Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
Int. j. morphol ; 34(4): 1482-1486, Dec. 2016. ilus
Article in English | LILACS | ID: biblio-840912

ABSTRACT

ICAM-1 which is expressed by endothelial cells and leukocytes are observed as first markers in diseases such as transplant rejection, diabetes and atherosclerosis and in infections caused by various pathogens. In the present study, it is aimed to reveal the age-related changes in the expression of ICAM-1 on rats. Therefore, a total of 30 albino rats were taken at the age of 6, 18 and 24 months without gender discrimination. Rats were fed with standard pellet feed during the study. Afterwards, rats were sacrificed and tissue samples were collected from their rats, and the samples were evaluated under the light microscope by staining with immnunohistochemical method. It was determined that the expression of both aortic endothelial cells and endothelial cells in the media layer had been significantly increased based on the age.


ICAM-1, que se expresa en las células endoteliales y los leucocitos, se observan como primeros marcadores de enfermedades como el rechazo de trasplante, la diabetes y la aterosclerosis y en las infecciones causadas por diversos patógenos. En el presente estudio, se pretende revelar los cambios relacionados con la edad en la expresión de ICAM-1 en ratas. Un total de 30 ratas albinas fueron seleccionadas con edades de 6, 18 y 24 meses, sin discriminación de sexo. Durante el estudio se administró a las ratas alimentación estándar de pellets. Posteriormente, los animales fueron sacrificados y se recogieron muestras de sus tejidos, los cuales fueron teñidos para inmunohistoquímica y se evaluaron a través de microscopio óptico. Se determinó que la expresión tanto de células endoteliales aórticas como de células endoteliales de la capa media se incrementó significativamente en función de la edad.


Subject(s)
Animals , Rats , Aorta/cytology , Endothelium, Vascular , Intercellular Adhesion Molecule-1/metabolism , Age Factors , Rats, Wistar
2.
Experimental & Molecular Medicine ; : e168-2015.
Article in English | WPRIM | ID: wpr-30206

ABSTRACT

Elevated plasma concentration of native low-density lipoprotein (nLDL) is associated with vascular smooth muscle cell (VSMC) activation and cardiovascular disease. We investigated the mechanisms of superoxide generation and its contribution to pathophysiological cell proliferation in response to nLDL stimulation. Lucigenin-induced chemiluminescence was used to measure nLDL-induced superoxide production in human aortic smooth muscle cells (hAoSMCs). Superoxide production was increased by nicotinamide adenine dinucleotide phosphate (NADPH) and decreased by NADPH oxidase inhibitors in nLDL-stimulated hAoSMC and hAoSMC homogenates, as well as in prepared membrane fractions. Extracellular signal-regulated kinase 1/2 (Erk1/2), protein kinase C-theta (PKCtheta) and protein kinase C-beta (PKCbeta) were phosphorylated and maximally activated within 3 min of nLDL stimulation. Phosphorylated Erk1/2 mitogen-activated protein kinase, PKCtheta and PKCbeta stimulated interactions between p47phox and p22phox; these interactions were prevented by MEK and PKC inhibitors (PD98059 and calphostin C, respectively). These inhibitors decreased nLDL-dependent superoxide production and blocked translocation of p47phox to the membrane, as shown by epifluorescence imaging and cellular fractionation experiments. Proliferation assays showed that a small interfering RNA against p47phox, as well as superoxide scavenger and NADPH oxidase inhibitors, blocked nLDL-induced hAoSMC proliferation. The nLDL stimulation in deendothelialized aortic rings from C57BL/6J mice increased dihydroethidine fluorescence and induced p47phox translocation that was blocked by PD98059 or calphostin C. Isolated aortic SMCs from p47phox-/- mice (mAoSMCs) did not respond to nLDL stimulation. Furthermore, NADPH oxidase 1 (Nox1) was responsible for superoxide generation and cell proliferation in nLDL-stimulated hAoSMCs. These data demonstrated that NADPH oxidase activation contributed to cell proliferation in nLDL-stimulated hAoSMCs.


Subject(s)
Animals , Humans , Aorta/cytology , Cell Line , Cell Proliferation , Cells, Cultured , Lipoproteins, LDL/metabolism , Mice, Inbred C57BL , Mitogen-Activated Protein Kinases/metabolism , Muscle, Smooth, Vascular/cytology , Myocytes, Smooth Muscle/cytology , NADPH Oxidases/metabolism , Phosphorylation , Protein Kinase C/metabolism , Signal Transduction , Superoxides/metabolism
3.
Experimental & Molecular Medicine ; : e180-2015.
Article in English | WPRIM | ID: wpr-186437

ABSTRACT

Leptin is a peptide hormone, which has a central role in the regulation of body weight; it also exerts many potentially atherogenic effects. Ferulic acid ethyl ester (FAEE) has been approved for antioxidant properties. The aim of this study was to investigate whether FAEE can inhibit the atherogenic effects of leptin and the possible molecular mechanism of its action. Both of cell proliferation and migration were measured when the aortic smooth muscle cell (A10 cell) treated with leptin and/or FAEE. Phosphorylated p44/42MAPK, cell cycle-regulatory protein (for example, cyclin D1, p21, p27), beta-catenin and matrix metalloproteinase-9 (MMP-9) proteins levels were also measured. Results demonstrated that leptin (10, 100 ng ml-1) significantly increased the proliferation of cells and the phosphorylation of p44/42MAPK in A10 cells. The proliferative effect of leptin was significantly reduced by the pretreatment of U0126 (0.5 muM), a MEK inhibitor, in A10 cells. Meanwhile, leptin significantly increased the protein expression of cyclin D1, p21, beta-catenin and decreased the expression of p27 in A10 cells. In addition, leptin (10 ng ml-1) significantly increased the migration of A10 cells and the expression of MMP-9 protein. Above effects of leptin were significantly reduced by the pretreatment of FAEE (1 and 10 muM) in A10 cells. In conclusion, FAEE exerts multiple effects on leptin-induced cell proliferation and migration, including the inhibition of p44/42MAPK phosphorylation, cell cycle-regulatory proteins and MMP-9, thereby suggesting that FAEE may be a possible therapeutic approach to the inhibition of obese vascular disease.


Subject(s)
Animals , Rats , Antioxidants/pharmacology , Aorta/cytology , Caffeic Acids/pharmacology , Cell Line , Cell Movement/drug effects , Cell Proliferation/drug effects , Leptin/metabolism , Matrix Metalloproteinase 9/metabolism , Muscle, Smooth, Vascular/cytology , Myocytes, Smooth Muscle/cytology , beta Catenin/metabolism
4.
Int. j. morphol ; 31(4): 1430-1438, Dec. 2013. ilus
Article in Spanish | LILACS | ID: lil-702329

ABSTRACT

Las células perivasculares tienen un origen común en las células madre embrionarias y en los vasos sanguíneos que proporcionan un nicho para la mantención de su troncalidad. La expresión de marcadores embrionarios y de células indiferenciadas, como también la gran variedad de fenotipos celulares generados desde los pericitos, podrían ser explicados por la capacidad de estas células de ser inducidas a un estado "stemness" cuando son tratadas con factores adecuados. Nuestros resultados describen la expresión de células con OCT-4 citoplasmático en una ubicación anatómica perivascular donde de su nicho se encuentra en la región intima de la aorta en rata. In vitro las células aisladas por el método de explante que promueve el aislamiento de células migratorias desde los tejidos muestran un fenotipo con un citoplasma alargado y que expresan aSMA, PDGFRa y b, siendo estos dos últimos marcadores específicos de pericitos. En estas células se presenta una tranlocación a la variante nuclear de OCT-4 que ha sido descrito como el principal regulador de los procesos de autorrenovación y pluripotencia. La expresión de OCT-4 confirma y amplía aún más las observaciones obtenidas en nuestras investigaciones anteriores y demuestra que células madre se encuentran en los vasos sanguíneos en un microambiente que, probablemente, les permite que sobrevivan y permanezcan en reposo como un tipo de célula troncal quiescente.


Perivascular cells have a common origin from embryonic stem cells and blood vessels provide a niche for the maintenance of their stemness. Embryonic markers expression of undifferentiated cells, as well as, the wide variety of cellular phenotypes generated from pericytes, could be explained by the ability of these cells to be induced to a state of "stemness" when treated with appropriate factors. Our findings describe the expression of cells with cytoplasmic OCT-4 in perivascular anatomical location where their niche region is in the intima of the aorta in rats. In vitro isolated cells by explant method that promotes the isolation of migratory cells from tissues show an elongated cytoplasm phenotype, expressing aSMA, PDGFRa & b where the last two are specific markers of pericytes. These cells present a translocated nuclear variant of OCT-4 that has been described as the master regulator of self-renewal processes and pluripotency. The expression of OCT-4 further confirms and extends the observations obtained in our previous research and proves that stem cells found in the blood vessels in a microenvironment that probably allows them to survive and remain at rest as a type of quiescent stem cell.


Subject(s)
Animals , Rats , Aorta/cytology , Pericytes , Stem Cell Niche , Transcription Factors , Translocation, Genetic , Cells, Cultured , Flow Cytometry , Fluorescent Antibody Technique, Indirect , Polymerase Chain Reaction , Rats, Sprague-Dawley
5.
Experimental & Molecular Medicine ; : 684-692, 2011.
Article in English | WPRIM | ID: wpr-190966

ABSTRACT

Recent epidemiologic studies clearly showed that early intensive glucose control has a legacy effect for preventing diabetic macrovascular complications. However, the cellular and molecular processes by which high glucose leads to macrovascular complications are poorly understood. Vascular smooth muscle cell (VSMC) dysfunction due to high glucose is a characteristic of diabetic vascular complications. Activation of nuclear factor-kappaB (NF-kappaB) may play a key role in the regulation of inflammation and proliferation of VSMCs. We examined whether VSMC proliferation and plasminogen activator inhibitor-1 (PAI-1) expression induced by high glucose were mediated by NF-kappaB activation. Also, we determined whether selective inhibition of NF-kappaB would inhibit proliferation and PAI-1 expression in VSMCs. VSMCs of the aorta of male SD rats were treated with various concentrations of glucose (5.6, 11.1, 16.7, and 22.2 mM) with or without an inhibitor of NF-kappaB or expression of a recombinant adenovirus vector encoding an IkappaB-alpha mutant (Ad-IkappaBalphaM). VSMC proliferation was examined using an MTT assay. PAI-1 expression was assayed by real-time PCR and PAI-1 protein in the media was measured by ELISA. NF-kappaB activation was determined by immunohistochemical staining, NF-kappaB reporter assay, and immunoblotting. We found that glucose stimulated VSMC proliferation and PAI-1 expression in a dose-dependent manner up to 22.2 mM. High glucose (22.2 mM) alone induced an increase in NF-kappaB activity. Treatment with inhibitors of NF-kappaB such as MG132, PDTC or expression of Ad-IkappaB-alphaM in VSMCs prevented VSMC proliferation and PAI-1 expression induced by high glucose. In conclusion, inhibition of NF-kappaB activity prevented high glucose-induced VSMC proliferation and PAI-1 expression.


Subject(s)
Animals , Male , Rats , Aorta/cytology , Cardiovascular Diseases/prevention & control , Cell Proliferation/drug effects , Cells, Cultured , Diabetes Complications/prevention & control , Gene Expression Regulation/drug effects , Glucose/immunology , Leupeptins/pharmacology , Muscle, Smooth, Vascular/cytology , Myocytes, Smooth Muscle/cytology , NF-kappa B/antagonists & inhibitors , Plasminogen Activator Inhibitor 1/genetics , Proline/analogs & derivatives , Rats, Sprague-Dawley , Thiocarbamates/pharmacology
6.
Yonsei Medical Journal ; : 420-428, 2011.
Article in English | WPRIM | ID: wpr-95678

ABSTRACT

PURPOSE: Dexmedetomidine, a full agonist of alpha2B-adrenoceptors, is used for analgesia and sedation in the intensive care units. Dexmedetomidine produces an initial transient hypertension due to the activation of post-junctional alpha2B-adrenoceptors on vascular smooth muscle cells (SMCs). The aims of this in vitro study were to identify mitogen-activated protein kinase (MAPK) isoforms that are primarily involved in full, alpha2B-adrenoceptor agonist, dexmedetomidine-induced contraction of isolated rat aortic SMCs. MATERIALS AND METHODS: Rat thoracic aortic rings without endothelium were isolated and suspended for isometric tension recording. Cumulative dexmedetomidine (10(-9) to 10(-6) M) dose-response curves were generated in the presence or absence of extracellular signal-regulated kinase (ERK) inhibitor PD 98059, p38 MAPK inhibitor SB 203580, c-Jun NH2-terminal kinase (JNK) inhibitor SP 600125, L-type calcium channel blocker (verapamil and nifedipine), and alpha2-adrenoceptor inhibitor atipamezole. Dexmedetomidine-induced phosphorylation of ERK, JNK, and p38 MAPK in rat aortic SMCs was detected using Western blotting. RESULTS: SP 600125 (10(-6) to 10(-5) M) attenuated dexmedetomidine-evoked contraction in a concentration-dependent manner, whereas PD 98059 had no effect on dexmedetomidine-induced contraction. SB 203580 (10(-5) M) attenuated dexmedetomidine-induced contraction. Dexmedetomidine-evoked contractions were both abolished by atipamezole and attenuated by verapamil and nifedipine. Dexmedetomidine induced phosphorylation of JNK and p38 MAPK in rat aortic SMCs, but did not induce phosphorylation of ERK. CONCLUSION: Dexmedetomidine-induced contraction involves a JNK- and p38 MAPK-mediated pathway downstream of alpha2-adrenoceptor stimulation in rat aortic SMCs. In addition, dexmedetomidine-induced contractions are primarily dependent on calcium influx via L-type calcium channels.


Subject(s)
Animals , Male , Rats , Adrenergic alpha-2 Receptor Agonists/pharmacology , Anthracenes/pharmacology , Aorta/cytology , Dexmedetomidine/pharmacology , Enzyme Inhibitors/pharmacology , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Flavonoids/pharmacology , Imidazoles/pharmacology , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , Muscle Contraction , Muscle, Smooth, Vascular/drug effects , Protein Isoforms/antagonists & inhibitors , Pyridines/pharmacology , Rats, Sprague-Dawley , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors
7.
Journal of Korean Medical Science ; : 514-520, 2008.
Article in English | WPRIM | ID: wpr-201065

ABSTRACT

The understanding of main mechanisms that determine the ability of immune privilege related to Sertoli cells (SCs) will provide clues for promoting a local tolerogenic environment. In this study, we evaluated the property of humoral and cellular immune response modulation provided by porcine SCs. Porcine SCs were resistant to human antibody and complement-mediated formation of the membrane attack complex (38.41+/-2.77% vs. 55.02+/-5.44%, p=0.027) and cell lysis (42.95+/-1.75% vs. 87.99 +/-2.25%, p<0.001) compared to immortalized aortic endothelial cells, suggesting that porcine SCs are able to escape cellular lysis associated with complement activation by producing one or more immunoprotective factors that may be capable of inhibiting membrane attack complex formation. On the other hand, porcine SCs and their culture supernatant suppressed the up-regulation of CD40 expression (p<0.05) on DCs in the presence of LPS stimulation. These novel findings, as we know, suggest that immune modulatory effects of porcine SCs in the presence of other antigen can be obtained from the first step of antigen presentation. These might open optimistic perspectives for the use of porcine SCs in tolerance induction eliminating the need for chronic immunosuppressive drugs.


Subject(s)
Animals , Humans , Male , Mice , Antibodies, Heterophile/immunology , Antibody Formation/immunology , CD40 Antigens/immunology , Aorta/cytology , Cell Line, Transformed , Cell Survival/immunology , Complement Membrane Attack Complex/immunology , Complement System Proteins/immunology , Dendritic Cells/cytology , Endothelial Cells/cytology , Epitopes/immunology , Immune Tolerance/immunology , Immunity, Cellular/immunology , Mice, Inbred C57BL , Sertoli Cells/cytology , Swine , Tissue Engineering , Transplantation, Heterologous
8.
Experimental & Molecular Medicine ; : 267-277, 2007.
Article in English | WPRIM | ID: wpr-201428

ABSTRACT

In vascular smooth muscle cells (VSMCs), induction of the heme oxygenase-1 (HO-1) confers vascular protection against cellular proliferation mainly via its up-regulation of the cyclin-dependent kinase inhibitor p21(WAF1/CIP1) that is involved in negative regulation of cellular proliferation. In the present study, we investigated whether the phytochemical curcumin and its metabolite tetrahydrocurcumin could induce HO-1 expression and growth inhibition in rat VSMCs and, if so, whether their antiproliferative effect could be mediated via HO-1 expression. At non-toxic concentrations, curcumin possessing two Michael-reaction acceptors induced HO-1 expression by activating antioxidant response element (ARE) through translocation of the nuclear transcription factor E2-related factor-2 (Nrf2) into the nucleus and also inhibited VSMC growth triggered by 5% FBS in a dose-dependent manner. In contrast, tetrahydrocurcumin lacking Michael-reaction acceptor showed no effect on HO-1 expression, ARE activation and VSMC growth inhibition. The antiproliferative effect of curcumin in VSMCs was accompanied by the increased expression of p21(WAF1/CIP1). Inhibition of VSMC growth and expression of p21(WAF1/CIP1) by curcumin were partially, but not completely, abolished when the cells were co- incubated with the HO inhibitor tin protoporphyrin. In human aortic smooth muscle cells (HASMCs), curcumin also inhibited growth triggered by TNF-alpha and increased p21(WAF1/CIP1) expression via HO-1-dependent manner. Our findings suggest that curcumin has an ability to induce HO-1 expression, presumably through Nrf2-dependent ARE activation, in rat VSMCs and HASMCs, and provide evidence that the antiproliferative effect of curcumin is considerably linked to its ability to induce HO-1 expression.


Subject(s)
Animals , Humans , Rats , Active Transport, Cell Nucleus , Aorta/cytology , Cell Nucleus/metabolism , Cell Proliferation/drug effects , Cells, Cultured , Curcumin/analogs & derivatives , Cyclin-Dependent Kinase Inhibitor p21/biosynthesis , Gene Expression Regulation , Heme Oxygenase (Decyclizing)/biosynthesis , Heme Oxygenase-1/biosynthesis , Metalloporphyrins/pharmacology , Muscle, Smooth, Vascular/drug effects , Myocytes, Smooth Muscle/drug effects , NF-E2-Related Factor 2/metabolism , Protoporphyrins/pharmacology , Regulatory Sequences, Nucleic Acid , Response Elements , Tumor Necrosis Factor-alpha/pharmacology
9.
Journal of Huazhong University of Science and Technology (Medical Sciences) ; (6): 135-6, 177, 2005.
Article in English | WPRIM | ID: wpr-641005

ABSTRACT

This study examined the effect of artesunate (Art) on the proliferation, DNA replication, cell cycles and apoptosis of vascular smooth muscle cells (VSMCs). Primary cultures of VSMCs were established from aortas of mice and artesunate of different concentrations was added into the medium. The number of VSMCs was counted and the curve of cell growth was recorded. The activity of VSMCs was assessed by using MTT method and inhibitory rate was calculated. DNA replication was evaluated by [3H]-TdR method and apoptosis by DNA laddering and HE staining. Flowmetry was used for simultaneous analysis of cell apoptosis and cell cycles. Compared with the control group, VSMCs proliferation in Art interfering groups were inhibited and [3H]-TdR incorprating rate were decreased as well as cell apoptosis was induced. The progress of cell cycle was blocked in G0/G1 by Art in a dose-dependent manner. It is concluded that Art inhibits VSMCs proliferation by disturbing DNA replication, inducing cell apoptosis and blocking cell cycle in G0/G1 phase.


Subject(s)
Aorta/cytology , Apoptosis/drug effects , Artemisinins/pharmacology , Cell Cycle/drug effects , Cells, Cultured , DNA Replication/drug effects , Muscle, Smooth, Vascular/cytology , Sesquiterpenes/pharmacology
10.
Experimental & Molecular Medicine ; : 18-26, 2005.
Article in English | WPRIM | ID: wpr-18135

ABSTRACT

NF-kappa B promotes cell survival against external stress such as radiation. We examined whether NF-kappa B decoy transfection enhances the antiproliferative effects of radiation on vascular smooth muscle cells (VSMCs) in vitro. The irradiation induced activation or nuclear translocation of NF-kappa B p65 in VSMCs was confirmed by immunofluorescence. NF-kB decoy transfection resulted in inhibition of the radiation-induced NF-kB activation in VSMCs and the subsequent reduction of transcription and translocation of ICAM, iNOS, and TNF-alpha, downstream molecules under the control of NF-kappa B. By using MTT assay, NF-kappa B decoy augmented the antiproliferative effects of radiation, where the effect of low dose radiation (2 and 8-Gy) of the cells transfected with NF-kappa B decoy was equivalent to the high dose (16-Gy) irradiated non-transfected cells at 48 h after irradiation: 1.06+/-0.16, 1.11+/-0.22, 1.20+/-0.25, respectively. The decrease in proliferation and survival of the radiation treated cells by flow cytometry analysis showed that NF-kappa B inhibition did not show any additive effects on the cell cycle of the irradiated VSMCs, while apoptosis was significantly increased after NF-kappa B decoy transfection in the irradiated VSMCs (apoptosis fraction: 13.33+/-2.08% vs. 26.29+/-7.43%, for radiation only vs. radiation+NF-kappa B decoy transfection, P < 0.05). In addition, at 48 h, NF-kappa B decoy transfection dose dependently (10 mM vs. 20 mM) inhibited proliferation of 16Gy-irradiated VSMCs, and showed greater antiproliferative efficacy than 100 mM sulfasalazine, a specific NF-kappa B inhibitor. These results indicate that NF-kappa B inhibition reduces proliferation and survival of irradiated VSMCs, likely by increased apoptosis rather than additive cell cycle arrest and suggest the possibility of adjunctive gene therapy using NF-kappa B decoy to improve efficacy and to decrease the adverse effects of intracoronary radiation therapy.


Subject(s)
Animals , Male , Rats , Aorta/cytology , Apoptosis , Cell Cycle/physiology , Cell Proliferation/radiation effects , Cells, Cultured , Gamma Rays , Intercellular Adhesion Molecule-1/metabolism , Muscle, Smooth, Vascular/cytology , Myocytes, Smooth Muscle/cytology , NF-kappa B/antagonists & inhibitors , Nitric Oxide Synthase/metabolism , Protein Transport , Rats, Sprague-Dawley , Transcription, Genetic , Transfection , Tumor Necrosis Factor-alpha/metabolism
11.
Braz. j. morphol. sci ; 21(1): 35-38, Jan.-Mar. 2004. ilus, tab
Article in English | LILACS | ID: lil-658764

ABSTRACT

The segmental microscopic architecture of the aortic wall of guinea pigs and albino rats was studied at the thoracicascending (T2-3), thoracic descending (T6-7) and abdominal infrarenal (L5-6) levels. Variables such as layer thickness,the number of elastic lamellae in the medial layer, and the diameter of the aortic segments were analyzedhistomorphometrically. The aortic wall of both species showed the usual elastic pattern, although variable amounts ofelastic lamellae, collagen fibers and smooth muscle cells were observed in the segmental analyses. A marked reductionin the number of elastic lamellae in the medial layer and in vascular diameter was observed in the abdominal aorticsegment of both species. Intimal folds, a gradual decrease in elastic lamellae from the ascending to the aorta abdominalaorta and a meshwork of adventitial collagen fibers and elastic lamellae were observed. These data indicate that thereare small but significant segmental variations in the aortic wall of these two species.


Subject(s)
Animals , Male , Female , Guinea Pigs , Rats , Aorta, Thoracic , Aorta/cytology , Collagen , Elastic Tissue/ultrastructure , Microscopy, Electron, Scanning , Rats, Inbred Strains
12.
Biol. Res ; 37(3): 419-430, 2004. graf
Article in English | LILACS | ID: lil-394436

ABSTRACT

In addition to the induction of cell proliferation and migration, bradykinin (BK) can increase c-fos mRNA expression, activate ERK 1/2 and generate reactive oxygen species (ROS) in vascular smooth muscle cells (VSMC). It is not known, however, whether BK can induce cellular proliferation and extracellular matrix production via redox-sensitive signaling pathways. We investigated the role(s) of ROS in proliferation, migration and collagen synthesis induced by BK in VSMC derived from Sprague Dawley rat aorta. BK (10 nM) increased VSMC proliferation by 30 % (n=5); this proliferation was inhibited by the antioxidants N-acetylcysteine (20 mM) and a-lipoic acid (LA, 250 mM). In addition, BK induced an increase in cell migration and in collagen levels that were blocked by LA. ROS production induced by BK (n=10) was significantly inhibited by bisindolylmaleimide (4mM) and by PD98059 (40mM). These results suggest that: 1) ROS participate in the mechanism(s) used by bradykinin to induce cellular proliferation; 2) bradykinin induces ROS generation through a pathway that involves the kinases PKC and MEK; and 3) ROS participate in the pathways mediating cell migration and the production of collagen as a response to treatment with bradykinin. To our knowledge, this is the first report describing mechanisms to explain the participation of ROS in the cellular proliferation and extracellular matrix pathway regulated by BK.


Subject(s)
Animals , Male , Rats , Antioxidants/pharmacology , Bradykinin/pharmacology , Cell Division/drug effects , Cell Movement/drug effects , Collagen/biosynthesis , Muscle, Smooth, Vascular/cytology , Reactive Oxygen Species , Aorta/cytology , Cells, Cultured , Rats, Sprague-Dawley
13.
Biocell ; 27(2): 189-196, Aug. 2003.
Article in English | LILACS | ID: lil-384244

ABSTRACT

Remodeling of large and small arteries contributes to the development and complications of hypertension. Artery structural changes in chronic sustained hypertension include vascular smooth muscle cells (VSMC) proliferation and extracellular matrix (ECM) modifications. Extracellular constituents such as proteoglycans (PGs), may modulate vascular stiffness and VSMC growth and differentiation. We examined the effect of growth factors on secreted and membrane-bound PGs synthesis by cultured aortic smooth muscle cells (SMC) from 12- to 14- week-old spontaneously hypertensive rats (SHR) and age-matched Wistar rats. After stimulation with platelet-derived growth factor (PDGF-BB), 10% fetal calf serum (FCS) or 0.1% FCS as control, PGs synthesis (dpm/ng DNA) was evaluated in the medium (M-ECM) and in the cell layer (P-ECM) by a double-isotopic label method using both [3H]-glucosamine and [35S]-sodium sulfate which are incorporated into all complex carbohydrates or only into sulfated dysaccharides, respectively. Data are presented as percent of the control (0.1% FCS). SHR VSMC displayed a significantly greater synthesis of M-ECM [3H]-PGs than Wistar rat cells, with both treatments, but no differences in M-ECM [35S] uptake were found in any case. In the P-ECM, both PDGF-BB and 10% FCS produced a greater effect on [3H]-PGs and sulfated PGs synthesis in VSMC from SHR. An important change seen in SHR cells was a significant decreased sulfation, assessed by [35S]/[3H] ratio, in basal and stimulation conditions. Present results indicate the existence of changes in PGS synthesis and modulation in VSMC from a conduit-artery of SHR and support the pathophysiological role proposed for matrix proteoglycans in the vascular wall changes associated to hypertension and related vascular diseases as atherosclerosis.


Subject(s)
Male , Aorta/metabolism , Hypertension/metabolism , Hypertrophy/metabolism , Extracellular Matrix/metabolism , Muscle, Smooth, Vascular/metabolism , Proteoglycans/metabolism , Aorta/cytology , Arteriosclerosis/metabolism , Arteriosclerosis/pathology , Arteriosclerosis/physiopathology , Cells, Cultured , Cell Division/drug effects , Cell Division/physiology , Platelet-Derived Growth Factor/metabolism , Platelet-Derived Growth Factor/pharmacology , Glucosamine/metabolism , Extracellular Matrix/drug effects , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular , Proteoglycans/drug effects , Proteoglycans , Rats , Rats, Inbred SHR , Sulfur Radioisotopes , Sulfates/metabolism
14.
Experimental & Molecular Medicine ; : 135-140, 2000.
Article in English | WPRIM | ID: wpr-105751

ABSTRACT

Thrombospondin-1 (TSP-1), a multifunctional extracellular matrix protein, inhibits neovascularization and is implicated in the regression of tumor growth and metastasis. We found that the synthesis of TSP-1 in porcine aortic endothelial (PAE) cells was decreased in a dose-dependent manner by phorbol 12-myristate 13-acetate (PMA) treatment in porcine aortic endothelial (PAE) cells. In this study, a responsive site on the TSP-1 promotor affected by PMA treatment in PAE was characterized. The level of TSP-1 mRNA was also decreased by PMA after 1 h and persisted that way for at least 24 h. PMA treatment and c-Jun overexpression suppressed the transcription of TSP-1 promotor-luciferase reporter gene. A deletion between -767 and -657 on the TSP-1 promotor neutralized the PMA-induced down-regulation. In addition, oligo a (-767 approximately -723) was responsive to PMA-induced repression, while oligo b (-734 approximately -689) and c (-700 approximately -656) was not. Electrophoretic mobility shift assays showed that this PMA responsive element specifically bound a nuclear protein and that the binding activity was diminished by PMA treatment in PAE cells but not in Hep 3B cells. In supershift assay, potential regulatory elements in this region, SP1 and GATA-1, were not responsive to the inhibition of TSP-1 expression by PMA. Our results suggest that the repression of TSP-1 synthesis by PMA is mediated by blocking a particular unknown nuclear protein binding to the responsive site (-767 approximately -735), which is regulated by c-Jun.


Subject(s)
Animals , Aorta/cytology , Cell Line , Down-Regulation , Endothelium, Vascular/drug effects , Endothelium, Vascular/cytology , Promoter Regions, Genetic , Proto-Oncogene Proteins c-jun/metabolism , Response Elements , Swine , Tetradecanoylphorbol Acetate/pharmacology , Thrombospondin 1/genetics , Thrombospondin 1/biosynthesis
15.
Yonsei Medical Journal ; : 740-755, 2000.
Article in English | WPRIM | ID: wpr-189804

ABSTRACT

In order to determine the precise mechanism of the interactions between different types of cells, which are common phenomena in tissues and organs, the importance of coculture techniques are becoming increasingly important. In the area of cardiology, artificial arteries have been developed, based on the understanding of physiological communication of the arterial smooth muscle cells (SMC), endothelial cells (EC), and the extracellular matrix (ECM). In the study of atherosclerosis, the modification of low-density lipoprotein (LDL), which result in the recruitment and accumulation of white blood cells, especially, monocytes/macrophages, and foam cell formation, are hypothesized. Although there are well known animal models, an in vitro model of atherogenesis with a precisely known atherogenesis mechanism has not yet been developed. In this paper, an arterial wall reconstruction model using rabbit primary cultivated aortic SMCs and ECs, was shown. In addition, human peripheral monocytes were used and the transmigration of monocytes was observed by scanning electron and laser confocal microscopy. Monocyte differentiation into macrophages was shown by immunohistochemistry and comprehensive gene expression analysis. With the modified form of LDL, the macrophages were observed to accumulate lipids with a foamy appearance and differentiate into the foam cells in the ECM between the ECs and SMCs in the area of our coculture model.


Subject(s)
Male , Rabbits , Animals , Aorta/physiology , Aorta/cytology , Arteriosclerosis/etiology , Cell Differentiation/physiology , Cell Movement , Coculture Techniques , Endothelium, Vascular/physiology , Endothelium, Vascular/cytology , Extracellular Matrix/metabolism , Foam Cells/ultrastructure , Foam Cells/cytology , Macrophages/physiology , Macrophages/cytology , Microscopy, Confocal , Microscopy, Electron , Microscopy, Electron, Scanning , Monocytes/ultrastructure , Monocytes/physiology , Muscle, Smooth, Vascular/physiology , Muscle, Smooth, Vascular/cytology , Myosins/metabolism , Protein Isoforms/metabolism
16.
Yonsei Medical Journal ; : 251-261, 1995.
Article in English | WPRIM | ID: wpr-52255

ABSTRACT

In order to investigate the anti-proliferative effect of 3-hydroxy-3-methylglutaryl coenzyme. A reductase inhibitor, we evaluated the effects of lovastatin on DNA replication and the proliferation of rat mesangial and aortic smooth muscle cells, both of which were mesenchymal origin cells. Proliferations were determined by measuring [3H]thymidine uptake, and counting the number of cells. Growth-arrested mesangial and aortic smooth muscle cells were exposed to platelet-derived growth factor (PDGF), endothelin (ET) and angiotensin II (Ang II) to stimulate mitogenesis. All agents exhibited dose-dependent stimulation of [3H] thymidine uptake. PDGF was more potent than the others. Ang II increased [3H] thymidine uptake without demonstrable mitogenic activity. Lovastatin inhibited PDGF (10 ng/ml in mesangial cell, 25 ng/ml in smooth muscle cell)-, ET (10(-7)M)- and Ang II (10(-7)M)-induced [3H] thymidine uptake significantly in a dose-dependent manner in both cells. The increase of cell number in response to PDGF and ET treatment were also inhibited at 10 microM of lovastatin. The inhibitory effect of lovastatin was largely overcome in the presence of exogenous mevalonate at 200 microM, with 75.5% restoration from lovastatin-induced inhibition on PDGF-induced [3H] thymidine uptake in mesangial cells (77.8% in aortic smooth muscle cells). However, the addition of cholesterol did not prevent inhibition by lovastatin. In conclusion, lovastatin had an inhibitory effect on mesangial and aortic smooth muscle cell proliferation, and mevalonate was essential for DNA replication in both types of cells. Lovastatin may reduce glomerular and atherosclerotic injury through an anti-proliferative effect on mesangial and vascular smooth muscle cells, in addition to lowering circulating lipids.


Subject(s)
Male , Rats , Angiotensin II/pharmacology , Animals , Aorta/cytology , Cell Division/drug effects , Cells, Cultured , Endothelins/pharmacology , Glomerular Mesangium/cytology , Lovastatin/pharmacology , Muscle, Smooth, Vascular/cytology , Platelet-Derived Growth Factor/pharmacology , Rats, Sprague-Dawley , Thymidine/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL