. In vitro studies demonstrate the effect of oxysterols on cytokine release by intestinal cells cultured alone. However, physiologically, the response of the intestinal epithelium to external agents occurs in the presence of dendritic cells (DCs). The aim of the study was to analyze the effect of 7-ketocholesterol on the barrier functions and IL-10 mRNA expression of Caco-2 cells in the presence of DCs, and secondly, on IL-10 mRNA expression in DCs. Caco-2 cells were co-cultured with monocyte-derived dendritic cells and induced with 7-ketocholesterol in a transwell system. DCs did not affect the transepithelial electrical resistance (TER) of the Caco-2 cell monolayer, but increased IL-10 mRNA expression in Caco-2 cells. 7-ketocholesterol decreased the TER of Caco-2 cells co-cultured with DCs and diminished IL-10 mRNA expression in Caco-2 cells induced by the presence of DCs. IL-10 mRNA expression fell in DCs co-cultured with Caco-2 cells after treatment with 7-ketocholesterol. Oxidized cholesterols present in gut mucosa may contribute to the decrease of epithelial barrier functions and the inappropriate development of an inflammatory response to food compounds.


Subject(s)
Dendritic Cells/drug effects , Interleukin-10/genetics , Intestinal Mucosa/drug effects , Ketocholesterols/pharmacology , Caco-2 Cells/drug effects , Coculture Techniques/instrumentation , Coculture Techniques/methods , Dendritic Cells/cytology , Epithelial Cells/cytology , Epithelial Cells/drug effects , Humans , Intestinal Mucosa/cytology , Occludin/genetics , RNA, Messenger , Zonula Occludens-1 Protein/genetics
16.
Inflamm Res ; 62(12): 1015-23, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24077843

ABSTRACT

OBJECTIVE AND DESIGN: The damage of barrtier tissues, such as the vascular endothelium and intestinal epithelium, may lead to disturbances of local immune homeostasis. The aim of the study was to assess and compare the effect of oxidized cholesterols (7-ketocholesterol and 25-hydroxycholesterol) on the barrier properties of human primary aortic endothelium (HAEC) and intestinal epithelium Caco-2 cells using a realtime cell electric impedance sensing system (RTCA-DP). MATERIALS AND METHODS: HAEC and Caco-2 cells were stimulated with 7-ketocholesterol and 25-hydroxycholesterol by the RTCA-DP system. Apoptosis was assessed by flow cytometry and cell monolayer morphology was assessed under a light microscope. RESULTS: 7-ketocholesterol decreased impedance (nCI) in both the endothelium and epithelium. However, the decrease was more profound in the endothelium. Similarly, although 25-hydroxycholesterol decreased nCI in both the endothelium and epithelium, the effect was weaker than that of 7-ketocholesterol, which caused extensive damage to the endothelial monolayer, while 25-hydroxycholesterol caused partial damage and did not affect the epithelial monolayer. 7-ketocholesterol, but not 25-hydroxycholesterol, increased endothelial cell apoptosis and decreased the viability of endothelial cells. However, 7-ketocholesterol and 25-hydroxycholesterol decreased epithelial cell apoptosis and increased viability. CONCLUSION: Oxidized cholesterols destroy the HAEC, but not the Caco-2 epithelial barrier, via cell apoptosis dependent on the site of oxidation. Damage to the endothelium by oxidized cholesterol may disrupt local homeostasis and provide open access to inner parts of the vascular wall for lipids, other peripheral blood-derived agents, and immune cells, leading to inflammation and atherogenesis.


Subject(s)
Endothelial Cells/drug effects , Epithelial Cells/drug effects , Hydroxycholesterols/pharmacology , Ketocholesterols/pharmacology , Aorta/cytology , Apoptosis/drug effects , Caco-2 Cells , Cell Survival/drug effects , Cells, Cultured , Humans , Intestinal Mucosa/cytology
17.
Arch Med Sci ; 9(1): 159-65, 2013 Feb 21.
Article in English | MEDLINE | ID: mdl-23515919

ABSTRACT

For many years atherosclerosis was believed to be the passive accumulation of cholesterol in vessel walls. Today the picture is more complex, as immune processes occur in atherogenesis. Considerable attention is focused on the particular role of adaptive immune responses orchestrated by T cell subsets. Since the role of Th1/Th2 balance and Th1 cell domination in atherogenesis is already known, the involvement of regulatory T lymphocytes and recently described Th17 cells raises new concerns. On one hand, each of these cells may specifically drive responses of vascular wall tissues and immune cells; however, they are subject to the control of a plethora of tissue- and pathogen-derived agents. Due to ineffective tissue regeneration, remodeling of the vascular wall occurs. The understanding of the immune regulatory network gives perspectives of innovative immunomodulatory therapies of atherosclerosis and the prevention of its complications, such as coronary artery disease.

18.
Med Sci Monit ; 18(8): BR331-336, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22847194

ABSTRACT

BACKGROUND: The prevalence of hypertension is growing at an alarming rate. Increasing attention is being focussed on the oxidative stress accompanying this disease. In this study we examined the impact of this disease on some parameters of erythrocytes and human blood plasma. MATERIAL/METHODS: We examined the impact of hypertension on some parameters of erythrocytes and human plasma. The study involved 13 patients with hypertension and 19 healthy subjects. We determined lipid peroxidation, SH groups concentration, antioxidants enzymes activity, ATPase activity, total antioxidant capacity, total cholesterol level and erythrocyte membrane fluidity. RESULTS: We found an increased level of lipid peroxidation and the concentration of SH groups in membrane proteins in patients with hypertension, and a decrease in the activity of catalase and superoxide dysmutase. No changes were observed in glutathione peroxidase and ATPase activity, level of total antioxidant capacity, total cholesterol level and fluidity of erythrocyte membranes. CONCLUSIONS: These results suggest the existence of an impaired oxidative balance in hypertensive human erythrocytes.


Subject(s)
Erythrocytes/pathology , Erythrocytes/physiology , Hypertension/pathology , Hypertension/physiopathology , Adenosine Triphosphatases/blood , Antioxidants/metabolism , Catalase/blood , Cholesterol/metabolism , Erythrocyte Membrane/metabolism , Erythrocytes/enzymology , Female , Glutathione Peroxidase/blood , Humans , Hypertension/blood , Hypertension/enzymology , Lipid Peroxidation , Male , Membrane Fluidity , Middle Aged , Superoxide Dismutase/blood
19.
Eur J Nutr ; 51(5): 549-56, 2012 Aug.
Article in English | MEDLINE | ID: mdl-21850495

ABSTRACT

PURPOSE: A diet rich in berries is believed to play a distinct role in the prevention of metabolic diseases associated with obesity. So far, there have been no published clinical observations evaluating the influence of Aronia melanocarpa on hemostasis. The aim of our study was to investigate the effects of A. melanocarpa extract (AM) supplementation on platelet aggregation, clot formation, and lysis in patients with metabolic syndrome (MS). METHODS: Middle-aged non-medicated subjects with MS (n = 38) and 14 healthy volunteers were included in this study. Patients with MS were treated with 100 mg of AM three times daily for 2 months. RESULTS: We observed a significant reduction in the concentration of TC, LDL-C, and TG after AM supplementation. Beneficial changes in coagulation parameters were also observed. After 1 month of AM administration, we noticed significant inhibition of platelet aggregation. However, this effect became less pronounced after 2 months of supplementation. In the case of coagulation induced by endogenic thrombin, a significant decrease in the overall potential for coagulation was induced after 1 or 2 months of supplementation. Moreover, after 1 month of AM extract supplementation, we observed a beneficial reduction in the overall potential for clot formation and fibrinolysis. CONCLUSIONS: We observed the normalization of hemostasis parameters in MS patients after both 1 and 2 months of AM administration. After 1 month of AM supplementation, we found favorable changes in regards to the overall potential for plasma clotting, clot formation, and lysis, as well as in the lipid profiles of subjects.


Subject(s)
Blood Coagulation/drug effects , Dietary Supplements , Fibrinolysis/drug effects , Metabolic Syndrome/drug therapy , Photinia/chemistry , Plant Extracts/administration & dosage , Platelet Aggregation/drug effects , Adult , Aged , Blood Platelets/drug effects , Blood Platelets/metabolism , Cholesterol, LDL/blood , Diet , Female , Humans , Male , Metabolic Syndrome/physiopathology , Middle Aged
SELECTION OF CITATIONS
SEARCH DETAIL
...