26
|
Wong KL, Wu KC, Wu RSC, Chou YH, Cheng TH, Hong HJ. Tetramethylpyrazine inhibits angiotensin II-increased NAD(P)H oxidase activity and subsequent proliferation in rat aortic smooth muscle cells. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2007; 35:1021-1035. [PMID: 18186588 DOI: 10.1142/s0192415x0700548x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] [Imported: 06/09/2025]
Abstract
Tetramethylpyrazine (TMP) is the major component extracted from the Chinese herb, Chuanxiong, which is widely used in China for the treatment of cardiovascular problems. The aims of this study were to examine whether TMP may alter angiotenisn II (Ang II)-induced proliferation and to identify the putative underlying signaling pathways in rat aortic smooth muscle cells. Cultured rat aortic smooth muscle cells were preincubated with TMP and then stimulated with Ang II, [3H]-thymidine incorporation and the ET-1 expression was examined. Ang II increased DNA synthesis which was inhibited by TMP (1-100 microM). TMP inhibited the Ang II-induced ET-1 mRNA levels and ET-1 secretion. TMP also inhibited Ang II-increased NAD(P)H oxidase activity, intracellular reactive oxygen species (ROS) levels, and the ERK phosphorylation. Furthermore, TMP and antioxidants such as Trolox and diphenylene iodonium decreased Ang II-induced ERK phosphorylation, and activator protein-1 reporter activity. In summary, we demonstrate for the first time that TMP inhibits Ang II-induced proliferation and ET-1, partially by interfering with the ERK pathway via attenuation of Ang II-increased NAD(P)H oxidase and ROS generation. Thus, this study delivers important new insight in the molecular pathways that may contribute to the proposed beneficial effects of TMP in cardiovascular disease.
Collapse
MESH Headings
- Angiotensin II/pharmacology
- Animals
- Aorta, Thoracic/drug effects
- Aorta, Thoracic/metabolism
- Aorta, Thoracic/pathology
- Cell Proliferation/drug effects
- Cells, Cultured
- DNA/metabolism
- Endothelin-1/metabolism
- Extracellular Signal-Regulated MAP Kinases/metabolism
- Male
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- NADPH Oxidases/drug effects
- NADPH Oxidases/metabolism
- Pyrazines/pharmacology
- RNA, Messenger/metabolism
- Rats
- Rats, Sprague-Dawley
- Reactive Oxygen Species/metabolism
- Signal Transduction/drug effects
- Transcription Factor AP-1/metabolism
- Vasodilator Agents/pharmacology
Collapse
|
|
18 |
11 |
27
|
Leung YM, Wong KL, Lin CH, Chao CC, Chou CH, Chang LY, Chen SW, Cheng TH, Kuo YH. Dependence of 6beta-acetoxy-7alpha-hydroxyroyleanone block of Kv1.2 channels on C-type inactivation. Cell Mol Life Sci 2010; 67:147-156. [PMID: 19865797 PMCID: PMC11115866 DOI: 10.1007/s00018-009-0178-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2009] [Revised: 09/27/2009] [Accepted: 10/06/2009] [Indexed: 11/30/2022] [Imported: 06/09/2025]
Abstract
Voltage-gated K(+) (Kv) channels exhibit slow or C-type inactivation during continuous depolarization. A selective pharmacological agent targeting C-type inactivation is hitherto lacking. Here, we report that 6beta-acetoxy-7alpha-hydroxyroyleanone (AHR), a diterpenoid compound isolated from Taiwania cryptomerioides, can selectively modify C-type inactivation of Kv1.2 channels. Extracellular, but not intracellular, AHR (50 muM) dramatically accelerated the slow decay of Kv currents and left-shifted the steady-state inactivation curve. AHR blocked Kv currents with an IC(50) of 17.7 muM. AHR did not affect the kinetics and voltage-dependence of Kv1.2 channel activation. Channel block by AHR was independent of intracellular K(+) concentration. In addition, effect of AHR was much attenuated in a Kv1.2 V370G mutant defective in C-type inactivation. Therefore, block of Kv1.2 channels by AHR did not appear to involve direct occlusion of the outer pore but depended on C-type inactivation. AHR could thus be a probe targeting Kv channel C-type inactivation gate.
Collapse
|
research-article |
15 |
11 |
28
|
Leung YM, Wong KL, Cheng KS, Kuo CS, Su TH, Chen YW, Cheng TH. Inhibition of voltage-gated K+ channels and Ca2+ channels by diphenidol. Pharmacol Rep 2012; 64:739-744. [PMID: 22814027 DOI: 10.1016/s1734-1140(12)70869-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2011] [Revised: 01/05/2012] [Indexed: 11/24/2022] [Imported: 06/09/2025]
Abstract
BACKGROUND Although diphenidol has long been deployed as an anti-emetic and anti-vertigo drug, its mechanism of action remains unclear. In particular, little is known as to how diphenidol affects neuronal ion channels. Recently, we showed that diphenidol blocked neuronal voltage-gated Na(+) channels, causing spinal blockade of motor function, proprioception and nociception in rats. In this work, we investigated whether diphenidol could also affect voltage-gated K(+) and Ca(2+) channels. METHODS Electrophysiological experiments were performed to study ion channel activities in two neuronal cell lines, namely, neuroblastoma N2A cells and differentiated NG108-15 cells. RESULTS Diphenidol inhibited voltage-gated K(+) channels and Ca(2+) channels, but did not affect store-operated Ca(2+) channels. CONCLUSION Diphenidol is a non-specific inhibitor of voltage-gated ion channels in neuronal cells.
Collapse
|
|
13 |
11 |
29
|
Tsai TY, Leong IL, Cheng KS, Shiao LR, Su TH, Wong KL, Chan P, Leung YM. Lysophosphatidylcholine-induced cytotoxicity and protection by heparin in mouse brain bEND.3 endothelial cells. Fundam Clin Pharmacol 2018; 33:52-62. [PMID: 29974515 DOI: 10.1111/fcp.12399] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2017] [Revised: 06/28/2018] [Accepted: 07/03/2018] [Indexed: 11/29/2022] [Imported: 06/09/2025]
|
|
7 |
10 |
30
|
Cheng TH, Chen JJ, Chen CH, Wong KL. Effects of propofol on cyclic strain-induced endothelin-1 expression in human umbilical vein endothelial cells. Anesthesiology 2009; 110:74-80. [PMID: 19104173 DOI: 10.1097/aln.0b013e318190b51c] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] [Imported: 06/09/2025]
Abstract
BACKGROUND Propofol is one of the most popular intravenous induction agents of general anesthesia. Experimental results revealed that propofol exerted hypotensive and antioxidative effects. However, the intracellular mechanism of propofol remains to be delineated. The aims of this study were to test the hypothesis that propofol may alter strain-induced endothelin-1 (ET-1) secretion and nitric oxide production, and to identify the putative underlying signaling pathways in human umbilical vein endothelial cells. METHODS Cultured human umbilical vein endothelial cells were exposed to cyclic strain in the presence of propofol, and ET-1 expression was examined by Northern blotting and enzyme-linked immunosorbent assay kit. Activation of extracellular signal-regulated protein kinase, endothelial nitric oxide synthase, and protein kinase B were assessed by Western blot analysis. RESULTS The authors show that propofol inhibits strain-induced ET-1 expression, strain-increased reactive oxygen species formation, and extracellular signal-regulated protein kinase phosphorylation. On the contrary, nitric oxide production, endothelial nitric oxide synthase activity, and protein kinase B phosphorylation were enhanced by propofol treatment. Furthermore, in the presence of PTIO, a nitric oxide scavenger, and KT5823, a specific inhibitor of cyclic guanosine monophosphate-dependent protein kinase, the inhibitory effect of propofol on strain-induced extracellular signal-regulated protein kinase phosphorylation and ET-1 release was reversed. CONCLUSIONS The authors demonstrate for the first time that propofol inhibits strain-induced ET-1 secretion and enhances strain-increased nitric oxide production in human umbilical vein endothelial cells. Thus, this study delivers important new insight into the molecular pathways that may contribute to the proposed hypotensive effects of propofol in the cardiovascular system.
Collapse
|
Comparative Study |
16 |
10 |
31
|
Preventive Treatment with Ketamine Attenuates the Ischaemia-Reperfusion Response in a Chronic Postischaemia Pain Model. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2015; 2015:380403. [PMID: 26161236 PMCID: PMC4487903 DOI: 10.1155/2015/380403] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/18/2014] [Accepted: 12/21/2014] [Indexed: 01/30/2023] [Imported: 06/09/2025]
Abstract
Ischemia and inflammation may be pathophysiological mechanisms of complex regional pain syndrome (CRPS). Ketamine has proposed anti-inflammatory effects and has been used for treating CRPS. This study aimed to evaluate anti-inflammatory and analgesic effects of ketamine after ischaemia-reperfusion injury in a chronic postischaemia pain (CPIP) model of CRPS-I. Using this model, ischemia was induced in the hindlimbs of male Sprague-Dawley rats. Ketamine, methylprednisolone, or saline was administered immediately after reperfusion. Physical effects, (oedema, temperature, and mechanical and cold allodynia) in the bilateral hindpaws, were assessed from 48 hours after reperfusion. Fewer (56%) rats in the ketamine group developed CPIP at the 48th hour after reperfusion (nonsignificant). Ketamine treated rats showed a significantly lower temperature in the ischaemic hindpaw compared to saline (P < 0.01) and methylprednisolone (P < 0.05) groups. Mechanical and cold allodynia were significantly lower in the ischaemic side in the ketamine group (P < 0.05). Proinflammatory cytokines TNF-α and IL-2 were significantly lower at the 48th hour after reperfusion in ketamine and methylprednisolone groups, compared to saline (all P < 0.05). In conclusion, immediate administration of ketamine after an ischaemia-reperfusion injury can alleviate pain and inflammation in the CPIP model and has potential to treat postischaemic pain.
Collapse
|
Journal Article |
10 |
9 |
32
|
Gong CL, Wong KL, Cheng KS, Kuo CS, Chao CC, Tsai MF, Leung YM. Inhibitory effects of magnolol on voltage-gated Na+ and K+ channels of NG108-15 cells. Eur J Pharmacol 2012; 682:73-8. [PMID: 22374258 DOI: 10.1016/j.ejphar.2012.02.013] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2011] [Revised: 01/31/2012] [Accepted: 02/09/2012] [Indexed: 11/19/2022] [Imported: 06/09/2025]
Abstract
Magnolol, a polyphenolic compound isolated from Houpu, a Chinese herb from the bark of Magnolia officinalis, has been reported to have in vitro and in vivo neuroprotective effects. In spite of these reported beneficial effects, studies on the direct impact of magnolol on neuronal ion channels have been scarce. Whether magnolol affects voltage-gated Na(+) channels (VGSC) and voltage-gated K(+) (Kv) channels is unknown. Using the whole-cell voltage-clamp method, we studied the effects of magnolol on voltage-gated ion channels in neuronal NG108-15 cells. Magnolol inhibited VGSC channels with mild state-dependence (IC(50) of 15 and 30 μM, at holding potentials of -70 and -100 mV, respectively). No frequency-dependence was observed in magnolol block. Magnolol caused a left-shift of 18 mV in the steady-state inactivation curve but did not affect the voltage-dependence of activation. Magnolol inhibited Kv channels with an IC(50) of 21 μM, and it caused a 20-mV left-shift in the steady-state inactivation curve without affecting the voltage-dependence of activation. In conclusion, magnolol is an inhibitor of both VGSC and Kv channels and these inhibitory effects may in part contribute to some of the reported neuroprotective effects of magnolol.
Collapse
|
Journal Article |
13 |
8 |
33
|
Tsai CC, Lai TY, Huang WC, Yang T, Liu IM, Wong KL, Chan P, Cheng JT. Tetramethylpyrazine as potassium channel opener to lower calcium influx into cultured aortic smooth muscle cells. PLANTA MEDICA 2003; 69:557-558. [PMID: 12865978 DOI: 10.1055/s-2003-40638] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023] [Imported: 06/09/2025]
Abstract
In the present study, the effect of tetramethylpyrazine (TMP) on calcium (Ca 2+) influx was investigated in cultured vascular smooth muscle (A7r5) cells using Fura-2 as an indicator. The increase of Ca 2+ concentration in A7r5 cells produced by vasopressin or phenylephrine was attenuated by TMP from 0.01 micromol/L to 1 mmol/L. The decrease in the intracellular potassium concentration in A7r5 cells by TMP from 0.01 micromol/L to 10 micromol/L was characterized using PBFI/AM. Inhibitors specific to the small conductance calcium-activated potassium (SKCa ) channel or the ATP-sensitive potassium (K ATP ) channel abolished the actions of TMP. The obtained results indicate that the decrease of Ca 2+ influx into A7r5 cells by TMP is mainly mediated by the opening of potassium channels.
Collapse
|
|
22 |
7 |
34
|
Chao HH, Chen CH, Liu JC, Lin JW, Wong KL, Cheng TH. L-Carnitine attenuates angiotensin II-induced proliferation of cardiac fibroblasts: role of NADPH oxidase inhibition and decreased sphingosine-1-phosphate generation. J Nutr Biochem 2010; 21:580-588. [PMID: 19447019 DOI: 10.1016/j.jnutbio.2009.03.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2008] [Revised: 02/16/2009] [Accepted: 03/09/2009] [Indexed: 11/23/2022] [Imported: 06/09/2025]
Abstract
The heart is unable to synthesize L-carnitine and is strictly dependent on the L-carnitine provided by the blood stream; however, additional studies are needed to better understand the mechanism of L-carnitine supplementation to the heart. The aim of this study was to evaluate the effects of L-carnitine on angiotensin II (Ang II)-induced cardiac fibroblast proliferation and to explore its intracellular mechanism(s). Cultured rat cardiac fibroblasts were pretreated with L-carnitine (1-30 mM) then stimulated with Ang II (100 nM). Ang II increased fibroblast proliferation and endothelin-1 expression, which were partially inhibited by L-carnitine. L-carnitine also attenuated Ang II-induced NADPH oxidase activity, reactive oxygen species formation, extracellular signal-regulated kinase phosphorylation, activator protein-1-mediated reporter activity and sphingosine-1-phosphate generation. In addition, L-carnitine increased prostacyclin (PGI(2)) generation in cardiac fibroblasts. siRNA transfection of PGI(2) synthase significantly reduced L-carnitine-induced PGI(2) and its anti-proliferation effects on cardiac fibroblasts. Furthermore, blockading potential PGI(2) receptors, including immunoprecipitation (IP) receptors and peroxisome proliferator-activated receptors alpha (PPAR alpha) and delta, revealed that siRNA-mediated blockage of PPAR alpha considerably reduced the anti-proliferation effect of L-carnitine. In summary, these results suggest that L-carnitine attenuates Ang II-induced effects (including NADPH oxidase activation, sphingosine-1-phosphate generation and cell proliferation) in part through PGI(2) and PPAR alpha-signaling pathways.
Collapse
|
|
15 |
7 |
35
|
Kang FC, Wang SC, So EC, Chang MM, Wong KL, Cheng KS, Chen YC, Huang BM. Propofol may increase caspase and MAPK pathways, and suppress the Akt pathway to induce apoptosis in MA‑10 mouse Leydig tumor cells. Oncol Rep 2019; 41:3565-3574. [PMID: 31002349 DOI: 10.3892/or.2019.7129] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2018] [Accepted: 04/17/2019] [Indexed: 11/06/2022] [Imported: 06/09/2025] Open
Abstract
In the western world, there is an increasing trend of occurrence in testicular cancer. Treatment of malignant testicular cancer is primarily combined surgery with various chemical drugs. Propofol has been frequently used as an anesthetic and sedative induction agent, which could modulate different γ‑aminobutyric acid receptors in the central nervous system. Studies demonstrated that propofol activates endoplasmic reticulum stress to induce apoptosis in lung cancer. However, it remains elusive whether propofol regulates caspase and/or mitogen‑activated protein kinase (MAPK) pathways to induce apoptosis in Leydig tumor cells. In the present study, MA‑10 mouse Leydig tumor cells were treated with propofol, and possible signal pathways associated with apoptosis were investigated. Results demonstrated that increasing dosage of propofol (300‑600 µM) for 24 h significantly decreased cell viability in MA‑10 cells (P<0.05). In flow cytometry analysis, the amount of sub‑G1 phase cell numbers in MA‑10 cells was significantly increased by propofol (P<0.05). Additionally, Annexin V/propidium iodide double staining further confirmed that propofol could induce MA‑10 cell apoptosis. Furthermore, cleaved caspase‑8, ‑9 and ‑3, and/or poly(ADP‑ribose) polymerase were significantly activated following treatment of propofol in MA‑10 cells. In addition, c‑Jun N‑terminal kinase, extracellular signal‑regulated kinase 1/2, and p38 were significantly activated by propofol in MA‑10 cells (P<0.05), indicating that propofol may induce apoptosis through the MAPK pathway. Additionally, propofol diminished the phosphorylation of Akt to activate apoptosis in MA‑10 cells. In conclusion, propofol may induce MA‑10 cell apoptosis by activating caspase and MAPK pathways, and inhibiting the Akt pathway in MA‑10 cells, demonstrating that propofol may be a potential anticancer agent against Leydig cell cancer.
Collapse
|
Journal Article |
6 |
7 |
36
|
So EC, Lin YX, Tseng CH, Pan BS, Cheng KS, Wong KL, Hao LJ, Wang YK, Huang BM. Midazolam induces apoptosis in MA-10 mouse Leydig tumor cells through caspase activation and the involvement of MAPK signaling pathway. Onco Targets Ther 2014; 7:211-21. [PMID: 24611016 PMCID: PMC3928462 DOI: 10.2147/ott.s56084] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] [Imported: 06/09/2025] Open
Abstract
Purpose The present study aims to investigate how midazolam, a sedative drug for clinical use with cytotoxicity on neuronal and peripheral tissues, induced apoptosis in MA-10 mouse Leydig tumor cells. Methods The apoptotic effect and underlying mechanism of midazolam to MA-10 cells were investigated by flow cytometry assay and Western blotting methods. Results Data showed that midazolam induced the accumulation of the MA-10 cell population in the sub-G1 phase and a reduction in the G2/M phase in a time- and dose-dependent manner, suggesting an apoptotic phenomenon. Midazolam could also induce the activation of caspase-8, -9, and -3 and poly (ADP-ribose) polymerase proteins. There were no changes in the levels of Bax and cytochrome-c, whereas Bid was significantly decreased after midazolam treatment. Moreover, midazolam decreased both pAkt and Akt expression. In addition, midazolam stimulated the phosphorylation of p38 and c-Jun NH2-terminal kinase but not extracellular signal-regulated kinase. Conclusion Midazolam could induce MA-10 cell apoptosis through the activation of caspase cascade, the inhibition of pAkt pathway, and the induction of p38 and c-Jun NH2-terminal kinase pathways.
Collapse
|
|
11 |
7 |
37
|
Kang FC, Wang SC, Chang MM, Pan BS, Wong KL, Cheng KS, So EC, Huang BM. Midazolam activates caspase, MAPKs and endoplasmic reticulum stress pathways, and inhibits cell cycle and Akt pathway, to induce apoptosis in TM3 mouse Leydig progenitor cells. Onco Targets Ther 2018; 11:1475-1490. [PMID: 29588601 PMCID: PMC5858853 DOI: 10.2147/ott.s154442] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] [Imported: 06/09/2025] Open
Abstract
Background Midazolam (MDZ) has powerful hypnosis, amnesia, anti-anxiety and anticonvulsant effects. Studies have shown that prenatally developmental toxicity of diazepam can be observed in many organs/tissues. However, it remains elusive in male reproductive system. Materials and methods TM3 mouse Leydig progenitor cell line was used to determine whether MDZ has any unfavorable effects. Results Midazolam significantly decreased cell viability in dose- and time-dependent manners in TM3 cells. In flow cytometry analysis, midazolam significantly increased subG1 phase cell numbers, and annexin V/PI double staining assay further confirmed that MDZ induced apoptosis in TM3 cells. Moreover, MDZ significantly induced the expression of caspase-8 and -3 proteins and the phosphorylation of JNK, ERK1/2 and p38. Besides, MDZ didn’t activate Akt pathway in TM3 cells. Furthermore, the expressions of p-EIF2α, ATF4, ATF3 and CHOP were induced by midazolam, suggesting that midazolam could induce apoptosis through endoplasmic reticulum (ER) stress in TM3 cells. Additionally, the expressions of cyclin A, cyclin B and CDK1 were inhibited by midazolam through the regulation of p53 in TM3 cells, indicating that midazolam could regulate cell cycle to induce apoptosis. Conclusion Midazolam could activate caspase, MAPKs and ER stress pathways and impede Akt pathway and cell cycle to induce apoptosis in TM3 mouse Leydig progenitor cells.
Collapse
|
Journal Article |
7 |
7 |
38
|
Wong KL, So EC, Chen CC, Wu RSC, Huang BM. Regulation of steroidogenesis by Cordyceps sinensis mycelium extracted fractions with (hCG) treatment in mouse Leydig cells. ACTA ACUST UNITED AC 2007; 53:75-7. [PMID: 17453685 DOI: 10.1080/01485010600915236] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] [Imported: 06/09/2025]
Abstract
The in vitro effect of extracted fractions of Cordyceps sinensis (CS) mycelium on hCG-treated testosterone production from purified normal mouse Leydig cells was examined. Different fractions extracted from CS (F1-water soluble polysaccharide, F2- water soluble protein and F3- poorly water soluble polysaccharide, and protein) were added to Leydig cells with hCG, and the production of testosterone was determined by radioimmunoassay (RIA). Testosterone productions stimulated by hCG in mouse Leydig cells were suppressed by F2 at 10 mg/ml and F3 at doses from 3 to 10 mg/ml, respectively. F2 and F3 at 10 mg/ml did inhibit dbcAMP-stimulated testosterone productions which indicated that F2 and F3 might affect steroidogenesis at the site after the formation of cyclic AMP. Finally, cycloheximide inhibited F2- and F3-treated mouse Leydig cell testosterone production.
Collapse
|
Research Support, Non-U.S. Gov't |
18 |
6 |
39
|
Chen YH, Wong KL, Shieh JP, Chuang YC, Yang YC, So EC. Use of condoms as blade covers during laryngoscopy, a method to reduce possible cross infection among patients. J Infect 2006; 52:118-23. [PMID: 15904960 DOI: 10.1016/j.jinf.2005.03.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2005] [Accepted: 03/03/2005] [Indexed: 10/25/2022] [Imported: 06/09/2025]
Abstract
OBJECTIVES Laryngoscope blades are in close contact with mucous membranes and can possibly contaminated with virulent or readily transmissible organisms. As laryngoscopy is often required during endotracheal intubation, proper cleaning and sterilization of the laryngoscope blade is crucial to prevent cross-contamination among patients. METHODS We tested the effectiveness of latex condom using as a laryngoscope blade cover during endotracheal intubation. Both control (no condom) and study group blades were rinsed with sterile saline after intubation. The rinse was sent for bacteria culture, and appearance of bacterial colonization was counted as positive. A water leak test (WLT) was performed on used condoms to verify their integrity. RESULTS There were total 162 laryngoscopes studied with 83 (51.2%) scopes in the study group and 79 (48.8%) in the control group. Rate of positive bacterial culture were 13.3% and 88.6% in the study and control group, respectively. Although WLT (+) rate of 41% was found in the study group, a high negative culture rate (71.6%) was also noted among the WLT (+) group. CONCLUSIONS Condom when using as a blade cover during laryngoscopy is a simple, inexpansive and effective way in reducing cross contamination among patients.
Collapse
|
|
19 |
6 |
40
|
Leong IL, Tsai TY, Wong KL, Shiao LR, Cheng KS, Chan P, Leung YM. Valproic acid inhibits ATP-triggered Ca 2+ release via a p38-dependent mechanism in bEND.3 endothelial cells. Fundam Clin Pharmacol 2018; 32:499-506. [PMID: 29752814 DOI: 10.1111/fcp.12381] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Revised: 04/22/2018] [Accepted: 05/04/2018] [Indexed: 01/06/2023] [Imported: 06/09/2025]
Abstract
Valproic acid (VA) is currently used to treat epilepsy and bipolar disorder. It has also been demonstrated to promote neuroprotection and neurogenesis. Although beneficial actions of VA on brain blood vessels have also been demonstrated, the effects of VA on brain endothelial cell (EC) Ca2+ signaling are hitherto unreported. In this report, we examined the effects of VA on agonist-triggered Ca2+ signaling in mouse cortical bEND.3 EC. While VA (100 μm) did not cause an acute inhibition of ATP-triggered Ca2+ signaling, a 30-min VA treatment strongly suppressed ATP-triggered intracellular Ca2+ release; however, such treatment did not affect Ca2+ release triggered by cyclopiazonic acid, an inhibitor of SERCA Ca2+ pump, suggesting there was no reduction in Ca2+ store size. VA-activated p38 signaling, and VA-induced inhibition of ATP-triggered Ca2+ release was prevented by SB203580, a p38 inhibitor, suggesting VA caused the inhibition by activating p38. Remarkably, VA treatment did not affect acetylcholine-triggered Ca2+ release, suggesting VA may not inhibit inositol 1,4,5-trisphosphate-induced Ca2+ release per se, and may not act directly on Gq or phospholipase C. Taken together, our results suggest VA treatment, via a p38-dependent mechanism, led to an inhibition of purinergic receptor-effector coupling.
Collapse
|
Journal Article |
7 |
5 |
41
|
A new mask designed for patients implanted with a nasogastric tube. Med Eng Phys 2007; 30:1020-3. [PMID: 18006357 DOI: 10.1016/j.medengphy.2007.09.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2007] [Revised: 09/26/2007] [Accepted: 09/28/2007] [Indexed: 11/20/2022] [Imported: 06/09/2025]
Abstract
Pulmonary ventilation of patients implanted with a nasogastric tube is often difficult for medical personnel, since air leakage through the facemask and nasogastric tube interface is inevitable. Here we designed and tested a special facemask to improve ventilation in these patients. Forty patients with ASA class I-II were randomly assigned to two groups (study and control, n=20 each patients/group). All patients received abdominal surgery with a nasogastric tube inserted before operation. Before induction arterial lines were set up under local anesthesia. Blood gas samples were taken before, during and 1 min after endotracheal intubation. Haemodynamic data were obtained from the artery lines. Inspiratory and expiratory tidal volumes were measured by the Wright's spirometer connected to the anesthesia machine. For induction we used the new mask for the study group and the conventional mask for the controls. Blood gas values and leakage volumes were compared statistically (unpaired t-test, significant levels set at p<0.05). Air leakage was significantly reduced under the new mask with a concomitant improvement in PaO(2). There was however no change in terms of haemodynamic and PaCO(2) between the two groups. Results indicated the advantage of the new mask for patients with nasogastric tubes.
Collapse
|
Journal Article |
18 |
5 |
42
|
Chen CY, Hour MJ, Shiao LR, Wong KL, Leung YM, Chan P, So EC. Quercetin depletes intracellular Ca 2+ stores and blunts ATP-triggered Ca 2+ signaling in bEnd.3 endothelial cells. Fundam Clin Pharmacol 2019; 34:213-221. [PMID: 31618480 DOI: 10.1111/fcp.12514] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Revised: 09/11/2019] [Accepted: 10/14/2019] [Indexed: 12/21/2022] [Imported: 06/09/2025]
Abstract
Quercetin is a flavonol polyphenol widely found in many vegetables, grains, and fruits. Quercetin has been shown to inhibit proliferation and invasion of various glioma cells and is regarded as a potential anticancer agent against glioma. However, whether and how this drug could affect brain blood vessels and endothelial cells (EC) are less understood. Further, there is hitherto no report on how quercetin affects brain EC Ca2+ homeostasis. In this report, we investigated the effects of quercetin on Ca2+ homeostasis in mouse brain bEnd.3 EC. We demonstrated that quercetin raised cytosolic Ca2+ level in a concentration-dependent manner. Quercetin-triggered Ca2+ signal composed of both internal Ca2+ release and extracellular Ca2+ influx. Quercetin caused Ca2+ release from the endoplasmic reticulum, and consistently, inhibition of inositol 1,4,5-trisphosphate receptor (IP3R) by xestospongin C (XeC) suppressed quercetin-triggered Ca2+ release. Quercetin also caused Ca2+ release from lysosomes, an observation in concordance with the inhibition of quercetin-triggered Ca2+ release by trans-Ned-19, a blocker of two-pore channels. As quercetin depleted intracellular Ca2+ storage, it suppressed ATP-induced Ca2+ release and thereby blunted ATP-triggered Ca2+ signaling. In addition, quercetin co-treatment significantly suppressed ATP-stimulated nitric oxide release. Our work therefore showed, for the first time, quercetin perturbed intracellular Ca2+ stores and strongly suppressed ATP-triggered response in bEnd.3 cells.
Collapse
|
Journal Article |
6 |
4 |
43
|
Tsai TY, Lou SL, Wong KL, Wang ML, Su TH, Liu ZM, Yeh LJ, Chan P, Gong CL, Leung YM. Suppression of Ca2+ influx in endotoxin-treated mouse cerebral cortex endothelial bEND.3 cells. Eur J Pharmacol 2015; 755:80-7. [PMID: 25771453 DOI: 10.1016/j.ejphar.2015.03.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2014] [Revised: 03/01/2015] [Accepted: 03/03/2015] [Indexed: 12/17/2022] [Imported: 06/09/2025]
|
|
10 |
4 |
44
|
Chen CY, Lin WC, Wong KL, Cheng KS, Leung YM, Yang SE. Gossypol stimulates opening of a Ca2+
- and Na+
-permeable but Ni2+
- and Co2+
-impermeable pore in bEND.3 endothelial cells. Clin Exp Pharmacol Physiol 2018; 45:788-796. [PMID: 29498086 DOI: 10.1111/1440-1681.12929] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Revised: 02/13/2018] [Accepted: 02/17/2018] [Indexed: 12/29/2022] [Imported: 06/09/2025]
|
|
7 |
4 |
45
|
Wu KC, Wong KL, Wang ML, Shiao LR, Leong IL, Gong CL, Cheng KS, Chan P, Leung YM. Eicosapentaenoic acid triggers Ca 2+ release and Ca 2+ influx in mouse cerebral cortex endothelial bEND.3 cells. J Physiol Sci 2018; 68:33-41. [PMID: 27873157 PMCID: PMC10717322 DOI: 10.1007/s12576-016-0503-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2016] [Accepted: 11/11/2016] [Indexed: 11/29/2022] [Imported: 06/09/2025]
Abstract
Eicosapentaenoic acid (EPA), an omega-3 fatty acid abundant in fish oil, protects endothelial cells (EC) from lipotoxicity and triggers EC NO release. The latter is related to an elevation of cytosolic Ca2+. Although EPA has been shown to cause human EC cytosolic Ca2+ elevation, the mechanism is unclear. Microfluorimetric imaging was used here to measure free cytosolic Ca2+ concentration. EPA was shown to cause intracellular Ca2+ release in mouse cerebral cortex endothelial bEND.3 cells; interestingly, the EPA-sensitive intracellular Ca2+ pool(s) appeared to encompass and was larger than the Ca2+ pool mobilized by sarcoplasmic-endoplasmic reticulum Ca2+-ATPase inhibition by cyclopiazonic acid. EPA also opened a Ca2+ influx pathway pharmacologically distinct from store-operated Ca2+ influx. Surprisingly, EPA-triggered Ca2+ influx was Ni2+-insensitive; and EPA did not trigger Mn2+ influx. Further, EPA-triggered Ca2+ influx did not involve Na+-Ca2+ exchangers. Thus, our results suggest EPA triggered unusual mechanisms of Ca2+ release and Ca2+ influx in EC.
Collapse
|
research-article |
7 |
4 |
46
|
Tsai TY, Leong IL, Shiao LR, Wong KL, Shao L, Chan P, Leung YM. Tannic acid, a vasodilator present in wines and beverages, stimulates Ca2+ influx via TRP channels in bEND.3 endothelial cells. Biochem Biophys Res Commun 2020; 526:117-121. [PMID: 32197839 DOI: 10.1016/j.bbrc.2020.03.078] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2020] [Accepted: 03/13/2020] [Indexed: 10/24/2022] [Imported: 06/09/2025]
|
|
5 |
4 |
47
|
Chow LWC, Wong KL, Shiao LR, Wu KC, Leung YM. Polyamine stimulation perturbs intracellular Ca2+ homeostasis and decreases viability of breast cancer BT474 cells. ACTA ACUST UNITED AC 2020; 75:65-73. [PMID: 32092040 DOI: 10.1515/znc-2019-0119] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2019] [Accepted: 01/21/2020] [Indexed: 11/15/2022] [Imported: 06/09/2025]
Abstract
Intracellular polyamines such as spermine and spermidine are essential to cell growth in normal and especially in cancer cells. However, whether extracellular polyamines affect cancer cell survival is unknown. We therefore examined the actions of extracellular polyamines on breast cancer BT474 cells. Our data showed that spermine, spermidine, and putrescine decreased cell viability by apoptosis. These polyamines also elicited Ca2+ signals, but the latter were unlikely triggered via Ca2+-sensing receptor (CaSR) as BT474 cells have been demonstrated previously to lack CaSR expression. Spermine-elicited Ca2+ response composed of both Ca2+ release and Ca2+ influx. Spermine caused a complete discharge of the cyclopiazonic acid (CPA)-sensitive Ca2+ pool and, expectedly, endoplasmic reticulum (ER) stress. The Ca2+ influx pore opened by spermine was Mn2+-impermeable, distinct from the CPA-triggered store-operated Ca2+ channel, which was Mn2+-permeable. Spermine cytotoxic effects were not due to oxidative stress, as spermine did not trigger reactive oxygen species formation. Our results therefore suggest that spermine acted on a putative polyamine receptor in BT474 cells, causing cytotoxicity by Ca2+ overload, Ca2+ store depletion, and ER stress.
Collapse
|
|
5 |
3 |
48
|
Tsai TY, Chan P, Gong CL, Wong KL, Su TH, Shen PC, Leung YM, Liu ZM. Parthenolide-Induced Cytotoxicity in H9c2 Cardiomyoblasts Involves Oxidative Stress. ACTA CARDIOLOGICA SINICA 2016; 31:33-41. [PMID: 27122844 DOI: 10.6515/acs20140422b] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] [Imported: 06/09/2025]
Abstract
BACKGROUND Cardiac cellular injury as a consequence of ischemia and reperfusion involves nuclear factor-κB (NF-κ B), amongst other factors, and NF-κ B inhibitors could substantially reduce myocardial infarct size. Parthenolide, a sesquiterpene lactone compound which could inhibit NF-κ B, has been shown to ameliorate myocardial reperfusion injury but may also produce toxic effects in cardiomyocytes at high concentrations. The aim of this study was to examine the cytotoxic effects of this drug on H9c2 cardiomyoblasts, which are precursor cells of cardiomyocytes. METHODS Cell viability and apoptosis were examined by MTT and TUNEL assay, respectively, and protein expression was analyzed by western blot. Reactive oxygen species (ROS) production was measured using DCFH-DA as dye. Cytosolic Ca(2+) concentration and mitochondrial membrane potential were measured microfluorimetrically using, respectively, fura 2 and rhodamine 123 as dyes. RESULTS Parthenolide caused apoptosis at 30 μ M, as judged by TUNEL assay and Bax and cytochrome c translocation. It also caused collapse of mitochondrial membrane potential and endoplasmic reticulum stress. Parthenolide triggered ROS formation, and vitamin C (antioxidant) partially alleviated parthenolide-induced cell death. CONCLUSIONS The results suggested that parthenolide at high concentrations caused cytotoxicity in cardiomyoblasts in part by inducing oxidative stress, and demonstrated the imperative for cautious and appropriate use of this agent in cardioprotection. KEY WORDS Cardiomyoblast; Endoplasmic reticulum stress; Oxidative stress; Parthenolide; Reperfusion injury.
Collapse
|
Journal Article |
9 |
3 |
49
|
Antagonism of Ca 2+-sensing receptors by NPS 2143 is transiently masked by p38 activation in mouse brain bEND.3 endothelial cells. Naunyn Schmiedebergs Arch Pharmacol 2019; 392:823-832. [PMID: 30826858 DOI: 10.1007/s00210-019-01637-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Accepted: 02/22/2019] [Indexed: 10/27/2022] [Imported: 06/09/2025]
Abstract
Ca2+-sensing receptors (CaSR) are G protein-coupled receptors which are activated by a rise in extracellular Ca2+. CaSR activation has been known to inhibit parathyroid hormone release and stimulate calcitonin release from parathyroid glands and thyroid parafollicular C cells, respectively. The roles of CaSR in other cell types including endothelial cells (EC) are much less understood. In this work, we demonstrated protein and functional expression of CaSR in mouse cerebral EC (bEND.3). Unexpectedly, CaSR response (high Ca2+-elicited cytosolic [Ca2+] elevation) was unaffected by edelfosine or U73122 but strongly suppressed by SK&F 96365, ruthenium red, and 2-aminoethoxydiphenyl borate (2-APB), suggesting involvement of TRPV and TRPC channels but not Gq-phospholipase C. Acute application of NPS2143, a negative allosteric modulator of CaSR, suppressed CaSR response. However, a 40-min NPS2143 pre-treatment surprisingly enhanced CaSR response. After 4-24 h of application, this enhancement faded away and suppression of CaSR response was observed again. Similar results were obtained when La3+ and Sr2+ were used as CaSR agonists. The transient NPS 2143 enhancement effect was abolished by SB203580, a p38 inhibitor. Consistently, NPS 2143 triggered a transient p38 activation. Taken together, results suggest that in bEND.3 cells, NPS 2143 caused acute suppression of CaSR response, but then elicited a transient enhancement of CaSR response in a p38-dependent manner. NPS 2143 effects on CaSR in bEND.3 cells therefore depended on drug exposure time. These findings warrant cautious use of this agent as a CaSR modulator and potential cardiovascular drug.
Collapse
|
Research Support, Non-U.S. Gov't |
6 |
3 |
50
|
Tsai KF, Shen CJ, Cheung CW, Wang TL, Chow LWC, Leung YM, Wong KL. Lipotoxicity in human lung alveolar type 2 A549 cells: Mechanisms and protection by tannic acid. CHINESE J PHYSIOL 2021; 64:289-297. [PMID: 34975122 DOI: 10.4103/cjp.cjp_68_21] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] [Imported: 06/09/2025] Open
Abstract
Palmitic acid (PA) is a saturated free fatty acid which, when being excessive, accounts for lipotoxicity. Using human lung A549 cells as a model for lung alveolar type 2 epithelial cells, we found that challenge of A549 cells with PA resulted in apoptotic cell death, as reflected by positive annexin V and PI staining, and also appearance of cleaved caspase-3. PA treatment also caused depletion of intracellular Ca2+ store, endoplasmic reticulum (ER) stress, and oxidative stress. Tannic acid (TA), a polyphenol present in wines and many beverages, alleviated PA-induced ER stress, oxidative stress and apoptotic death. Thus, our results suggest PA lipotoxicity in lung alveolar type 2 epithelial cells could be protected by TA.
Collapse
|
|
4 |
2 |