• Title/Summary/Keyword: Nonselective cation channel

Search Result 29, Processing Time 0.029 seconds

Effects of Dangkwisoo-San, Ginger and Curcumin on Transient Receptor Potential Melastatin 7 Channels (당귀수산, 생강, 커큐민의 대사성 질환과 관련된 일과성 수용체 전압 이온통로조절에 관한 연구)

  • Kim, Byung Joo
    • Journal of Korean Medicine for Obesity Research
    • /
    • v.18 no.1
    • /
    • pp.10-18
    • /
    • 2018
  • Objectives: Metabolic syndrome is correlated with increased cardiovascular risk and characterized by several factors, including visceral obesity, hypertension, insulin resistance, and dyslipidemia. Several members of a large family of nonselective cation entry channels, e.g., transient receptor potential (TRP) melastatin 7 (TRPM7) channels have been associated with the development of cardiovascular diseases. The purpose of this study was to investigate the effects of Dangkwisoo-san, ginger and curcumin on TRPM7 channel. Methods: Human embryonic kidney (HEK) 293 cells stably transfected with the TRPM7 expression vectors were maintained in Dulbecco's modified Eagle's medium (DMEM) supplemented with 10% fetal bovine serum (FBS), 1% penicillin/streptomycin, $5{\mu}g/mL$ blasticidin, and 0.4 mg/mL zeocin in a humidified 20% $O_2$/10% $CO_2$ atmosphere at $37^{\circ}C$. Whole-cell patch clamp recordings were obtained using an Axopatch 700B amplifier and pClamp v.10.4 software, and signals were digitalized at 5 kHz using Digidata 1422A. Results: Dangkwisoo-san extract (100, 200, 300, 400, and $500{\mu}g/mL$) inhibited the outward and inward TRPM7 whole-cell currents at dose dependent manner and the half maximal inhibitory concentration $(IC)_{50}$ of Dangkwisoo-san was $218.3{\mu}g/mL$. Also, ginger extract (100, 200, 300, 400, and $500{\mu}g/mL$) inhibited the outward and inward of TRPM7 whole-cell currents in a dose dependent manner and the $IC_{50}$ of ginger was $877.2{\mu}g/mL$. However, curcumin had no effects on TRPM7 whole-cell currents. Conclusions: These results suggest that both Dangkwisoo-san and ginger have good roles to inhibit the TRPM7 channel, suggesting that Dangkwisoo-san and ginger are considered one of the candidate agents for the treatment of metabolic syndrome such as cardiovascular disease.

Endothelial Ca2+ signaling-dependent vasodilation through transient receptor potential channels

  • Hong, Kwang-Seok;Lee, Man-Gyoon
    • The Korean Journal of Physiology and Pharmacology
    • /
    • v.24 no.4
    • /
    • pp.287-298
    • /
    • 2020
  • Ca2+ signaling of endothelial cells plays a critical role in controlling blood flow and pressure in small arteries and arterioles. As the impairment of endothelial function is closely associated with cardiovascular diseases (e.g., atherosclerosis, stroke, and hypertension), endothelial Ca2+ signaling mechanisms have received substantial attention. Increases in endothelial intracellular Ca2+ concentrations promote the synthesis and release of endothelial-derived hyperpolarizing factors (EDHFs, e.g., nitric oxide, prostacyclin, or K+ efflux) or directly result in endothelial-dependent hyperpolarization (EDH). These physiological alterations modulate vascular contractility and cause marked vasodilation in resistance arteries. Transient receptor potential (TRP) channels are nonselective cation channels that are present in the endothelium, vascular smooth muscle cells, or perivascular/sensory nerves. TRP channels are activated by diverse stimuli and are considered key biological apparatuses for the Ca2+ influx-dependent regulation of vasomotor reactivity in resistance arteries. Ca2+-permeable TRP channels, which are primarily found at spatially restricted microdomains in endothelial cells (e.g., myoendothelial projections), have a large unitary or binary conductance and contribute to EDHFs or EDH-induced vasodilation in concert with the activation of intermediate/small conductance Ca2+-sensitive K+ channels. It is likely that endothelial TRP channel dysfunction is related to the dysregulation of endothelial Ca2+ signaling and in turn gives rise to vascular-related diseases such as hypertension. Thus, investigations on the role of Ca2+ dynamics via TRP channels in endothelial cells are required to further comprehend how vascular tone or perfusion pressure are regulated in normal and pathophysiological conditions.

Overexpression in Arabidopsis of a Plasma Membrane-targeting Glutamate Receptor from Small Radish Increases Glutamate-mediated Ca2+ Influx and Delays Fungal Infection

  • Kang, Seock;Kim, Ho Bang;Lee, Hyoungseok;Choi, Jin Young;Heu, Sunggi;Oh, Chang Jae;Kwon, Soon Il;An, Chung Sun
    • Molecules and Cells
    • /
    • v.21 no.3
    • /
    • pp.418-427
    • /
    • 2006
  • Ionotropic glutamate receptors (iGluRs) are ligand-gated nonselective cation channels that mediate fast excitatory neurotransmission. Although homologues of the iGluRs have been identified in higher plants, their roles are largely unknown. In this work we isolated a full-length cDNA clone (RsGluR) encoding a putative glutamate receptor from small radish. An RsGluR:mGFP fusion protein was localized to the plasma membrane. In Arabidopsis thaliana overexpressing the fulllength cDNA, glutamate treatment triggered greater $Ca^{2+}$ influx in the root cells of transgenic seedlings than in those of the wild type. Transgenic plants exhibited multiple morphological changes such as necrosis at their tips and the margins of developing leaves, dwarf stature with multiple secondary inflorescences, and retarded growth, as previously observed in transgenic Arabidopsis overexpressing AtGluR3.2 [Kim et al. (2001)]. Microarray analysis showed that jasmonic acid (JA)-responsive genes including defensins and JA-biosynthetic genes were up-regulated. RsGluR overexpression also inhibited growth of a necrotic fungal pathogen Botrytis cinerea possibly due to up-regulation of the defensins. Based on these results, we suggest that RsGluR is a glutamate-gated $Ca^{2+}$ channel located in the plasma membrane of higher plants and plays a direct or indirect role in defense against pathogen infection by triggering JA biosynthesis.

Activation of K+ channel by 1-EBIO rescues the head and neck squamous cell carcinoma cells from Ca2+ ionophore-induced cell death

  • Yin, Ming Zhe;Park, Seok-Woo;Kang, Tae Wook;Kim, Kyung Soo;Yoo, Hae Young;Lee, Junho;Hah, J. Hun;Sung, Myung Hun;Kim, Sung Joon
    • The Korean Journal of Physiology and Pharmacology
    • /
    • v.20 no.1
    • /
    • pp.25-33
    • /
    • 2016
  • Ion channels in carcinoma and their roles in cell proliferation are drawing attention. Intracellular $Ca^{2+}$ ($[Ca^{2+}]_i$)-dependent signaling affects the fate of cancer cells. Here we investigate the role of $Ca^{2+}$-activated $K^+$ channel (SK4) in head and neck squamous cell carcinoma cells (HNSCCs) of dif-ferent cell lines; SNU-1076, OSC-19 and HN5. Treatment with $1{\mu}M$ ionomycin induced cell death in all the three cell lines. Whole-cell patch clamp study suggested common expressions of $Ca^{2+}$-activated $Cl^-$ channels (Ano-1) and $Ca^{2+}$-activated nonselective cation channels (CAN). 1-EBIO, an activator of SK4, induced outward $K^+$ current (ISK4) in SNU-1076 and OSC-19. In HN5, ISK4 was not observed or negligible. The 1-EBIO-induced current was abolished by TRAM-34, a selective SK4 blocker. Interestingly, the ionomycin-induced cell death was effectively prevented by 1-EBIO in SNU-1076 and OSC-19, and the rescue effect was annihilated by combined TRAM-34. Con-sistent with the lower level of ISK4, the rescue by 1-EBIO was least effective in HN5. The results newly demonstrate the role of SK4 in the fate of HNSCCs under the $Ca^{2+}$ overloaded condition. Pharmacological modulation of SK4 might provide an intriguing novel tool for the anti-cancer strategy in HNSCC.

Oxidized Low-density Lipoprotein- and Lysophosphatidylcholine-induced $Ca^{2+}$ Mobilization in Human Endothelial Cells

  • Kim, Moon-Young;Liang, Guo-Hua;Kim, Ji-Aee;Choi, Soo-Seung;Choi, Shin-Ku;Suh, Suk-Hyo
    • The Korean Journal of Physiology and Pharmacology
    • /
    • v.13 no.1
    • /
    • pp.27-32
    • /
    • 2009
  • The effects of oxidized low-density lipoprotein(OxLDL) and its major lipid constituent lysophosphatidylcholine(LPC) on $Ca^{2+}$ entry were investigated in cultured human umbilical endothelial cells(HUVECs) using fura-2 fluorescence and patch-clamp methods. OxLDL or LPC increased intracellular $Ca^{2+}$ concentration($[Ca^{2+}]_i$), and the increase of $[Ca^{2+}]_i$ by OxLDL or by LPC was inhibited by $La^{3+}$ or heparin. LPC failed to increase $[Ca^{2+}]_i$ in the presence of an antioxidant tempol. In addition, store-operated $Ca^{2+}$ entry(SOC), which was evoked by intracellular $Ca^{2+}$ store depletion in $Ca^{2+}$-free solution using the sarcoplasmic reticulum $Ca^{2+}$ pump blocker, 2, 5-di-t-butyl-l,4-benzohydroquinone(BHQ), was further enhanced by OxLDL or by LPC. Increased SOC by OxLDL or by LPC was inhibited by U73122. In voltage-clamped cells, OxLDL or LPC increased $[Ca^{2+}]_i$ and simultaneously activated non-selective cation(NSC) currents. LPC-induced NSC currents were inhibited by 2-APB, $La^{3+}$ or U73122, and NSC currents were not activated by LPC in the presence of tempol. Furthermore, in voltage-clamped HUVECs, OxLDL enhanced SOC and evoked outward currents simultaneously. Clamping intracellular $Ca^{2+}$ to 1 ${\mu}M$ activated large-conductance $Ca^{2+}$-activated $K^+(BK_{ca})$ current spontaneously, and this activated $BK_{ca}$ current was further enhanced by OxLDL or by LPC. From these results, we concluded that OxLDL or its main component LPC activates $Ca^{2+}$-permeable $Ca^{2+}$-activated NSC current and $BK_{ca}$ current simultaneously, thereby increasing SOC.

Phosphorylation as a Signal Transduction Pathway Related with N-channel Inactivation in Rat Sympathetic Neurons (N형 칼슘통로 비활성화와 연계된 세포 신호전달 체계로서의 인산화과정)

  • Lim Wonil;Goo Yong Sook
    • Progress in Medical Physics
    • /
    • v.15 no.4
    • /
    • pp.220-227
    • /
    • 2004
  • In N-type $Ca^{2+}$ channels, the mechanism of inactivation - decline of inward current during a depolarizing voltage step- is still controversial between voltage-dependent inactivation and $Ca^{2+}$ -dependent inactivation. In the previous paper we demonstrated that fast component of inactivation of N-type calcium channels does not involve classic $Ca^{2+}$ -dependent mechanism and the slowly inactivating component could result from a $Ca^{2+}$ -dependent process. However, there should be signal transduction pathway which enhances inactivation no matter what the inactivation mechanism is. We have investigated the effect of phosphorylation on calcium channels of rat sympathetic neurons. Intracellular dialysis with the phosphatase inhibitors okadaic acid markedly enhanced the inactivation. The rapidly inactivating component is N-type calcium current, which is blocked by $\omega$-conotoxin GVIA. Staurosporine, a nonselective protein kinase inhibitor, prevented the action of okadaic acid, suggesting that protein phosphorylation is involved. More specifically lavendustin C, inhibitor of CaM kinase II, prevented the action of okadaic acid, suggesting that calmodulin dependent pathway is involved in inactivation process. It is not certain to this point whether phosphorylation process is inactivation itself. Molecular biological research regarding binding site should be followed to address the question of how the divalent cation binding site is related to phoshorylation process.

  • PDF

Effects of Olanzapine on the Pacemaker Potentials of Interstitial Cells of Cajal in Murine Small Intestine (생쥐 소장 카할세포에서 발생하는 향도잡이 기능의 올란자핀 효능에 관한 연구)

  • Kim, Byung Joo
    • Journal of Life Science
    • /
    • v.29 no.9
    • /
    • pp.1010-1015
    • /
    • 2019
  • The interstitial cells of Cajal (ICCs) are the pacemaker cells in the gastrointestinal (GI) tract. In the present study, the effects of olanzapine, an atypical antipsychotic agent, on pacemaker potentials in cultured ICCs from the small intestine of the mouse were investigated. The whole-cell patch-clamp configuration was used to record pacemaker potentials from cultured ICCs. Olanzapine produced pacemaker depolarizations in a concentration-dependent manner in current clamp mode. Methoctramine, a muscarinic $M_2$ receptor antagonist, did not inhibit olanzapine-induced pacemaker depolarizations, whereas 4-diphenylacetoxy-N-methylpiperidine methiodide (4-DAMP) muscarinic $M_3$ receptor antagonist did inhibit it. When guanosine 5'-[${\beta}$-thio] diphosphate (GDP-${\beta}$-S; 1 mM) was in the pipette solution, olanzapine-induced pacemaker depolarization was blocked. Also, low $Na^+$ solution externally eliminated the generation of pacemaker potentials and inhibited the olanzapine-induced pacemaker depolarizations. Additionally, the nonselective cation channel blocker, flufenamic acid, inhibited the olanzapine-induced pacemaker depolarizations. Pretreatment with U-73122, an active phospholipase C (PLC) inhibitor, also eliminated the generation of pacemaker potentials and suppressed the olanzapine-induced pacemaker depolarizations. These results suggested that olanzapine modulates the pacemaker potentials through muscarinic $M_3$ receptor activation by G protein-dependent external $Na^+$ and PLC pathway in the ICCs. Therefore, olanzapine could affect intestinal motility through ICCs.

Effects of Alisma canaliculatum Extract in Pacemaker Potential of Intestinal Interstitial cells of Cajal in mice (생쥐 소장 및 대장 카할세포의 자발적 탈분극에서 택사의 효과에 관한 비교연구)

  • Kwon, Hyo Eun;Park, Dong Suk;Kim, Jeong Nam;Kim, Byung Joo
    • Herbal Formula Science
    • /
    • v.30 no.2
    • /
    • pp.37-44
    • /
    • 2022
  • Objectives : The purpose of this study was to examine the effects of Alisma canaliculatum Extract (ACE) on pacemaker potentials of small and large intestinal interstitial Cells of Cajal (ICC) in mice. Methods : We used enzymatic digestions to dissociate the ICC in the small and large intestine in mice. The whole-cell patch-clamp method was used to record pacemaker potentials in ICC. Results : 1. The ICC generated the pacemaker potentials in small intestine in mice. ACE (0.1-1mg/ml) induced membrane depolarization and decreased frequency with concentration-dependent manners. 2. Pretreatment with a Ca2+ free solution, Na+ 5 mM solution or 2-APB, a nonselective cation channel blocker, stopped the small intestinal ICC pacemaker potentials. In the case of Ca2+-free solution, Na+ 5 mM solution or 2-APB, ACE had no effects on the membrane depolarizations in small intestinal ICC. 3. The ICC generated the pacemaker potentials in large intestine in mice. Membrane depolarization appears regularly in the small intestine, but irregularly in the large intestine. ACE induced membrane depolarization (0.1-1mg/ml) and increased frequency (0.1-0.5mg/ml). 4. Pretreatment with a Ca2+ free solution, Na+ 5 mM solution or 2-APB, stopped the large intestinal ICC pacemaker potentials. In the case of Ca2+-free solution, Na+ 5 mM solution or 2-APB, ACE depolarized the membrane depolarizations in large intestinal ICC. 5. In mice, intestinal transit rate (ITR) values were dose-dependently decreased by the intragastric administration of ACE. Conclusions : These results suggest that ACE can regulate the pacemaker activity of ICC and the reaction by ACE is different from the small and large intestinal ICC, and the control of the intestinal motion by ACE may be caused by many complex processes.

Characterization of Trigeminal Ganglion Neurons Expressing Transient Receptor Potential Ankyrin 1 (TRPA1) in the Rat (흰쥐의 삼차신경절에서 Transient receptor potential ankyrin 1 (TRPA1)의 발현 특성에 관한 연구)

  • Paik, Sang-Kyoo;Na, Yeon-Kyung;Kim, Yun-Sook
    • Applied Microscopy
    • /
    • v.42 no.1
    • /
    • pp.27-33
    • /
    • 2012
  • Transient receptor potential ankyrin 1 (TRPA1), responding to noxious cold (${\leq}17^{\circ}C$) and pungent compounds, is implicated in nociception, but little is known about the coexpression of TRPA1 and other channels or receptors involved in the nociception in craniofacial regions. To address this issue, we characterized the TRPA1-immunopositive (+) neurons in the rat trigeminal ganglion (TG) and investigated their colocalization with other proteins known to be expressed in nociceptive neurons, such as transient receptor potential vanilloid (TRPV1) and $P2X_3$ receptor, using light microscopic immunofluorescence labeling method with TRPA1 and TRPV1 or $P2X_3$ antisera. The majority of TRPA1+ neurons costained for TRPV1 (TRPV1+/TRPA1+; 58.8%, 328/558) and 41.2% only expressed TRPA1 but not TRPV1. The TRPV1+/TRPA1+ neurons were small and medium sized. In addition, we investigated the colocalization of TRPA1 with $P2X_3$, a nonselective cation channel activated by ATP that may be released in the extracellular space as a result of tissue damage and inflammation. Among all TRPA1+ TG neurons, 26.1% (310/1186) costained for $P2X_3$, whereas 73.9% (876/1186) of TRPA1+ neurons did not coexpress $P2X_3$. $P2X_3$+/TRPA1+ neurons were predominantly small and medium sized. These results suggest that TRPA1+ neurons coexpressing TRPV1 or $P2X_3$ are involved in specific roles in the transmission and processing of orofacial nociceptive information by noxious cold, heat, and inflammation.