• Title/Summary/Keyword: ODQ

Search Result 77, Processing Time 0.022 seconds

Nitric Oxide-Induced Apoptosis of Human Dental Pulp Cells Is Mediated by the Mitochondria-Dependent Pathway

  • Park, Min Young;Jeong, Yeon Jin;Kang, Gi Chang;Kim, Mi-Hwa;Kim, Sun Hun;Chung, Hyun-Ju;Jung, Ji Yeon;Kim, Won Jae
    • The Korean Journal of Physiology and Pharmacology
    • /
    • v.18 no.1
    • /
    • pp.25-32
    • /
    • 2014
  • Nitric oxide (NO) is recognized as a mediator and regulator of inflammatory responses. NO is produced by nitric oxide synthase (NOS), and NOS is abundantly expressed in the human dental pulp cells (HDPCs). NO produced by NOS can be cytotoxic at higher concentrations to HDPCs. However, the mechanism by which this cytotoxic pathway is activated in cells exposed to NO is not known. The purpose of this study was to elucidate the NO-induced cytotoxic mechanism in HDPCs. Sodium nitroprusside (SNP), a NO donor, reduced the viability of HDPCs in a dose- and time-dependent manner. We investigated the in vitro effects of nitric oxide on apoptosis of cultured HDPCs. Cells showed typical apoptotic morphology after exposure to SNP. Besides, the number of Annexin V positive cells was increased among the SNP-treated HDPCs. SNP enhanced the production of reactive oxygen species (ROS), and N-acetylcysteine (NAC) ameliorated the decrement of cell viability induced by SNP. However, a soluble guanylate cyclase inhibitor (ODQ) did not inhibited the decrement of cell viability induced by SNP. SNP increased cytochrome c release from the mitochondria to the cytosol and the ratio of Bax/Bcl-2 expression levels. Moreover, SNP-treated HDPCs elevated activities of caspase-3 and caspase-9. While pretreatment with inhibitors of caspase (z-VAD-fmk, z-DEVD-fmk) reversed the NO-induced apoptosis of HDPCs. From these results, it can be suggested that NO induces apoptosis of HDPCs through the mitochondria-dependent pathway mediated by ROS and Bcl-2 family, but not by the cyclic GMP pathway.

Mechanism of L-NAME-Resistant Endothelium-Dependent Relaxation Induced by Acetylcholine in Rabbit Renal Artery

  • Yeon, Dong-Soo;Ahn, Duck-Sun;Lee, Young-Ho;Kwon, Seong-Chun
    • The Korean Journal of Physiology and Pharmacology
    • /
    • v.4 no.6
    • /
    • pp.471-477
    • /
    • 2000
  • In the rabbit renal artery, acetylcholine $(ACh,\;1\;nM{\sim}10\;{\mu}M)$ induced endothelium-dependent relaxation of arterial rings precontracted with norepinephrine $(NE,\;1\;{\mu}M)$ in a dose-dependent manner. $N^G-nitro- L-arginine$ (L-NAME, 0.1 mM), an inhibitor of NO synthase, or ODQ $(1\;{\mu}M),$ a soluble guanylate cyclase inhibitor, partially inhibited the ACh-induced endothelium-dependent relaxation. The ACh-induced relaxation was abolished in the presence of 25 mM KCl and L-NAME. The cytochrome P450 inhibitors, 7- ethoxyresorufin $(7-ER,\;10\;{\mu}M),$ miconazole $(10\;{\mu}M),$ or 17-octadecynoic acid $(17-ODYA,\;10\;{\mu}M),$ failed to inhibit the ACh-induced relaxation in the presence of L-NAME. 11,12-epoxyeicosatrienoic acid $(11,12-EET,\;10\;{\mu}M)$ had no relaxant effect. The ACh-induced relaxation observed in the presence of L-NAME was significantly reduced by a combination of iberiotoxin $(0.3\;{\mu}M)$ and apamin $(1\;{\mu}M),$ and almost completely blocked by 4-aminopyridine (5 mM). The ACh-induced relaxation was antagonized by $P_{2Y}$ receptor antagonist, cibacron blue $(10\;and\;100\;{\mu}M),$ in a dose-dependent manner. Furthermore, 2-methylthio-ATP (2MeSATP), a potent $P_{2Y}$ agonist, induced the endothelium-dependent relaxation, and this relaxation was markedly reduced by either the combination of iberiotoxin and apamin or by cibacron blue. In conclusion, in renal arteries isolated from rabbit, ACh produced non-NO relaxation that is mediated by an EDHF. The results also suggest that ACh may activate the release of ATP from endothelial cells, which in turn activates $P_{2Y}$ receptor on the endothelial cells. Activation of endothelial $P_{2Y}$ receptors induces a release of EDHF resulting in a vasorelaxation via a mechanism that involves activation of both the voltage-gated $K^+$ channels and the $Ca^{2+}-activated\;K^+\;channels$. The results further suggest that EDHF does not appear to be a cytochrome P450 metabolite.

  • PDF

Study on the Mechanism of Vascular Relaxation of Methanol Extract of Rose multiflora Radix (장미근(薔薇根) 메탄올 추출물의 혈관이완 기전에 대한 연구)

  • Kim, Dae-Joong;Jo, Nam-Geun;Lee, Jun-Kyoung;Cao, Li-Hua;Lee, Hyuck;An, Jung-Seok;Um, Jae-Yeon;Joe, Gye-One;Na, Han-Il;Kyung, Eun-Ho;Kang, Dae-Gil;Lee, Ho-Sub
    • Journal of Physiology & Pathology in Korean Medicine
    • /
    • v.21 no.2
    • /
    • pp.408-413
    • /
    • 2007
  • Vascular tone plays an important role in the regulation of blood pressure. In the present study, the methanol extract of Rosae multiflora Radix (MRM) induced dose-dependent relaxation of phenylephrine-precontracted aorta, which was abolished by removal of functional endothelium. Pretreatment of the endothelium-intact aortic tissues with $N^G$-nitro-L-arginine methly ester (L-NAME) or 1H-[1,2,4]-oxadiazole-[4,3-${\alpha}$]-quinoxalin-1-one (ODQ) inhibited the relaxation induced by MRM, respectively. But, the relaxation effect of MRM was not blocked by indomethacine, glibenclamide, tetraethylammonium (TEA), verapamil, diltiazem, atropine, and propranolol, respectively. Moreover, incubation of endothelium-intact aortic rings with MRM increased the production of cGMP. Taken together, the present results suggest that MRM relaxes vascular smooth muscle via endothelium-dependent nitric oxide/cGMP signaling. These results would be useful for further study to MRM on animal models with cardiovascular diseases.

Inhibitory Effects of Naeso-san on Pacemaker Potentials in Interstitial Cells of Cajal of Murine Small Intestine (생쥐 소장 카할세포의 내향성 향도잡이 전압에 미치는 내소산의 억제효과에 관한 연구)

  • Hong, Noo Ri;Ahn, Tae Seok;Park, Hyun Soo;Chae, Han;Kwon, Young Kyu;Kim, Byung Joo
    • Journal of Physiology & Pathology in Korean Medicine
    • /
    • v.28 no.6
    • /
    • pp.630-635
    • /
    • 2014
  • The purpose of this study was to investigate the effects of Naeso-san in interstitial cells of Cajal (ICCs) in murine small intestine. First, we isolated ICCs from murine small intestine. After that, we cultured these cells for 1 days. The patch-clamp technique was applied on ICCs that formed network-like structures in culture (1 days). Spontaneous rhythms were routinely recorded from cultured ICCs under current-clamp conditions, and the ICCs within networks displayed more robust electrical rhythms (pacemaker potentials). To understand the relationship between Naeso-san and pacemaker activity in ICCs, we examined the effects of Naeso-san on pacemaker potentials of ICCs. In current clamp mode (I = 0), the addition of Naeso-san (10 mg/ml - 50 mg/ml) decreased the amplitude and frequency of the pacemaker potentials of ICCs in a dose dependent manner. However, these effects were blocked by intracellular $GDP{\beta}S$, a G-protein inhibitor, and glibenclamide, a specific ATP-sensitive K+ channels blocker. Pretreatment with SQ-22536, an adenylate cyclase inhibitor, did not block the Naeso-san induced effects, whereas pretreatment with ODQ, a guanylate cyclase inhibitor, or L-NAME, an inhibitor of nitric oxide (NO) synthase blocked the Naeso-san induced effects. Our findings provide insight into unraveling the modulation of Naeso-san in pacemaker potentials of ICCs and developing therapeutic agents against gastrointestinal motility disorders.

Pituitary Adenylate Cyclase-activating Polypeptide Inhibits Pacemaker Activity of Colonic Interstitial Cells of Cajal

  • Wu, Mei Jin;Kee, Keun Hong;Na, Jisun;Kim, Seok Won;Bae, Youin;Shin, Dong Hoon;Choi, Seok;Jun, Jae Yeoul;Jeong, Han-Seong;Park, Jong-Seong
    • The Korean Journal of Physiology and Pharmacology
    • /
    • v.19 no.5
    • /
    • pp.435-440
    • /
    • 2015
  • This study aimed to investigate the effect of pituitary adenylate cyclase-activating peptide (PACAP) on the pacemaker activity of interstitial cells of Cajal (ICC) in mouse colon and to identify the underlying mechanisms of PACAP action. Spontaneous pacemaker activity of colonic ICC and the effects of PACAP were studied using electrophysiological recordings. Exogenously applied PACAP induced hyperpolarization of the cell membrane and inhibited pacemaker frequency in a dose-dependent manner (from 0.1 nM to 100 nM). To investigate cyclic AMP (cAMP) involvement in the effects of PACAP on ICC, SQ-22536 (an inhibitor of adenylate cyclase) and cell-permeable 8-bromo-cAMP were used. SQ-22536 decreased the frequency of pacemaker potentials, and cell-permeable 8-bromo-cAMP increased the frequency of pacemaker potentials. The effects of SQ-22536 on pacemaker potential frequency and membrane hyperpolarization were rescued by co-treatment with glibenclamide (an ATP-sensitive $K^+$ channel blocker). However, neither $N^G$-nitro-L-arginine methyl ester (L-NAME, a competitive inhibitor of NO synthase) nor 1H-[1,2,4]oxadiazolo[4,3-${\alpha}$]quinoxalin-1-one (ODQ, an inhibitor of guanylate cyclase) had any effect on PACAP-induced activity. In conclusion, this study describes the effects of PACAP on ICC in the mouse colon. PACAP inhibited the pacemaker activity of ICC by acting through ATP-sensitive $K^+$ channels. These results provide evidence of a physiological role for PACAP in regulating gastrointestinal (GI) motility through the modulation of ICC activity.

Mechanism underlying NO-induced apoptosis in human gingival fibroblasts

  • Hwang, In-Nam;Jeong, Yeon-Jin;Jung, Ji-Yeon;Lee, Jin-Ha;Kim, Kang-Moon;Kim, Won-Jae
    • International Journal of Oral Biology
    • /
    • v.34 no.1
    • /
    • pp.7-14
    • /
    • 2009
  • Nitric oxide (NO) acts as an intracellular messenger at the physiological level but can be cytotoxic at high concentrations. The cells within periodontal tissues, such as gingival and periodontal fibroblasts, contain nitric oxide syntheses and produce high concentrations of NO when exposed to bacterial lipopolysaccharides and cytokines. However, the cellular mechanisms underlying NO-induced cytotoxicity in periodontal tissues are unclear at present. In our current study, we examined the NO-induced cytotoxic mechanisms in human gingival fibroblasts (HGF). Cell viability and the levels of reactive oxygen species (ROS) were determined using a MTT assay and a fluorescent spectrometer, respectively. The morphological changes in the cells were examined by Diff-Quick staining. Expression of the Bcl-2 family and Fas was determined by RT-PCR or western blotting. The activity of caspase-3, -8 and -9 was assessed using a spectrophotometer. Sodium nitroprusside (SNP), a NO donor, decreased the cell viability of the HGF cells in a dose- and time-dependent manner. SNP enhanced the production of ROS, which was ameliorated by NAC, a free radical scavenger. ODQ, a soluble guanylate cyclase inhibitor, did not block the SNP-induced decrease in cell viability. SNP also caused apoptotic morphological changes, including cell shrinkage, chromatin condensation, and DNA fragmentation. The expression of Bax, a member of the proapoptotic Bcl-2 family, was upregulated in the SNP-treated HGF cells, whereas the expression of Bcl-2, a member of the anti-apoptotic Bcl-2 family, was downregulated. SNP augmented the release of cytochrome c from the mitochondria into the cytosol and enhanced the activity of caspase-8, -9, and -3. SNP also upregulated Fas, a component of the death receptor assembly. These results suggest that NO induces apoptosis in human gingival fibroblast via ROS and the Bcl-2 family through both mitochondrial- and death receptor-mediated pathways. Our data also indicate that the cyclic GMP pathway is not involved in NO-induced apoptosis.

Nitric Oxide (NO) Inhibites the Neuronal Activities in the Rat Nucleus Tractus Solitarius

  • Kim, Mi-Won;Park, Mun-Sung;Ryu, Sun-Youl;Jung, Ji-Yeon;Kim, Sun-Hun;Kim, Min-Seok;Kim, Won-Jae;Jeong, Yeon Jin
    • The Korean Journal of Physiology and Pharmacology
    • /
    • v.8 no.1
    • /
    • pp.7-15
    • /
    • 2004
  • Nitric oxide (NO) system has been implicated in a wide range of physiological functions in the nervous system. However, the role of NO in regulating the neural activity in the gustatory zone of nucleus tractus solitarius (NTS) has not been established. The present study was aimed to investigate the role of NO in the gustatory NTS neurons. Sprague-Dawley rats, weighing about 50 g, were used. Whole cell patch recording and immunohistochemistry were done to determine the electrophysiological characteristics of the rostral gustatory nucleus of the tractus solitaries and distribution of NO synthases (NOS). Neuronal NOS (nNOS) immunoreactivity was strongly detected along the solitary tract extending from rostral to caudal medulla. Resting membrane potentials of NTS neurons were $-49.2{\pm}2\;mV$ and action potential amplitudes were $68.5{\pm}2\;mV$ with a mean duration measured at half amplitude of $1.7{\pm}0.3\;ms$. Input resistance, determined from the response to a 150 ms, -100 pA hyperpolarizing current pulse, was $385{\pm}15\;M{\Omega}$, Superfusion of SNAP or SNP, NO donors, produced either hyperpolarization (68%), depolarization (5%), or no effect (27%). The hyperpolarization was mostly accompanied by a decrease in input resistance. The hyperpolarization caused by SNAP or SNP increased the time to initiate the first action potential, and decreased the number of action potentials elicited by current injection. SNP or SNAP also markedly decreased the number of firing neural discharges of the spontaneous NTS neural activity under zero current. Superfusion of L-NAME, a NOS inhibitor, slightly depolarized the membrane potential and increased the firing rate of NTS neurons induced by current injection. ODQ, a soluble guanylate cyclase inhibitor, ameliorated the SNAP-induced changes in membrane potential, input resistance and firing rates. 8-Br-cGMP, a non-degradable cell-permeable cGMP, hyperpolarized the membrane potential and decreased the number of action potentials. It is suggested that NO in the gustatory NTS has an inhibitory role on the neural activity of NTS through activating soluble guanylate cyclase.