• 제목/요약/키워드: Non-selective cation channel

검색결과 21건 처리시간 0.029초

Ononis spinosa alleviated capsaicin-induced mechanical allodynia in a rat model through transient receptor potential vanilloid 1 modulation

  • Jaffal, Sahar Majdi;Al-Najjar, Belal Omar;Abbas, Manal Ahmad
    • The Korean Journal of Pain
    • /
    • 제34권3호
    • /
    • pp.262-270
    • /
    • 2021
  • Background: Transient receptor potential vanilloid 1 (TRPV1) is a non-selective cation channel implicated in pain sensation in response to heat, protons, and capsaicin (CAPS). It is well established that TRPV1 is involved in mechanical allodynia. This study investigates the effect of Ononis spinosa (Fabaceae) in CAPS-induced mechanical allodynia and its mechanism of action. Methods: Mechanical allodynia was induced by the intraplantar (ipl) injection of 40 ㎍ CAPS into the left hind paw of male Wistar rats. Animals received an ipl injection of 100 ㎍ O. spinosa methanolic leaf extract or 2.5% diclofenac sodium 20 minutes before CAPS injection. Paw withdrawal threshold (PWT) was measured using von Frey filament 30, 90, and 150 minutes after CAPS injection. A molecular docking tool, AutoDock 4.2, was used to study the binding energies and intermolecular interactions between O. spinosa constituents and TRPV1 receptor. Results: The ipsilateral ipl injection of O. spinosa before CAPS injection increased PWT in rats at all time points. O. spinosa decreased mechanical allodynia by 5.35-fold compared to a 3.59-fold decrease produced by diclofenac sodium. The ipsilateral pretreatment with TRPV1 antagonist (300 ㎍ 4-[3-Chloro-2-pyridinyl]-N-[4-[1,1-dimethylethyl] phenyl]-1-piperazinecarboxamide [BCTC]) as well as the β2-adrenoreceptor antagonist (150 ㎍ butoxamine) attenuated the action of O. spinosa. Depending on molecular docking results, the activity of the extract could be attributed to the bindings of campesterol, stigmasterol, and ononin compounds to TRPV1. Conclusions: O. spinosa alleviated CAPS-induced mechanical allodynia through 2 mechanisms: the direct modulation of TRPV1 and the involvement of β2 adrenoreceptor signaling.

TRPM7 Is Essential for RANKL-Induced Osteoclastogenesis

  • Yang, Yu-Mi;Jung, Hwi-Hoon;Lee, Sung Jun;Choi, Hyung-Jun;Kim, Min Seuk;Shin, Dong Min
    • The Korean Journal of Physiology and Pharmacology
    • /
    • 제17권1호
    • /
    • pp.65-71
    • /
    • 2013
  • The transient receptor potential melastatin type 7 (TRPM7) channel is a widely expressed non-selective cation channel with fusion to the C-terminal alpha kinase domain and regarded as a key regulator of whole body $Mg^{2+}$ homeostasis in mammals. However, the roles of TRPM7 during osteoclastogenesis in RAW264.7 cells and bone marrow-derived monocyte/macrophage precursor cells (BMMs) are not clear. In the present study, we investigate the roles of TRPM7 in osteoclastogenesis using methods of small interfering RNA (siRNA), RT-PCR, patch-clamp, and calcium imaging. RANKL (receptor activator of NF-${\kappa}B$ ligand) stimulation did not affect the TRPM7 expression and TRPM7-mediated current was activated in HEK293, RAW264.7, and BMM cells by the regulation of $Mg^{2+}$. Knock-down of TRPM7 by siTRPM7 reduced intracellular $Ca^{2+}$ concentration ($[Ca^{2+}]_i$) increases by 0 mM $[Mg^{2+}]_e$ in HEK293 cells and inhibited the generation of RANKL-induced $Ca^{2+}$ oscillations in RAW264.7 cells. Finally, knock-down of TRPM7 suppressed RANKL-mediated osteoclastogenesis such as activation and translocation of NFATc1, formation of multinucleated cells, and the bone resorptive activity, sequentially. These results suggest that TRPM7 plays an essential role in the RANKL-induced $[Ca^{2+}]_i$ oscillations that triggers the late stages of osteoclastogenesis.

Functional Expression of TRPV 4 Cation Channels in Human Mast Cell Line (HMC-1)

  • Kim, Kyung-Soo;Shin, Dong-Hoon;Nam, Joo-Hyun;Park, Kyung-Sun;Zhang, Yin-Hua;Kim, Woo-Kyung;Kim, Sung-Joon
    • The Korean Journal of Physiology and Pharmacology
    • /
    • 제14권6호
    • /
    • pp.419-425
    • /
    • 2010
  • Mast cells are activated by specific allergens and also by various nonspecific stimuli, which might induce physical urticaria. This study investigated the functional expression of temperature sensitive transient receptor potential vanilloid (TRPV) subfamily in the human mast cell line (HMC-1) using whole-cell patch clamp techniques. The temperature of perfusate was raised from room temperature (RT, $23{\sim}25^{\circ}C$) to a moderately high temperature (MHT, $37{\sim}39^{\circ}C$) to activate TRPV3/4, a high temperature (HT, $44{\sim}46^{\circ}C$) to activate TRPV1, or a very high temperature (VHT, $53{\sim}55^{\circ}C$) to activate TRPV2. The membrane conductance of HMC-1 was increased by MHT and HT in about 50% (21 of 40) of the tested cells, and the I/V curves showed weak outward rectification. VHT-induced current was 10-fold larger than those induced by MHT and HT. The application of the TRPV 4 activator $3{\alpha}$-phorbol 12,13-didecanoate ($4{\alpha}$ PDD, $1\;{\mu}M$) induced weakly outward rectifying currents similar to those induced by MHT. However, the TRPV3 agonist camphor or TRPV1 agonist capsaicin had no effect. RT-PCR analysis of HMC-1 demonstrated the expression of TRPV4 as well as potent expression of TRPV2. The $[Ca^{2+}]_c$ of HMC-1 cells was also increased by MHT or by $4{\alpha}$ PDD. In summary, our present study indicates that HMC-1 cells express $Ca^{2+}$-permeable TRPV4 channels in addition to the previously reported expression of TRPV2 with a higher threshold of activating temperature.

Activation of transient receptor potential vanilloid 3 by the methanolic extract of Schisandra chinensis fruit and its chemical constituent γ-schisandrin

  • Nam, Yuran;Kim, Hyun Jong;Kim, Young-Mi;Chin, Young-Won;Kim, Yung Kyu;Bae, Hyo Sang;Nam, Joo Hyun;Kim, Woo Kyung
    • The Korean Journal of Physiology and Pharmacology
    • /
    • 제21권3호
    • /
    • pp.309-316
    • /
    • 2017
  • Transient receptor potential vanilloid 3 (TRPV3) is a non-selective cation channel with modest permeability to calcium ions. It is involved in intracellular calcium signaling and is therefore important in processes such as thermal sensation, skin barrier formation, and wound healing. TRPV3 was initially proposed as a warm temperature sensor. It is activated by synthetic small-molecule chemicals and plant-derived natural compounds such as camphor and eugenol. Schisandra chinensis (Turcz.) Baill (SC) has diverse pharmacological properties including antiallergic, anti-inflammatory, and wound healing activities. It is extensively used as an oriental herbal medicine for the treatment of various diseases. In this study, we investigated whether SC fruit extracts and seed oil, as well as four compounds isolated from the fruit can activate the TRPV3 channel. By performing whole-cell patch clamp recording in HEK293T cells overexpressing TRPV3, we found that the methanolic extract of SC fruit has an agonistic effect on the TRPV3 channel. Furthermore, electrophysiological analysis revealed that ${\gamma}$-schisandrin, one of the isolated compounds, activated TRPV3 at a concentration of $30{\mu}M$. In addition, ${\gamma}$-schisandrin (${\sim}100{\mu}M$) increased cytoplasmic $Ca^{2+}$ concentrations by approximately 20% in response to TRPV3 activation. This is the first report to indicate that SC extract and ${\gamma}$-schisandrin can modulate the TRPV3 channel. This report also suggests a mechanism by which ${\gamma}$-schisandrin acts as a therapeutic agent against TRPV3-related diseases.

The Mechanism of t-Butylhydroperoxide-Induced Apoptosis in IMR-32 Human Neuroblastoma Cells

  • Kim, Jung-Ae;Lee, Yong-Soo;Huh, Keun
    • The Korean Journal of Physiology and Pharmacology
    • /
    • 제3권1호
    • /
    • pp.19-27
    • /
    • 1999
  • Apoptosis has been implicated in the pathophysiological mechanisms of various neurodegenerative diseases. In a variety of cell types, oxidative stress has been demonstrated to play an important role in the apoptotic cell death. However, the exact mechanism of oxidative stress-induced apoptosis in neuronal cells is not known. In this study, we induced oxidative stress in IMR-32 human neuroblastoma cells with tert- butylhydroperoxide (TBHP), which was confirmed by significantly reduced glutathione content and glutathione reductase activity, and increased glutathione peroxidase activity. TBHP induced decrease in cell viability and increase in DNA fragmentation, a hallmark of apoptosis, in a dose-dependent manner. TBHP also induced a sustained increase in intracellular $Ca^{2+}$ concentration, which was completely prevented either by EGTA, an extracellular $Ca^{2+}$ chelator or by flufenamic acid (FA), a non-selective cation channel (NSCC) blocker. These results indicate that the TBHP-induced intracellular $Ca^{2+}$ increase may be due to $Ca^{2+}$ influx through the activation of NSCCs. In addition, treatment with either an intracellular $Ca^{2+}$ chelator (BAPTA/AM) or FA significantly suppressed the TBHP-induced apoptosis. Moreover, TBHP increased the expression of p53 gene but decreased c-myc gene expression. Taken together, these results suggest that the oxidative stress-induced apoptosis in neuronal cells may be mediated through the activation of intracellular $Ca^{2+}$ signals and altered expression of p53 and c-myc.

  • PDF

Onion peel extract and its constituent, quercetin inhibits human Slo3 in a pH and calcium dependent manner

  • Wijerathne, Tharaka Darshana;Kim, Ji Hyun;Kim, Min Ji;Kim, Chul Young;Chae, Mee Ree;Lee, Sung Won;Lee, Kyu Pil
    • The Korean Journal of Physiology and Pharmacology
    • /
    • 제23권5호
    • /
    • pp.381-392
    • /
    • 2019
  • Sperm function and male fertility are closely related to pH dependent $K^+$ current (KSper) in human sperm, which is most likely composed of Slo3 and its auxiliary subunit leucine-rich repeat-containing protein 52 (LRRC52). Onion peel extract (OPE) and its major active ingredient quercetin are widely used as fertility enhancers; however, the effect of OPE and quercetin on Slo3 has not been elucidated. The purpose of this study is to investigate the effect of quercetin on human Slo3 channels. Human Slo3 and LRRC52 were co-transfected into HEK293 cells and pharmacological properties were studied with the whole cell patch clamp technique. We successfully expressed and measured pH sensitive and calcium insensitive Slo3 currents in HEK293 cells. We found that OPE and its key ingredient quercetin inhibit Slo3 currents. Inhibition by quercetin is dose dependent and this degree of inhibition decreases with elevating internal alkalization and internal free calcium concentrations. Functional moieties in the quercetin polyphenolic ring govern the degree of inhibition of Slo3 by quercetin, and the composition of such functional moieties are sensitive to the pH of the medium. These results suggest that quercetin inhibits Slo3 in a pH and calcium dependent manner. Therefore, we surmise that quercetin induced depolarization in spermatozoa may enhance the voltage gated proton channel (Hv1), and activate non-selective cation channels of sperm (CatSper) dependent calcium influx to trigger sperm capacitation and acrosome reaction.

바닐로이드 수용체 TRPV1의 막수송과정에서의 Rab11의 역할 (Role of Rab11 on Membrane Trafficking of Rat Vanilloid Receptor, TRPV1)

  • 엄기범;이순열
    • 한국산학기술학회논문지
    • /
    • 제12권7호
    • /
    • pp.3096-3102
    • /
    • 2011
  • 바닐로이드 수용체 TRPV1(transient receptor potential vanilloid 1)은 캡사이신, pH, 열 등의 통증 유발물질에 의해 활성화되는 비특이적 양이온 채널로서 통증발현에 핵심적인 막 단백질이다. TRPV1의 막 수송에 관한 연구가 미미한 가운데 FIP3(family of Rab11 interacting protein 3)가 TRPV1 채널과 결합하여 막수송에 관여한다고 보고되었다. FIP3는 Rab11과 결합하는 단백질인데 최근 Rab11 단백질이 여러 채널 단백질의 막수송에 직접적으로 또는 간접적으로 중요하다고 보고되었다. 그러므로 본 연구에서는 Rab11이 TRPV1의 막 수송에서의 역할을 알아보기 위하여 세포 생물학적, 생화학적으로 알아보았다. 공촛점 현미경을 통하여 확인한 결과 Rab11은 실제로 세포내에서 TRPV1과 동일한 위치에서 발현되어 있음을 확인하였다. 하지만 생화학적인 방법인 GST-pulldown을 실시하였을 때 TRPV1과 Rab11간에는 서로 직접적인 결합은 하지 않는 것으로 나타났다. 비록 직접적인 결합은 하지 않지만 Rab11이 TRPV1의 막 수송에 관여한다고 가정하고 Rab11의 TRPV1의 막수송에서의 역할을 더 자세히 알아보기 위하여 세포내 Rab11a의 발현을 siRNA를 사용하여 Rab11a의 발현을 50%수준으로 저해한 후 TRPV1의 세포막으로의 이동을 알아본 결과 Rab11 발현 저해 시 세포막에 이동된 TRPV1이 현저히 감소함을 확인할 수 있었다. 이 결과로부터 Rab11이 아마도 FIP3을 포함하는 방법으로 TRPV1의 막 수송에 영향을 주는 것으로 결론지을 수 있다.

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
    • /
    • 제13권1호
    • /
    • 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.

Involvement of Intracellular Ca2+-and PI3K-Dependent ERK Activation in TCDD-Induced Inhibition of Cell Proliferation in SK-N-SH Human Neuronal Cells

  • Yang, Seun-Ah;Lee, Yong-Soo;Jin, Da-Qing;Jung, Jae-Wook;Park, Byung-Chul;Lee, Yoon-Seok;Paek, Seung-Hwan;Jeong, Tae-Cheon;Choi, Han-Gon;Yong, Chul-Soon;Yoo, Bong-Kyu;Kim, Jung-Ae
    • Biomolecules & Therapeutics
    • /
    • 제13권2호
    • /
    • pp.78-83
    • /
    • 2005
  • 2,3,7,8-Tetrachlorodibenzo-p-dioxin(TCDD) has previously shown to induce neurotoxicity through intracellular $Ca^{2+}$ increase in rat neurons. In this study we investigated the role and signaling pathway of intracellular $Ca^{2+}$ in TCDD-induced inhibition of neuronal cell proliferation in SK-N-SH human neuronal cells. We found that TCDD(10nM) rapidly increased the level of intracellular $Ca^{2+}$, which was completely blocked by the extracellular $Ca^{2+}$ chelation with EGTA (1 mM) or by pretreatment of the cells with the non-selective cation channel blocker. flufenamic acid (200 ${\mu}M$). However, pretreatment of the cells with dantrolene (25 ${\mu}M$) and TMB-8(10 ${\mu}M$), intracellular $Ca^{2+}$-release blockers, or a voltage-sensitive $Ca^{2+}$ channel blocker, varapamil (100 ${\mu}M$), failed to block the TCDD-induced $Ca^{2+}$ increase in the cells. In addition, TCDD induced a rapid and transient activation of phatidvlinositol 3-kinase (PI3K) and extracellular signal-regulated kinase 1/2(ERK1/2), which was ingnificantly blocked by the pretreatment with BAPTA, an intracellular $Ca^{2+}$ chelator, and LY294002, a PI3K inhibitor. Furthermore, inhibitors of PI3K, ERK, or an intracellular $Ca^{2+}$ chelator further potentiated the anti-proliferative effect of TCDD in the cells. Collectively, the results suggest that intracellular $Ca^{2+}$ and PI3K-dependent activation of ERK 1/2 may be involved in the TCDD-induced inhibition of cell proliferation in SK-N-SH human neuronal cells.

A Cannabinoid Receptor Agonist N-Arachidonoyl Dopamine Inhibits Adipocyte Differentiation in Human Mesenchymal Stem Cells

  • Ahn, Seyeon;Yi, Sodam;Seo, Won Jong;Lee, Myeong Jung;Song, Young Keun;Baek, Seung Yong;Yu, Jinha;Hong, Soo Hyun;Lee, Jinyoung;Shin, Dong Wook;Jeong, Lak Shin;Noh, Minsoo
    • Biomolecules & Therapeutics
    • /
    • 제23권3호
    • /
    • pp.218-224
    • /
    • 2015
  • Endocannabinoids can affect multiple cellular targets, such as cannabinoid (CB) receptors, transient receptor potential cation channel, subfamily V, member 1 (TRPV1) and peroxisome proliferator-activated receptor ${\gamma}$($PPAR{\gamma}$). The stimuli to induce adipocyte differentiation in hBM-MSCs increase the gene transcription of the $CB_1$ receptor, TRPV1 and $PPAR{\gamma}$. In this study, the effects of three endocannabinoids, N-arachidonoyl ethanolamine (AEA), N-arachidonoyl dopamine (NADA) and 2-arachidonoyl glycerol (2-AG), on adipogenesis in hBM-MSCs were evaluated. The adipocyte differentiation was promoted by AEA whereas inhibited by NADA. No change was observed by the treatment of non-cytotoxic concentrations of 2-AG. The difference between AEA and NADA in the regulation of adipogenesis is associated with their effects on $PPAR{\gamma}$ transactivation. AEA can directly activate $PPAR{\gamma}$. The effect of AEA on $PPAR{\gamma}$ in hBM-MSCs may prevail over that on the $CB_1$ receptor mediated signal transduction, giving rise to the AEA-induced promotion of adipogenesis. In contrast, NADA had no effect on the $PPAR{\gamma}$ activity in the $PPAR{\gamma}$ transactivation assay. The inhibitory effect of NADA on adipogenesis in hBM-MSCs was reversed not by capsazepine, a TRPV1 antagonist, but by rimonabant, a $CB_1$ antagonist/inverse agonist. Rimonabant by itself promoted adipogenesis in hBM-MSCs, which may be interpreted as the result of the inverse agonism of the $CB_1$ receptor. This result suggests that the constantly active $CB_1$ receptor may contribute to suppress the adipocyte differentiation of hBM-MSCs. Therefore, the selective $CB_1$ agonists that are unable to affect cellular $PPAR{\gamma}$ activity inhibit adipogenesis in hBM-MSCs.