Generic placeholder image

CNS & Neurological Disorders - Drug Targets

Editor-in-Chief

ISSN (Print): 1871-5273
ISSN (Online): 1996-3181

Review Article

The Role of Traditional Chinese Herbal Medicines and Bioactive Ingredients on Ion Channels: A Brief Review and Prospect

Author(s): Yian Huang, Shumei Ma, Yan Wang, Renjie Yan, Sheng Wang, Nan Liu, Ben Chen, Jia Chen and Li Liu*

Volume 18, Issue 4, 2019

Page: [257 - 265] Pages: 9

DOI: 10.2174/1871527317666181026165400

Price: $65

Abstract

Traditional Chinese Medicines (TCMs), particularly the Chinese herbal medicines, are valuable sources of medicines and have been used for centuries. The term “TCMs” both represents to the single drug agent like Salvia miltiorrhiza, Ligusticum chuanxiong and Angelica sinensis, and those herbal formulas like Jingshu Keli, Wenxin Keli and Danzhen powder. In recent years, the researches of TCMs developed rapidly to understand the scientific basis of these herbs. In this review, we collect the studies of TCM and their containing bioactive compounds, and attempt to provide an overview for their regulatory effects on different ion channels including Ca2+, K+, Na+, Cl- channels and TRP, P2X receptors. The following conditions are used to limit the range of our review. (i) Only the herbal materials are included in this review and the animal- and mineral-original TCMs are excluded. (ii) The major discussions in this review focus on single TCM agent and the herbal formulas are only discussed for a little. (iii) Those most famous herbal medicines like Capsicum annuum (pepper), Curcuma longa (ginger) and Cannabis sativa (marijuana) are excluded. (iv) Only those TCM herbs with more than 5 research papers confirming their effects on ion channels are discussed in this review. Our review discusses recently available scientific evidences for TCMs and related bioactive compounds that have been reported with the modulatory effects on different ion channels, and thus provides a new ethnopharmacological approach to understand the usage of TCMs.

Keywords: Traditional chinese medicine, potassium channel, calcium channel, ion channels, single chemical entity, ethnomedicines.

Next »
Graphical Abstract
[1]
Imamura T, Ishizuka O, Aizawa N, et al. Gosha-jinki-gan reduces transmitter proteins and sensory receptors associated with C fiber activation induced by acetic acid in rat urinary bladder. Neurourol Urodyn 2008; 27: 832-7.
[2]
Matsumura Y, Yokoyama Y, Hirakawa H, Shigeto T, Futagami M, Mizunuma H. The prophylactic effects of a traditional Japanese medicine, goshajinkigan, on paclitaxel-induced peripheral neuropathy and its mechanism of action. Mol Pain 2014; 10: 61.
[3]
Mizuno K, Kono T, Suzuki Y, et al. Goshajinkigan, a traditional Japanese medicine, prevents oxaliplatin-induced acute peripheral neuropathy by suppressing functional alteration of TRP channels in rat. J Pharmacol Sci 2014; 125: 91-8.
[4]
Huang J, Tao J, Xue X, et al. Gua Lou Gui Zhi decoction exerts neuroprotective effects on post-stroke spasticity via the modulation of glutamate levels and AMPA receptor expression. Int J Mol Med 2013; 31: 841-8.
[5]
Chen X, Li H, Huang M, et al. Effect of Gua Lou Gui Zhi decoction on focal cerebral ischemia-reperfusion injury through regulating the expression of excitatory amino acids and their receptors. Mol Med Rep 2014; 10: 248-54.
[6]
Wang X, Wang X, Gu Y, Wang T, Huang C. Wenxin Keli attenuates ischemia-induced ventricular arrhythmias in rats: Involvement of Ltype calcium and transient outward potassium currents. Mol Med Rep 2013; 7: 519-24.
[7]
Wang T, Lu M, Du Q, et al. An integrated anti-arrhythmic target network of a Chinese medicine compound, Wenxin Keli, revealed by combined machine learning and molecular pathway analysis. Mol Biosyst 2017; 13: 1018-30.
[8]
Imamura T, Ishizuka O, Zhong C, et al. An extract (THC-002) of Ba-Wei-Die-Huang-Wan inhibits expression of tachykinins, and P2X3 and TRPV1 receptors, and inhibits ATP-induced detrusor overactivity in spontaneously hypertensive rats. Neurourol Urodyn 2009; 28: 529-34.
[9]
Lee WC, Wu CC, Chuang YC, Tain YL, Chiang PH. Ba-Wei-Die-Huang-Wan (Hachimi-jio-gan) can ameliorate cyclophosphamide-induced ongoing bladder overactivity and acidic adenosine triphosphate solution-induced hyperactivity on rats prestimulated bladder. J Ethnopharmacol 2016; 184: 1-9.
[10]
Kamei J, Miyata S, Ohsawa M. Involvement of the benzodiazepine system in the anxiolytic-like effect of Yokukansan (Yi-gan san). Prog Neuropsychopharmacol Biol Psychiatry 2009; 33: 1431-7.
[11]
Kawakami Z, Kanno H, Ueki T, et al. Neuroprotective effects of yokukansan, a traditional Japanese medicine, on glutamate-mediated excitotoxicity in cultured cells. Neuroscience 2009; 159: 1397-407.
[12]
Kawakami Z, Ikarashi Y, Kase Y. Isoliquiritigenin is a novel NMDA receptor antagonist in kampo medicine yokukansan. Cell Mol Neurobiol 2011; 31: 1203-12.
[13]
Wakabayashi M, Hasegawa T, Yamaguchi T, et al. Yokukansan, a traditional Japanese medicine, adjusts glutamate signaling in cultured keratinocytes. BioMed Res Int 2014; 2014364092
[14]
Hao S, Bao YM, Zhao RG, et al. Effects of resibufogenin on voltage-gated sodium channels in cultured rat hippocampal neurons. Neurosci Lett 2011; 501: 112-6.
[15]
Song T, Chu X, Zhang X, et al. Bufalin, a bufanolide steroid from the parotoid glands of the Chinese toad, inhibits L-type Ca(2+) channels and contractility in rat ventricular myocytes. Fundam Clin Pharmacol 2017; 31: 340-6.
[16]
Liu ZR, Ye P, Ji YH. Exploring the obscure profiles of pharmacological binding sites on voltage-gated sodium channels by BmK neurotoxins. Protein Cell 2011; 2: 437-44.
[17]
Zou X, He Y, Qiao J, Zhang C, Cao Z. The natural scorpion peptide, BmK NT1 activates voltage-gated sodium channels and produces neurotoxicity in primary cultured cerebellar granule cells. Toxicon 2016; 109: 33-41.
[18]
He Y, Zou X, Li X, et al. Activation of sodium channels by alpha-scorpion toxin, BmK NT1, produced neurotoxicity in cerebellar granule cells: an association with intracellular Ca(2+) overloading. Arch Toxicol 2017; 91: 935-48.
[19]
Chen K, Yu B. Certain progress of clinical research on Chinese integrative medicine. Chin Med J (Engl) 1999; 112: 934-7.
[20]
Liao JF, Wang HH, Chen MC, Chen CC, Chen CF. Benzodiazepine binding site-interactive flavones from Scutellaria baicalensis root. Planta Med 1998; 64: 571-2.
[21]
Hui KM, Wang XH, Xue H. Interaction of flavones from the roots of Scutellaria baicalensis with the benzodiazepine site. Planta Med 2000; 66: 91-3.
[22]
Wang H, Hui KM, Xu S, Chen Y, Wong JT, Xue H. Two flavones from Scutellaria baicalensis Georgi and their binding affinities to the benzodiazepine site of the GABAA receptor complex. Pharmazie 2002; 57: 857-8.
[23]
Wang F, Xu Z, Ren L, Tsang SY, Xue H. GABA A receptor subtype selectivity underlying selective anxiolytic effect of baicalin. Neuropharmacology 2008; 55: 1231-7.
[24]
Hui KM, Huen MS, Wang HY, et al. Anxiolytic effect of wogonin, a benzodiazepine receptor ligand isolated from Scutellaria baicalensis Georgi. Biochem Pharmacol 2002; 64: 1415-24.
[25]
de Carvalho RS, Duarte FS, de Lima TC. Involvement of GABAergic non-benzodiazepine sites in the anxiolytic-like and sedative effects of the flavonoid baicalein in mice. Behav Brain Res 2011; 221: 75-82.
[26]
Park HG, Yoon SY, Choi JY, et al. Anticonvulsant effect of wogonin isolated from Scutellaria baicalensis. Eur J Pharmacol 2007; 574: 112-9.
[27]
Liu X, Hong SI, Park SJ, et al. The ameliorating effects of 5,7-dihydroxy-6-methoxy-2(4-phenoxyphenyl)-4H-chromene-4-one, an oroxylin A derivative, against memory impairment and sensorimotor gating deficit in mice. Arch Pharm Res 2013; 36: 854-63.
[28]
Zhang SQ, Obregon D, Ehrhart J, et al. Baicalein reduces beta-amyloid and promotes nonamyloidogenic amyloid precursor protein processing in an Alzheimer’s disease transgenic mouse model. J Neurosci Res 2013; 91: 1239-46.
[29]
Sun X, Chan LN, Gong X, Sucher NJ. N-methyl-D-aspartate receptor antagonist activity in traditional Chinese stroke medicines. Neurosignals 2003; 12: 31-8.
[30]
Tang W, Sun X, Fang JS, Zhang M, Sucher NJ. Flavonoids from Radix Scutellariae as potential stroke therapeutic agents by targeting the second postsynaptic density 95 (PSD-95)/disc large/zonula occludens-1 (PDZ) domain of PSD-95. Phytomedicine 2004; 11: 277-84.
[31]
Lin YL, Dai ZK, Lin RJ, et al. Baicalin, a flavonoid from Scutellaria baicalensis Georgi, activates large-conductance Ca2+-activated K+ channels via cyclic nucleotide-dependent protein kinases in mesenteric artery. Phytomedicine 2010; 17: 760-70.
[32]
Chang Y, Lu CW, Lin TY, Huang SK, Wang SJ. Baicalein, a Constituent of Scutellaria baicalensis, Reduces Glutamate Release and Protects Neuronal Cell Against Kainic Acid-Induced Excitotoxicity in Rats. Am J Chin Med 2016; 44: 943-62.
[33]
Shih HC, Hsu CS, Yang LL. In vitro study of the tocolytic effect of oroxylin A from Scutellaria baicalensis root. J Biomed Sci 2009; 16: 27.
[34]
Sui F, Zhang CB, Yang N, et al. Anti-nociceptive mechanism of baicalin involved in intervention of TRPV1 in DRG neurons in vitro. J Ethnopharmacol 2010; 129: 361-6.
[35]
Deng J, Wang DX, Liang AL, Tang J, Xiang DK. Effects of baicalin on alveolar fluid clearance and alpha-ENaC expression in rats with LPS-induced acute lung injury. Can J Physiol Pharmacol 2017; 95: 122-8.
[36]
Li C, Lin G, Zuo Z. Pharmacological effects and pharmacokinetics properties of Radix Scutellariae and its bioactive flavones. Biopharm Drug Dispos 2011; 32: 427-45.
[37]
Lei X, Chen J, Liu CX, Lin J, Lou J, Shang HC. Status and thoughts of Chinese patent medicines seeking approval in the US market. Chin J Integr Med 2014; 20: 403-8.
[38]
Sun X, Chan LN, Sucher NJ. Magnesium as NMDA receptor blocker in the traditional Chinese medicine Danshen. Phytomedicine 2005; 12: 173-7.
[39]
Wang W, Hu GY, Wang YP. Selective modulation of L-type calcium current by magnesium lithospermate B in guinea-pig ventricular myocytes. Life Sci 2006; 78: 2989-97.
[40]
Shou Q, Pan Y, Xu X, et al. Salvianolic acid B possesses vasodilation potential through NO and its related signals in rabbit thoracic aortic rings. Eur J Pharmacol 2012; 697: 81-7.
[41]
Gao Y, Zhang K, Zhu F, et al. Salvia miltiorrhiza (Danshen) inhibits L-type calcium current and attenuates calcium transient and contractility in rat ventricular myocytes. J Ethnopharmacol 2014; 158 Pt A: 397-403.
[42]
Li RW, Yang C, Shan L, et al. Relaxation effect of a novel Danshensu/tetramethylpyrazine derivative on rat mesenteric arteries. Eur J Pharmacol 2015; 761: 153-60.
[43]
Cai Q, Huang SY, Tan JZ. Effects of sodium tanshinone B on the protein expression of NMDAR1 in rat hippocampal subfields following focal ischemia/reperfusion injury. Zhongguo Zhong Xi Yi Jie He Za Zhi 2012; 32: 1073-6.
[44]
Gu M, Zhang G, Su Z, Ouyang F. Identification of major active constituents in the fingerprint of Salvia miltiorrhiza Bunge developed by high-speed counter-current chromatography. J Chromatogr A 2004; 1041: 239-43.
[45]
Fei YX, Wang SQ, Yang LJ, et al. Salvia miltiorrhiza Bunge (Danshen) extract attenuates permanent cerebral ischemia through inhibiting platelet activation in rats. J Ethnopharmacol 2017; 207: 57-66.
[46]
Yang Y, Cai F, Li PY, et al. Activation of high conductance Ca(2+)-activated K(+) channels by sodium tanshinoneII-A sulfonate (DS-201) in porcine coronary artery smooth muscle cells. Eur J Pharmacol 2008; 598: 9-15.
[47]
Shan H, Li X, Pan Z, et al. Tanshinone IIA protects against sudden cardiac death induced by lethal arrhythmias via repression of microRNA-1. Br J Pharmacol 2009; 158: 1227-35.
[48]
Ng CF, Koon CM, Cheung DW, et al. The anti-hypertensive effect of Danshen (Salvia miltiorrhiza) and Gegen (Pueraria lobata) formula in rats and its underlying mechanisms of vasorelaxation. J Ethnopharmacol 2011; 137: 1366-72.
[49]
Yoon JY, Ahn SH, Oh H, et al. A novel Na+ channel agonist, dimethyl lithospermate B, slows Na+ current inactivation and increases action potential duration in isolated rat ventricular myocytes. Br J Pharmacol 2004; 143: 765-73.
[50]
Liu AD, Cai GH, Wei YY, et al. Anxiolytic effect of essential oils of Salvia miltiorrhiza in rats. Int J Clin Exp Med 2015; 8: 12756-64.
[51]
Zhu J, Xu Y, Ren G, et al. Tanshinone IIA Sodium sulfonate regulates antioxidant system, inflammation, and endothelial dysfunction in atherosclerosis by downregulation of CLIC1. Eur J Pharmacol 2017; 815: 427-36.
[52]
Sun S, Yin Y, Yin X, et al. Anti-nociceptive effects of Tanshinone IIA (TIIA) in a rat model of complete Freund’s adjuvant (CFA)-induced inflammatory pain. Brain Res Bull 2012; 88: 581-8.
[53]
Chen RC, Sun GB, Ye JX, Wang J, Zhang MD, Sun XB. Salvianolic acid B attenuates doxorubicin-induced ER stress by inhibiting TRPC3 and TRPC6 mediated Ca(2+) overload in rat cardiomyocytes. Toxicol Lett 2017; 276: 21-30.
[54]
Kimura M, Kimura I, Takahashi K. The neuromuscular blocking actions of coclaurine derivatives and of paeoniflorin derivatives. Planta Med 1982; 45: 136.
[55]
Kimura M, Kimura I, Takahashi K, et al. Blocking effects of blended paeoniflorin or its related compounds with glycyrrhizin on neuromuscular junctions in frog and mouse. Jpn J Pharmacol 1984; 36: 275-82.
[56]
Kimura M, Kimura I, Nojima H. Depolarizing neuromuscular blocking action induced by electropharmacological coupling in the combined effect of paeoniflorin and glycyrrhizin. Jpn J Pharmacol 1985; 37: 395-9.
[57]
Wang RR, Li N, Zhang YH, Ran YQ, Pu JL. The effects of paeoniflorin monomer of a Chinese herb on cardiac ion channels. Chin Med J (Engl) 2011; 124: 3105-11.
[58]
Tsai TY, Wu SN, Liu YC, Wu AZ, Tsai YC. Inhibitory action of L-type Ca2+ current by paeoniflorin, a major constituent of peony root, in NG108-15 neuronal cells. Eur J Pharmacol 2005; 523: 16-24.
[59]
Chen YF, Lee MM, Fang HL, Yang JG, Chen YC, Tsai HY. Paeoniflorin inhibits excitatory amino acid agonist-and high-dose morphine-induced nociceptive behavior in mice via modulation of N-methyl-D-aspartate receptors. BMC Complement Altern Med 2016; 16: 240.
[60]
Gu J, Chen J, Yang N, et al. Combination of Ligusticum chuanxiong and Radix Paeoniae ameliorate focal cerebral ischemic in MCAO rats via endoplasmic reticulum stress-dependent apoptotic signaling pathway. J Ethnopharmacol 2016; 187: 313-24.
[61]
Zhang GQ, Hao XM, Chen SZ, Zhou PA, Cheng HP, Wu CH. Blockade of paeoniflorin on sodium current in mouse hippocampal CA1 neurons. Acta Pharmacol Sin 2003; 24: 1248-52.
[62]
Cao BY, Yang YP, Luo WF, et al. Paeoniflorin, a potent natural compound, protects PC12 cells from MPP+ and acidic damage via autophagic pathway. J Ethnopharmacol 2010; 131: 122-9.
[63]
Gu XS, Wang F, Zhang CY, et al. Neuroprotective Effects of Paeoniflorin on 6-OHDA-Lesioned Rat Model of Parkinson’s Disease. Neurochem Res 2016; 41: 2923-36.
[64]
Jin SN, Wen JF, Wang TT, Kang DG, Lee HS, Cho KW. Vasodilatory effects of ethanol extract of Radix Paeoniae Rubra and its mechanism of action in the rat aorta. J Ethnopharmacol 2012; 142: 188-93.
[65]
Sui F, Zhou HY, Meng J, et al. A Chinese Herbal Decoction, Shaoyao-Gancao Tang, Exerts analgesic effect by down-regulating the TRPV1 channel in a rat model of arthritic pain. Am J Chin Med 2016; 44: 1363-78.
[66]
Tsai CC, Lai TY, Huang WC, Liu IM, Cheng JT. Inhibitory effects of potassium channel blockers on tetramethylpyrazine-induced relaxation of rat aortic strip in vitro. Life Sci 2002; 71: 1321-30.
[67]
Tsai CC, Lai TY, Huang WC, et al. Tetramethylpyrazine as potassium channel opener to lower calcium influx into cultured aortic smooth muscle cells. Planta Med 2003; 69: 557-8.
[68]
Liang SD, Gao Y, Xu CS, Xu BH, Mu SN. Effect of tetramethylpyrazine on acute nociception mediated by signaling of P2X receptor activation in rat. Brain Res 2004; 995: 247-52.
[69]
Liang S, Xu C, Li G, Gao Y. P2X receptors and modulation of pain transmission: focus on effects of drugs and compounds used in traditional Chinese medicine. Neurochem Int 2010; 57: 705-12.
[70]
Zhang ZG, Zhang XL, Wang XY, Luo ZR, Song JC. Inhibition of acid sensing ion channel by ligustrazine on angina model in rat. Am J Transl Res 2015; 7: 1798-811.
[71]
Zhou ZY, Xu JQ, Zhao WR, et al. Ferulic acid relaxed rat aortic, small mesenteric and coronary arteries by blocking voltage-gated calcium channel and calcium desensitization via dephosphorylation of ERK1/2 and MYPT1. Eur J Pharmacol 2017; 815: 26-32.
[72]
Zhong J, Pollastro F, Prenen J, Zhu Z, Appendino G, Nilius B. Ligustilide: a novel TRPA1 modulator. Pflugers Arch 2011; 462: 841-9.
[73]
Sun XH, Ding JP, Li H, et al. Activation of large-conductance calcium-activated potassium channels by puerarin: the underlying mechanism of puerarin-mediated vasodilation. J Pharmacol Exp Ther 2007; 323: 391-7.
[74]
Choo MK, Park EK, Yoon HK, Kim DH. Antithrombotic and antiallergic activities of daidzein, a metabolite of puerarin and daidzin produced by human intestinal microflora. Biol Pharm Bull 2002; 25: 1328-32.
[75]
Miao WN, Shen YJ, Zeng XR. [Effect of Puerarin on K+ channel of isolated ventricular myocyte in guinea pig]. Zhongguo Ying Yong Sheng Li Xue Za Zhi 2002; 18: 155-8.
[76]
Deng Y, Ng ES, Yeung JH, et al. Mechanisms of the cerebral vasodilator actions of isoflavonoids of Gegen on rat isolated basilar artery. J Ethnopharmacol 2012; 139: 294-304.
[77]
Lee S, Gim H, Shim JH, et al. The traditional herbal medicine, Ge-Gen-Tang, inhibits pacemaker potentials by nitric oxide/cGMP dependent ATP-sensitive K(+) channels in cultured interstitial cells of Cajal from mouse small intestine. J Ethnopharmacol 2015; 170: 201-9.
[78]
Xu H, Zhao M, Liang S, et al. The Effects of Puerarin on Rat Ventricular Myocytes and the Potential Mechanism. Sci Rep 2016; 6: 35475.
[79]
Xu C, Xu W, Xu H, et al. Role of puerarin in the signalling of neuropathic pain mediated by P2X3 receptor of dorsal root ganglion neurons. Brain Res Bull 2012; 87: 37-43.
[80]
Liu S, Yu S, Xu C, et al. Puerarin alleviates aggravated sympathoexcitatory response induced by myocardial ischemia via regulating P2X3 receptor in rat superior cervical ganglia. Neurochem Int 2014; 70: 39-49.
[81]
Liu S, Zhang C, Shi Q, et al. Puerarin blocks the signaling transmission mediated by P2X3 in SG and DRG to relieve myocardial ischemic damage. Brain Res Bull 2014; 101: 57-63.
[82]
Campbell DT. Modified kinetics and selectivity of sodium channels in frog skeletal muscle fibers treated with aconitine. J Gen Physiol 1982; 80: 713-31.
[83]
Grishchenko II, Naumov AP, Zubov AN. Gating and selectivity of aconitine-modified sodium channels in neuroblastoma cells. Neuroscience 1983; 9: 549-54.
[84]
Ghiasuddin SM, Soderlund DM. Mouse brain synaptosomal sodium channels: activation by aconitine, batrachotoxin, and veratridine, and inhibition by tetrodotoxin. Comp Biochem Physiol C 1984; 77: 267-71.
[85]
Friese J, Gleitz J, Gutser UT, et al. Aconitum sp. alkaloids: the modulation of voltage-dependent Na+ channels, toxicity and antinociceptive properties. Eur J Pharmacol 1997; 337: 165-74.
[86]
Gutser UT, Friese J, Heubach JF, et al. Mode of antinociceptive and toxic action of alkaloids of Aconitum spec. Naunyn Schmiedebergs Arch Pharmacol 1998; 357: 39-48.
[87]
Poletto CJ, Van Doren CL. Elevating pain thresholds in humans using depolarizing prepulses. IEEE Trans Biomed Eng 2002; 49: 1221-4.
[88]
Ameri A. Effects of the alkaloids 6-benzoylheteratisine and heteratisine on neuronal activity in rat hippocampal slices. Neuropharmacology 1997; 36: 1039-46.
[89]
Luo J, Min S, Wei K, Cao J. Ion channel mechanism and ingredient bases of Shenfu Decoction’s cardiac electrophysiological effects. J Ethnopharmacol 2008; 117: 439-45.
[90]
Wang C, Sun D, Liu C, et al. Mother root of Aconitum carmichaelii Debeaux exerts antinociceptive effect in Complet Freund’s Adjuvant-induced mice: roles of dynorpin/kappa-opioid system and transient receptor potential vanilloid type-1 ion channel. J Transl Med 2015; 13: 284.
[91]
Wang C, Liu C, Wan H, et al. Wu-tou decoction inhibits chronic inflammatory pain in mice: participation of TRPV1 and TRPA1 ion channels. BioMed Res Int 2015; 2015328707
[92]
Xie S, Jia Y, Liu A, Dai R, Huang L. Hypaconitine-induced QT prolongation mediated through inhibition of KCNH2 (hERG) potassium channels in conscious dogs. J Ethnopharmacol 2015; 166: 375-9.
[93]
Kite GC, Howes MJ, Leon CJ, Simmonds MS. Liquid chromatography/mass spectrometry of malonyl-ginsenosides in the authentication of ginseng. Rapid Commun Mass Spectrom 2003; 17: 238-44.
[94]
Choi SJ, Kim TH, Shin YK, et al. Effects of a polyacetylene from Panax ginseng on Na+ currents in rat dorsal root ganglion neurons. Brain Res 2008; 1191: 75-83.
[95]
Huang LF, Shi HL, Gao B, et al. Decichine enhances hemostasis of activated platelets via AMPA receptors. Thromb Res 2014; 133: 848-54.
[96]
Nabavi SF, Sureda A, Habtemariam S, Nabavi SM. Ginsenoside Rd and ischemic stroke; a short review of literatures. J Ginseng Res 2015; 39: 299-303.
[97]
Xu L, Huang SP. Effect of the ginsenoside Rb1 on the spontaneous contraction of intestinal smooth muscle in mice. World J Gastroenterol 2012; 18: 5462-9.
[98]
Yan S, Li Z, Li H, Arancio O, Zhang W. Notoginsenoside R1 increases neuronal excitability and ameliorates synaptic and memory dysfunction following amyloid elevation. Sci Rep 2014; 4: 6352.
[99]
Qi D, Zhu Y, Wen L, Liu Q, Qiao H. Ginsenoside Rg1 restores the impairment of learning induced by chronic morphine administration in rats. J Psychopharmacol 2009; 23: 74-83.
[100]
Wang S, Li M, Guo Y, et al. Effects of Panax notoginseng ginsenoside Rb1 on abnormal hippocampal microenvironment in rats. J Ethnopharmacol 2017; 202: 138-46.
[101]
Guo S, Chen Y, Pang C, et al. Ginsenoside Rb1, a novel activator of the TMEM16A chloride channel, augments the contraction of guinea pig ileum. Pflugers Arch 2017; 469: 681-92.
[102]
Kang TH, Murakami Y, Matsumoto K, et al. Rhynchophylline and isorhynchophylline inhibit NMDA receptors expressed in Xenopus oocytes. Eur J Pharmacol 2002; 455: 27-34.
[103]
Kang TH, Murakami Y, Takayama H, et al. Protective effect of rhynchophylline and isorhynchophylline on in vitro ischemia-induced neuronal damage in the hippocampus: putative neurotransmitter receptors involved in their action. Life Sci 2004; 76: 331-43.
[104]
Gui L, Li ZW, Du R, et al. Inhibitory effect of rhynchophylline on human ether-a-go-go related gene channel. Sheng Li Xue Bao 2005; 57: 648-52.
[105]
Zhou J, Zhou S. Antihypertensive and neuroprotective activities of rhynchophylline: The role of rhynchophylline in neurotransmission and ion channel activity. J Ethnopharmacol 2010; 132: 15-27.
[106]
Gan R, Dong G, Yu J, Wang X, Fu S, Yang S. Protective effects of isorhynchophylline on cardiac arrhythmias in rats and guinea pigs. Planta Med 2011; 77: 1477-81.
[107]
He Y, Zeng SY, Zhou SW, et al. Effects of rhynchophylline on GluN1 and GluN2B expressions in primary cultured hippocampal neurons. Fitoterapia 2014; 98: 166-73.
[108]
Li J, Liu W, Peng Q, et al. Effect of rhynchophylline on conditioned place preference on expression of NR2B in methamphetamine-dependent mice. Biochem Biophys Res Commun 2014; 452: 695-700.
[109]
Jiang M, Chen Y, Li C, et al. Inhibiting effects of rhynchophylline on zebrafish methamphetamine dependence are associated with amelioration of neurotransmitters content and down-regulation of TH and NR2B expression. Prog Neuropsychopharmacol Biol Psychiatry 2016; 68: 31-43.
[110]
Kobayashi Y, Nakano Y, Kizaki M, Hoshikuma K, Yokoo Y, Kamiya T. Capsaicin-like anti-obese activities of evodiamine from fruits of Evodia rutaecarpa, a vanilloid receptor agonist. Planta Med 2001; 67: 628-33.
[111]
Moon TC, Murakami M, Kudo I, et al. A new class of COX-2 inhibitor, rutaecarpine from Evodia rutaecarpa. Inflamm Res 1999; 48: 621-5.
[112]
Lee CM, Gu JA, Rau TG, et al. Synthetic Fluororutaecarpine Inhibits Inflammatory Stimuli and Activates Endothelial Transient Receptor Potential Vanilloid-Type 1. Molecules 2017; 22.
[113]
Wang T, Wang Y, Kontani Y, et al. Evodiamine improves diet-induced obesity in a uncoupling protein-1-independent manner: involvement of antiadipogenic mechanism and extracellularly regulated kinase/mitogen-activated protein kinase signaling. Endocrinology 2008; 149: 358-66.
[114]
De Petrocellis L, Schiano Moriello A, Fontana G, et al. Effect of chirality and lipophilicity in the functional activity of evodiamine and its analogues at TRPV1 channels. Br J Pharmacol 2014; 171: 2608-20.
[115]
Iwaoka E, Wang S, Matsuyoshi N, et al. Evodiamine suppresses capsaicin-induced thermal hyperalgesia through activation and subsequent desensitization of the transient receptor potential V1 channels. J Nat Med 2016; 70: 1-7.
[116]
Wang S, Yamamoto S, Kogure Y, Zhang W, Noguchi K, Dai Y. Partial Activation and Inhibition of TRPV1 Channels by Evodiamine and Rutaecarpine, Two Major Components of the Fruits of Evodia rutaecarpa. J Nat Prod 2016; 79: 1225-30.
[117]
Wu D, Hu Z. Rutaecarpine induces chloride secretion across rat isolated distal colon. J Pharmacol Exp Ther 2008; 325: 256-66.
[118]
Hu J, Pang WS, Han J, Zhang K, Zhang JZ, Chen LD. Gualou Guizhi decoction reverses brain damage with cerebral ischemic stroke, multi-component directed multi-target to screen calcium-overload inhibitors using combination of molecular docking and protein-protein docking. J Enzyme Inhib Med Chem 2018; 33: 115-25.

Rights & Permissions Print Cite
© 2024 Bentham Science Publishers | Privacy Policy