Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
1.
The Korean Journal of Physiology and Pharmacology ; : 69-79, 2020.
Article in English | WPRIM | ID: wpr-787137

ABSTRACT

Aging is one of the risk factors for the development of cardiovascular diseases. During the progression of cellular senescence, cells enter a state of irreversible growth arrest and display resistance to apoptosis. As a flavonoid, quercetin induces apoptosis in various cells. Accordingly, we investigated the relationship between quercetin-induced apoptosis and the inhibition of cellular senescence, and determined the mechanism of oxidative stress-induced vascular smooth muscle cell (VSMC) senescence. In cultured VSMCs, hydrogen peroxide (H₂O₂) dose-dependently induced senescence, which was associated with increased numbers of senescence-associated β-galactosidase-positive cells, decreased expression of SMP30, and activation of p53-p21 and p16 pathways. Along with senescence, expression of the anti-apoptotic protein Bcl-2 was observed to increase and the levels of proteins related to the apoptosis pathway were observed to decrease. Quercetin induced apoptosis through the activation of AMP-activated protein kinase. This action led to the alleviation of oxidative stress-induced VSMC senescence. Furthermore, the inhibition of AMPK activation with compound C and siRNA inhibited apoptosis and aggravated VSMC senescence by reversing p53-p21 and p16 pathways. These results suggest that senescent VSMCs are resistant to apoptosis and quercetin-induced apoptosis attenuated the oxidative stress-induced senescence through activation of AMPK. Therefore, induction of apoptosis by polyphenols such as quercetin may be worthy of attention for its anti-aging effects.


Subject(s)
Aging , AMP-Activated Protein Kinases , Apoptosis , Cardiovascular Diseases , Cellular Senescence , Hydrogen Peroxide , Muscle, Smooth, Vascular , Polyphenols , Quercetin , Risk Factors , RNA, Small Interfering
2.
Yeungnam University Journal of Medicine ; : 77-82, 2012.
Article in Korean | WPRIM | ID: wpr-147275

ABSTRACT

AMP-activated protein kinase (AMPK) is an important cellular fuel sensor. Its activation requires phosphorylation at Thr-172, which resides in the activation loop of the alpha1 and alpha2 subunits. Several AMPK upstream kinases are capable of phosphorylating AMPK at Thr-172, including LKB1 and CaMKKbeta (Ca2+/calmodulin-dependent protein kinase kinasebeta). AMPK has been implicated in the regulation of physiological signals, such as in the inhibition of cholesterol fatty acid, and protein synthesis, and enhancement of glucose uptake and blood flow. AMPK activation also exhibits several salutary effects on the vascular function and improves vascular abnormalities. AMPK is modulated by numerous hormones and cytokines that regulate the energy balance in the whole body. These hormone and cytokines include leptin, adiponectin, ghrelin, and even thyroid hormones. Moreover, AMPK is activated by several drugs and xenobiotics. Some of these are in being clinically used to treat type 2 diabetes (e.g., metformin and thiazolidinediones), hypertension (e.g., nifedipine and losartan), and impaired blood flow (e.g., aspirin, statins, and cilostazol). I reviewed the precise mechanisms of the AMPK activation pathway and AMPK-modulating drugs.


Subject(s)
Adiponectin , AMP-Activated Protein Kinases , Aspirin , Calcium-Calmodulin-Dependent Protein Kinase Kinase , Cholesterol , Cytokines , Ghrelin , Glucose , Hypertension , Leptin , Metformin , Nifedipine , Phosphorylation , Phosphotransferases , Protein Kinases , Thyroid Hormones , Xenobiotics
3.
Endocrinology and Metabolism ; : 109-115, 2012.
Article in Korean | WPRIM | ID: wpr-57558

ABSTRACT

AMP-activated protein kinase (AMPK) is an important cellular fuel sensor. Activation of AMPK requires phosphorylation at threonine (Thr)-172, which resides in the activation loop of the alpha1 and alpha2 subunits. Several AMPK upstream kinases are capable of phosphorylating AMPK at Thr-172, including LKB1 and CaMKKbeta. AMPK has been implicated in the regulation of physiological signals, such as inhibition of cholesterol, fatty acid, protein synthesis, and enhancement of glucose uptake and blood flow. AMPK activation also exhibits several salutary effects on vascular function and improves vascular abnormalities. AMPK is activated by numerous drugs and xenobiotics. Some of these are in clinical use for the treatment of type 2 diabetes (e.g., metformin and thiazolidinediones), hypertension (e.g., nifedipine and losartan), and impaired blood flow (e.g., aspirin, statins, and cilostazol). Plant-derived xenobiotics or nutraceuticals that were claimed to have health benefits in diabetes or cancer have been reported to activate AMPK. These include resveratrol from red wine, epigallocatechin gallate from green tea, capsaicin from peppers, berberine, which is a yellow dye of the genus berberis, genistein from soy bean, and ginsenoside from ginseng panax. AMPK is also modulated by numerous hormones and cytokines that regulate energy balance at the whole body level, including leptin, adiponectin, ghrelin, and even thyroid hormones. This work shows that the precise mechanisms of AMPK kinase and AMPK interaction.


Subject(s)
Adiponectin , AMP-Activated Protein Kinases , Aspirin , Berberine , Berberis , Calcium-Calmodulin-Dependent Protein Kinase Kinase , Capsaicin , Catechin , Cholesterol , Cytokines , Dietary Supplements , Genistein , Ghrelin , Glucose , Hypertension , Insurance Benefits , Leptin , Metformin , Nifedipine , Panax , Phosphorylation , Phosphotransferases , Protein Kinases , Soybeans , Stilbenes , Tea , Threonine , Thyroid Hormones , Wine , Xenobiotics
4.
The Korean Journal of Physiology and Pharmacology ; : 203-210, 2011.
Article in English | WPRIM | ID: wpr-727881

ABSTRACT

Cilostazol is a selective inhibitor of phosphodiesterase 3 that increases intracellular cAMP levels and activates protein kinase A, thereby inhibiting vascular smooth muscle cell (VSMC) proliferation. We investigated whether AMP-activated protein kinase (AMPK) activation induced by heme oxygenase-1 (HO-1) is a mediator of the beneficial effects of cilostazol and whether cilostazol may prevent cell proliferation and reactive oxygen species (ROS) production by activating AMPK in VSMC. In the present study, we investigated VSMC with various concentrations of cilostazol. Treatment with cilostazol increased HO-1 expression and phosphorylation of AMPK in a dose- and time-dependent manner. Cilostazol also significantly decreased platelet-derived growth factor (PDGF)-induced VSMC proliferation and ROS production by activating AMPK induced by HO-1. Pharmacological and genetic inhibition of HO-1 and AMPK blocked the cilostazol-induced inhibition of cell proliferation and ROS production.These data suggest that cilostazol-induced HO-1 expression and AMPK activation might attenuate PDGF-induced VSMC proliferation and ROS production.


Subject(s)
AMP-Activated Protein Kinases , Cell Proliferation , Cyclic AMP-Dependent Protein Kinases , Heme , Heme Oxygenase-1 , Muscle, Smooth, Vascular , Phosphorylation , Platelet-Derived Growth Factor , Reactive Oxygen Species , Tetrazoles
5.
The Korean Journal of Physiology and Pharmacology ; : 299-304, 2010.
Article in English | WPRIM | ID: wpr-728368

ABSTRACT

Losartan is a selective angiotensin II (Ang II) type 1 (AT1) receptor antagonist which inhibits vascular smooth muscle cells (VSMCs) contraction and proliferation. We hypothesized that losartan may prevent cell proliferation by activating AMP-activated protein kinase (AMPK) in VSMCs. VSMCs were treated with various concentrations of losartan. AMPK activation was measured by Western blot analysis and cell proliferation was measured by MTT assay and flowcytometry. Losartan dose- and time-dependently increased the phosphorylation of AMPK and its downstream target, acetyl-CoA carboxylase (ACC) in VSMCs. Losartan also significantly decreased the Ang II- or 15% FBS-induced VSMC proliferation by inhibiting the expression of cell cycle associated proteins, such as p-Rb, cyclin D, and cyclin E. Compound C, a specific inhibitor of AMPK, or AMPK siRNA blocked the losartan-induced inhibition of cell proliferation and the G0/G1 cell cycle arrest. These data suggest that losartan-induced AMPK activation might attenuate Ang II-induced VSMC proliferation through the inhibition of cell cycle progression.


Subject(s)
Acetyl-CoA Carboxylase , AMP-Activated Protein Kinases , Angiotensin II , Blotting, Western , Cell Cycle , Cell Cycle Checkpoints , Cell Proliferation , Contracts , Cyclin D , Cyclin E , Cyclins , Losartan , Muscle, Smooth, Vascular , Phosphorylation , Proteins , RNA, Small Interfering
6.
The Korean Journal of Physiology and Pharmacology ; : 123-129, 2009.
Article in English | WPRIM | ID: wpr-728745

ABSTRACT

Aprotinin is used clinically in cardiopulmonary bypass surgery to reduce transfusion requirements and the inflammatory response. The mechanism of action for the anti-inflammatory effects of aprotinin is still unclear. We examined our hypothesis whether inhibitory effects of aprotinin on cytokine-induced inducible nitric oxide synthase (iNOS) expression (IL-1beta plus TNF-alpha), reactive oxygen species (ROS) generation, and vascular smooth muscle cell (VSMC) proliferation were due to HO-1 induction in rat VSMCs. Aprotinin induced HO-1 protein expression in a dose-dependent manner, which was potentiated during inflammatory condition. Aprotinin reduced cytokine mixture (CM)-induced iNOS expression in a dose dependent manner. Furthermore, aprotinin reduced CM-induced ROS generation, cell proliferation, and phosphorylation of JNK but not of P38 and ERK1/2 kinases. Aprotinin effects were reversed by pre-treatment with the HO-1 inhibitor, tin protoporphyrin IX (SnPPIX). HO-1 is therefore closely involved in inflammatory-stimulated VSMC proliferation through the regulation of ROS generation and JNK phosphorylation. Our results suggest a new molecular basis for aprotinin anti-inflammatory properties.


Subject(s)
Animals , Rats , Aprotinin , Cardiopulmonary Bypass , Cell Proliferation , Inflammation , Metalloporphyrins , Muscle, Smooth, Vascular , Nitric Oxide Synthase Type II , Phosphorylation , Phosphotransferases , Protoporphyrins , Reactive Oxygen Species , Tin
7.
The Korean Journal of Physiology and Pharmacology ; : 309-313, 2009.
Article in English | WPRIM | ID: wpr-727519

ABSTRACT

Spontaneous hypertensive rats (SHR) are an established model of genetic hypertension. Vascular smooth muscle cells (VSMC) from SHR proliferate faster than those of control rats (Wistar-Kyoto rats; WKY). We tested the hypothesis that induction of heme oxygenase (HO)-1 induced by aprotinin inhibits VSMC proliferation through cell cycle arrest in hypertensive rats. Aprotinin treatment inhibited VSMC proliferation in SHR more than in normotensive rats. These inhibitory effects were associated with cell cycle arrest in the G1 phase. Tin protoporphyrin IX (SnPPIX) reversed the anti-proliferative effect of aprotinin in VSMC from SHR. The level of cyclin D was higher in VSMC of SHR than those of WKY. Aprotinin treatment downregulated the cell cycle regulator, cyclin D, but upregulated the cyclin-dependent kinase inhibitor, p21, in VSMC of SHR. Aprotinin induced HO-1 in VSMC of SHR, but not in those of control rats. Furthermore, aprotinin-induced HO-1 inhibited VSMC proliferation of SHR. Consistently, VSMC proliferation in SHR was significantly inhibited by transfection with the HO-1 gene. These results indicate that induction of HO-1 by aprotinin inhibits VSMC proliferation through cell cycle arrest in hypertensive rats.


Subject(s)
Animals , Rats , Aprotinin , Cell Cycle , Cell Cycle Checkpoints , Cell Proliferation , Cyclin D , G1 Phase , Heme , Heme Oxygenase (Decyclizing) , Heme Oxygenase-1 , Hypertension , Metalloporphyrins , Muscle, Smooth, Vascular , Phosphotransferases , Protoporphyrins , Tin , Transfection
8.
The Korean Journal of Physiology and Pharmacology ; : 275-280, 2008.
Article in English | WPRIM | ID: wpr-728376

ABSTRACT

A brief ischemic insult induces significant protection against subsequent massive ischemic events. The molecular mechanisms known as preconditioning (PC)-induced ischemic tolerance are not completely understood. We investigated whether kinetic changes of cyclooxygenase (COX)-2 during reperfusion time-periods after PC were related to ischemic tolerance. Rats were given PC by occlusion of middle cerebral artery (MCAO) for 10 min and sacrificed after the indicated time-periods of reperfusion (1, 2, 4, 8, 12, 18 or 24 h). In PC-treated rats, focal ischemia was induced by occlusion of MCA for 24 h and brain infarct volume was then studied to determine whether different reperfusion time influenced the damage. We report that the most significant protection against focal ischemia was obtained in rats with 8 h reperfusion after PC. Administration of indomethacin (10 mg/kg, oral) or rofecoxib (5 mg/kg, oral) 48 h prior to PC counteracted the effect of PC. Immunohistochemical analysis showed that COX-2 and HO-1 protein were induced in PC-treated rat brain, which was significantly inhibited by rofecoxib. Taken together, we concluded that the kinetic changes of COX-2 expression during the reperfusion period after PC might be partly responsible for ischemic tolerance.


Subject(s)
Animals , Rats , Brain , Heme Oxygenase (Decyclizing) , Indomethacin , Ischemia , Ischemic Preconditioning , Lactones , Middle Cerebral Artery , Prostaglandin-Endoperoxide Synthases , Reperfusion , Stroke , Sulfones
9.
The Korean Journal of Physiology and Pharmacology ; : 189-195, 2007.
Article in English | WPRIM | ID: wpr-728208

ABSTRACT

To study the protective effects of antioxidants on the radiation damages of the cells, vascular smooth muscle cells (VSMC) from thoracic aorta of Sprague-Dawley rats were cultured and irradiated with gamma-ray. Cell viability was measured by direct cell counting and MTT assay, and flow cytometry was performed to measure fractional distributions of the cells. Gamma-ray irradiation inhibited cell proliferations accompanied with decreased G1 phase and increased S- and G2/M phases, and the maximum effects were observed at 1500 or 2000 cGy. Submaximal concentrations of antioxidants, such as allopurinol, vitamin C, N-acetylcycteine (NAC), lipoic acid, dihydrolipoic acid and rebamipide tended to increase the cell viability suppressed by low dose of radiation (500 cGy), and enalapril and vitamin E increased it significantly. Allopurinol, vitamin E, NAC, lipoic acid, captopril and enalapril significantly increased G1 phase. Allopurinol and vitamin E tended to increase c-Myc expression, detected by Western blot, that was reduced by the radiation, and enalapril increased it significantly. The cell viability and c-Myc expression were highly correlated (r=0.97) with each other. These results suggest that antioxidants, especially enalapril and vitamin E, recover the viability of VSMC from gamma-radiation injury, through a mechanism which includes increase of c-Myc protein expression.


Subject(s)
Animals , Rats , Allopurinol , Antioxidants , Aorta , Aorta, Thoracic , Ascorbic Acid , Blotting, Western , Captopril , Cell Count , Cell Cycle , Cell Survival , Enalapril , Flow Cytometry , G1 Phase , Gamma Rays , Muscle, Smooth, Vascular , Rats, Sprague-Dawley , Thioctic Acid , Vitamin E , Vitamins
10.
Yeungnam University Journal of Medicine ; : 36-44, 2006.
Article in Korean | WPRIM | ID: wpr-102197

ABSTRACT

BACKGROUND: There is controversy regarding whether COX-2 specific inhibitors are associated with elevation of blood pressure. We compared the effects of aspirin, indomethacin, and celecoxib for vascular reactivity induced by phenylephrine. We also tested the effects of indomethacin and NO donor on COX-1 and COX-2 protein expression, as well as nitrite production in culture medium of vascular smooth muscle cells. MATERILAS AND METHODS: In this experiment, we used the isometric tension study for vascular reactivity. After 45 minutes of pretreatment with aspirin, indomethacin, celecoxib, and phenylephrine induced contractions were tested. COX-1 and COX-2 protein expressions were analyzed by Western blot and nitrite production by the Griess reaction. RESULTS: Although celecoxib pretreatment caused enhanced arterial contraction, aspirin pretreatment induced more potent arterial contraction than celecoxib in the isometric tension study of rabbit femoral artery. COX-1 protein expression was unchanged by indomethacin, SNP and NOR-3; COX-2 protein expression was increased by the addition of indomethacin, SNP, and NOR-3. Especially, NOR-3, a NO donor, significantly increased COX-2 protein expression with unstimulated conditions as well as LPS stimulation. Induction of nitrite production was higher with NOR-3 treatment than SNP treatment with LPS stimulation. CONCLUSION: These results suggest that aspirin caused more potent vascular contraction than celecoxib and indomethacin. COX-2 expression in VSMC depended on the types of NO donor and LPS stimulation.


Subject(s)
Humans , Anti-Inflammatory Agents, Non-Steroidal , Aspirin , Blood Pressure , Blotting, Western , Cyclooxygenase Inhibitors , Femoral Artery , Indomethacin , Muscle, Smooth, Vascular , Phenylephrine , Prostaglandin-Endoperoxide Synthases , Tissue Donors , Celecoxib
11.
Yeungnam University Journal of Medicine ; : 62-70, 2006.
Article in Korean | WPRIM | ID: wpr-102194

ABSTRACT

BACKGROUND: Benign prostatic hyperplasia (BPH) is the most common benign tumor in older men; the etiology of this disease remains poorly understood. Testosterone and dihydrotestosterone (DHT) both act as androgen via a single androgen receptor. Testosterone is converted to DHT by 5alpha-reductase in prostatic stromal cells. Progesterone has been reported to inhibit DHT conversion; howevwe, its effect on prostatic stromal cells remains to be elucidated. MATERILAS AND METHODS: In this experiment, we investigated the effect of progesterone on androgen receptor expression induced by DHT. We also tested the effect of progesterone on cyclooxygenase-2 (COX-2) expression, as well as prostate stromal cell proliferation using the cell count kit-8. RESULTS: Progesterone did not cause an increase of prostate stromal cell proliferation. The mRNA expression of the androgen receptor and COX-2 were not changed by progesterone; the expressions of androgen receptor and COX-2 proteins were decreased by progesterone in prostate stromal cells. CONCLUSION: These results suggest that in prostate stromal cells, progesterone decreases androgen receptor protein expression, which results in decrement of COX-2 protein expression. This effect might be mediated by post-transcriptional regulation.


Subject(s)
Humans , Male , Cell Count , Cyclooxygenase 2 , Dihydrotestosterone , Progesterone , Prostate , Prostatic Hyperplasia , Receptors, Androgen , RNA, Messenger , Stromal Cells , Testosterone
12.
The Korean Journal of Physiology and Pharmacology ; : 161-165, 2006.
Article in English | WPRIM | ID: wpr-728563

ABSTRACT

NO and cyclooxygenase-2 (COX-2) are contributes to vascular inflammation induced by various stimulation. The mechanism, which explains a linkage between NO and COX-2, could be of importance in promoting pathophysiological conditions of vessel. We investigated the effects of NO donors on the COX-1 and COX-2 mRNA/protein expression, as well as the nitrite production in culture medium of vascular smooth muscle cell (VSMC). VSMC was primarily cultured from thoracic aorta of rat. In this experiments, COX-1 and COX-2 mRNA/protein expressions were analysed and nitrite productions were investigated using Griess reagent. VSMC did not express COX-2 protein in basal condition (Non-lipopolysaccharide (LPS) stimulated). In LPS-stimulated experiments, after 3 hours of NO donor pretreatment, LPS 10 microgram/ml was treated for 24 hours. COX-1 protein expressions were unchanged by SNP and NOR-3. NOR-3 significantly increased COX-2 mRNA/protein expression under LPS stimulation. In contrast, SNP did not increase COX-2 mRNA/protein expression under LPS stimulation. Nitrite production was higher in NOR-3 treatment than SNP treatment under LPS stimulation. These results suggest that the expression of COX-2 in VSMC is regulated by NOR-3, COX-2 expressions were depending on the types of NO donor and LPS stimulation in VSMC.


Subject(s)
Animals , Humans , Rats , Aorta, Thoracic , Cyclooxygenase 1 , Cyclooxygenase 2 , Inflammation , Muscle, Smooth, Vascular , Nitric Oxide , Tissue Donors
13.
The Korean Journal of Physiology and Pharmacology ; : 215-221, 2005.
Article in English | WPRIM | ID: wpr-728727

ABSTRACT

We investigated the effects of testosterone and dihydrotestosterone on inflammatory response of iNOS and COX-2 expression in rat vascular smooth muscle cells. Rat vascular smooth muscle cells (VSMC) stimulated with bacterial lipopolysaccharide (LPS; 10microgram/ml) for 24 hours were incubated with increasing amounts of testosterone and dihydrotestosterone (1 and 100 nM). LPS was found to induce inflammatory response of iNOS and COX-2 mRNA and protein in VSMC. These processes were affected by male sex steroid hormones. For 3 hours, however, pretreatment of the cells with 100 nM each of testosterone and dihydrotestosterone suppressed LPS induced iNOS and COX-2 protein expression. RT-PCR analysis revealed that testosterone and dihydrotestosterone did not inhibit mRNA expression of iNOS and COX-2 stimulated by 24 hours of LPS incubation. Proliferation rate was slower in VSMC treated with testosterone and dihydrotestosterone. Testosterone enhanced androgen receptor expression, and LPS significantly reduced androgen receptor protein expression in VSMC. These results indicate that the expression of both iNOS and COX-2 proteins was suppressed by testosterone and dihydrotestosterone in LPS stimulated VSMC and leading to reduction of vascular inflammation.


Subject(s)
Animals , Humans , Male , Rats , Dihydrotestosterone , Gonadal Steroid Hormones , Inflammation , Muscle, Smooth, Vascular , Receptors, Androgen , RNA, Messenger , Testosterone
14.
Journal of the Korean Society for Vascular Surgery ; : 24-29, 2004.
Article in Korean | WPRIM | ID: wpr-48604

ABSTRACT

PURPOSE: iNOS expression in vascular smooth muscle cells (VSMC) causes the development of septic shock, and multiple organ dysfunction syndrome (MODS). For the inhibition of iNOS expression, glucocorticoids are known to inhibit iNOS expression but immunosuppression decreases its clinical availability. Recently, aspirin was reported to inhibit iNOS expression, but the mechanism and effectiveness are still unclear. In this investigation, on aspirin, several non steroidal antiinflammatory drugs (NSAIDs) were applied to clarify the inhibitory mechanism of iNOS expression and NO production in lipopolysaccharide (LPS) treated VSMCs. METHOD: VSMCs were primarily cultured from rat aorta and confirmed by immunocytochemistry of anti-smooth muscle myosin antibody. LPS, an inducer of iNOS, and NSAIDs, such as aspirin, indomethacin, ketoprofen sodium salicylate and acetaminophen were used. The concentrations of nitrite in culture media following the addition of LPS with a 1-hour pretreatment of NSAIDs were measured by spectrophotometry with griess reaction. Western blot and RT-PCR for iNOS protein and iNOS mRNA, respectively, were performed. RESULT: Acetaminophen had no effect on the inhibition of nitrite production. NSAIDs, especially ketoprofen and sodium salicylate, showed a significant inhibitory effect on nitrite production. In their mechanism, all the NSAIDs in present study inhibited iNOS mRNA and protein expression. CONCLUSION: These results suggest that the inhibitory mechanism on iNOS expression of NSAIDs is due to the inhibition of iNOS mRNA expression and subsequent inhibition of iNOS protein expression.


Subject(s)
Animals , Rats , Acetaminophen , Anti-Inflammatory Agents, Non-Steroidal , Aorta , Aspirin , Blotting, Western , Culture Media , Glucocorticoids , Immunohistochemistry , Immunosuppression Therapy , Indomethacin , Ketoprofen , Multiple Organ Failure , Muscle, Smooth, Vascular , Myosins , RNA, Messenger , Shock, Septic , Sodium Salicylate , Spectrophotometry
15.
The Korean Journal of Physiology and Pharmacology ; : 343-351, 2001.
Article in English | WPRIM | ID: wpr-727418

ABSTRACT

This study was undertaken to investigate the mechanism of lipopolysaccharide (LPS) and nitric oxide (NO) as a regulator of vascular smooth muscle cell (VSMC) proliferation. VSMC was primarily cultured from rat aorta and confirmed by the immunocytochemistry with anti-smooth muscle myosin antibody. The number of viable VSMCs were counted, and lactate dehydrogenase (LDH) activity was measured to assess the degree of cell death. Concentrations of nitrite in the culture medium were measured as an indicator of NO production. LPS was introduced into the medium to induce the inducible nitric oxide synthase (iNOS) in VSMC, and Western blot for iNOS protein and RT-PCR for iNOS mRNA were performed to confirm the presence of iNOS. Inhibitors of iNOS and soluble guanylate cyclase (sGC), sodium nitroprusside (SNP) and L-arginine were employed to observe the action of LPS on the iNOS-NO-cGMP signalling pathway. LPS and SNP decreased number of VSMCs and increased the nitrite concentration in the culture medium, but there was no significant change in LDH activity. A cell permeable cGMP derivative, 8-Bromo-cGMP, decreased the number of VSMCs with no significant change in LDH activity. L-arginine, an NO substrate, alone tended to reduce cell count without affecting nitrite concentration or LDH level. Aminoguanidine, an iNOS specific inhibitor, inhibited LPS-induced reduction of cell numbers and reduced the nitrite concentration in the culture medium. LY 83583, a guanylate cyclase inhibitor, suppressed the inhibitory actions of LPS and SNP on VSMC proliferation. LPS increased amounts of iNOS protein and iNOS mRNA in a concentration-dependent manner. These results suggest that LPS inhibits the VSMC proliferation via production of NO by inducing iNOS gene expression. The cGMP which is produced by subsequent activation of guanylate cyclase would be a major mediator in the inhibitory action of iNOS-NO signalling on VSMC proliferation.


Subject(s)
Animals , Rats , Aorta , Arginine , Blotting, Western , Cell Count , Cell Death , Gene Expression , Guanylate Cyclase , Immunohistochemistry , L-Lactate Dehydrogenase , Muscle, Smooth, Vascular , Myosins , Nitric Oxide , Nitric Oxide Synthase Type II , Nitroprusside , RNA, Messenger
16.
The Korean Journal of Physiology and Pharmacology ; : 369-378, 2000.
Article in English | WPRIM | ID: wpr-728140

ABSTRACT

This study was undertaken to investigate an involvement of nitroxergic innervation in gastric smooth muscle of rat. Isometric tension study, the measurement of single cell length, NADPH diaphorase stain of smooth muscle layers and neuronal nitric oxide synthase (nNOS) western blotting were performed. Sodium nitroprusside (SNP), a nitric oxide donor, relaxed the muscle strips precontracted by acetylcholine (ACh) in a concentration-dependent manner. Pretreatment of L-arginine decreased the contraction induced by electric field stimulation (EFS). Pretreatment of NG-nitro-L-arginine methyl ester (L-NAME), a NOS inhibitor, increased the EFS-induced contractions. LY 83583, a guanylate cyclase (GC) inhibitor, reversed the inhibitory actions of L-arginine on the muscle contractions. The effects of L-Arginine, L-NAME and LY 83583 on ACh-induced contractions were not significant. L-arginine reduced the EFS-induced contraction in circular muscle, whereas L-NAME enhanced the EFS-induced contraction in longitudinal strips. By EFS, the phasic contractions appeared approximately 20~25 seconds later. L-NAME significantly shortened the delay time to about 2~3 seconds. In single cell study, ACh contracted gastric smooth muscle cells, SNP relaxed the cells, and the latter also inhibited the ACh-induced contraction. LY 83583 enhanced the ACh-induced contraction and antagonized SNP-induced relaxation. NADPH diaphorase activity was assessed by a histochemistry, nitroblue tetrazolium (NTB) staining. Positive staining was observed in both circular and longitudinal muscle layers. L-arginine increased the staining, while L-NAME decreased the staining. Western blotting for nNOS proved the presence of nNOS in rat gastric smooth muscle. EFS and additional Ca2+ increased nNOS protein expression. These results suggest that in rat stomach, both circular and longitudinal muscle layers are innervated with nitroxergic nerves which relax the gastric smooth muscle via NO-cGMP pathway.


Subject(s)
Animals , Humans , Rats , Acetylcholine , Arginine , Blotting, Western , Guanylate Cyclase , Muscle Contraction , Muscle, Smooth , Myocytes, Smooth Muscle , NADPH Dehydrogenase , NG-Nitroarginine Methyl Ester , Nitric Oxide , Nitric Oxide Synthase Type I , Nitroblue Tetrazolium , Nitroprusside , Relaxation , Stomach , Tissue Donors
17.
The Korean Journal of Physiology and Pharmacology ; : 439-445, 1999.
Article in English | WPRIM | ID: wpr-727851

ABSTRACT

This study is to investigate the mechanism of inhibitory effect of imipramine on the calcium utilization in single cells isolated from canine detrusor. 2 mm thick smooth muscle chops were incubated in 0.12% collagenase solution at 36degreeC, and aerated with 95% O2/5% CO2, and then cell suspension was examined Acetylcholine (ACh) evoked a concentration-dependent contraction of the isolated detrusor cells in normal physiologic salt solution (PSS), and the ACh-induced contraction was significantly inhibited by imipramine. In Ca2+-free PSS, ACh-induced contraction was less than those in normal PSS and it was not affected by the pretreatment with imipramine. Ca2+-induced contraction in Ca2+-free PSS was supressed by imipramine, but addition of A 23187, a calcium ionophore, overcomed the inhibitory effect of imipramine. High potassium-depolarization (40 mM KCl) evoked cell contraction, which was inhibited by imipramine. Caffeine, a releasing agent of the stored Ca2+ from sarcoplasmic reticulum, evoked a contraction of the cells that was not blocked by the pretreatment with imipramine. These results suggest that imipramine inhibits the influx of calcium in the detrusor cells through both the receptor-operated- and voltage-gated-calcium channels, but does not affect the release of calcium from intracellular storage site.


Subject(s)
Acetylcholine , Caffeine , Calcimycin , Calcium , Collagenases , Imipramine , Muscle, Smooth , Sarcoplasmic Reticulum
18.
Yeungnam University Journal of Medicine ; : 318-325, 1999.
Article in Korean | WPRIM | ID: wpr-197100

ABSTRACT

BACKGROUND: Anticholinesterase drug inhibits acetylcholinesterase(AChE), induce accumulation of acetylcholine(ACh) near cholinergic receptors and cholinergic stimulation. This experiment was performed to study the effects of anticholinesterase drugs on gastric motility and the effect of ethanal on anticholinesterase drug-induced motility change. MATERIALS AND METHODS: After excision of stomach, 2x10mm circular musele strips were made, which were then fixed to the isolated muscle chamber. An isometric tension transducer was used to measure the contraction change of the gastric smooth muscle strips after drug addition. RESULTS: Fenthion, and irreversible anticholinesterase drug, increased ACh induced contraction of gastric smooth muscle strips and PAM, a cholinesterase activator, antagnized this action. Physostigmine, a reversible anticholinesterase drug, also increased the ACh induced contraction. The gastric motility was decreased by PAM. Ethanol, which is known to induce smooth muscle relaxation, inhibited the increase of contraction by fenthion. CONCLUSION: These results indicate that irreversible and reversible anticholinesterase drugs increase gastric motility and antagonized by cholinesterase activating drugs. And when exposed to both ethanol and anticholinesterase drug, gastric motility was decreased by the smooth muscle relaxation effect by ethanal.


Subject(s)
Acetaldehyde , Cholinesterase Inhibitors , Cholinesterases , Ethanol , Fenthion , Muscle, Smooth , Physostigmine , Receptors, Cholinergic , Relaxation , Stomach , Transducers
19.
Yeungnam University Journal of Medicine ; : 326-332, 1999.
Article in Korean | WPRIM | ID: wpr-197099

ABSTRACT

BACKGROUND: In korea the agricultural community widely uses organophosphorous, and organophosphorous poisonings are increasing every year. We compared change in activity of acetylcholinesterase and pseudocholinesterase by organophosphorous and by the interaction of ethanol and organophosphorous. We also compared the effect of reversible anticholinesterase drugs, physostigmine and neostigmine. The object of this study is to investigate the effects of several anticholinesterase drugs and on how ethanol influences the activity of cholinesterase. MATERIALS AND METHODS: Fifteen male university students were randomly selected, and blood samples were taken from the antecubital vein. The acetylcholinesterase in the RBC and the pseudocholinesterase in the serum were extracted and separated. The enzyme activity change was measured by the electrometric method. After adding acetylcholine, the pH change was measured with a pH meter. RESULTS AND CONCLUSION: Our results indicated that reversible anticholinesterase drugs decreased the cholinesterase activity more efficiently than organophosphorous. The acetyl cholinesterase and pseudocholinosterase activity were decreased by ethanol. When ethanol was added, oxime a cholinesterase activator, increased acetylcholinesterase activity but dose not increased pseudocholinesterase activity.


Subject(s)
Humans , Male , Acetylcholine , Acetylcholinesterase , Cholinesterase Inhibitors , Cholinesterases , Ethanol , Hydrogen-Ion Concentration , Korea , Neostigmine , Physostigmine , Poisoning , Butyrylcholinesterase , Veins
20.
Korean Journal of Medicine ; : 804-813, 1998.
Article in Korean | WPRIM | ID: wpr-117129

ABSTRACT

No abstract available.


Subject(s)
Thioacetamide
SELECTION OF CITATIONS
SEARCH DETAIL