-
为选出临床常用中药,我们对国家专利数据库中治疗HF的中药复方专利进行挖掘分析。登录国家知识产权局中国专利公布公告网站(中国专利公布公告 cnipa.gov.cn),在“中国专利公布公告”的“高级查询”界面勾选“发明公布”与“发明授权”,分别将“心力衰竭和中药”作为“名称”项目进行检索,检索时间为2023年2月。本研究纳入的116项中药复方专利中,中药复方组成的药物最多有50味中药,最少有2味中药,共有418味中药。运用频次统计分析发现,出现频次最高的药物是黄芪(55次)。频次≥10的中药见表1。
表 1 中国专利数据库中治疗HF的高频次中药
药名 频次 药名 频次 药名 频次 黄芪 55 白术 21 三七 12 丹参 40 当归 21 干姜 11 茯苓 40 麦冬 21 鸡血藤 11 附子 38 甘草 20 桃仁 11 人参 31 泽泻 19 猪苓 11 葶苈子 28 红参 16 红花 10 桂枝 28 党参 15 肉桂 10 川芎 23 五味子 15 益母草 10
Research progress on natural medicines and active compounds in the treatment of heart failure
-
摘要: 心衰是各种心血管疾病的终末阶段,也是导致患者死亡的主要原因。长期以来,天然药物一直用于治疗心衰,且效果显著。对国家专利数据库中的中药复方专利数据进行数据挖掘,选出临床治疗心衰的常见中药,并对其含有的经典单一活性成分进行分析,为开发治疗心衰的天然药物提供理论依据。Abstract: Heart failure is the terminal stage of various cardiovascular diseases and a leading cause of death. For a long time, natural medicines have been used to treat heart failure(HF) with remarkable effects. In this paper, the Traditional Chinese Medicine compound patents in the national patent database were mined, common Traditional Chinese Medicines for the clinical treatment of HF were selected, and the single active ingredient contained in them was analyzed, which provided some valuable tips for the development of drugs for the treatment of heart failure.
-
Key words:
- heart failure /
- natural medicine /
- monomer compounds /
- pharmacological action
-
表 1 中国专利数据库中治疗HF的高频次中药
药名 频次 药名 频次 药名 频次 黄芪 55 白术 21 三七 12 丹参 40 当归 21 干姜 11 茯苓 40 麦冬 21 鸡血藤 11 附子 38 甘草 20 桃仁 11 人参 31 泽泻 19 猪苓 11 葶苈子 28 红参 16 红花 10 桂枝 28 党参 15 肉桂 10 川芎 23 五味子 15 益母草 10 -
[1] XU L H, CHEN L Y, GU G Y, et al. Natural products from traditional Chinese medicine for the prevention and treatment of heart failure: progress and perspectives[J]. Rev Cardiovasc Med, 2022, 23(2):60. doi: 10.31083/j.rcm2302060 [2] WANG A Z, ZHAO W, YAN K T, et al. Mechanisms and efficacy of traditional Chinese medicine in heart failure[J]. Front Pharmacol, 2022, 13:810587. doi: 10.3389/fphar.2022.810587 [3] FU S F, ZHANG J H, GAO X M, et al. Clinical practice of traditional Chinese medicines for chronic heart failure[J]. Heart Asia, 2010, 2(1):24-27. doi: 10.1136/ha.2009.001123 [4] ZANG Y B, WAN J J, ZHANG Z, et al. An updated role of astragaloside IV in heart failure[J]. Biomed Pharmacother, 2020, 126:110012. doi: 10.1016/j.biopha.2020.110012 [5] WANG Y, WANG Q Y, LI C, et al. A review of Chinese herbal medicine for the treatment of chronic heart failure[J]. Curr Pharm Des, 2017, 23(34):5115-5124. [6] MA X, ZHANG K, LI H X, et al. Extracts from Astragalus membranaceus limit myocardial cell death and improve cardiac function in a rat model of myocardial ischemia[J]. J Ethnopharmacol, 2013, 149(3):720-728. doi: 10.1016/j.jep.2013.07.036 [7] ZHOU R J, CHEN H J, CHEN J P, et al. Extract from Astragalus membranaceus inhibit breast cancer cells proliferation via PI3K/AKT/mTOR signaling pathway[J]. BMC Complementary Altern Med, 2018, 18(1):83. doi: 10.1186/s12906-017-2057-9 [8] FRANK A, BONNEY M, BONNEY S, et al. Myocardial ischemia reperfusion injury: from basic science to clinical bedside[J]. Semin Cardiothorac Vasc Anesth, 2012, 16(3):123-132. doi: 10.1177/1089253211436350 [9] YIN B, HOU X W, LU M L. Astragaloside IV attenuates myocardial ischemia/reperfusion injury in rats via inhibition of calcium-sensing receptor-mediated apoptotic signaling pathways[J]. Acta Pharmacol Sin, 2019, 40(5):599-607. doi: 10.1038/s41401-018-0082-y [10] FISCHER P, HILFIKER-KLEINER D. Survival pathways in hypertrophy and heart failure: the gp130-STAT3 axis[J]. Basic Res Cardiol, 2007, 102(4):279-297. doi: 10.1007/s00395-007-0658-z [11] DENISE H K, PRAPHULLA S, GUNNAR K, et al. Continuous glycoprotein-130-mediated signal transducer and activator of transcription-3 activation promotes inflammation, left ventricular rupture, and adverse outcome in subacute myocardial infarction[J]. Circulation, 2010, 122(2):145-155. doi: 10.1161/CIRCULATIONAHA.109.933127 [12] ZHAO P, WANG Y, ZENG S, et al. Protective effect of astragaloside IV on lipopolysaccharide-induced cardiac dysfunction via downregulation of inflammatory signaling in mice[J]. Immunopharmacol Immunotoxicol, 2015, 37(5):428-433. doi: 10.3109/08923973.2015.1080266 [13] ZHOU R N, SONG Y L, RUAN J Q, et al. Pharmacokinetic evidence on the contribution of intestinal bacterial conversion to beneficial effects of astragaloside IV, a marker compound of astragali Radix, in traditional oral use of the herb[J]. Drug Metab Pharmacokinet, 2012, 27(6):586-597. doi: 10.2133/dmpk.DMPK-11-RG-160 [14] SUN C H, JIANG M M, ZHANG L, et al. Cycloastragenol mediates activation and proliferation suppression in concanavalin A-induced mouse lymphocyte pan-activation model[J]. Immunopharmacol Immunotoxicol, 2017, 39(3):131-139. doi: 10.1080/08923973.2017.1300170 [15] TAN S F, WANG G F, GUO Y P, et al. Preventive effects of a natural anti-inflammatory agent, astragaloside IV, on ischemic acute kidney injury in rats[J]. Evid Based Complement Alternat Med, 2013, 2013:284025. [16] GUO H T, CALLAWAY J B, TING J P-Y. Inflammasomes: mechanism of action, role in disease, and therapeutics[J]. Nat Med, 2015, 21(7):677-687. doi: 10.1038/nm.3893 [17] WANG J, WU M L, CAO S P, et al. Cycloastragenol ameliorates experimental heart damage in rats by promoting myocardial autophagy via inhibition of AKT1-RPS6KB1 signaling[J]. Biomed Pharmacother, 2018, 107:1074-1081. doi: 10.1016/j.biopha.2018.08.016 [18] LI M, HAN B, ZHAO H, et al. Biological active ingredients of astragali radix and its mechanisms in treating cardiovascular and cerebrovascular diseases[J]. Phytomedicine, 2022, 98:153918. doi: 10.1016/j.phymed.2021.153918 [19] ZHANG L, FAN C D, JIAO H C, et al. Calycosin alleviates doxorubicin-induced cardiotoxicity and pyroptosis by inhibiting NLRP3 inflammasome activation[J]. Oxid Med Cell Longev, 2022, 2022:1733834. [20] CHEN G H, XU H L, XU T, et al. Calycosin reduces myocardial fibrosis and improves cardiac function in post-myocardial infarction mice by suppressing TGFBR1 signaling pathways[J]. Phytomedicine, 2022, 104:154277. doi: 10.1016/j.phymed.2022.154277 [21] RÍOS J L, ANDÚJAR I, RECIO M C, et al. Lanostanoids from fungi: a group of potential anticancer compounds[J]. J Nat Prod, 2012, 75(11):2016-2044. doi: 10.1021/np300412h [22] ESTEBAN C I. Interés medicinal de Poria Cocos (= Wolfiporia extensa)[J]. Rev Iberoam De Micología, 2009, 26(2):103-107. [23] RÍOS J L. Chemical constituents and pharmacological properties of Poria cocos[J]. Planta Med, 2011, 77(7):681-691. doi: 10.1055/s-0030-1270823 [24] VAN DER POL A, VAN GILST W H, VOORS A A, et al. Treating oxidative stress in heart failure: past, present and future[J]. Eur J Heart Fail, 2019, 21(4):425-435. doi: 10.1002/ejhf.1320 [25] LI X, CHEN S, LI J E, et al. Chemical composition and antioxidant activities of polysaccharides from Yingshan cloud mist tea[J]. Oxidative Med Cell Longev, 2019, 2019:1915967. [26] LIU X F, WANG X Q, XU X F, et al. Purification, antitumor and anti-inflammation activities of an alkali-soluble and carboxymethyl polysaccharide CMP33 from Poria cocos[J]. Int J Biol Macromol, 2019, 127:39-47. doi: 10.1016/j.ijbiomac.2019.01.029 [27] WEI C Y, WANG H Z, SUN X, et al. Pharmacological profiles and therapeutic applications of pachymic acid (Review)[J]. Exp Ther Med, 2022, 24(3):547. doi: 10.3892/etm.2022.11484 [28] LI F F, YUAN Y, LIU Y, et al. Pachymic acid protects H9c2 cardiomyocytes from lipopolysaccharide-induced inflammation and apoptosis by inhibiting the extracellular signal-regulated kinase 1/2 and p38 pathways[J]. Mol Med Rep, 2015, 12(2):2807-2813. doi: 10.3892/mmr.2015.3712 [29] HUANG Y C, CHANG W L, HUANG S F, et al. Pachymic acid stimulates glucose uptake through enhanced GLUT4 expression and translocation[J]. Eur J Pharmacol, 2010, 648(1-3):39-49. doi: 10.1016/j.ejphar.2010.08.021 [30] LUO C M, YI F L, XIA Y L, et al. Comprehensive quality evaluation of the lateral root of Aconitum carmichaelii Debx. (Fuzi): simultaneous determination of nine alkaloids and chemical fingerprinting coupled with chemometric analysis[J]. J Sep Sci, 2019, 42(5):980-990. [31] LIN S Q, LIU K X, WU W K, et al. Study on pretreatment of FPS-1 in rats with hepatic ischemia-reperfusion injury[J]. Am J Chin Med, 2009, 37(2):323-337. doi: 10.1142/S0192415X09006874 [32] LU Y J, ZHOU J, XU C Q, et al. JAK/STAT and PI3K/AKT pathways form a mutual transactivation loop and afford resistance to oxidative stress-induced apoptosis in cardiomyocytes[J]. Cell Physiol Biochem, 2008, 21(4):305-314. doi: 10.1159/000129389 [33] LU X H, ZHANG L, LI P Y, et al. The protective effects of compatibility of Aconiti Lateralis Radix Praeparata and Zingiberis Rhizoma on rats with heart failure by enhancing mitochondrial biogenesis via Sirt1/PGC-1α pathway[J]. Biomed Pharmacother, 2017, 92:651-660. doi: 10.1016/j.biopha.2017.05.117 [34] WU H, LIU X, GAO Z Y, et al. Anti-myocardial infarction effects of Radix aconiti lateralis preparata extracts and their influence on small molecules in the heart using matrix-assisted laser desorption/ionization–mass spectrometry imaging[J]. Int J Mol Sci, 2019, 20(19):4837. doi: 10.3390/ijms20194837 [35] CHAN T F, CHAN J C, TOMLINSON B, et al. Chinese herbal medicines revisited: a Hong Kong perspective[J]. Lancet, 1993, 342(8886-8887):1532-1534. doi: 10.1016/S0140-6736(05)80091-1 [36] LIU M, CAO Y, LV D Y, et al. Effect of processing on the alkaloids in Aconitum tubers by HPLC-TOF/MS[J]. J Pharm Anal, 2017, 7(3):170-175. doi: 10.1016/j.jpha.2017.01.001 [37] HONERJÄGER P, MEISSNER A. The positive inotropic effect of aconitine[J]. Naunyn-Schmiedeberg's Arch Pharmacol, 1983, 322(1):49-58. doi: 10.1007/BF00649352 [38] WU M P, ZHANG Y S, ZHOU Q M, et al. Higenamine protects ischemia/reperfusion induced cardiac injury and myocyte apoptosis through activation of β2-AR/PI3K/AKT signaling pathway[J]. Pharmacol Res, 2016, 104:115-123. doi: 10.1016/j.phrs.2015.12.032 [39] LOPASCHUK G D, KARWI Q G, TIAN R, et al. Cardiac energy metabolism in heart failure[J]. Circ Res, 2021, 128(10):1487-1513. doi: 10.1161/CIRCRESAHA.121.318241 [40] WANG D, LU C Y, TENG L S, et al. Therapeutic effects of Chinese herbal medicine against neuroendocrinological diseases especially related to hypothalamus-pituitary-adrenal and hypothalamus-pituitary-gonadal axis[J]. Pak J Pharm Sci, 2014, 27(3-Suppl):741-754. [41] TSAI M Y, YANG R C, WU H T, et al. Anti-angiogenic effect of Tanshinone IIA involves inhibition of matrix invasion and modification of MMP-2/TIMP-2 secretion in vascular endothelial cells[J]. Cancer Lett, 2011, 310(2):198-206. doi: 10.1016/j.canlet.2011.06.031 [42] LI X, DU J R, YU Y, et al. Tanshinone IIA inhibits smooth muscle proliferation and intimal hyperplasia in the rat carotid balloon-injured model through inhibition of MAPK signaling pathway[J]. J Ethnopharmacol, 2010, 129(2):273-279. doi: 10.1016/j.jep.2010.03.021 [43] REN Z H, TONG Y H, XU W, et al. Tanshinone II A attenuates inflammatory responses of rats with myocardial infarction by reducing MCP-1 expression[J]. Phytomedicine, 2010, 17(3-4):212-218. doi: 10.1016/j.phymed.2009.08.010 [44] TAKAHASHI M. NLRP3 inflammasome as a novel player in myocardial infarction[J]. Int Heart J, 2014, 55(2):101-105. doi: 10.1536/ihj.13-388 [45] MA Y Z, LI H, YUE Z B, et al. Cryptotanshinone attenuates cardiac fibrosis via downregulation of COX-2, NOX-2, and NOX-4[J]. J Cardiovasc Pharmacol, 2014, 64(1):28-37. doi: 10.1097/FJC.0000000000000086 [46] JIN Y C, KIM C W, KIM Y M, et al. Cryptotanshinone, a lipophilic compound of Salvia miltiorrriza root, inhibits TNF-alpha-induced expression of adhesion molecules in HUVEC and attenuates rat myocardial ischemia/reperfusion injury in vivo[J]. Eur J Pharmacol, 2009, 614(1-3):91-97. doi: 10.1016/j.ejphar.2009.04.038 [47] ZHANG Y S, CHEN L, LI F, et al. Cryptotanshinone protects against adriamycin-induced mitochondrial dysfunction in cardiomyocytes[J]. Pharm Biol, 2016, 54(2):237-242. doi: 10.3109/13880209.2015.1029052 [48] MA S T,YANG D C,WANG K Y,et.al. Cryptotanshinone attenuates isoprenaline-induced cardiac fibrosis in mice associated with upregulation and activation of matrix metalloproteinase-2[J]. Mol Med Report, 2012, 6(1):145-150. [49] SONG Q T, CHU X, ZHANG X, et al. Mechanisms underlying the cardioprotective effect of Salvianic acid A against isoproterenol-induced myocardial ischemia injury in rats: possible involvement of L-type calcium channels and myocardial contractility[J]. J Ethnopharmacol, 2016, 189:157-164. doi: 10.1016/j.jep.2016.05.038 [50] ANDRASSY M, VPLZ H C, IGWE J C, et al. High-mobility group box-1 in ischemia-reperfusion injury of the heart[J]. Circulation, 2008, 117(25): 3216-3226. [51] XIANG J, ZHANG C L, DI T T, et al. Salvianolic acid B alleviates diabetic endothelial and mitochondrial dysfunction by down-regulating apoptosis and mitophagy of endothelial cells[J]. Bioengineered, 2022,13(2): 3486-3502. [52] ZHOU Z, LIU Y, MIAO A D, et al. Salvianolic acid B attenuates plasminogen activator inhibitor type 1 production in TNF-alpha treated human umbilical vein endothelial cells[J]. J Cell Biochem, 2005, 96(1):109-116. doi: 10.1002/jcb.20567 [53] VAN DER POL A, VAN GILST W H, VOORS A A, et al. Treating oxidative stress in heart failure: past, present 1and future[J]. Eur J Heart Fail, 2019, 21(4): 425-435. [54] LI C Y, YANG P, JIANG Y L, et al. Ginsenoside Rb1 attenuates cardiomyocyte apoptosis induced by myocardial ischemia reperfusion injury through mTOR signal pathway[J]. Biomed Pharmacother, 2020, 125:109913. doi: 10.1016/j.biopha.2020.109913 [55] WU Y, XIA Z Y, MENG Q T, et al. Shen-Fu injection preconditioning inhibits myocardial ischemia-reperfusion injury in diabetic rats: activation of eNOS via the PI3K/Akt pathway[J]. J Biomed Biotechnol, 2011, 2011:384627. [56] HUANG L Z, ZHAO H F, HUANG B K, et al. Acanthopanax senticosus: review of botany, chemistry and pharmacology[J]. Pharmazie, 2011, 66(2):83-97. [57] NIU J, PI Z F, YUE H, et al. Effect of ginseng polysaccharide on the urinary excretion of type 2 diabetic rats studied by liquid chromatography–mass spectrometry[J]. Analyt Teehnot. Biomed Life Sci, 2012, 907:7-12. doi: 10.1016/j.jchromb.2012.08.012 [58] LI C, CAI J P, GENG J S, et al. Purification, characterization and anticancer activity of a polysaccharide from Panax ginseng[J]. Int J Biol Macromol, 2012, 51(5):968-973. doi: 10.1016/j.ijbiomac.2012.06.031 [59] KIM M H, BYON Y Y, KO E J, et al. Immunomodulatory activity of ginsan, a polysaccharide of Panax ginseng, on dendritic cells[J]. Korean J Physiol Pharmacol, 2009, 13(3):169-173. doi: 10.4196/kjpp.2009.13.3.169