Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 50
Filter
1.
Int J Immunogenet ; 43(4): 218-25, 2016 Aug.
Article in English | MEDLINE | ID: mdl-27317472

ABSTRACT

Coronary artery disease (CAD) remains a major cause of death in developed countries. Both environmental and, less known, genetic factors contribute to progression of CAD to myocardial infarction (MI). Immune system is activated in patients with CAD through dendritic cells (DCs), which present plaque antigens to T lymphocytes. Production of proinflammatory cytokines by activated T cells contributes to plaque rupture in MI. Chemokine receptor 7 (CCR7) on DCs is required for their chemotaxis from plaque to lymph nodes. This makes possible an interaction of DCs with T lymphocytes and initiation of specific immune response. We hypothesized that single nucleotide polymorphisms (SNPs) in CCR7 gene locus are associated with previous MI in patients with CAD. To test this hypothesis, we genotyped six SNPs from the CCR7 gene locus in 300 consecutive patients, admitted for elective coronary angiography. We performed univariate-, multivariate- (including potential confounders) and haplotype-based tests of association of SNPs with previous MI and results of angiography. Allele A of rs17708087 SNP was associated with previous MI. This association remained significant after adjustment for age, sex, smoking, hypercholesterolaemia and drugs used by patients (odds ratio 2.13, 95% confidence interval: 1.13-3.86). Therefore, we conclude that CCR7 gene locus harbours a polymorphism that modifies risk of MI in patients with CAD. Replication of this association could be sought in a prospective cohort of initially healthy individuals.


Subject(s)
Coronary Artery Disease/genetics , Genetic Association Studies , Genetic Predisposition to Disease , Myocardial Infarction/genetics , Receptors, CCR7/genetics , Aged , Alleles , Coronary Angiography , Coronary Artery Disease/diagnostic imaging , Coronary Artery Disease/pathology , Female , Genotype , Haplotypes , Humans , Male , Middle Aged , Myocardial Infarction/diagnostic imaging , Myocardial Infarction/pathology , Polymorphism, Single Nucleotide , Risk Factors
2.
Clin Exp Immunol ; 181(1): 126-32, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25707554

ABSTRACT

Immune cells may take part in the renin-angiotensin-aldosterone system (RAAS), which plays a pivotal role in the regulation of vascular tone and blood pressure. The aim of the study was to analyse the expression and activity of angiotensin-converting enzyme type 1 (ACE1) and ACE2 in human monocytes (MO) and their subsets. The highest relative level of ACE1-, as well as ACE2-mRNA expression, was observed in CD14(++)CD16(-) (classical) MO. Moreover, in these cells, mean level of ACE2-mRNA was almost two times higher than that of ACE1-mRNA (11.48 versus 7.073 relative units, respectively). In peripheral blood mononuclear cells (PBMC), MO and classical MO, ACE1 and ACE2 protein expression was stronger compared to other MO subpopulations. The highest level of Ang II generated from Ang I in vitro was observed in classical MO. In this setting, generation of Ang-(1-9) by PBMC and classical MO was higher when compared to the whole MO population (P < 0.05). The generation rate of vasoprotective Ang-(1-7) was comparable in all analysed cell populations. However, in CD14(+)CD16(++) (non-classical) MO, formation of Ang-(1-7) was significantly greater than Ang II (P < 0.001). We suggest that in physiological conditions MO (but also lymphocytes forming the rest of PBMC pool) may be involved in the regulation of vessel wall homeostasis via the RAAS-related mechanisms. Moreover, non-classical MO, which are associated preferentially with the vascular endothelium, express the vasoprotective phenotype.


Subject(s)
Monocytes/enzymology , Peptidyl-Dipeptidase A/metabolism , Angiotensin-Converting Enzyme 2 , GPI-Linked Proteins/immunology , Healthy Volunteers , Humans , Lipopolysaccharide Receptors/immunology , Monocytes/immunology , Peptidyl-Dipeptidase A/genetics , RNA, Messenger/biosynthesis , Receptors, IgG/immunology , Renin-Angiotensin System/immunology
3.
Horm Metab Res ; 47(8): 600-4, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25565097

ABSTRACT

Multifunctional peptide oxytocin currently undergoes intensive research due to its proposed anti-obesity properties. Until now, little is known about regulation of oxytocin receptor in metabolically active tissues in obesity. The aim of the present study was to measure expression of oxytocin receptor upon obese phenotype with respect to the variety among adipose tissue and skeletal muscles with distinct anatomical localisation. Total homogenates were prepared from epididymal, retroperitoneal and inguinal adipose tissues as well as quadriceps and soleus muscle from lean and obese Zucker rats. Oxytocin receptor protein was determined by immunoblot. Interestingly, elevated oxytocin receptor was observed in epididymal adipose tissue of obese rats in contrast to its downregulation in subcutaneous and no change in retroperitoneal fat. In lean animals, oxytocin receptor protein was expressed at similar levels in all adipose depots. This uniformity was not observed in the case of skeletal muscle in which fibre type composition seems to be determinant of oxytocin receptor expression. Quadriceps muscle with the predominance of glycolytic fibres exhibits higher oxytocin receptor expression than almost exclusively oxidative soleus muscle. Oxytocin receptor protein levels were decreased in both skeletal muscles analysed upon obese phenotype. The present work demonstrates that even under identical endocrine circumstances, oxytocin receptor is differentially regulated in adipose tissue of obese rats depending on fat depot localisation. These results also imply which tissues may be preferentially targeted by oxytocin treatment in metabolic disease.


Subject(s)
Adipose Tissue/metabolism , Muscle, Skeletal/metabolism , Obesity/metabolism , Receptors, Oxytocin/metabolism , Animals , Disease Models, Animal , Male , Quadriceps Muscle/metabolism , Rats , Rats, Zucker
4.
J Physiol Pharmacol ; 70(4)2019 Aug.
Article in English | MEDLINE | ID: mdl-31642813

ABSTRACT

The tissue renin-angiotensin system (RAS) plays an important role in the development and progression of many diseases. It has been confirmed that angiotensin II (ANG II) participates in the proliferation and angiogenesis of breast cancer. Moreover, some RAS dysregulations in cancer have been observed. Recent studies on the role of two opposite axes of angiotensinogen metabolism - ACE (angiotensin-converting enzyme)/ANGII/AT1R (angiotensin receptor type 1) and ACE-2/ANG 1-7/MAS (mitochondrial assembly) - indicate their importance in tumor growth and invasion, but studies describing the metabolic pathways in breast cancer and the role of newer angiotensins, such as ANG 1-12, remain lacking. In this study, the metabolism of angiotensinogen fragments in three breast cancer lines, namely, MDA-MB-231, MCF-7, and T-47D, compared with normal breast tissue cells (PCS-600) was estimated. Incubation of the cancer cells with angiotensinogen resulted in the prevalent formation of ANG 1-7. A difference in the ability to form ANG II was observed between cell lines. In normal breast cells, the strong predominance of the ACE-2/ANG 1-7/MAS pathway was detected. In cancer cells, differences in angiotensinogen metabolism depending on cancer line were observed; the prevalence of the ACE/ANG II/AT1R pathway was shown. Expressions of the RAS component were dysregulated in cancer cells and differed between cell lines. In conclusion, the ability of breast cancer cells to produce numerous angiotensin peptide metabolites was demonstrated. The metabolism of angiotensinogen differed between various types of breast cancer cells. The obtained results indicate the greater importance of the classical pathway - ACE/ANG II/AT1R - in breast cancer cells. The production of ANG 1-12 seems to be marginal in breast tissue, but a tendency for the higher formation of this peptide in cancer cells was observed. The production of ANG 1-7 was significantly lower in cancer cells, whereas the expression of MAS receptor was higher than that in the control. This finding suggests that substances with MAS receptor agonist activity could be useful in the treatment of breast cancer, but this requires further investigations.


Subject(s)
Angiotensin I/metabolism , Angiotensinogen/metabolism , Breast Neoplasms/metabolism , Peptide Fragments/metabolism , Breast/cytology , Breast/metabolism , Cell Line , Female , Humans , Proto-Oncogene Mas , Proto-Oncogene Proteins/metabolism , Receptors, G-Protein-Coupled/metabolism
5.
Amino Acids ; 35(2): 359-64, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18157587

ABSTRACT

Taurine chloramine (TauCl) and taurine bromamine (TauBr), products of myeloperoxidase halide system, exert anti-inflammatory properties. TauCl was demonstrated to inhibit the production of a variety of pro-inflammatory mediators including cyclooxygenase-2 (COX-2) dependent production of prostaglandin E(2) (PGE(2)). Recently we have demonstrated that both major leukocyte haloamines, TauCl and TauBr, induced expression of HO-1 in non-activated and LPS-activated J774.2 macrophages. In this study, we have shown that TauCl and TauBr, at non-cytotoxic concentrations, inhibited the production of (PGE(2)) without altering the expression of COX-2 protein, in LPS/IFN-gamma stimulated J774.2 cells. The inhibitory effect of TauCl and TauBr was reversed by chromium III mesoporhyrin (CrMP), an inhibitor of HO-1 activity. Our data suggest that HO-1 might participate in anti-inflammatory effects of TauCl/TauBr possibly by inhibition of COX-2 activity and decrease of PGE(2) production.


Subject(s)
Dinoprostone/biosynthesis , Heme Oxygenase-1/physiology , Macrophages/drug effects , Taurine/analogs & derivatives , Animals , Blotting, Western , Cell Survival/drug effects , Cells, Cultured , Cyclooxygenase 2/analysis , Cyclooxygenase 2/drug effects , Cyclooxygenase 2/metabolism , Dose-Response Relationship, Drug , Down-Regulation/drug effects , Down-Regulation/physiology , Heme Oxygenase-1/analysis , Heme Oxygenase-1/drug effects , Macrophages/metabolism , Mice , Mitogen-Activated Protein Kinase Kinases/drug effects , Mitogen-Activated Protein Kinase Kinases/metabolism , Nitric Oxide Synthase Type II/analysis , Nitric Oxide Synthase Type II/drug effects , Nitric Oxide Synthase Type II/metabolism , Taurine/pharmacology
6.
J Physiol Pharmacol ; 69(1): 15-21, 2018 Feb.
Article in English | MEDLINE | ID: mdl-29769417

ABSTRACT

Adenosine triphosphate (ATP) is an essential substrate metabolite in human beings. Mitochondrial oxidative phosphorylation provides > 95% of ATP with the remainder derived from glycolysis or tricarboxylic acid cycle (TCA). In normal hearts, acetyl-CoA is synthesized from the ß-oxidation of free fatty acids (FFA) and the oxidation of pyruvate. Pyruvate is synthesized from glycolysis and can be submitted either for decarboxylation to acetyl-CoA or for dehydrogenation to lactate. Moreover, pyruvate, as well as lactate, plays a key role in aerobic glucose metabolism which is highly dependent on ubiquitous regulatory mechanisms. Many recent advances in molecular biology, genetics, and physiology have revealed new insights into the metabolic flux of lactate. The initial perception characterized by increased lactate production and accumulation in peripheral tissues in anaerobic conditions has been recently contested. The paradigm of increased lactate concentration in the anaerobic setting is discussed according to contemporary reports. Nevertheless, the clinical role of lactate as a prognostic factor in cardiovascular diseases is undisturbed, especially in the field of innovative technology of left/bi ventricular-assist devices and biochips where it reassured its diagnostic and prognostic impact on the cardiovascular system.


Subject(s)
Lactic Acid/metabolism , Shock, Cardiogenic/metabolism , Animals , Humans
7.
Cell Mol Biol (Noisy-le-grand) ; 53(4): 51-60, 2007 May 15.
Article in English | MEDLINE | ID: mdl-17531161

ABSTRACT

The goal of this study was to characterize the impact of induction or inhibition of the heme-HO system on renal apoptosis in clipped and non-clipped kidneys from 2K1C hypertensive rats. Male Sprague-Dawley rats had a 0.25 mm silver clip placed around the left renal artery. Four groups of rats were studied: sham operated animals, 2K1C control rats, 2K1C rats received weekly injections of CoPP (5 mg/100 g body wt, administered subcutaneously), and 2K1C rats pretreated with SnMP (5 mg/ 100g body wt, administered intraperitoneally three times a week). The animals were sacrificed three weeks after surgery. We measured systolic blood pressure, plasma renin activity, non-clipped and clipped kidney HO-1 and HO-2 protein expression, HO activity, heme content, nitrotyrosine levels, and activation of selected pro- and anti-apoptotic proteins. Systolic blood pressure and plasma renin activity were significantly higher in 2K1C rats compared to sham rats. Compared to kidneys from sham animals, clipped kidneys from 2K1C rats showed a significant increase in HO-1 expression with increases in HO activity (26%), heme content (47%) and nitrotyrosine levels (49%), accompanied by an increase in caspase-3 and caspase-9 activity. In contrast, non-clipped kidneys from 2K1C rats showed no differences in HO-1 expression, HO activity, heme content, nitrotyrosine levels and caspase activity compared to sham rats. In clipped kidneys from 2K1C rats, inhibition of HO activity by SnMP augmented caspase-3 and caspase-9 activity and decreased expression of the anti-apoptotic Bcl-2 protein, while induction of HO-1 with CoPP strongly inhibited the activity of both caspases and increased the induction of Bcl-2 and Bcl-xl proteins. These findings demonstrate that the clipped kidneys responded to decreased renal perfusion pressure and increased oxidative stress by activation of the heme-HO system, which exerts antiapoptotic action via mechanisms involving decreased caspase-3 and caspase-9 activity, and increased expression of antiapoptotic molecules.


Subject(s)
Apoptosis/physiology , Heme Oxygenase-1/genetics , Hypertension, Renovascular/genetics , Animals , Caspases/metabolism , Gene Expression Regulation , Heme/analysis , Heme Oxygenase (Decyclizing)/metabolism , Heme Oxygenase-1/metabolism , Hypertension, Renovascular/enzymology , Hypertension, Renovascular/etiology , Hypertension, Renovascular/pathology , Kidney/chemistry , Kidney/enzymology , Kidney/pathology , Kidney/surgery , Male , Proto-Oncogene Proteins c-bcl-2/metabolism , Rats , Rats, Sprague-Dawley , Tyrosine/analogs & derivatives , Tyrosine/analysis , bcl-X Protein/metabolism
8.
J Physiol Pharmacol ; 58(3): 583-8, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17928652

ABSTRACT

Recently, we have shown that MK-886 - an inhibitor of five lipoxygenase activating protein (FLAP) inhibits atherosclerosis in apolipoprotein E / LDL receptor - double knockout mice. We, therefore, wanted to find out if other FLAP inhibitor - BAYx1005 given at a dose of 1.88 mg per 100 mg of body weight per day during 16 weeks, could also attenuate atherogenesis. In apoE/LDLR - DKO mouse model BAYx1005 inhibited atherogenesis, measured both by "en face" method (23.84 +/- 2.7% vs. 15.16 +/- 1.4%) and "cross-section" method (497236 +/- 31516 microm(2) vs. 278107 +/- 21824 microm(2)). This is the first report that shows the effect of BAYx1005 on atherogenesis in gene-targeted mice.


Subject(s)
Apolipoproteins E/genetics , Atherosclerosis/drug therapy , Quinolines/pharmacology , Receptors, LDL/genetics , Animals , Aorta/drug effects , Aorta/metabolism , Aorta/pathology , Apolipoproteins E/metabolism , Atherosclerosis/genetics , Atherosclerosis/metabolism , Azo Compounds/chemistry , Female , Lipoxygenase Inhibitors/chemistry , Lipoxygenase Inhibitors/pharmacology , Lipoxygenase Inhibitors/therapeutic use , Mice , Mice, Inbred C57BL , Mice, Knockout , Molecular Structure , Quinolines/chemistry , Quinolines/therapeutic use , Receptors, LDL/metabolism , Staining and Labeling/methods
9.
J Physiol Pharmacol ; 58(3): 529-40, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17928648

ABSTRACT

The metabolism of renin-angiotensin system (RAS) is more complicated than previously expected and understanding the biological phenomena regulated by variety of angiotensin metabolites requires their precise and possibly comprehensive quantitation. Physiological concentrations of angiotensins (Ang) in biological fluids are low, therefore their accurate measurements require very sensitive and specific analytical methods. In this study we developed an accurate and reproducible method of quantitation of angiotensin metabolites through coupling of liquid chromatography and electrospray ionization - mass spectrometry (LC-ESI-MS). With this method main angiotensin metabolites (Ang I, II, III, IV, 1-9, 1-7, 1-5) can be reliably measured in organ bath of rat tissues (aorta, renal artery, periaortal adipose tissue) and in medium of cultured endothelial cells (EA.hy926), exposed to Ang I for 15 minutes, in the absence or in the presence of angiotensin converting enzyme inhibitor, perindoprilat. Presented LC-ESI-MS method proved to be a quick and reliable solution to comprehensive analysis of angiotensin metabolism in biological samples.


Subject(s)
Angiotensins/analysis , Angiotensins/metabolism , Chromatography, Liquid/methods , Spectrometry, Mass, Electrospray Ionization/methods , Amino Acid Sequence , Angiotensin-Converting Enzyme Inhibitors/pharmacology , Angiotensins/standards , Animals , Cell Line , Cell Line, Tumor , Culture Media, Conditioned/analysis , Culture Media, Conditioned/metabolism , Endothelial Cells/cytology , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Humans , Hybridomas , Indoles/pharmacology , Male , Models, Biological , Organ Culture Techniques/methods , Rats , Rats, Inbred WKY , Reference Standards , Reproducibility of Results , Time Factors
10.
J Physiol Pharmacol ; 68(4): 609-617, 2017 Aug.
Article in English | MEDLINE | ID: mdl-29151078

ABSTRACT

Ghrelin, an acylated 28-amino acid polypeptide, was primary isolated from the stomach, and the stomach is a main source of circulating ghrelin. Ghrelin strongly and dose-dependently stimulates release of growth hormone from the anterior pituitary, as well as increases food intake and fat deposition. Previous studies showed that ghrelin exhibits protective and therapeutic effect in different parts of the gastrointestinal system, including the oral cavity. The aim of present study was to examine the role of growth hormone and insulin-like growth factor-1 (IGF-1) in the healing of gingival ulcers. Studies were performed on rats with the intact pituitary gland and hypophysectomized rats. In anesthetized rats, chronic ulcers of the gum were induced by acetic acid. Rats were treated intraperitoneally twice a day with saline or ghrelin (4, 8 or 16 nmol/kg/dose) for six days. In pituitary-intact rats, administration of ghrelin significantly increased serum concentration of growth hormone and IGF-1 and this effect was associated with a significant increase in the healing rate of gingival ulcers. Moreover, treatment with ghrelin increased mucosal blood flow and DNA synthesis in the gum, while a local inflammation was decreased what was observed as a reduction in mucosal concentration of pro-inflammatory interleukin-1ß. Hypophysectomy decreased serum level of growth hormone below a detection limit; whereas serum concentration of IGF-1 was reduced by 90%. On the other hand, removal of the pituitary gland was without any significant effect on the healing rate of gingival ulcers or on the ulcer-induced increase in DNA synthesis and concentration of pro-inflammatory interleukin-1ß in gingival mucosa. Administration of ghrelin failed to affect serum level of growth hormone and IGF-1 in hypophysectomized rats, and was without any effect on the healing rate of gingival ulcers, mucosal blood flow, DNA synthesis or concentration of interleukin-1ß in gingival mucosa. Neither induction of gingival ulcers nor hypophysectomy nor administration of ghrelin significantly affected serum concentration of pro-inflammatory interleukin-1ß. We concluded that endogenous growth hormone and IGF-1 were involved in the therapeutic effect of exogenous ghrelin in the healing of gingival mucosa damage.


Subject(s)
Ghrelin/pharmacology , Gingiva/drug effects , Gingival Diseases/drug therapy , Growth Hormone/metabolism , Insulin-Like Growth Factor I/metabolism , Oral Ulcer/drug therapy , Wound Healing/drug effects , Animals , Gingiva/metabolism , Gingival Diseases/metabolism , Inflammation/drug therapy , Inflammation/metabolism , Interleukin-1beta/metabolism , Male , Mouth Mucosa/drug effects , Mouth Mucosa/metabolism , Oral Ulcer/metabolism , Rats , Rats, Wistar
11.
Br J Pharmacol ; 174(22): 4055-4069, 2017 Nov.
Article in English | MEDLINE | ID: mdl-27935022

ABSTRACT

BACKGROUND AND PURPOSE: Inflammation plays a key role in atherosclerosis. The protective role of angiotensin 1-7 (Ang-(1-7)) in vascular pathologies suggested the therapeutic use of low MW, non-peptide Ang-(1-7) mimetics, such as AVE0991. The mechanisms underlying the vaso-protective effects of AVE0991, a Mas receptor agonist, remain to be explored. EXPERIMENTAL APPROACH: We investigated the effects of AVE0991 on the spontaneous atherosclerosis in apolipoprotein E (ApoE)-/- mice, in the context of vascular inflammation and plaque stability. KEY RESULTS: AVE0991 has significant anti-atherosclerotic properties in ApoE-/- mice and increases plaque stability, by reducing plaque macrophage content, without effects on collagen. Using the descending aorta of chow-fed ApoE-/- mice, before significant atherosclerotic plaque develops, we gained insight to early events in atherosclerosis. Interestingly, perivascular adipose tissue (PVAT) and adventitial infiltration with macrophages and T-cells precedes atherosclerotic plaque or the impairment of endothelium-dependent NO bioavailability (a measure of endothelial function). AVE0991 inhibited perivascular inflammation, by reducing chemokine expression in PVAT and through direct actions on monocytes/macrophages inhibiting their activation, characterized by production of IL-1ß, TNF-α, CCL2 and CXCL10, and differentiation to M1 phenotype. Pretreatment with AVE0991 inhibited migration of THP-1 monocytes towards supernatants of activated adipocytes (SW872). Mas receptors were expressed in PVAT and in THP-1 cells in vitro, and the anti-inflammatory effects of AVE0991 were partly Mas dependent. CONCLUSIONS AND IMPLICATIONS: The selective Mas receptor agonist AVE0991 exhibited anti-atherosclerotic and anti-inflammatory actions, affecting monocyte/macrophage differentiation and recruitment to the perivascular space during early stages of atherosclerosis in ApoE-/- mice. LINKED ARTICLES: This article is part of a themed section on Targeting Inflammation to Reduce Cardiovascular Disease Risk. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v174.22/issuetoc and http://onlinelibrary.wiley.com/doi/10.1111/bcp.v82.4/issuetoc.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Atherosclerosis/drug therapy , Imidazoles/therapeutic use , Angiotensin I , Animals , Aorta/drug effects , Aorta/immunology , Aorta/pathology , Atherosclerosis/immunology , Atherosclerosis/pathology , Cell Line , Cell Line, Tumor , Cytokines/genetics , Female , Humans , Leukocytes/drug effects , Leukocytes/immunology , Macrophages/drug effects , Macrophages/immunology , Mice, Inbred C57BL , Mice, Knockout, ApoE , Peptide Fragments , Plaque, Atherosclerotic , Proto-Oncogene Mas , Proto-Oncogene Proteins/agonists , Receptors, G-Protein-Coupled/agonists
12.
J Physiol Pharmacol ; 57(4): 649-60, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17229988

ABSTRACT

Production of arachidonic acid (AA) metabolites - prostacyclin (PGI(2)) in large vessels and prostaglandin E(2) (PGE(2)) in microcirculation is intrinsically involved in maintenance of vascular wall homeostasis. EA.hy 926 is a hybrid cell line, is derived by fusion of HUVEC with A549 cells. The aim of this study was to examine the production of prostacyclin and PGE2 in resting and IL-1beta-stimulated EA.ha 926 cells, in comparison with its progenitor cells. Non-stimulated EA.hy 926 cells has been found to produce much lower amounts of prostacyclin than resting HUVEC. Resting hybrid cells produced more PGE(2) than prostacyclin, despite they expressed high levels of COX-1 and PGI(2) synthase. On the contrary to HUVEC and A549, EA.hy 926 cells did not respond to IL-1beta with COX-2 induction and increase of prostaglandin production, however they did it in response to lysophosphatidylcholine (LPC). The characteristics of EA.hy 926 cells in terms of the pattern of prostanoid formation could facilitate studies on endothelial metabolism and role of these important lipid mediators.


Subject(s)
Dinoprostone/biosynthesis , Endothelial Cells/metabolism , Epoprostenol/biosynthesis , Hybrid Cells/metabolism , Interleukin-1beta/pharmacology , Blotting, Western , Cell Line, Tumor , Cyclooxygenase 1/biosynthesis , Cyclooxygenase 2/biosynthesis , Endothelial Cells/drug effects , Humans , Hybrid Cells/drug effects , Reverse Transcriptase Polymerase Chain Reaction
13.
J Physiol Pharmacol ; 57(1): 109-17, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16601319

ABSTRACT

Activation of both poly (ADP-ribose) polymerase (PARP) and inducible nitric oxide synthase (NOS-2) have been implicated in the pathogenesis of various forms of inflammation, therefore compounds which may simultaneously inhibit both pathways are of potential therapeutic interest. We tested the influence of potent inhibitor of PARP, 1, 5-isoquinolinediol (ISO), on NOS-2 induction in model of mouse macrophages (cell line J774.2) stimulated with lipopolysaccharide (1 microg/ml). Pretreatment with ISO (1-300 microM) resulted in dose-dependent inhibition of accumulation of NOS-2-derived nitrite in culture medium (IC(50) = 9,3 microM) as well as inhibition of NOS-2 protein induction in cultured J774.2 cells; ISO given 10 hours after LPS did not influence activity of NOS-2. Interestingly, another PARP inhibitor, 3-aminobenzamide (3-AB, 10-3000 microM), did not influence 24-hr nitrite accumulation in J774.2 cell culture, either administered 15 minutes prior to LPS or 10 hrs after LPS. Scavenging of reactive oxygen species by use of mixture of SOD and catalase (SOD/Cat, 100/300 - 1000/3000 U/ml) as well as cell permeable SOD-mimetic [Mn(III)TBAP, 1- 100 microM], did not influence NOS-2 induction in J774.2 cells. In summary, we identified 1, 5-isoquinoline as potent inhibitor of induction of NOS-2 in LPS-treated mouse macrophages. The exact mechanism of inhibitory action of this compound on NOS-2 induction requires further investigation.


Subject(s)
Enzyme Inhibitors/pharmacology , Nitric Oxide Synthase Type II/antagonists & inhibitors , Poly(ADP-ribose) Polymerase Inhibitors , Animals , Benzamides/pharmacology , Cell Line , Cell Respiration , Isoquinolines , Lipopolysaccharides , Macrophages/drug effects , Macrophages/metabolism , Mice , Nitric Oxide Synthase Type II/metabolism , Nitrites/metabolism , Quinolines/pharmacology
14.
Physiol Res ; 65(4): 561-570, 2016 11 08.
Article in English | MEDLINE | ID: mdl-26988149

ABSTRACT

We used mass spectrometry to quantitate production of angiotensinogen metabolites in renal artery of 3- and 7-month-old Wistar-Kyoto (WKY) and Spontaneously Hypertensive Rats (SHR). Tissue fragments were incubated for 15 min in oxygenated buffer, with added angiotensin I. Concentrations of angiotensins I (ANG I), II (ANG II), III (ANG III), IV (ANG IV), angiotensin (1-9) [ANG (1-9)], angiotensin (1-7) [ANG (1-7)], and angiotensin (1-5) [ANG (1-5)], excreted into the buffer during experiment, were measured using liquid chromatography-mass spectrometry (LC/MS) and expressed per mg of dry tissue. Effects of pretreatment with 10 microM perindoprilat on the production of ANG I metabolites were quantitated. Background production of any of ANG I metabolites differed neither between WKY and SHR rats nor between 3- and 7-month-old rats. Perindoprilat pretreatment of renal arteries resulted, as expected, in decrease of ANG II production. However, renal arteries of 7-month-old SHR rats were resistant to ACE inhibitor and did not change ANG II production in response to perindoprilat. In renal arteries, taken from 3-month-old rats, pretreated with perindoprilat, incubation with ANG I, resulted in the level of ANG (1-9) significantly higher in SHR than WKY rats. Our conclusion is that in SHR rats, sensitivity of renal artery ACE to perindoprilat inhibition changes with age.


Subject(s)
Angiotensin I/metabolism , Hypertension/metabolism , Indoles/pharmacology , Peptide Fragments/metabolism , Renal Artery/metabolism , Aging/metabolism , Animals , In Vitro Techniques , Male , Rats, Inbred SHR , Rats, Inbred WKY , Renal Artery/drug effects
15.
J Physiol Pharmacol ; 67(4): 531-541, 2016 Aug.
Article in English | MEDLINE | ID: mdl-27779474

ABSTRACT

Increasing evidence indicates a role of oxytocin in controlling energy metabolism. The aim of his study was to investigate oxytocin effects on obese phenotype in leptin-resistant Zucker fatty rats, focusing on glucose and lipid metabolism. Zucker fatty rats and their lean controls were treated with oxytocin (3.6 µg/100g body weight/day) by osmotic minipumps implanted subcutaneously for 2 weeks. Two-hours intraperitoneal glucose tolerance test was performed in fasting rats. Oxytocin decreased food intake in both phenotypes while body weight gain reduced only in obese animals. In obese rats oxytocin impaired hepatic insulin extraction and enhanced liver triglyceride accumulation. Moreover, in the skeletal muscle of lean rats oxytocin treatment downregulated insulin signal transduction by decreasing of insulin receptor substrate 1 protein level and stimulating of its serine phosphorylation. Concurrently, the gene expression of insulin receptor substrate 1 in the skeletal muscle and adipose tissue was downregulated by oxytocin. In obese rats, oxytocin reduced adipocyte size and normalised mRNA levels of both fatty acid binding protein 4 and fatty acid synthase but attenuated gene expression of glucose transporter 4. The present study in Zucker fatty rats demonstrated ambivalent effects of oxytocin treatment with predominantly negative impact on skeletal muscle insulin pathway in lean animals.


Subject(s)
Adipose Tissue/drug effects , Liver/drug effects , Muscle, Skeletal/drug effects , Obesity/metabolism , Oxytocin/pharmacology , Adipose Tissue/metabolism , Adipose Tissue/pathology , Animals , C-Peptide/blood , Eating/drug effects , Fatty Acid Synthase, Type I/genetics , Fatty Acid-Binding Proteins/genetics , Glucose Tolerance Test , Insulin/blood , Leptin/blood , Liver/metabolism , Liver/pathology , Male , Muscle, Skeletal/metabolism , Obesity/blood , Obesity/pathology , Oxytocin/blood , Oxytocin/pharmacokinetics , RNA, Messenger/metabolism , Rats, Zucker , Receptors, Oxytocin/genetics , Triglycerides/metabolism
16.
J Diabetes Res ; 2016: 4846819, 2016.
Article in English | MEDLINE | ID: mdl-27803936

ABSTRACT

Purpose. Products of angiotensin (ANG) I metabolism may predispose to vascular complications of diabetes mellitus. Methods. Diabetes was induced with streptozotocin (75 mg/kg i.p.). Rat aorta fragments, isolated 4 weeks later, were pretreated with perindoprilat (3 µM), thiorphan (3 µM), or vehicle and incubated for 15 minutes with ANG I (1 µM). Products of ANG I metabolism through classical (ANG II, ANG III, and ANG IV) and alternative (ANG (1-9), ANG (1-7), and ANG (1-5)) pathways were measured in the buffer, using liquid chromatography-mass spectrometry. Results. Incubation with ANG I resulted in higher concentration of ANG II (P = 0.02, vehicle pretreatment) and lower of ANG (1-9) (P = 0.048, perindoprilat pretreatment) in diabetes. Preference for the classical pathway is suggested by higher ANG III/ANG (1-7) ratios in vehicle (P = 0.03), perindoprilat (P = 0.02), and thiorphan pretreated (P = 0.02) diabetic rat. Within the classical pathway, ratios of ANG IV/ANG II (P = 0.01) and of ANG IV/ANG III (P = 0.049), but not of ANG III/ANG II are lower in diabetes. Conclusions. Diabetes in rats led to preference toward deleterious (ANG II, ANG III) over protective (ANG IV, ANG (1-9), and ANG (1-7)) ANG I metabolites.


Subject(s)
Angiotensin II/metabolism , Angiotensin I/metabolism , Aorta/metabolism , Diabetes Mellitus, Experimental/metabolism , Angiotensin I/drug effects , Angiotensin II/drug effects , Angiotensin-Converting Enzyme Inhibitors/pharmacology , Animals , Aorta/drug effects , Chromatography, Liquid , Indoles/pharmacology , Mass Spectrometry , Peptide Fragments/drug effects , Peptide Fragments/metabolism , Protease Inhibitors/pharmacology , Rats , Rats, Sprague-Dawley , Thiorphan/pharmacology
17.
J Physiol Pharmacol ; 67(1): 75-91, 2016 Feb.
Article in English | MEDLINE | ID: mdl-27010897

ABSTRACT

The inhibition of angiotensin-converting enzyme (ACE) or the blockade of angiotensin (Ang) AT-1 receptors affords protection against acute gastric mucosal injury, but whether the major metabolite of renin-angiotensin system (RAS), Ang-(1-7), accelerates the healing process of preexisting gastric ulcers remains unknown. Previous studies documented that Ang-(1-7) acting via its own Mas receptor exerts vascular responses opposing those of Ang II. We studied the effects of the Ang-(1-7)/Mas receptor axis on the healing rate of acetic-acid-induced gastric ulcers with or without the blockade of Mas receptors by A 779 and compared it with the effects of activation and blockade of the AT-1 receptor by the treatment with Ang II and losartan, respectively, the inhibition of ACE by lisinopril, the NO/cNOS inhibition by L-NAME and inhibition of prostaglandin/COX system by indomethacin in the presence of Ang-(1-7). Additionally, ex vivo metabolism of Ang I in gastric tissue was assessed by LC/MS method. At day 9 after ulcer induction, the area of these ulcers and the accompanying changes in total gastric blood flow (GBF) were determined as were gastric mucosal blood flow (GMBF) at ulcer margin and gastric oxygen uptake (GVO2). The gastric mucosal expression of mRNAs for constitutive nitric oxide synthase (cNOS), superoxide dismutase (SOD), and pro-inflammatory cytokines interleukin 1ß (IL-1ß) and tumor necrosis factor alpha (TNF-α) and plasma level of both cytokines were determined by RT-PCR and ELISA. The 9 days treatment with Ang II dose-dependently increased the area of gastric ulcers and this effect was accompanied by a significant fall in the GBF, GVO2 and GMBF at ulcer margin. In contrast, treatment with Ang-(1-7) which produced a significant rise in the luminal content of NO significantly reduced the area of gastric ulcer and significantly increased the GBF, GVO2 and the GMBF at ulcer margin. Similar GMBF changes and significant reduction the area of gastric ulcer was observed in rats with gastric ulcers treated with the agonist of Mas receptor, AVE 0991. These effects of Ang-(1-7) and AVE 0991 were eliminated by blockade of the Mas receptor with A779. Similarly to Ang-(1-7), treatment with losartan or lisinopril significantly reduced the area of gastric ulcers and the accompanying increase in the GMBF at ulcer margin and these effects were significantly attenuated by a concomitant administration of L-NAME and indomethacin. The rate of healing of ulcers was associated with a decrease in ex vivo Ang-(1-7) formation and this effect was attenuated by lisinopril. The treatment with Ang-(1- 7) or AVE 0991 increased the expression of mRNA for cNOS and SOD and downregulated that of IL-1ß and TNF-α followed by the decrease in the plasma IL-1ß and TNF-α levels. We conclude that the Ang-(1-7)/Mas receptor system accelerates the healing of preexisting gastric ulcers via an increase in the gastric macro- and microcirculations, and an increase in gastric tissue oxygenation. These effects are mediated by PG and NO derived from overexpression of cNOS, an increase in the expression of antioxidizing enzyme SOD 2 and an anti-inflammatory action involving the inhibition of expression and release of pro-inflammatory cytokines IL-1ß and TNF-α. Our results seem to underlie the importance of the Ang-(1-7), AT-1 and Mas receptors in the regulation of local vascular and metabolic effects associated with mechanism of gastric ulcer healing.


Subject(s)
Angiotensin I/metabolism , Cytokines/metabolism , Nitric Oxide/metabolism , Peptide Fragments/metabolism , Prostaglandins/metabolism , Proto-Oncogene Proteins/metabolism , Receptors, G-Protein-Coupled/metabolism , Renin-Angiotensin System/drug effects , Stomach Ulcer/metabolism , Angiotensin II/metabolism , Angiotensin-Converting Enzyme Inhibitors/pharmacology , Animals , Enzyme Inhibitors/pharmacology , Gastric Mucosa/drug effects , Gastric Mucosa/metabolism , Imidazoles/pharmacology , Indomethacin/pharmacology , Interleukin-1beta/metabolism , Lisinopril/pharmacology , Losartan/pharmacology , Male , NG-Nitroarginine Methyl Ester/pharmacology , Nitric Oxide Synthase/metabolism , Proto-Oncogene Mas , Rats , Rats, Wistar , Superoxide Dismutase/metabolism , Tumor Necrosis Factor-alpha/metabolism
18.
Article in English | MEDLINE | ID: mdl-15626597

ABSTRACT

Antiplatelet thienopyridines (ticlopidine, clopidogrel) and their thienopyrimidinone congeners, induce prostacyclin-dependent thrombolysis in vivo. Here we tested whether thienopyridines (ticlopidine, clopidogrel, and its enantiomer without antiplatelet properties) and structurally related thienopyrimidinones release NO from coronary endothelium in the isolated guinea pig heart, perfused according to Langendorff technique. The involvement of endothelium-derived NO in coronary vasodilation induced by these agents was assessed by effect of L-N(G)-nitro-arginine methyl ester (L-NAME). In addition, effect of thienopyridines or thienopyrimidinones on nitrite accumulation in cultured endothelium was assayed. Tienopyridines (10-100 micromol L(-1)) and thienopyrimidinones (10-30 micromol L(-1)) produced concentration-dependent increase in coronary flow comparable to that induced by acetylcholine (0.1 micromol L(-1)) or bradykinin (3 nmol L(-1)) which was inhibited by L-NAME (by 50-70%) but not by indomethacin. Furthermore, thienopyridines and thienopyrimidinones caused NO release from cultured endothelial cells. In conclusion, both thienopyridines independently from their antiplatelet action and their thienopyrimidinone congeners that are devoid of antiplatelet action stimulate coronary endothelium to release NO. Endothelial action of these compounds merits further investigation.


Subject(s)
Endothelium, Vascular/drug effects , Pyridines/pharmacology , Pyrimidines/pharmacology , Animals , Coronary Circulation/drug effects , Coronary Vessels/drug effects , Coronary Vessels/metabolism , Endothelium, Vascular/metabolism , Guinea Pigs , Heart , In Vitro Techniques , Nitric Oxide/metabolism , Platelet Aggregation Inhibitors/pharmacology
19.
J Physiol Pharmacol ; 56(2): 299-311, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15985710

ABSTRACT

Nitric oxide (NO), depending on the amount, time and source of generation may exert both, protective and deleterious actions during endotoxic acute lung injury (ALI). Evaluation of the expression and localization of NOS isoforms in the lung of lipopolysaccharide (LPS)-treated rats may contribute to understanding the role of NO in pathogenesis of ALI. Tissue samples (lung, heart, liver, kidney and spleen) as well as peripheral blood polymorphonuclear cells (PMNs) were collected from control male Wistar rats and LPS - treated animals, 15, 30, 60, 120 and 180 min after LPS injection (2 mg kg(-1) min(-1) for 10 minutes, i.v.). Levels of NOS-2 and NOS-3 mRNA and protein in tissues and PMNs were estimated by RT-PCR, Northern blotting and Western blotting. Additionally, myeloperoxidase (MPO) activity in tissue samples was assayed. NOS-3 mRNA as well as protein were detected in lungs of control animals; pulmonary NOS-3 expression was not influenced by LPS. The induction of NOS-2 mRNA in rat lungs and in PMNs isolated from peripheral blood was observed 15 minutes after LPS challenge. In contrast, increase of NOS-2 mRNA in the heart, kidneys, liver and spleen was observed 2-3 hours after LPS injection. In all tissues rise in NOS-2 mRNA was followed after 1-2 hours by increase of NOS-2 protein. Importantly, progressive leukocyte sequestration in the lung parenchyma that started as early as 15 min after LPS injection was revealed only in the lungs; in other organs no significant changes in MPO activity were detected up to 180 min after LPS injection. In conclusion, infusion of LPS caused much more rapid expression of NOS-2 in lungs as compared to the heart, kidneys, liver and spleen. Early induction of NOS-2 may depend on the LPS-stimulated rapid neutrophil sequestration within lung vasculature and fast induction of NOS-2 in sequestrated neutrophils.


Subject(s)
Endotoxemia/enzymology , Leukocytes/physiology , Lung/enzymology , Nitric Oxide Synthase/genetics , Animals , Endotoxemia/blood , Male , Nitric Oxide Synthase/analysis , Nitric Oxide Synthase Type II , Nitric Oxide Synthase Type III , Peroxidase/metabolism , RNA, Messenger/analysis , Rats , Rats, Wistar
20.
J Physiol Pharmacol ; 56(4): 627-35, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16391419

ABSTRACT

It is widely appreciated that inflammation and oxidant stress contribute to atherogenesis. Curcumin, a polyphenolic natural compound has been reported to possess anti-inflammatory and anti-oxidant actions. We hypothesized that curcumin could inhibit the development of atherosclerosis in the apoE/LDLR-double knockout mice fed with Western diet (21% fat, 0.15% cholesterol w/w, without cholic acid). Curcumin (purity>or=98%), premixed with diet, was given for 4 months at a dose of 0.3 mg/ per day/ per mouse. In this model curcumin inhibited atherogenesis, measured both by "en face" method (25,15+/-2,9% vs. 19,2+/-0,6%, p<0,05) and "cross-section" method (565867+/-39764 microm2 vs. 299201+/-20373 microm2, p<0,05). Importantly, curcumin influenced neither the concentrations of cholesterol and triglycerides in blood nor animal body weight. To our knowledge, this is the first report that shows the anti-atherogenic effect of low dose of curcumin in fine model of atherosclerosis: gene-targeted apoE/LDLR-double knockout mice.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Atherosclerosis/prevention & control , Curcumin/pharmacology , Animals , Aorta/drug effects , Aorta/pathology , Apolipoproteins E/genetics , Atherosclerosis/chemically induced , Atherosclerosis/pathology , Diet, Atherogenic , Female , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, LDL/genetics
SELECTION OF CITATIONS
SEARCH DETAIL