Skip to content
BY-NC-ND 4.0 license Open Access Published by De Gruyter Open Access September 18, 2021

Traditional Chinese medicine in the treatment of high incidence diseases in cold areas: the thrombotic diseases

  • Xueqing Tang , Xin Liu , Monayo Seth Mikaye , Hongrui Zhao and Yong Zhang EMAIL logo
From the journal Frigid Zone Medicine

Abstract

Thrombotic diseases are the leading causes of death worldwide, especially in cold climates. Traditional Chinese medicine (TCM)-based therapies have gained increasing popularity worldwide, but also raised some concerns about its efficacy, safety profile and exact mechanisms. TCM has been traditionally used in the management of thrombosis and convincingly proven effective in modifying thrombosis progression, particularly the platelet function, coagulation system and fibrinolytic system. This review article focuses on TCM regulation of thrombosis with brief discussion on the fundamental aspects and relevant background information for better understanding of the subject. In addition to its antithrombotic effects, we will dive insight into the cellular and molecular mechanisms of TCM as pharmacological regulators of platelet aggregation, coagulation, and fibrinolysis. With increasing awareness and understanding of the benefits and potentials of TCM, TCM products will in no doubt gain its broader applications in the treatment of thrombosis and associated disorders, which in turn will deepen our understanding of its pharmacological and molecular mechanisms. Finally, current review provides a perspective view on the future directions to TCM research on thrombosis.

1 Introduction

Thrombosis is a critical medical event that accounts for considerable morbidity and mortality all over the world[1], especially in cold climates. Clinical studies identified thrombosis induced by temperature drop as risk factor of myocardial infarction[2] and stroke[3]. It has been documented that low ambient temperature is associated with increased levels of circulating markers for blood coagulation including fibrinogen, plasminogen activator inhibitor 1 (PAI-1), von Willebrand Factor (vWF) and sCD40L. Moreover, in human umbilical vein endothelial cells (HUVECs), exposure to low temperatures induced expression of pro-thrombotic factors and inflammatory adhesion molecules[4], indicating that cold could cause endothelial injury.

Thrombosis is a complex process, which is accompanied by activation of platelet adhesion, aggregation, secretion function, intrinsic and extrinsic coagulation systems and inhibition of fibrinolysis, leading to blood coagulation and fibrin formation[5]. An array of antithrombotic drugs, including antiplatelets, anticoagulants and thrombolytic drugs, have been developed based on the mechanisms of thrombosis and has been in clinical use for the treatment of thrombotic diseases for a long time. Of the antiplatelet agents, cilostazol, clopidogrel and aspirin have been in clinical use worldwide. However, some of the antiplatelet agents can elicit various side effects including headache[6], internal bleeding, prolonged bleeding time, gastrointestinal bleeding and palpitation[7, 8], which impedes their applications for cardiovascular protection. Anticoagulants, such as heparin, have been used in preventing myocardial infarction since the early 20th century owing to their efficacy in inhibiting blood clotting. Although it had been discovered a century ago, it took a long time before this agent finally got into clinical applications. Anticoagulants can cause severe side effects such as bleeding, headache, fever and nausea[9]; thus, their clinical application has been constrained. Compared with antiplatelets and anticoagulants, fibrinolytic drugs have a major advantage in that they can enzymatically break down existing blood clots, thereby finding their wide use in the treatment of cardiovascular diseases[10]. Among fibrinolytic drugs, tissue-type plasminogen activator (t-PA) was approved in 1996 by the United States Food and Drug Administration (FDA) for intravenous injection against thrombosis. However, the major disadvantages of these thrombolytic drugs are their high cost and several side effects such as bleeding, nausea and hemorrhage, which limit their clinical use[11]. Therefore, to minimize the side effects and widen their applications, improvement of antithrombotic agents by the processes such as purification need to be carried out; or otherwise, new anticoagulants need to be developed.

Many TCM formulations have been used for thousands of years in clinical practice because of their proven efficacy, wide indications, excellent safety profile, and low toxicity[12]. In recent years, the use of TCM-based therapies has gained its ever-increasing acceptance worldwide[13] and broad applications as such anti-inflammatory, immunomodulatory[14], anti-atherosclerotic[15], anti-cancer[16], and anti-diabetic drugs[17]. TCM can also be sued for emergent public health crisis as manifested by its outstanding effectiveness against COVID-19, which has led to many deaths associated with blood clotting, and inclusion in the guideline for diagnosis and treatment of COVID-19[18]. The first case of successful cure of a COVID-19 patient who was discharged from the hospital following TCM treatment was reported on January 24th superscript 2020. Soon after, another case of a cured patient who received TCM therapy was reported[19]. These facts motivate more active application of TCM to patients with COVID-19 pneumonia[19].

Moreover, TCM is also frequently used for the clinical treatment of blood stasis in China. Many studies have focused on the antiplatelet, anticoagulant and fibrinolytic efficacy of TCM in preventing and treating thrombosis. In this article, we provide an overview on TCM preparations that are used for thrombotic events and comment on how they affect platelet aggregation, anticoagulation and fibrinolysis. As a quick reference guide, Table 1 summaries the effects of TCM and their active components with regard to thrombosis.

Table 1

Summary of TCMs and their effects on thrombosis.

Common name Latin name Active compounds Mechanism Reference
Safflower Carthamus tinctorius SY, HSYA Reduced platelet aggregation; inhibited PAF receptor; inhibited serotonin release; decreased intraplatelet Ca2+; inhibited TXA2 production; reduced GP IIb/IIIa expression 25, 26, 27, 28, 29, 30
Aconite Radix Aconiti praeparata Higenamine Reduced platelet aggregation; inhibited α2-adrenergic receptor; inhibited TXA2 receptor 32, 33, 34
Andrographis Andrographis paniculata AP1, AP3 Reduced platelet aggregation; decreased intraplatelet Ca2+; increase cGMP levels 38, 39, 40
Hawthorn Crataegus pinnatifida Eriodectyol Reduced platelet aggregation; inhibited TXA2 production; inhibited serotonin release 42, 43, 44, 45
Motherwort Leonurus japonicus. Spirolabdane diterpenoids Reduced platelet aggregation 47, 48, 49, 50
- Angelica pubescens Coumarin Reduced platelet aggregation; inhibited 5-LOX and COX-1 activity 52, 53, 54
Dong quai Angelica sinensis SF, LIG Reduced platelet aggregation; inhibited TXA2 and TXB2 production; suppressed 6-keto PGF generation; reduced vWF antigen and α-granule membrane protein 55, 56, 57, 58, 59, 60, 61, 62, 63
Corydalis Corydalis yanhusuo DHC, THB Reduced platelet aggregation; impaired thromboxane formation and phosphoinositides breakdown; decreased intraplatelet Ca2+; bound to ADP receptors P2Y1 and P2Y12, thrombin receptor PAR1 65, 66, 67
Hops Humulus lupulus L. XN Reduced platelet aggregation; inhibited TXA2 production; decreased intraplatelet Ca2+; decreased ROS accumulation and platelet mtDNA release 69, 70, 71
Liquorice Glycyrrhiza glabra Glycyrrhetinic acid, GL Inhibited thrombin; inhibited FXa 78, 79, 80
Feces Trogopterus Kaempferol coumaroyl rhamnosides Inhibited thrombin 83, 84
- Zanthoxylum nitidum var. tomentosum Toddalolactone Inhibited PAI-1 92
Pillbug Porcellio scaber Latreille PSLTro01 Activated plasminogen 94
Earthworm Eisenia fetida/Lumbricus rubellus EfP, LrP Activated plasminogen; directly degraded fibrin 95, 96, 97, 98, 99, 100, 101
Ginkgo Ginkgo biloba L. Ginkgolides, BB Reduced platelet aggregation; inhibited PAF receptor and glycine receptor; decreased intraplatelet Ca2+; inhibited TXA2 production; increase cAMP and cGMP levels 103, 104, 105, 106
Ginkgetin, isoginkgetin, bilobetin, amentoflavone Inhibited thrombin 107
EGb761 Increased TM and t-PA expression and activity 108, 109
Asian Ginseng Panax ginseng Ginsenoside Rgl, Rp3, Ro, Rp1, gintonin Reduced platelet aggregation; decreased intraplatelet Ca2+; inhibited TXA2 production; inhibited serotonin release; inhibited ATP release; impaired GPVI signaling 111, 112, 113, 114, 115, 116
Ginsenosides Rg2, Rg3 Inhibited FXa 117
Ginsenosides Rg1 Increased plasminogen activator secretion 118
Notoginseng Panax notoginseng Ginsenoside Rg1, notoginsenoside Fc Reduced platelet aggregation 119, 120, 121, 122
Notoginsenoside R1 Increased t-PA and u-PA expression; decreased PAI-1 activity 123
Danshen Salvia miltiorrhiza Bge. SA Reduced platelet aggregation; decreased TXB2 and vWF levels; increased 6-keto-PGF levels; increased cAMP level 125, 126, 127
Tanshinones Enhanced AT-III and PC activities; inhibited FXa; inhibited thrombin; inhibited FVIIa 127, 128, 129
- Decreased plasma PAI-1 levels; increased t-PA levels 127
- Coptis chinensis BBR Reduced platelet aggregation; inhibited TXA2 production; inhibited α2-adrenergic receptor 132, 133
BBR Inhibited thrombin 134
Chuanxiong Ligusticum wallichii Tetramethylpyra-zine, SF Reduced platelet aggregation; inhibited TXA2 production; reduced GP IIb/IIIa expression; reduced NO production; increased cAMP level 136, 137, 138, 139
- Inhibited thrombin 140
Ginger Zingiber officinale Gingerol, zingerone Reduced platelet aggregation; inhibited LOX and COX activity; inhibited TXA2 production; decreased intraplatelet Ca2+; reduced P-selectin and PAC-1 expression 142, 143, 144, 145, 146
Zingerone Inhibited FXa production and activity 146
- Celastrus orbiculatus NST-50 Reduced platelet aggregation; inhibited TXB2 production; increased 6-keto PGF generation 149
NST-50 Decreased PAI-1 levels; increased t-PA levels 149
Baical skullcap root Scutellaria baicalensis Georgi OroA, baicalin, WGN, WGNS Reduced platelet aggregation 151, 152, 153, 154
OroA, baicalin, WGN, WGNS Inhibited thrombin production and activity; inhibited FXa production and activity; inhibited vitamin K reductases; inhibited TF production and activity 152, 153, 154, 155, 156
OroA, baicalin, WGN, WGNS Decreased PAI-1 levels 152, 153, 154
- Dioscorea zingiberensis TSSN, diosgenin Reduced platelet aggregation 158, 159, 160
- Inhibited FVIII 160
Madder Rubia cordifolia - Reduced platelet aggregation; inhibited TXB2 production; increased 6-keto PGF generation 162
Purpurin Inhibited FVII 163
Purpurin Increased t-PA levels; activated pro-urokinase 162, 163
Astragalus Astragalus membranaceus AGS-IV Bound to prothrombin and AT-III 165
AGS-IV Decreased PAI-1 levels; increased t-PA levels 166
Peony root Paeoniae lactiflora Paeoniflorin Reduced platelet aggregation; inhibited TXA2 and TXB2 production; increased 6-keto PGF generation; modulated vWF-GP Ib interaction 167, 168, 169, 170
Paeoniflorin Increased t-PA activity; increased u-PA levels 169, 171
Turmeric Curcuma Longa Curcumin, curdione Reduced platelet aggregation; inhibited TXB2 production; decreased intraplatelet Ca2+; inhibited dense granule secretion; inhibited P-selectin expression; increased cAMP levels 173, 174, 175, 176, 177, 178
Curcumin Inhibited thrombin; inhibited FXa; inhibited FVII synthesis 179, 180
Curcumin Increased u-PA levels 181
Yellow mealworm Tenebrio molitor - Reduced platelet aggregation; decreased intraplatelet Ca2+; inhibited P-selectin and PAC-1 expression; improved NO production; inhibited ET-1 secretion 183
- Inhibited FXa production and activity 183
  1. safflower yellow; HSYA: hydroxysafflor yellow A; AP1: andrographolide; AP3: 14-deoxy-11,12 didehydroandrographolide; PAF: platelet activating factor; TXA2: thromboxane A2; GP: glycoprotein; cGMP: cyclic guanosine monophosphate; LOX: lipoxygenase; COX: cyclooxygenase; SF: sodium ferulate; LIG: Z-ligustilide; TXB2: thromboxane B2; PG: prostaglandins; vWF: von Willebrand Factor; DHC: dehydrocorydaline; THB: canadine; ADP: adenosine diphosphate; ATP: adenosine triphosphate; XN: xanthohumol; GL: glycyrrhizin; PAI-1: plasminogen activator inhibitor 1; EfP: E. fetida proteases; LrP: L. rubellus proteases; BB: bilobalide; TM: thrombomodulin; t-PA: tissue type plasminogen activator; u-PA: urokinase type plasminogen activator; SA: salvianolic acid; BBR: berberine; OroA: oroxylin A; WGN: wogonin; WGNS: wogonoside; TSSN: total steroidal saponin; AGS-IV: astragaloside IV; cAMP: cyclic adenosine monophosphate; AT-III: antithrombin III; PC: protein C; ET-1: endothelin-1

Antiplatelet drugs

Platelets play a key role in blood clotting and normal hemostasis. When vascular injury occurs, platelets initially tether to the subendothelial extracellular matrix through multiple receptors, including the glycoprotein (GP) Ib/V/IX complex binding to vWF as well as glycoprotein VI (GPVI) and αIIβ1 receptors on the platelet surface binding to the collagen components of the extracellular matrix. After adhesion, platelets undergo spreading, activation, and eventual aggregation to form a thrombus. Activated platelets also release granules and/or signals to facilitate the activation of nearby platelets and recruit them to the localized thrombus. This is done through positive feedback initiated by the formation of secondary signals mediated by cyclooxygenase (COX)-1 and 12-lipoxygenase (LOX) and granule secretion of small molecules known to activate the platelets, such as adenosine triphosphate (ATP), adenosine diphosphate (ADP), epinephrine, and serotonin. The most highly investigated small molecule released from the dense granule is ADP, which further signals the platelet purinergic receptors P2Y1 and P2Y12, and it is this pathway that is targeted clinically for prevention of occlusive thrombosis. Following platelet activation, COX-1 converts arachidonic acid (AA) into free fatty acids prostaglandins (PG)E2 and thromboxane A2 (TXA2), and 12-LOX catalyzes the formation of eicosanoids [12(S)-hydroxyeicosatetraenoic acid (12(S)-HETE)]. These bioactive lipid products interact with their respective G protein-coupled receptors (GPCRs) to reinforce platelet activation[20]. Platelet adhesion and their subsequent aggregation and recruitment to the site of injury form the platelet thrombus consisting of a “core” of tightly packed P-selectin-positive platelets surrounded by a “shell” of loosely packed platelets[21-22].

As a result, platelets can be an effective target for the prophylaxis and treatment of thrombosis and the associated diseases. The following section introduces some specific TCM drugs for their effects on platelets and the known mechanisms of action (Figure 1).

Fig. 1 TCMs and their effects on platelet.
Fig. 1

TCMs and their effects on platelet.

Safflower

Carthamus tinctorius, known as safflower, is widely utilized in TCM for various medical conditions, namely dysmenorrhea, amenorrhea, postpartum abdominal pain and mass, trauma and pain of joints[23]. Safflower is a major fraction of Xuebijing Injection, which is used for the syndrome of static blood and poison of COVID-19[24].

Safflower yellow (SY), one of the flavonoids isolated from safflower, inhibited the ADP-induced platelet aggregation in rats. In rabbit models, SY inhibited platelets adhesion, reduced the serotonin release, and lowered the level of free intraplatelet Ca2+ induced by platelet activating factor (PAF)[25-26]. Hydroxysafflor yellow A (HSYA), another active flavonoid isolated from safflower can inhibit PAF receptor activity and suppress the PAF-induced adhesion of platelets in rabbits[27]. HSYA can also suppress the production of TXA2 without affecting plasma PGI2 concentrations[28]. In another study, the aqueous extract of safflower was found to exhibit antithrombotic activity against venous thrombosis and pulmonary embolism in vivo as indicated by significant decrease in the thrombus weight[29].

In a clinical trial, patients with acute coronary syndrome were treated with safflower injection in addition to routine Western medicine therapy with aspirin, clopidogrel, statins, percutaneous coronary intervention, etc. As a result, their clinical symptoms of angina and electrocardiogram were improved. Ex vivo analysis later revealed reduced expression of GP IIb/IIIa, which might be a mechanism by which safflower suppresses platelet aggregation[30].

Numerous studies have demonstrated the efficacy of safflower in activating blood circulation and removing the stasis. Yet, further studies are needed for better understanding of the pharmacological properties and underlying mechanisms of safflower. Moreover, new bioactive components need to be identified to strengthen and expand the therapeutic effectiveness of safflower as TCM therapeutics. In addition, more experiments including in vitro, in vivo and clinical studies should be encouraged to identify the possible side effects, toxicity and interactions with other drugs.

Aconite

Aconite (Radix Aconiti praeparata) has been used in TCM practice in many Asian regions for the treatment of various diseases such as collapse, syncope, painful joints, edema, and bronchial asthma[31].

Higenamine was initially isolated from aconite by water and identified as the active cardiotonic component of aconite. In both the acute mouse thrombosis model and the rat arterio-venous shunt (AV-shunt) model, oral administration of higenamine increased the rate of recovery and decreased the weight of thrombus. Mechanically, higenamine into inhibited both human and rat platelet aggregation induced by ADP, collagen and epinephrine[32]. Higenamine could also directly block α2-adrenergic receptor leading to the inhibition of platelet aggregation[33]. In another study, higenamine was shown to antagonize AA-induced platelet aggregation likely by direct blocking of the TXA2 receptor[34].

Existing evidence indicates aconite a valuable therapeutic TCM for thrombotic disorders. However, the underlying mechanisms of aconite have not been elucidated. Additionally, some of the studies were conducted with small sample-size and the results might not be entirely reliable. Thus, there is a need for in-depth investigations on the mechanisms of aconite action, as well as well-designed preclinical studies and clinical trials to test the safety and clinical value of the drug.

Andrographis

Andrographis paniculate, as a traditional medicine in India, China and Scandinavia, has been widely used for common cold and digestive disorders. It exhibits a wide spectrum of biological activities of therapeutic importance, including antibacterial, antiviral[35, 36] and anti-inflammatory[37] properties.

In vitro studies revealed that Andrographis paniculate could reduce platelet aggregation. One study found that the alcohol extract of Andrographis paniculate and its active diterpenoids, andrographolide (AP1) and 14-deoxy-11,12 didehydroandrographolide (AP3), inhibited thrombin-induced platelet aggregation[38]. Moreover, Andrographis paniculate and andrographolide inhibited PAF-induced human blood platelet aggregation in a dose-dependent manner[39]. In another research, andrographolide was found to inhibit collagen-induced platelet aggregation and relative Ca2+ mobilization. The antiplatelet activity of andrographolide could be attributed to an increase in cyclic guanosine monophosphate (cGMP)/PKG to inhibit the p38 MAPK/hydroxyl radical/NF-κB/ERK2 cascade in human platelets[40].

Hawthorn

Hawthorn (Crataegus pinnatifida), widely distributed in the northeast part of China was well-known as both a medicine and food material. In TCM, hawthorn has been used for digestive problems, hyperlipidemia, poor circulation, and dyspnea[41].

In vivo, hawthorn significantly inhibited carrageenan-induced mice tail thrombosis[42]. Eriodectyol, isolated from the leaves of hawthorn, has been tested in vivo for its antithrombotic activity in transgenic zebrafish. As a result, eriodectyol showed significant antithrombotic activity[43]. An in vitro study reported that hawthorn extract inhibited ADP-induced platelet aggregation and [14C] serotonin release[44]. The hydro-alcoholic extract of the flowery heads of hawthorn inhibited the in vitro biosynthesis of TXA2[45].

More well-designed studies are needed for obtaining more accurate results to decipher the pharmacological characteristics of hawthorn. Hawthorn holds the potential as a therapeutic agent against thrombosis and as a good candidate for the development of new antithrombotic compounds.

Motherwort

Leonurus japonicus, commonly called Chinese motherwort, is an herbaceous flowering plant native to Asia. For thousands of years in China, the aerial part of Leonurus japonicus has been used to treat menoxenia, dysmenorrhea, amenorrhea, and other diseases in women. In a recent study, motherwort was reported to produce beneficial effects on the cardiovascular system [46].

Previous investigations on motherwort have led to the isolation of many diterpenoids, particularly the spirolabdane diterpenoids, by ethanol extraction[47]. It has been reported that bis-spirolabdane diterpenoids 1, 2 significantly reduced ADP-induced in vitro platelet aggregation, which is related to the absolute configuration of the spiral ring[48]. Qualitatively the same result was reported by another in vitro study with a sesquiterpenoid extracted from motherwort by EtOH[49]. Although not typically used intravenously, a clinical study with 105 patients found a decrease in platelet aggregation after 15 days of daily intravenous infusions of motherwort[50].

Based on the results from both basic and clinical studies, motherwort might be recommended as a drug for thrombosis treatment. However, further researches are required to uncover the possible adverse effects and optimal dosages, as well as other pharmacodynamic and pharmacokinetic characteristics of this medicine.

Angelica pubescens

Angelica pubescens has long been used as a traditional Chinese herbal medicine for its applications in the treatment of infections, inflammation, and pain[51]. Certain coumarin constituents could be isolated from Angelica pubescens by ethanol extraction. Linoleic acid, osthol, osthenol and two polyacetylenes were found to be the most active compounds responsible for the inhibitory activity of the dichloromethane extract from the roots of Angelica pubescens on 5-LOX and COX-1 in vitro[52]. As one of the main bioactive coumarin constituents, osthole could inhibit platelet aggregation in vitro and interfere with calcium influx and cyclic nucleotide phosphodiesterases[53]. Columbianadin (CBN), another bioactive coumarin, acted against platelet activation through inhibition of the PLCγ2-PKC cascade and the activation of Akt and ERKs/JNKs. In experimental mice, CBN increased the occlusion time of thrombotic platelet plug formation[54].

Besides Angelica pubescens, many other TCM preparations also contain coumarin constituents, indicating that coumarin is contained in multiple sources. More reliable basic and clinical studies can help work out the mechanisms and side effects of coumarin for its clinical applications.

Dong quai

Dong quai (Angelica sinensis) is a Chinese herbal medicine used for the treatment of menstrual cramping, irregular menses, and menopausal symptoms. It is a constituent of Xuebijing Injection, which is used for the treatment of static blood syndrome and poisoning in warm diseases associated with COVID-19[24]. Dong quai contains a number of antithrombotic constituents. Sodium ferulate (SF), or 3-methoxy-4-hydroxy-cinamate sodium, is an active aqueous extract from Angelica sinensis, Cimicifuga heracleifolia, Lignsticum chuangxiong, and other plants[55]. SF inhibits experimental thrombosis in rat carotid-jugular extracorporeal shunt model[56]. SF inhibits in vitro platelet aggregation induced by ADP thrombin or collagen[57]. The mechanism of SF action appears to be the inhibition of cyclooxygenase and TXA2 synthase. Inhibition of TXA2 production decreases TXA2/prostaglandin I2 (PGI2) ratio to shift the balance away from platelet aggregation[58, 59]. In rabbit platelets, SF inhibits the generation of thromboxane B2 (TXB2) from [14C] AA in a dose-dependent manner[60]. SF also suppresses the generation of 6-keto PGF in rabbit aorta. However, SF has a stronger inhibitory effect on TXB2 generation[61]. In addition, Z-ligustilide (LIG) is a characterized 3-n-alkylphthalide constituent of Angelica sinensis. A study showed that LIG significantly reduced arterial thrombus weight in an AV-shunt thrombosis in rats and inhibited platelet aggregation induced by ADP ex vivo[62].

In a 3-week clinical study, intravenous administration of Dong quai reduced the rate of platelet aggregation, vWF antigen, α-granule membrane protein, and TXB2[63].

Numerous investigations have provided the evidence for the efficacy of Dong quai, especially its major constituents SF, in suppressing thrombosis and the related diseases and delineated the underlying mechanisms for the pharmacological actions of SF.

Corydalis

Corydalis yanhusuo is a perennial herb which is widely distributed in the northeastern region of China. The tuber of Corydalis yanhusuo has been used as a local TCM for thousands of years[64]. Protopine isolated from the methanol extract of Corydalis tuber was reported to inhibit in vitro platelet aggregation by impairing thromboxane formation and phosphoinositides breakdown and decreasing intracellular calcium concentration[65]. The platelet bio-specific extraction combined with HPLC analysis found that potential active antiplatelet aggregation components in Corydalis yanhusuo were dehydrocorydaline (DHC) and canadine (THB). The platelet aggregation stimulated by thrombin, ADP and AA were inhibited by DHC and THB. In addition, DHC inhibited platelet aggregation likely by a mechanism involving the ADP receptors P2Y1 and P2Y12, and the effect of THB on platelet function might be related to its binding to thrombin receptor PAR1 mediating the Gi signaling pathway[66-67]. These studies on extracts of Corydalis yanhusuo have revealed that Corydalis yanhusuo holds great promise as an anti- thrombosis agent.

Hops

Hops (Humulus lupulus L.) has been used in TCM to treat insomnia, restlessness, dyspepsia, and lack of an appetite[68].

Xanthohumol (XN) isolated by alcohol is the major prenylated flavonoid of the hops. Using ferric chloride (FeCl3)-induced carotid artery injury, inferior vena cava ligation model, platelet function tests demonstrated that XN uniquely prevented both venous and arterial thrombosis by inhibiting platelet activation[69]. In vitro experiments reported that XN attenuated ADP- and collagen-induced platelet reactivity in whole blood[70-71]. It appears that XN inhibits the PI3-kinase/Akt, p38 MAPK, and PLCγ2-PKC cascades, leading to inhibition of the TXA2 formation, thereby inhibition of [Ca2+]i and platelet aggregation[71]. Another study showed that XN was highly effective in inhibiting platelet activation by decreasing ROS accumulation and platelet mitochondrial DNA (mtDNA) release without incurring a bleeding risk[69]. These findings suggested XN a promising new class of antiplatelet agents.

Dong quai inhibits thrombosis by impeding platelet adhesion. Safflower, aconite, andrographis, hawthorn, Angelica pubescens, dong quai, corydalis and hops have inhibitory effects on platelet activation, thereby preventing thrombus formation. Besides, safflower, aconite, andrographis, hawthorn, motherwort, angelica pubescens, dong quai, corydalis and hops reduce platelet aggregation to inhibit thrombosis.

Anticoagulation drugs

Coagulation is the main portion of the blood-clotting pathway involving the production of thrombin that leads to conversion of fibrinogen to fibrin[72]. The formation of the prothrombin activator complex, which is necessary for thrombin production and activation, occurs through two different pathways: the intrinsic pathway and the extrinsic pathway[73]. In the extrinsic pathway, tissue factor (TF) binds to factor II (FVII) to initiate the process of coagulation via promoting the proteolysis of FVII and conversion into FVIIa. The TF/FVIIa complex proteolytically cleaves traces of factor IX (FIX) and factor X (FX) into FIXa and FXa, respectively. This allows FXa to associate with cofactor FVa to form a prothrombinase complex and initiate blood coagulation. This pathway is formerly known as the common pathway, which develops through conversion of prothrombin to thrombin, followed by activation of fibrinogen to fibrin (monomers), the labile association of fibrin monomers, and the final formation of covalent bounds between adjacent fibrin strands[74]. The intrinsic pathway only serves as an amplification loop initiated by the extrinsic pathway. It begins with FXII, high-molecular-weight (HMW) kininogen, prekallekerin and FXI, which results in activation of FXI. Activated FXI then activates FIX which in turn acts with its cofactor (FVIII) to form a prothrombinase complex on a phospholipid surface to activate FX[75] eventually inducing the common pathway.

Anticoagulation medications are widely used in clinical practice, as cardiac-related emergencies are on the rise[76]. In recent studies, several TCM drugs have been found to have anticoagulation potentials (Figure 2).

Fig. 2 TCMs and their anticoagulation effects.
Fig. 2

TCMs and their anticoagulation effects.

Licorice

Liquorice or licorice is an important medicinal herb with nutritional and therapeutic values. It is mainly obtained from the root of Glycyrrhiza glabra. It is traditionally used for treating asthma, hoarseness of voice, cough, and lung diseases[77]. Recently, licorice has also been used for COVID-19 treatment as a major component of Jinhua Qinggan Granules, Lianhua Qingwen Capsule and Fangfeng Tongsheng Pills[24].

Glycyrrhizin (GL) is a natural compound obtained from the aqueous extract of the root of licorice. GL has been previously identified as a thrombin inhibitor[78]. Intravenous administration of GL caused a dose-dependent reduction in thrombus size in a venous thrombosis model of rats that combines stasis and hypercoagulability. GL was also found to prevent thrombosis in an AV-shunt model[79]. Glycyrrhetinic acid, a pentacyclic triterpene derived from licorice by ethanol, inhibits FXa noncompetitively. Intragastric administration of glycyrrhetinic acid reduced thrombus weight in an in vivo study[80].

Trogopterus feces

The dried excrement of flying squirrels (Trogopterus xanthipes), also known as “wulingzhi”, has a long history as an important pharmaceutical ingredient of TCM[81]. Trogopterus feces have been reported to promote blood circulation and resolve stasis[82].

A chemical investigation of the ethyl acetate extract has led to the isolation of six fatty acid esters, two aliphatic alcohols and two diterpenes. In an anticoagulative assay, three kaempferol coumaroyl rhamnosides elicited significant antithrombin activities[83]. In another study, four fatty acid esters isolated from Trogopterus feces were tested for their antithrombin activities using the thrombin time (TT) method assay. The results showed that two of the compounds significantly prolonged TT with a good dose-effect relationship[84]. However, in vivo validation of the pharmacological effects of Trogopterus feces is still missing.

Liquorice and Trogopterus feces inhibit thrombin, contributing to prevention of thrombosis. Moreover, liquorice also hinders thrombosis by inhibiting FX activation.

Fibrinolytic drugs

Dissolution of clot is controlled by the fibrinolytic system. Briefly, the production of plasmin from plasminogen with the aid of plasminogen activators leads to accelerated degradation of blood clots, along with the generation of fibrin degradation products, including D-dimers[85]. Plasminogen activators includes t-PA and urokinase type plasminogen activator (u-PA)[86]. t-PA is stored in the endothelium of the vessel wall and released at the site of a fibrin thrombus, while u-PA is mainly found in urine and secreted by kidney and urinary tract epithelial cells[87]. Besides, fibrinolytic process is also regulated by PAI-1, thrombinactivatable fibrinolysis inhibitor (TAFI), α2-antiplasmin, and α2-macroglobulin[88].

Fibrinolytics have been widely used clinically. Plasminogen activators were introduced into clinical practice in the 1980s for indications of myocardial infarction and in the 1990s for ischemic stroke[89]. Large clinical trials showed the benefits of recombinant tissue t-PA in acute ischemic stroke[90]. But because of the high cost, the clinical application of recombinant tissue t-PA is limited. Recent studies uncovered that certain TCM drugs possessed strong activating effects on the fibrinolytic system. The following contents and Figure 3 describe a few representative TCM drugs with fibrinolytic potentials and the mechanisms for their actions.

Fig. 3 TCMs and their effects on fibrinolytic system.
Fig. 3

TCMs and their effects on fibrinolytic system.

Fig. 4 TCMs and their multiple effects on thrombosis.
Fig. 4

TCMs and their multiple effects on thrombosis.

Zanthoxylum nitidum var. tomentosum

The root and rhizome of Z. nitidum var. tomentosum are commonly used in Chinese folk medicine to treat traumatic injury, rheumatic, and snakebite[91]. A study found that ethanol extract from Z. nitidum var. tomentosum exhibited PAI-1 inhibitory activity and identified toddalolactone as the main active component by chromogenic assay, with its effect confirmed by clot lysis assay. Toddalolactone was found to prevent the formation of the PAI-1/u-PA complex. Intraperitoneal administration of toddalolactone for 2 weeks significantly reduced the thrombus weight and intravascular thrombosis, resulting in improved blood circulation and blood flow in a mouse model of FeCl3-induced artery thrombosis[92]. Inspired by these findings, we speculate that PAI-1 inhibition is an important pharmacological basis for TCM to improve blood circulation and for the future development of new compounds for antithrombotic therapy.

Porcellio scaber Latreille

The terrestrial isopod, Porcellio scaber Latreille, is a cosmopolitan non-conglobating species, which has been used as a traditional Chinese animal medicine for thousands of years. Existing evidence supports that the extracts obtained from P. scaber Latreille can produce anti-inflammation, antioxidant and anti-tumor activities[93]. PSLTro01, a single-chain protein isolated from P. scaber Latreille by water extraction, inhibited carrageenan-induced tail thrombosis in an in vivo model[94]. Analysis of the fibrinolytic activity of PSLTro01 with plasminogen-rich and plasminogen-free fibrin plates showed that PSLTro01 does not directly degrade fibrin but acts as a plasminogen activator[94]. These findings suggest P. scaber Latreille as a new source of fibrinolytic agents for thrombolytic medicine.

Earthworm

Earthworm (Eisenia fetida and Lumbricus rubellus) has been used as a drug in clinics for at least the past two thousand years. As a Chinese traditional animal medicine, earthworm is described to have the antipyretic and diuretic effects for jaundice. In the Guidelines for the Diagnosis and Treatment of Novel Coronavirus Pneumonia (Trial Version 7), earthworm is recommended for syndrome of cold-damp stagnation of lung[24]. Earthworms are rich sources of proteases with thrombolytic potential. Recently, groups of fibrinolytic isozymes have been isolated from different earthworm species[95], such as E. fetida proteases (EfP)[96] and L. rubellus proteases (LrP)[97]. L. rubellus produces six LrP variants and E. fetida yields seven variants of fibrinolytic enzymes[96, 97]. These enzymes have better specificity towards fibrin in comparison with plasminogen activators such as t-PA and u-PA. LrPs are direct-acting fibrinolytic enzymes which activate plasminogen. In vivo animal models indicate that they can be orally administered for therapeutic purpose[98]. The main advantage of LrPs is their good safety profiles with a low risk of bleeding disorders[99]. Boluoke® is a commercially available LrPs approved by the China Food and Drug Administration. EfPs belong to serine proteases, a few of which are quite similar to LrPs with respect to functions. EfP-III-1 has the strongest fibrinolytic activity among the isozymes and high stability as well[100], and it acts as a t-PA-like activator to initiate the plasmin-antithrombus pathway[101].

Pillbug and earthworm facilitate plasminogen activation, thereby thrombolysis. In addition, Zanthoxylum nitidum var. tomentosum inhibits PAI-1 leading to thrombolysis activation.

Multiple pathway drugs

Ginkgo

Ginkgo biloba has been used in TCM for thousands of years. Its seeds are used for chronic cough, asthma, leucorrhoea, and urinary incontinence or frequency, and its leaves for cardiovascular disease, such as angina, hyperlipidemia, hypertension and cerebral vasospasm[102]. Ginkgo inhibits platelet aggregation. Ginkgolides are one of the main active components of ginkgo extract by alcohol. Among them, ginkgolide A (GA), ginkgolide B (GB), and ginkgolide C (GC) possess inhibitory effects on platelet aggregation with GB being the most potent antagonist of PAF receptor and glycine receptor[103-104]. In addition, GB can also activate matrix metalloproteinase-9, increase intracellular cyclic adenosine monophosphate (cAMP) and cGMP production, inhibit the intracellular Ca2+ elevation, and prevent TXA2 formation in collagen-stimulated platelets, leading to inhibition of platelet aggregation[105]. Bilobalide (BB) is another active component of alcohol extract from ginkgo. In a zebrafish thrombosis model, BB and ginkgolides significantly increased zebrafish heart red blood cells (RBC) intensity, implying the reduced thrombosis in zebrafish[106].

In addition, ginkgo extracts also have effects on the coagulation system. For example, the biflavones isolated from ginkgo, including ginkgetin, isoginkgetin, bilobetin and amentoflavone, can produce strong inhibitory effects on human thrombin[107].

The antithrombotic effect of ginkgo could be ascribed to the activation of the thrombolytic system. The ethanol extract from ginkgo promotes thrombolysis as streptokinase does[108]. EGb761, an extract of ginkgo leaves, significantly upregulates the expression of Krüppel-like factor 2 (KLF2) in human endothelial cells to increase the protein levels and activities of thrombomodulin (TM) and t-PA conferring its antithrombotic effect[109].

These findings provide convincing evidence for the beneficial effects of Ginkgo extract on thrombotic diseases. The widespread use of Ginkgo extract as an herbal supplement for cardiovascular and cerebrovascular conditions, cancer, arthritis, etc. suggests that it could be taken as a source for the discovery and development of novel drugs for better thrombosis treatment.

Panax ginseng

Panax ginseng, as a traditional Chinese herbal medicine, has been used for the treatment of human diseases particularly cardiovascular disease for thousands of years in China[110]. It has recently been used as one of major components of the prescription for the syndrome of internal blockade and external collapse in patients with severe COVID-19. Panax ginseng restores pulse to relieve desertion, promotes fluid production and tranquilizes the mind[24].

Panax ginseng extract isolated by water and alcohol was shown to effectively prevent carotid arterial thrombosis in rats[111]. In line with the above observations, Panax ginseng extract inhibits the U46619 (a stable synthetic analog of the endoperoxide prostaglandin PGH2 acting as a thromboxane A2 (TXA2) receptor agonist)-, AA-, collagen- and thrombin-induced platelet aggregation in rabbits and reduces serotonin secretion in vitro[111]. Ginsenoside Rgl, an alcohol extracts from Panax ginseng inhibited adrenaline- and thrombin-induced platelet aggregation and serotonin release, which might result in the reduction of [Ca2+]i elevation[112]. Ginsenoside-Rp3, a derivative of ginsenoside-Re, is a natural ginsenoside fraction isolated from Panax ginseng. Ginsenoside-Rp3 has been found to protect mice from thrombosis evidently by inhibiting collagen-, ADP-, and thrombin-induced platelet aggregation[113]. Mechanically, ginsenoside Rp3 inhibited granule secretion, integrin αIIbβ3 activation, MAPK signaling, Src, PLCγ2, and PI3K/Akt activation, and VASP stimulation[113]. Ginsenoside Ro reduced TXA2 production by inhibiting AA release via decreasing the phosphorylation of cPLA, p38-mitogen-activated protein kinase, and c-Jun N-terminal kinase1[114]. Another Panax ginseng extract Ginsenoside Rp1 was found to inhibit arteriovenous shunt thrombus formation in rats and ex vivo (rat) platelet aggregation and ATP secretion[115]. The signaling mechanisms for Ginsenoside Rp1 actions involve inhibition of collagen-induced platelet activation through modulation of early GPVI signaling events and stimulation of VASP and inhibition of ERK2 and p38-MAPK signaling as well[115]. Gintonin, a recently discovered non-saponin fraction of Panax ginseng, was found to inhibit agonist-induced platelet activation and thrombus formation through suppressing GPVI signaling, including activation of SFK, Syk, PLCγ2, MAPK, and PI3K/Akt[116].

Moreover, Panax ginseng and part of its major functional components (ginsenosides) also have anticoagulation effects as indicated by the delay of human blood clotting time[117]. For instance, chromogenic substrates assay showed ginsenosides Rg2 and Rg3 exhibited anti-FXa activities. And they were further molecularly docked with human FXa protein[117]. Additionally, Panax ginseng can activate the thrombolytic system. Ginsenoside Rgl significantly increased the secretion of plasminogen activator in HUVECs, indicating its thrombolytic property[118].

These studies demonstrate that ginsenosides and other active constituents confer the therapeutic efficacy of ginseng by regulating different signaling pathways. Future studies may be directed to delineate the specific anti thrombotic mechanism for each of the active constituents.

Panax notoginseng

Panax notoginseng has a long history of use in TCM owing to its hemostatic and cardiovascular effects[119]. Panax notoginseng is widely described for its antiplatelet activity. Raw and steamed Panax notoginseng significantly inhibited platelet aggregation and plasma coagulation in rats and collagen-induced platelet aggregation under in vitro conditions[119]. Panax notoginseng saponins (PNS), the principal constituents derived from Panax notoginseng by alcohol extraction, mainly contain ginsenoside Rg1, ginsenoside Rb1, notoginsenoside R1 and the like. A study showed that PNS suppressed thrombin-induced platelet aggregation in vitro and effectively improved hypercoagulable state in rats. PNS-induced activation of PPAR-γ and its downstream PI3K/Akt/eNOS pathway is believed to play the central role[120]. Ginsenoside Rg1 from Panax notoginseng attenuated arterial thrombus formation in the mouse model and inhibited platelet aggregation induced by ADP, collagen, thrombin, and U46619 via the inhibition of PKC and ERK pathway[121]. Notoginsenoside Fc, another main composition of PNS was found to suppress thrombin-, collagen- and ADP-induced platelet aggregation by inhibiting PLCγ2, DAG-PKC-TXA2 and IP3-[Ca2+]i. In the in vivo study, FeCl3 induced thrombosis in rat femoral artery was significantly alleviated by administration of notoginsenoside Fc[122].

A study reported that notoginsenoside R1 increased the synthesis of t-PA and decreased the activity of PAI-1 in cultured human endothelial cells. Also, in cultured human pulmonary artery smooth muscle cells, notoginsenoside R1 increased the fibrinolytic potentials by increasing the production of t-PA and u-PA[123].

The findings from the above-described studies form the scientific basis for practical uses of the over-ground part of Panax notoginseng. More attention should be paid to the efficacy and safety of Panax notoginseng in clinical use for the treatment of thrombotic disease.

Danshen

Danshen is the dried root of Salvia miltiorrhizae and is mainly used to treat and prevent cardiovascular disease, hyperlipidemia, and cerebrovascular disease throughout the world[124]. It is included in the Guidelines for the Diagnosis and Treatment of Novel Coronavirus Pneumonia (Trial Version 7) and recommended for COVID-19 patients with qi-yin deficiency syndrome[24].

One of the beneficial effects of Danshen is the inhibition of platelet aggregation. Salvianolic acid (SA) is one of water-soluble phenolic acids extracted from Danshen, and it reportedly improved regional cerebral blood flow after ischemia in rat thrombosis model[125]. SA significantly inhibited platelet aggregation induced by collagen, ADP, and AA both in vitro and in vivo[125]. In another study, salvianolic acid A (SAA) significantly reduced thrombus weight and increased cAMP level in a rat model of arteriovenous shunt. In vitro, pretreatment with SAA significantly inhibited platelet aggregation induced by various agonists and increased cAMP level in platelets activated by ADP[126]. Danshen extracts by ethyl acetate inhibited ADP-induced platelet aggregation, reduced the weight of FeCl3-induced thrombus in rat common carotid artery, decreased plasma TXB2 and vWF levels, and increased 6-keto-PGF1α levels[127].

Danshen can also elicit anticoagulation effects and prevent thrombosis. In a thrombosis model induced by FeCl3 in rat common carotid artery, ethanol extracts from Danshen enhanced the activities of antithrombin III (AT-III) and protein C (PC)[127]. EA extracts also showed strong activity against thrombin and FXa. Four marker compounds cryptotanshinone, tanshinone I, dihydrotanshinone I and tanshinone IIA with potential thrombin/FXa inhibitory activity have been identified from Danshen. Molecular docking study showed that all these four tanshinones could interact with certain key amino acid residues of the thrombin/FXa active cavities[128]. The fatty acids isolated from Danshen (linolenic, linoleic, and oleic acids) could bind to FVIIa and consequently prevent binding of soluble TF to FVIIa[129].

The antithrombotic effect of Danshen might be due to its role in activating the thrombolytic system. Ethanol extracts of Danshen decreased the plasma level of PAI-1 but increased that of t-PA to reduce FeCl3-induced common carotid artery thrombus in rats[127]. Although extensive researches have reported the inhibitory effects of Danshen in thrombosis, further in-depth investigations on the mechanisms of actions produced by different Danshen extracts are required, and preclinical studies and clinical trials are needed for validating their therapeutic efficacy and safety profile.

Coptis chinensis

Coptis chinensis is a well-recognized traditional herb and widely used in food and medicinal applications. Processed Coptis chinensis products can fight against severe skin disease, dysentery, gastroenteritis and diabetes[130]. In the recent public health crisis caused by SARS-CoV-2, Coptis chinensis finds its application to treat COVID-19 patients, especially those with syndrome of flaring heat in qifen and yingfen[24]. Berberine (BBR) is a natural alkaloid isolated from the plant Coptis chinensis and has gained its popularity as an antimicrobial for treating dysentery and infectious diarrhea[131]. A study documented that berberine has significant inhibitory effect on platelet aggregation induced by ADP, AA, collagen or calcium ionophore A23187 in rabbits. The mechanism for the effect has been ascribed to the inhibition of the synthesis of TXA2. In rabbits, the plasma prostacyclin level was reduced after intravenous administration of BBR[132]. BBR also inhibits platelet aggregation as a partial agonist of platelet α2 adrenoceptors[133].

Moreover, enzymatic assay confirmed that BBR can act as a thrombin inhibitor. Direct binding studies using surface plasmon resonance demonstrated that BBR interacts directly with thrombin. Competitive binding assay further unraveled that BBR binds to the same argartroban/thrombin interaction site[134].

Apparently, owing to its wide range of sources, low toxicity and cost, BBR has the potential to become a novel agent for the treatment of thrombotic disorders.

Chuanxiong

Ligusticum wallichii (Chuanxiong) is a Chinese medicinal herb that has been used orally with other herbs for heart and brain diseases for thousands of years[135].

The inhibitory effect of Chuanxiong on platelet aggregation has been reported. Tetramethylpyrazine, the key component responsible for the antiplatelet properties which is an alcohol extract of Chuanxiong, acts by inhibiting phosphoinositide breakdown and TXA2 formation[136], increasing intracellular cAMP and reducing GP IIb/IIIa expression[137]. Tetramethylpyrazine also increases nitric oxide production in human platelets via upregulating eNOS protein expression[138]. Another study showed that tetramethylpyrazine inhibited ADP-induced platelet aggregation via suppressing TXA2 secretion and intracellular Ca2+ mobilization in platelets following suppression of Akt phosphorylation[139]. SF can also be extracted from Ligusticum wallichii in addition to Angelica sinensis. And its antiplatelet effect has been discussed in the Angelica sinensis subsection.

Additionally, Chuanxiong can affect the coagulation system, as demonstrated by previous data showing that several compounds isolated from Chuanxiong act as thrombin inhibitors[140]. These findings provide preliminary evidence in support of Chuanxiong as a potent therapeutic herbal medicine against thrombotic vascular diseases.

Ginger

Ginger is the rhizome of Zingiber officinale, a perennial plant of the family Zingiberaceae. Ginger has broad applications as folk medicine in Chinese history for the treatment of several gastrointestinal and respiratory diseases owing to its stomachic, antiemetic, hemostatic and cardiotonic effects[141]. It is also included in the prescription for cold-dampness obstructing lung syndrome and stagnation of damp toxicity in lung syndrome for COVID-19 patients[24].

The most well-known traditional medicinal application of ginger is to promote the blood circulation via removing blood stasis with the mechanism related to its antiplatelet aggregation activity. An in vitro study demonstrated that the aqueous ginger extract inhibited platelet aggregation induced by ADP, epinephrine, collagen, and AA and reduced the synthesis of prostaglandin-endoperoxides, thromboxane and prostaglandins in platelets[142]. Additionally, ginger is effective in inhibiting the biosynthesis of eicosanoids with reduced TXB2 level in human[143]. Gingerol, the active component of ginger and its synthetic analogs G3-G6, were demonstrated to inhibit human platelet activation via inhibiting COX activity to mitigate AA-induced platelet serotonin release and aggregation[144]. Another in vitro study by the Chrono Log whole blood platelet aggregometer corroborated the antiplatelet effects of 8-gingerol, 8-shogaol, 8-paradol, and other gingerol analogues isolated from ginger by showing that these compounds elicit their antiplatelet activities in the presence of AA[145]. The underlying mechanism may be related to the inhibition of COX-1 enzyme to minimize the production of TXA2. Moreover, 8-paradol was proved to be the most potent inhibitor of COX-1 thereby platelet aggregation among the various gingerol analogues[145]. Zingerone, a phenolic alkanone found in Zingiber officinale, antagonizes thrombus formation in a FeCl3-induced carotid artery thrombosis model[146]. The results obtained from an ex vivo pulmonary thrombosis model showed that the formation of thrombi in the mouse lungs and associated mortality were significantly lower in mice treated with zingerone than in non-treated control animals[146]. Mechanically, zingerone inhibited platelet aggregation induced by ADP and U46619 likely by its suppressing the activation of PKC, mobilization of intracellular Ca2+, and expression of P-selectin and PAC-1[146].

Besides, inhibition of the coagulation system also contributed to the suppressive effect of zingerone on thrombosis. Zingerone inhibited the catalytic activity of FXa on its substrate S-2222, as well as FX production by FVIIa, in a dose-dependent manner[146].

Celastrus orbiculatus

Celastrus orbiculatus is used as a folk medicine for various types of diseases in northern China[147]. It has been long used to invigorate blood circulation, alleviate pain, tranquilize and allay excitement[148].

NST-50 is a bioactive fraction of C. orbiculatus fruit extracted by ethanol. A mouse study showed that NST-50 treatment improved acute pulmonary embolism in vivo[149]. Moreover, in a rat model of FeCl3-induced carotid arterial thrombus, NST-50 significantly reduced the wet weight of thrombus. This study also demonstrated that NST-50 decreased the plasma level of TXB2 but increased that of 6-keto-PGF1a. In vitro experiments exhibited that NST-50 inhibited platelet aggregation to evoke its antithrombotic efficacy[149]. Moreover, NST-50 treatment resulted in reciprocal changes of t-PA and PAI-1 with the former being significantly increased whereas the latter decreased in the FeCl3-induced carotid arterial thrombus rat model, indicating that NST-50 regulates the function of the fibrinolytic system to antagonize thrombus[149]. This study lays the pharmacological groundwork for the clinical applications of NST-50 to the treatment of acute coronary syndrome, venous thromboembolisms, and cerebrovascular thrombosis. The findings also indicate Celastrus orbiculatus as a natural source for the discovery and development of new antithrombotic compounds for clinical applications.

Scutellaria baicalensis Georgi

Scutellaria baicalensis Georgi is one of the most popular and multipurpose traditional Chinese medicinal herbs with a high flavonoid content. Scutellaria baicalensis Georgi has been routinely used in the treatment of bronchitis, hepatitis, allergy, inflammation, arteriosclerosis[150]. It is included in the Guidelines for the Diagnosis and Treatment of Novel Coronavirus Pneumonia (Trial Version 7), and can be used in COVID-19 patients with damp-heat stagnating in the lungs[24]. Studies reported in the literature clearly indicate that Scutellaria baicalensis Georgi and its flavonoid components can evoke strong antagonizing effects on blood clotting through all three conventional mechanisms: antiplatelet aggregation, anticoagulation, and fibrinolysis-promotion.

First, the antiplatelet effects of Scutellaria baicalensis Georgi have been demonstrated by a number of studies. In one study, Scutellaria flavonoid isolated by methanol was reported to significantly inhibit the platelet aggregation induced by ADP in rabbits[151]. Another study uncovered that oroxylin A (OroA), a flavonoid component of Scutellaria baicalensis Georgi, significantly inhibited platelet aggregation induced by thrombin in mice[152]. Still another flavonoid compound baicalin purified from Scutellaria baicalensis Georgi evoked strong inhibitory effect on platelet aggregation induced by thrombin, collagen and ADP, and on the FeCl3-induced thrombus formation in mice[153]. Furthermore, wogonin (WGN), a flavonoid isolated from Scutellaria baicalensis Georgi and wogonoside (WGNS) as a metabolite of wogonin both can inhibit mouse platelet aggregation induced by thrombin[154].

Second, the effect of Scutellaria baicalensis Georgi on the coagulation system has also been established. For example, treatment with OroA results in inhibition of the amidolytic activity of thrombin, the activation of thrombin from prothrombin, and the production and activity of FXa[152]. OroA can also suppress the function of quinone reductase in rat liver, which is one of the vitamin K reductases in the vitamin K cycling for the hepatic biosynthesis of certain blood coagulation factors[155]. Baicalin can suppress the generation and activity of thrombin and FXa[153]. Similarly, WGN and WGNS can also restrain the activities and production of thrombin and FXa, as well as FX activation by FVIIa[154]. At the molecular level, WGN inhibits the ERK/Egr-1- and JNK/AP-1-mediated transactivation of TF promoter activity, leading to downregulation of TF expression and activity induced by pro-inflammatory molecules[156].

Finally, Scutellaria baicalensis Georgi regulates the function of the fibrinolytic system. This is manifested by the data showing the inhibition of TNFα-induced production of PAI-1 and the significant reduction of the PAI-1 to t-PA ratio after treatment of OroA, baicalin, WGN or WGNS[152, 153, 154].

Dioscorea zingiberensis

Dioscorea zingiberensis, distributed widely in China, has been extensively used in TCM for the treatment of various diseases, including abdominal distention, deadly cold hand and foot, loss of appetite[157]. D. zingiberensis possesses significant inhibitory effects on platelet aggregation and thrombosis. The antithrombotic activity of the total steroidal saponin (TSSN) extracted from D. zingiberensis by alcohol was first demonstrated by a study conducted in a rat model of thrombosis created by ligating inferior vena cava and in a mouse model of pulmonary thrombosis[158]. In this study, TSSN was shown to significantly decrease ADP-induced platelet aggregation with both in vivo and in vitro models[158]. Diosgenin, one of the aglycone of steroidal saponins from D. zingiberensis, inhibited platelet aggregation whereby the bleeding time and clotting time were prolonged and thrombus formation was inhibited on inferior vena cava ligation thrombosis rat model and pulmonary thrombosis mice model[159]. Another investigation demonstrated that two steroidal saponins reduced platelet aggregation in vitro and in vivo[160]. The same study also revealed that these two steroidal saponins inhibited FVIII, indicating their effectiveness in antagonizing the coagulation system to curb thrombosis[160]. These results suggest that Dioscorea zingiberensis has the potential to become a therapeutic agent for thrombosis treatment.

Rubia cordifolia

Rubia cordifolia, is widely distributed in China, especially in northern China. The ethanol extract and aqueous extract of Rubia cordifolia were found to inhibit phenylhydrazine-induced thrombosis in AB strain zebrafish, indicating its antithrombotic potential [161].

The antithrombotic efficacy of Rubia cordifolia appears to be related to its antiplatelet property. In a rat model of blood stasis, Rubia cordifolia treatment decreased whole blood and plasma viscosity and increased fibrinogen content. As signaling mechanisms, Rubia cordifolia downregulated TXB2 level but upregulated 6-keto-PGF1α expression[162].

The anticoagulation effect of Rubia cordifolia also contributes to its antithrombotic efficacy. Purpurin, one of anthraquinones extracted from Rubia cordifolia by alcohol, was identified during the screening step as a specific inhibitor of spermidine-induced autoactivation of plasma hyaluronan-binding protein, a serine protease able to activate FVII[163].

Change to Furthermore, its antithrombotic activity is partially attributed to its ability to activate fibrinolysis. It has been reported that Rubia cordifolia enhanced the activity of t-PA[162], and purpurin stimulated pro-urokinase activation[163].

Astragalus

Astragalus membranaceus is an important medicinal herb widely cultivated in northern China. It has been formulated as an ingredient of herbal mixtures to treat patients with a deficiency in vitality which symptomatically presents as fatigue, anorexia, chronic diarrhea, fatigue, and abnormal uterine bleeding[164].

Astragaloside IV (AGS-IV) is one of the main active extracts from Astragalus membranaceus by alcohol. A drug-target network analysis suggests that AGS-IV could regulate the coagulation system by binding to F2 (prothrombin) and SERPINC1 (AT-III) to produce an antithrombotic effect[165]. Additionally, Astragalus membranaceus can also activate the fibrinolytic system by upregulating the expression of t-PA and downregulating the expression of PAI-1 in cultured HUVECs[166].

Paeonia lactiflora

The herb Paeonia lactiflora, mainly distributed in northern China, has been used in TCM for over 1,000 years to treat cramp, pain, giddiness, and congestion. It is included in the Guidelines for the Diagnosis and Treatment of Novel Coronavirus Pneumonia (Trial Version 7) as a medicine for patients with mild or severe novel coronavirus infection[24].

A study uncovered that ethanol extracts of Paeonia lactiflora reduced platelet aggregation[167]. Another study showed that Paeonia lactiflora significantly downregulated the plasma level of TXB2 and upregulated that of 6-keto-PGF1α[168]. Paeoniflorin, the major ethanol extract of Paeonia lactiflora, was found to decrease prostacyclin production in the culture medium of platelets and TXA2 production in platelets[169]. which indicated paeoniflorin, just like acetylsalicylic acid, could inhibit the metabolism of arachidonic acid, because both of them are the metabolic products of arachidonic acid. These effects may be partly responsible for the anti-thrombotic effect of paeoniflorin. Furthermore, paeoniflorin also inhibited shear stress-induced platelet aggregation through modulating the interaction between vWF and platelet GP Ib[170]. An increase in t-PA activity[169] and upregulation of u-PA via the MAPK signaling pathway[171] by paeoniflorin has also been documented, indicating the role of Paeonia lactiflora in modulating the fibrinolytic system.

The above in vitro studies of Paeonia lactiflora support the potential of paeoniflorin as a promising platelet inhibitor and thrombolytic activator from natural products with a good safety profile. Future studies are warranted the effects of Paeonia lactiflora on platelet aggregation and thrombolysis in vivo to validate its anti-thrombotic efficacy and safety.

Turmeric

Turmeric (Curcuma Longa) is a traditional India and China medicine for the treatments of various illnesses such as dermatologic diseases, infection, stress and depression[172]. A study demonstrated curcumin extracted from turmeric suppresses platelet aggregation provoked by adrenaline-, arachidonate-, and collagen and TXB2 generation[173]. Another study performed with rabbit platelets showed strong antiplatelet activity of curcumin in the presence of collagen, AA, thrombin, or PAF[174]. In an in vivo study, curcumin was shown to inhibit thrombin-induced platelet aggregation (human and rat) by altering the metabolism of eicosanoids[175]. In an earlier research, curcumin had been reported to restrain platelet aggregation induced by calcium ionophore A-23187 and other agonists known to increase cytosolic Ca2+[176]. Curcumin was also found to inhibit GPVI-mediated platelet activation and dense granule secretion induced by GPVI agonist convulxin by interfering with the kinase activity of Syk and the subsequent activation of PLCγ2[177]. A more recent study using the antiplatelet aggregation test claimed that curdione, a major compound from Curcuma species, preferentially inhibited PAF- and thrombin-induced platelet aggregation, repressed P-selectin expression, impeded intracellular Ca2+ mobilization, and elevated cAMP levels in PAF-activated platelets[178].

Moreover, curcumin possesses remarkable anticoagulation effects. The anticoagulant activity of curcumin is evidenced by the prolongation of blood clotting time. This is thought to occur due to an inhibition of thrombin or FXa[179]. Besides, curcumin selectively reduced ectopic hepatic FVII synthesis, but not FVII expression[180].

Furthermore, curcumin promotes the JNK/MAPK-mediated upregulation of u-PA gene expression in a dose-dependent manner to markedly promote fibrinolysis[181].

Tenebrio molitor

Tenebrio molitor is an edible insect that is widely prescribed as a TCM drug in China. It has been recently uncovered that Tenebrio molitor possesses antimicrobial, anticancer, and antihypertensive effects in addition to its antithrombotic actions[182].

The pharmacological effectiveness of Tenebrio molitor in encumbering platelet aggregation to counter thrombosis has been indicated by a study on two compounds isolated from Tenebrio molitor[183]. In vivo, either of the two compounds reduced FeCl3-induced thrombus formation and lowered the mortality of mice due to pulmonary thrombosis. These compounds inhibited platelet aggregation induced by ADP, U46619, and collagen. As for the possible signaling mechanisms, these compounds suppressed PKC activation, decreased cytosolic Ca2+ level, downregulated the expression of P-selectin and PAC-1, enhanced the production of NO, and inhibited the secretion of endothelin-1 (ET-1)[183]. Likewise, these compounds also inhibited the catalytic activity of FXa on its substrate S-2222 and FX activation by FVIIa in a concentration-dependent fashion indicating the role of Tenebrio molitor in modulating coagulation to manifest its antithrombotic action[183].

Ginkgo, Asian ginseng, notoginseng, danshen, Coptis chinensis, chuanxiong, ginger, Celastrus orbiculatus, baical skullcap root, Dioscorea zingiberensis, madder, peony root, turmeric and yellow mealworm have antiplatelet effects, contributing to preventing from thrombosis. Additionally, ginkgo, Asian ginseng, danshen, Coptis chinensis, chuanxiong, ginger, baical skullcap root, Dioscorea zingiberensis, madder, astragalus, turmeric and yellow mealworm hinder thrombosis by inhibiting coagulation. Furthermore, ginkgo, Asian ginseng, notoginseng, danshen, Celastrus orbiculatus, baical skullcap root, madder, astragalus, peony root and turmeric promote the activation of fibrinolytic system, resulting in reduced thrombus.

Discussion

Thrombosis is a multifactorial disease that affects a large proportion of the population, especially in cold areas. TCMs have been used for millennia for their health-promoting properties and therapeutic value for thrombosis. Certain TCMs produce their antiplatelet effects by multiple signaling pathways, such as increasing intraplatelet Ca2+, inhibiting thromboxane formation, reducing serotonin secretion. Some other TCMs activate or strengthen the anticoagulation system. For example, some TCMs inhibit production and/or activation of coagulation factors, some promote AT-III or PC synthesis and activation, and others inhibit vitamin K reductases. Besides, TCMs can also affect the fibrinolytic system to fulfill their antithrombotic effects. In this regard, some TCMs act to decrease PAI-1 expression and/or increase t-PA or u-PA expression and others directly act as fibrinolytic enzymes. Furthermore, we have also described that some TCMs have multiple effects on platelet function, coagulation, and fibrinolysis. An interesting and important point to note here is that many of the TCM with strong antithrombotic effectiveness are grown in the northern regions of China, such as Hawthorn, Asian Ginseng, Celastrus orbiculatus, Madder, Astragalus and Peony root. The findings documented by the studies published to date strongly support the notion that many ingredients and components of TCM are pharmacological modulators of thrombosis with promising therapeutic values and TCM is a natural resource and goldmine for new drug discovery as antithrombotic agents. The present literature review provides a full-scale view from different angles on the antithrombotic potential of TCM. We believe that the research data summarized in this article will help us gain comprehensive understanding of the antithrombotic pharmacology of TCM and provide a guidance for the development of TCM for its applications to antithrombotic therapy.

However, we must admit that we have made only the first step towards in-depth understanding of TCM in terms of its antithrombotic properties. Though with low toxicity in general, TCM can impose unwanted effects in patients. Therefore, more concrete evidence is required to accurately determine the level of risks associated with TCM applications, and more in-depth studies are also needed for improving our understanding of the mechanisms involved. Also, interactions between/among drugs are another issue which must be deciphered in the future investigations. With more research dedicated to understanding drug interactions, it may be possible not only to minimize the associated adverse effects, but also to facilitate the combinational therapy with multiple TCM components or formulations or between TCM and western medicine to yield synergetic therapeutic outcomes. In addition, translational studies on TCM and thrombosis will also witness their boom in the future, partially owing to our ever-increasing awareness of the importance of TCM as an alternative medicine and its likelihood of becoming part of modern medicine as well as the new strategies and technologies for isolating, purifying and identifying the active components from TCM.


#

These authors made equal contributions to this work.


Acknowledgements

This work was supported by the National Natural Science Foundation of China (81773735, 81903610, 91949130, 81961138018) and National Key R&D Program of China (2017YFC1702003).

  1. Contributions

    Yong Zhang proposed the conception for the review. Xueqing Tang and Xin Liu wrote the manuscript. Xueqing Tang prepared the table. Monayo Seth Mikaye gave critical discussions and revisions on manuscript. Hongrui Zhao gave revisions on manuscript. All authors read and approved the final version of the manuscript.

  2. Conflict of Interest

    Yong Zhang is an Editorial Board Member. The article was subject to the journal’s standard procedures, with peer review handled independently of this Member and his research groups. The authors declare that they have no other competing interests.

Abbreviations

TCM

traditional Chinese medicine

t-PA

tissue type plasminogen activator

FDA

Food and Drug A administration

GP

glycoprotein

vWF

von Willebrand Factor

COX

cyclooxygenase

LOX

lipoxygenase

ATP

adenosine triphosphate

ADP

adenosine diphosphate

AA

arachidonic acid

PG

prostaglandins

TXA2

thromboxane A2

12(S)-HETE

12(S)-hydroxyeicosatetraenoic acid

GPCRs

G protein-coupled receptors

SY

Safflower yellow

PAF

platelet activating factor

HSYA

hydroxysafflor yellow A

DIC

disseminated intravascular coagulation

AV-shunt

arteriovenous shunt

cGMP

cyclic guanosine monophosphate

CBN

columbianadin

SF

sodium ferulate

TXB2

thromboxane B2

LIG

ligustilide

DHC

dehydrocorydaline

THB

canadine

XN

xanthohumol

FeCl3

ferric chloride

TF

tissue factor

GL

glycyrrhizin

TT

thrombin time

u-PA

urokinasetype plasminogen activator

PAI-1

plasminogen activator inhibitor 1

TAFI

thrombinactivatable fibrinolysis inhibitor

EfP

E. fetida proteases

LrP

L. rubellus proteases

GA

ginkgolide A

GB

ginkgolide B

GC

ginkgolide C

cAMP

cyclic adenosine monophosphate

BB

bilobalide

RBC

red blood cells

KLF2

Krüppel-like factor 2

TM

thrombomodulin

HUVECs

human umbilical vein endothelial cells

PNS

panax notoginseng saponins

SA

salvianolic acid

SAA

salvianolic acid A

AT-III

antithrombin III

PC

protein C

BBR

berberine

OroA

Oroxylin A

WGN

wogonin

TSSN

total steroidal saponin

AGS-IV

astragaloside IV

ET-1

endothelin-1.

References

[1] Koupenova M, Kehrel B E, Corkrey H A, et al. Thrombosis and platelets: an update. Eur Heart J, 2017; 38(11): 785–791.10.1093/eurheartj/ehw550Search in Google Scholar PubMed

[2] Bhaskaran K, Hajat S, Haines A, et al. Short term effects of temperature on risk of myocardial infarction in England and Wales: time series regression analysis of the Myocardial Ischaemia National Audit Project (MINAP) registry. BMJ, 2010; 341: c3823.10.1136/bmj.c3823Search in Google Scholar PubMed PubMed Central

[3] Pant S, Badheka A O, Deshmukh A. Higher frequency of atrial fibrillation linked to colder seasons and air temperature on the day of ischemic stroke onset. J Stroke Cerebrovasc Dis, 2013; 22: 896.10.1016/j.jstrokecerebrovasdis.2013.06.026Search in Google Scholar PubMed

[4] Awad E M, Khan S Y, Sokolikova B, et al. Cold induces reactive oxygen species production and activation of the NF-kappa B response in endothelial cells and inflammation in vivo. J Thromb Haemost, 2013; 11(9): 1716–1726.10.1111/jth.12357Search in Google Scholar PubMed

[5] Rosendaal F R. Risk factors for venous thrombotic disease. Thromb Haemost, 1999; 82(9): 610–619.10.1055/s-0037-1615887Search in Google Scholar

[6] Birk S, Kruuse C, Petersen K A, et al. The headache-inducing effect of cilostazol in human volunteers. Cephalalgia, 2006; 26(11): 1304–1309.10.1111/j.1468-2982.2006.01218.xSearch in Google Scholar PubMed

[7] Francescone S, Halperin J L. “Triple therapy” or triple threat? Balancing the risks of antithrombotic therapy for patients with atrial fibrillation and coronary stents. J Am Coll Cardiol, 2008; 51(8): 826–827.10.1016/j.jacc.2007.11.034Search in Google Scholar PubMed

[8] Johnson S. Known knowns and known unknowns: risks associated with combination antithrombotic therapy. Thromb Res, 2008; 123(Supp l 1): S7–S11.10.1016/j.thromres.2008.08.011Search in Google Scholar PubMed

[9] Alban S. Adverse effects of heparin. Handb Exp Pharmacol, 2012: 211–263.10.1007/978-3-642-23056-1_10Search in Google Scholar PubMed

[10] Flemmig M, Melzig M F. Serine-proteases as plasminogen activators in terms of fibrinolysis. J Pharm Pharmacol, 2012; 64(8): 1025–1039.10.1111/j.2042-7158.2012.01457.xSearch in Google Scholar PubMed

[11] Disharoon D, Marr D W M, Neeves K B. Engineered microparticles and nanoparticles for fibrinolysis. J Thromb Haemost, 2019; 17(12): 2004–2015.10.1111/jth.14637Search in Google Scholar PubMed PubMed Central

[12] Lu A P, Jia H W, Xiao C, et al. Theory of traditional Chinese medicine and therapeutic method of diseases. World J Gastroenterol, 2004; 10(13): 1854–1856.10.3748/wjg.v10.i13.1854Search in Google Scholar PubMed PubMed Central

[13] Hsiao W L, Liu L. The role of traditional Chinese herbal medicines in cancer therapy--from TCM theory to mechanistic insights. Planta Med, 2010; 76(11): 1118–1131.10.1055/s-0030-1250186Search in Google Scholar PubMed

[14] Yang R, Yuan B C, Ma YS, et al. The anti-inflammatory activity of licorice, a widely used Chinese herb. Pharm Biol, 2017; 55(1): 5–18.10.1080/13880209.2016.1225775Search in Google Scholar PubMed PubMed Central

[15] Ma C, Xia R, Yang S, et al. Formononetin attenuates atherosclerosis via regulating interaction between KLF4 and SRA in apoE(-/-) mice. Theranostics, 2020; 10(3): 1090–1106.10.7150/thno.38115Search in Google Scholar PubMed PubMed Central

[16] Zhai B, Zhang N, Han X, et al. Molecular targets of beta-elemene, a herbal extract used in traditional Chinese medicine, and its potential role in cancer therapy: A review. Biomed Pharmacother, 2019; 114: 108812.10.1016/j.biopha.2019.108812Search in Google Scholar PubMed

[17] Bai L, Li X, He L, et al. Antidiabetic potential of flavonoids from traditional Chinese medicine: a review. Am J Chin Med, 2019; 47(5): 933–957.10.1142/S0192415X19500496Search in Google Scholar PubMed

[18] Ren J L, Zhang A H, Wang X J. Traditional Chinese medicine for COVID-19 treatment. Pharmacol Res, 2020; 155(8): 104743.10.1016/j.phrs.2020.104743Search in Google Scholar PubMed PubMed Central

[19] Li Y, Liu X, Guo L, et al. Traditional Chinese herbal medicine for treating novel coronavirus (COVID-19) pneumonia: protocol for a systematic review and meta-analysis. Syst Rev, 2020; 9(1): 75.10.1186/s13643-020-01343-4Search in Google Scholar PubMed PubMed Central

[20] Holinstat M. Normal platelet function. Cancer Metastasis Rev, 2017; 36(2): 195–198.10.1007/s10555-017-9677-xSearch in Google Scholar PubMed PubMed Central

[21] Stalker T J, Traxler E A, Wu J, et al. Hierarchical organization in the hemostatic response and its relationship to the platelet-signaling network. Blood, 2013; 121(10): 1875–1885.10.1182/blood-2012-09-457739Search in Google Scholar PubMed PubMed Central

[22] Welsh J D, Muthard R W, Stalker T J, et al. A systems approach to hemostasis: 4. How hemostatic thrombi limit the loss of plasma-borne molecules from the microvasculature. Blood, 2016; 127(12): 1598–1605.10.1182/blood-2015-09-672188Search in Google Scholar PubMed PubMed Central

[23] Delshad E, Yousefi M, Sasannezhad P, et al. Medical uses of carthamus tinctorius L. (Safflower): a comprehensive review from traditional medicine to modern medicine. Electron Physician, 2018; 10(4): 6672–6681.10.19082/6672Search in Google Scholar PubMed PubMed Central

[24] Chinese Center for Disease Control and Prevention. Guide Manual on Pharmacological Management of Coronavirus Diseases 2019 (COVID-19). Beijing: People's Medical Publishing House, 2020.Search in Google Scholar

[25] Zhou X, Tang L, Xu Y, et al. Towards a better understanding of medicinal uses of Carthamus tinctorius L. in traditional Chinese medicine: a phytochemical and pharmacological review. J Ethnopharmacol, 2014; 151(1): 27–43.10.1016/j.jep.2013.10.050Search in Google Scholar PubMed

[26] Memariani Z, Moeini R, Hamedi S S, et al. Medicinal plants with antithrombotic property in Persian medicine: a mechanistic review. J Thromb Thrombolysis, 2018; 45(1): 158–179.10.1007/s11239-017-1580-3Search in Google Scholar PubMed

[27] Zang B X, Jin M, Si N, et al. Antagonistic effect of hydroxysafflor yellow A on the platelet activating factor receptor. Yao Xue Xue Bao, 2002; 37(9): 696–699.Search in Google Scholar

[28] Zhu H B, Zhang L, Wang Z H, et al. Therapeutic effects of hydroxysafflor yellow A on focal cerebral ischemic injury in rats and its primary mechanisms. J Asian Nat Prod Res, 2005; 7(4): 607–613.10.1080/10286020310001625120Search in Google Scholar PubMed

[29] Li Y, Wang N. Antithrombotic effects of Danggui, Honghua and potential drug interaction with clopidogrel. J Ethnopharmacol, 2010; 128(3): 623–628.10.1016/j.jep.2010.02.003Search in Google Scholar PubMed

[30] Zhu Y F, Luo H M, Deng Z L, et al. Effects of the Chinese patent medicine, Honghua Injection, on platelet glycoprotein IIb/III a receptors in patients with acute coronary syndrome: a randomized controlled trial. Zhong Xi Yi Jie He Xue Bao, 2012; 10(3): 318–323.10.3736/jcim20120311Search in Google Scholar PubMed

[31] Zhang N, Lian Z, Peng X, et al. Applications of Higenamine in pharmacology and medicine. J Ethnopharmacol, 2017; 196(20): 242–252.10.1016/j.jep.2016.12.033Search in Google Scholar PubMed

[32] Yun-Choi H S, Pyo M K, Park K M, et al. Anti-thrombotic effects of higenamine. Planta Med 2001; 67(7): 619–622.10.1055/s-2001-17361Search in Google Scholar PubMed

[33] Yun-Choi H S, Pyo M K, Park K M, et al. Antithrombotic effects of YS-49 and YS-51--1-naphthylmethyl analogs of higenamine. Thromb Res, 2001; 104(4): 249–255.10.1016/S0049-3848(01)00372-3Search in Google Scholar

[34] Pyo M K, Kim J M, Jin J L, et al. Effects of higenamine and its 1-naphthyl analogs, YS-49 and YS-51, on platelet TXA2 synthesis and aggregation. Thromb res, 2007; 120(1): 81–86.10.1016/j.thromres.2006.07.006Search in Google Scholar

[35] Chang R S, Ding L, Chen G Q, et al. Dehydroandrographolide succinic acid monoester as an inhibitor against the human immunodeficiency virus. Proc Soc Exp Biol Med, 1991; 197(1): 59–66.10.3181/00379727-197-43225Search in Google Scholar

[36] Calabrese C, Berman S H, Babish J G, et al. A phase I trial of andrographolide in HIV positive patients and normal volunteers. Phytother Res, 2000; 14(5): 333–338.10.1002/1099-1573(200008)14:5<333::AID-PTR584>3.0.CO;2-DSearch in Google Scholar

[37] Shen Y C, Chen C F, Chiou W F. Andrographolide prevents oxygen radical production by human neutrophils: possible mechanism(s) involved in its anti-inflammatory effect. Br J Pharmacol, 2002; 135(2): 399–406.10.1038/sj.bjp.0704493Search in Google Scholar

[38] See D, Mason S, Roshan R. Increased tumor necrosis factor alpha (TNF-alpha) and natural killer cell (NK) function using an integrative approach in late stage cancers. Immunol Invest, 2002; 31: 137–153.10.1081/IMM-120004804Search in Google Scholar

[39] Kapil A, Koul I B, Banerjee S K, et al. Antihepatotoxic effects of major diterpenoid constituents of Andrographis paniculata. Biochem Pharmacol, 1993; 46(1): 182–185.10.1016/0006-2952(93)90364-3Search in Google Scholar

[40] Zhang X F, Tan B K. Antihyperglycaemic and anti-oxidant properties of Andrographis paniculata in normal and diabetic rats. Clin Exp Pharmacol Physiol, 2000; 27(5–6): 358–363.10.1046/j.1440-1681.2000.03253.xSearch in Google Scholar

[41] Dehghani S, Mehri S, Hosseinzadeh H. The effects of Crataegus pinnatifida (Chinese hawthorn) on metabolic syndrome: A review. Iran J Basic Med Sci, 2019; 22(5): 460–468.Search in Google Scholar

[42] Arslan R, Bor Z, Bektas N, et al. Antithrombotic effects of ethanol extract of Crataegus orientalis in the carrageenan-induced mice tail thrombosis model. Thromb Res, 2011; 127(3): 210–213.10.1016/j.thromres.2010.11.028Search in Google Scholar

[43] Song S J, Li L Z, Gao P Y, et al. Isolation of antithrombotic phenolic compounds from the leaves of crataegus pinnatifida. Planta Med, 2012; 78(18): 1967–1971.10.1055/s-0032-1327877Search in Google Scholar

[44] Rogers K L, Grice I D, Griffiths L R. Inhibition of platelet aggregation and 5-HT release by extracts of Australian plants used traditionally as headache treatments. Eur J Pharm Sci, 2000; 9(4): 355–363.10.1016/S0928-0987(99)00074-3Search in Google Scholar

[45] Vibes J, Lasserre B, Gleye J, et al. Inhibition of thromboxane A2 biosynthesis in vitro by the main components of Crataegus oxyacantha (Hawthorn) flower heads. Prostaglandins Leukot Essent Fatty Acids, 1994; 50(4): 173–175.10.1016/0952-3278(94)90141-4Search in Google Scholar

[46] Shang X, Pan H, Wang X, et al. Leonurus japonicus Houtt.: ethnopharmacology, phytochemistry and pharmacology of an important traditional Chinese medicine. J Ethnopharmacol, 2014; 152(1): 14–32.10.1016/j.jep.2013.12.052Search in Google Scholar

[47] Moon H I. Three diterpenes from Leonurus japonicus Houtt protect primary cultured rat cortical cells from glutamate-induced toxicity. Phytother Res, 2010; 24(8): 1256–1259.10.1002/ptr.3144Search in Google Scholar

[48] Xiong L, Zhou Q M, Peng C, et al. Bis-spirolabdane diterpenoids from Leonurus japonicus and their anti-platelet aggregative activity. Fitoterapia, 2015; 100(1):1–6.10.1016/j.fitote.2014.11.003Search in Google Scholar

[49] Xiong L, Zhou Q M, Peng C, et al. Sesquiterpenoids from the herb of Leonurus japonicus. Molecules, 2013; 18(5): 5051–5058.10.3390/molecules18055051Search in Google Scholar

[50] Zou Q Z, Bi R G, Li J M, et al. Effect of motherwort on blood hyperviscosity. Am J Chin Med, 1989; 17(1–2): 65–70.10.1142/S0192415X89000115Search in Google Scholar

[51] Chen D, Du Z, Lin Z, et al. The chemical compositions of angelica pubescens oil and its prevention of UV-B radiation-induced cutaneous photoaging. Chem Biodivers, 2018; 15(10): e1800235.10.1002/cbdv.201800235Search in Google Scholar

[52] Liu J H, Zschocke S, Reininger E, et al. Inhibitory effects of Angelica pubescens f. biserrata on 5-lipoxygenase and cyclooxygenase. Planta Med, 1998; 64(6): 525–529.10.1055/s-2006-957507Search in Google Scholar

[53] Hoult J R, Paya M. Pharmacological and biochemical actions of simple coumarins: natural products with therapeutic potential. Gen Pharmacol, 1996; 27(4): 713–722.10.1016/0306-3623(95)02112-4Search in Google Scholar

[54] Hou S M, Hsia C W, Tsai C L, et al. Modulation of human platelet activation and in vivo vascular thrombosis by columbianadin: regulation by integrin alphaIIbbeta3 inside-out but not outside-in signals. J Biomed Sci, 2020; 27(1): 60.10.1186/s12929-020-0619-5Search in Google Scholar

[55] Wang B H, Ou-Yang J P. Pharmacological actions of sodium ferulate in cardiovascular system. Cardiovasc Drug Rev, 2005; 23(2): 161–172.10.1111/j.1527-3466.2005.tb00163.xSearch in Google Scholar

[56] Xu L N, Ouyang R. Antithrombotic effect of sodium ferulate in rats (author's transl)]. Zhongguo Yao Li Xue Bao, 1981; 2(1): 35–37.Search in Google Scholar

[57] Yin Z Z, Zhang L Y, Xu L N. The effect of Dang-Gui (Angelica sinensis) and its ingredient ferulic acid on rat platelet aggregation and release of 5-HT (author's transl)]. Yao Xue Xue Bao, 1980; 15(6): 321–326.Search in Google Scholar

[58] Yin Z Z, Wang J P, Xu L N. Effect of sodium ferulate on malondialdehyde production from the platelets of rats. Zhongguo Yao Li Xue Bao, 1986; 7(4): 336–339.Search in Google Scholar

[59] Wang Z, Gao Y H, Huang R S, et al. Sodium ferulate is an inhibitor of thromboxane A2 synthetase. Zhongguo Yao Li Xue Bao, 1988; 9(5): 430–433.Search in Google Scholar

[60] Xu L N, Yu W G, Tian JY. Effect of sodium ferulate on C14-arachidonic acid metabolism in rabbit platelets. Zhong Xi Yi Jie He Za Zhi, 1988; 8(10): 614–615, 583.Search in Google Scholar

[61] Xu L N, Yu W G, Tian J Y, et al. Effect of sodium ferulate on arachidonic acid metabolism. Yao Xue Xue Bao, 1990; 25(6): 412–416.Search in Google Scholar

[62] Zhang L, Du J R, Wang J, et al. Z-ligustilide extracted from Radix Angelica Sinensis decreased platelet aggregation induced by ADP ex vivo and arterio-venous shunt thrombosis in vivo in rats. Yakugaku Zasshi, 2009; 129(7): 855–859.10.1248/yakushi.129.855Search in Google Scholar

[63] Dong W G, Liu S P, Zhu H H, et al. Abnormal function of platelets and role of angelica sinensis in patients with ulcerative colitis. World J Gastroenterol, 2004; 10(4): 606–609.10.3748/wjg.v10.i4.606Search in Google Scholar

[64] Chang S, Yang Z, Han N, et al. The antithrombotic, anticoagulant activity and toxicity research of ambinine, an alkaloid from the tuber of Corydalis ambigua var. amurensis. Regul Toxicol Pharmacol, 2018; 95: 175–181.10.1016/j.yrtph.2018.03.004Search in Google Scholar

[65] Ko F N, Wu T S, Lu S T, et al. Antiplatelet effects of protopine isolated from Corydalis tubers. Thromb Res, 1989; 56(2): 289–298.10.1016/0049-3848(89)90170-9Search in Google Scholar

[66] Tan C N, Zhang Q, Li C H, et al. Potential target-related proteins in rabbit platelets treated with active monomers dehydrocorydaline and canadine from Rhizoma corydalis. Phytomedicine, 2019; 54: 231–239.10.1016/j.phymed.2018.09.200Search in Google Scholar PubMed

[67] Li C H, Chen C, Zhang Q, et al. Differential proteomic analysis of platelets suggested target-related proteins in rabbit platelets treated with Rhizoma Corydalis. Pharm Biol, 2017; 55(1): 76–87.10.1080/13880209.2016.1229340Search in Google Scholar PubMed PubMed Central

[68] Zanoli P, Zavatti M. Pharmacognostic and pharmacological profile of Humulus lupulus L. J Ethnopharmacol, 2008; 116(3): 383–396.10.1016/j.jep.2008.01.011Search in Google Scholar PubMed

[69] Xin G, Wei Z, Ji C, et al. Xanthohumol isolated from Humulus lupulus prevents thrombosis without increased bleeding risk by inhibiting platelet activation and mtDNA release. Free Radic Biol Med, 2017; 108: 247–257.10.1016/j.freeradbiomed.2017.02.018Search in Google Scholar PubMed PubMed Central

[70] Luzak B, Kassassir H, Roj E, et al. Xanthohumol from hop cones (Humulus lupulus L.) prevents ADP-induced platelet reactivity. Arch Physiol Biochem, 2017; 123(1): 54–60.10.1080/13813455.2016.1247284Search in Google Scholar PubMed

[71] Lee Y M, Hsieh K H, Lu W J, et al. Xanthohumol, a Prenylated Flavonoid from Hops (Humulus lupulus), Prevents Platelet Activation in Human Platelets. Evid Based Complement Alternat Med, 2012: 852362.10.1155/2012/852362Search in Google Scholar PubMed PubMed Central

[72] Adams R L, Bird R J. Review article: Coagulation cascade and therapeutics update: relevance to nephrology. Part 1: Overview of coagulation, thrombophilias and history of anticoagulants. Nephrology (Carlton), 2009; 14(5): 462–470.10.1111/j.1440-1797.2009.01128.xSearch in Google Scholar PubMed

[73] Tanaka K A, Key N S, Levy J H. Blood coagulation: hemostasis and thrombin regulation. Anesth Analg, 2009; 108(5): 1433–1446.10.1213/ane.0b013e31819bcc9cSearch in Google Scholar PubMed

[74] Versteeg H H, Heemskerk J W, Levi M, et al. New fundamentals in hemostasis. Physiol Rev, 2013; 93(1): 327–358.10.1152/physrev.00016.2011Search in Google Scholar PubMed

[75] Ten Cate H, Hackeng T M, Garcia de Frutos P. Coagulation factor and protease pathways in thrombosis and cardiovascular disease. Thromb Haemost, 2017; 117(7): 1265–1271.10.1160/TH17-02-0079Search in Google Scholar PubMed

[76] Gelosa P, Castiglioni L, Tenconi M, et al. Pharmacokinetic drug interactions of the non-vitamin K antagonist oral anticoagulants (NOACs). Pharmacol Res, 2018; 135: 60–79.10.1016/j.phrs.2018.07.016Search in Google Scholar PubMed

[77] Pastorino G, Cornara L, Soares S, et al. Liquorice (Glycyrrhiza glabra): a phytochemical and pharmacological review. Phytother Res, 2018; 32(3): 2323–2339.10.1002/ptr.6178Search in Google Scholar PubMed PubMed Central

[78] Francischetti I M, Monteiro R Q, Guimaraes J A. Identification of glycyrrhizin as a thrombin inhibitor. Biochem Biophys Res Commun, 1997; 235(1): 259–263.10.1006/bbrc.1997.6735Search in Google Scholar PubMed

[79] Mendes-Silva W, Assafim M, Ruta B, et al. Antithrombotic effect of Glycyrrhizin, a plant-derived thrombin inhibitor. Thromb res, 2003; 112(1–2): 93–98.10.1016/j.thromres.2003.10.014Search in Google Scholar PubMed

[80] Jiang L, Wang Q, Shen S, et al. Discovery of glycyrrhetinic acid as an orally active, direct inhibitor of blood coagulation factor xa. Thromb Res, 2014; 133(3): 501–506.10.1016/j.thromres.2013.12.025Search in Google Scholar PubMed

[81] Xiao X, Zhou S H, Jiang N, et al. First record of Leptospira and Blastocystis infections in captive flying squirrels (Trogopterus xanthipes) from Enshi County, China. Acta Trop, 2019; 197: 105065.10.1016/j.actatropica.2019.105065Search in Google Scholar PubMed

[82] Baek S, Xia X, Min B S, et al. Trogopterins A-C: Three new neolignans from feces of Trogopterus xanthipes. Beilstein J Org Chem, 2014; 10: 2955–2962.10.3762/bjoc.10.313Search in Google Scholar PubMed PubMed Central

[83] Yang N Y, Tao W W, Duan J A. Antithrombotic flavonoids from the faeces of Trogopterus xanthipes. Nat Prod Res, 2010; 24: 1843–1849.10.1080/14786419.2010.482057Search in Google Scholar PubMed

[84] Yang N Y, Tao W W, Duan J A, et al. Four new fatty acid esters from the Feces of Trogopterus xanthipes. Lipids, 2009; 44(9): 849–853.10.1007/s11745-009-3329-zSearch in Google Scholar PubMed

[85] Kumar S S, Sabu A. Fibrinolytic enzymes for thrombolytic therapy. Adv Exp Med Biol, 2019; 1148: 345–381.10.1007/978-981-13-7709-9_15Search in Google Scholar PubMed

[86] Rabinstein A A. Treatment of Acute Ischemic Stroke. Continuum (Minneap Minn), 2017; 23(1): 62–81.10.1212/CON.0000000000000420Search in Google Scholar PubMed

[87] Gurewich V. Fibrinolysis: a misunderstood natural defense whose therapeutic potential is unknown. Cardiovasc Drugs Ther, 2019; 33: 749–753.10.1007/s10557-019-06923-8Search in Google Scholar PubMed

[88] Liesenborghs L, Verhamme P, Vanassche T. Staphylococcus aureus, master manipulator of the human hemostatic system. J Thromb Haemost, 2018; 16: 441–454.10.1111/jth.13928Search in Google Scholar PubMed

[89] Marder V J. Historical perspective and future direction of thrombolysis research: the re-discovery of plasmin. J Thromb Haemost, 2011; 9(Suppl 1): 364–373.10.1111/j.1538-7836.2011.04370.xSearch in Google Scholar PubMed

[90] Emberson J, Lees K R, Lyden P, et al. Effect of treatment delay, age, and stroke severity on the effects of intravenous thrombolysis with alteplase for acute ischaemic stroke: a meta-analysis of individual patient data from randomised trials. Lancet, 2014; 384(9958): 1929–1935.10.1016/S0140-6736(14)60584-5Search in Google Scholar

[91] Lu Q, Ma R, Yang Y, et al. Zanthoxylum nitidum (Roxb.) DC: Traditional uses, phytochemistry, pharmacological activities and toxicology. J Ethnopharmacol, 2020; 260(5): 112946.10.1016/j.jep.2020.112946Search in Google Scholar PubMed

[92] Yu B, Zhang G, Jin L, et al. Inhibition of PAI-1 activity by toddalolactone as a mechanism for promoting blood circulation and removing stasis by Chinese herb zanthoxylum nitidum var. tomentosum. Front Pharmacol, 2017; 8: 489.10.3389/fphar.2017.00489Search in Google Scholar PubMed PubMed Central

[93] Huber J, Fabritius H O, Griesshaber E, et al. Function-related adaptations of ultrastructure, mineral phase distribution and mechanical properties in the incisive cuticle of mandibles of porcellio scaber latreille, 1804. J Struct Biol, 2014; 188(1): 1–15.10.1016/j.jsb.2014.09.002Search in Google Scholar PubMed

[94] Tian Z, Li B, Guo L, et al. Purification and biochemical characterization of a novel fibrinolytic enzyme, PSLTro01, from a medicinal animal Porcellio scaber Latreille. Int J Biol Macromol, 2015; 80: 536–546.10.1016/j.ijbiomac.2015.06.046Search in Google Scholar PubMed

[95] Nakajima N, Ishihara K, Sugimoto M, et al. Chemical modification of earthworm fibrinolytic enzyme with human serum albumin fragment and characterization of the protease as a therapeutic enzyme. Biosci Biotechnol Biochem, 1996; 60(2): 293–300.10.1271/bbb.60.293Search in Google Scholar PubMed

[96] Wang F, Wang C, Li M, et al. Purification, characterization and crystallization of a group of earthworm fibrinolytic enzymes from Eisenia fetida. Biotechnol Lett, 2003; 25: 1105–1109.10.1023/A:1024196232252Search in Google Scholar

[97] Cho I H, Choi E S, Lim H G, et al. Purification and characterization of six fibrinolytic serine-proteases from earthworm Lumbricus rubellus. J Biochem Mol Biol, 2004; 37(2): 199–205.10.5483/BMBRep.2004.37.2.199Search in Google Scholar

[98] Lee C K, Shin J S, Kim B S, et al. Antithrombotic effects by oral administration of novel proteinase fraction from earthworm Eisenia andrei on venous thrombosis model in rats. Arch Pharm Res, 2007; 30(4): 475–480.10.1007/BF02980222Search in Google Scholar PubMed

[99] Tjandrawinata R R, Yunaidi D A, Susanto L W. The safety and tolerability of lumbrokinase DLBS1033 in healthy adult subjects. Drug Res (Stuttg), 2016; 66(6): 293–299.10.1055/s-0035-1569406Search in Google Scholar PubMed

[100] Fan Q, Wu C, Li L, et al. Some features of intestinal absorption of intact fibrinolytic enzyme III-1 from Lumbricus rubellus. Biochim Biophys Acta, 2001; 1526(3): 286–292.10.1016/S0304-4165(01)00140-4Search in Google Scholar

[101] Nakajima N, Sugimoto M, Ishihara K. Stable earthworm serine proteases: application of the protease function and usefulness of the earthworm autolysate. J Biosci Bioeng, 2000; 90(2): 174–179.10.1016/S1389-1723(00)80106-1Search in Google Scholar

[102] Mahady G B. Ginkgo biloba for the prevention and treatment of cardiovascular disease: a review of the literature. J Cardiovasc Nurs, 2002; 16(4): 21–32.10.1097/00005082-200207000-00004Search in Google Scholar PubMed

[103] Stromgaard K, Nakanishi K. Chemistry and biology of terpene trilactones from Ginkgo biloba. Angew Chem Int Ed Engl, 2004; 43(13): 1640–1658.10.1002/chin.200424256Search in Google Scholar

[104] Nakanishi K. Terpene trilactones from Gingko biloba: from ancient times to the 21st century. Bioorg Med Chem, 2005; 13(17): 4987–5000.10.1016/j.bmc.2005.06.014Search in Google Scholar PubMed

[105] Cho H J, Nam K S. Inhibitory effect of ginkgolide B on platelet aggregation in a cAMP- and cGMP-dependent manner by activated MMP-9. J Biochem Mol Biol, 2007; 40: 678–683.10.5483/BMBRep.2007.40.5.678Search in Google Scholar

[106] Li X Y, Li C, Liu T, et al. Chemical analysis, pharmacological activity and process optimization of the proportion of bilobalide and ginkgolides in Ginkgo biloba extract. J Pharm Biomed Anal, 2018; 160: 46–54.10.1016/j.jpba.2018.07.037Search in Google Scholar PubMed

[107] Chen T R, Wei L H, Guan X Q, et al. Biflavones from Ginkgo biloba as inhibitors of human thrombin. Bioorg Chem, 2019; 92(Suppl): 103199.10.1016/j.bioorg.2019.103199Search in Google Scholar PubMed

[108] Naderi G A, Asgary S, Jafarian A, et al. Fibrinolytic effects of Ginkgo biloba extract. Exp Clin Cardiol, 2005; 10(2): 85–87.Search in Google Scholar

[109] Chiu Y L, Tsai W C, Wu C H, et al. Ginkgo biloba induces thrombomodulin expression and tissue-type plasminogen activator secretion via the activation of kruppel-like factor 2 within endothelial cells. Am J Chin Med, 2020; 48(2): 357–372.10.1142/S0192415X20500184Search in Google Scholar PubMed

[110] Lee C H, Kim J H. A review on the medicinal potentials of ginseng and ginsenosides on cardiovascular diseases. J Ginseng Res, 2014; 38(3): 161–166.10.1016/j.jgr.2014.03.001Search in Google Scholar PubMed PubMed Central

[111] Jin Y R, Yu J Y, Lee J J, et al. Antithrombotic and antiplatelet activities of Korean red ginseng extract. Basic Clin Pharmacol Toxicol, 2007; 100(3): 170–175.10.1111/j.1742-7843.2006.00033.xSearch in Google Scholar PubMed

[112] Kimura Y, Okuda H, Arichi S. Effects of various ginseng saponins on 5-hydroxytryptamine release and aggregation in human platelets. J Pharm Pharmacol, 1988; 40(12): 838–843.10.1111/j.2042-7158.1988.tb06285.xSearch in Google Scholar PubMed

[113] Irfan M, Jeong D, Kwon H W, et al. Ginsenoside-Rp3 inhibits platelet activation and thrombus formation by regulating MAPK and cyclic nucleotide signaling. Vascul Pharmacol, 2018; 109: 45–55.10.1016/j.vph.2018.06.002Search in Google Scholar PubMed

[114] Shin J H, Kwon H W, Rhee M H, et al. Inhibitory effects of thromboxane A2 generation by ginsenoside Ro due to attenuation of cytosolic phospholipase A2 phosphorylation and arachidonic acid release. J Ginseng Res, 2019; 43(2): 236–241.10.1016/j.jgr.2017.12.007Search in Google Scholar PubMed PubMed Central

[115] Endale M, Lee W M, Kamruzzaman S M, et al. Ginsenoside-Rp1 inhibits platelet activation and thrombus formation via impaired glycoprotein VI signalling pathway, tyrosine phosphorylation and MAPK activation. Br J Pharmacol, 2012; 167(1): 109–127.10.1111/j.1476-5381.2012.01967.xSearch in Google Scholar PubMed PubMed Central

[116] Irfan M, Jeong D, Saba E, et al. Gintonin modulates platelet function and inhibits thrombus formation via impaired glycoprotein VI signaling. Platelets, 2019; 30: 589–598.10.1080/09537104.2018.1479033Search in Google Scholar PubMed

[117] Xiong L, Qi Z, Zheng B, et al. Inhibitory Effect of Triterpenoids from Panax ginseng on Coagulation Factor X. Molecules, 2017; 22: 649.10.3390/molecules22040649Search in Google Scholar PubMed PubMed Central

[118] Ushio Y. Effect of ginsenoside Rgl on the release of enzymes by cultured endothelial cells. Am J Chin Med, 1992; 20(1): 91–101.10.1142/S0192415X92000102Search in Google Scholar PubMed

[119] Lau A J, Toh D F, Chua T K, et al. Antiplatelet and anticoagulant effects of Panax notoginseng: comparison of raw and steamed Panax notoginseng with Panax ginseng and Panax quinquefolium. J Ethnopharmacol, 2009; 125(3): 380–386.10.1016/j.jep.2009.07.038Search in Google Scholar PubMed

[120] Shen Q, Li J, Zhang C, et al. Panax notoginseng saponins reduce high-risk factors for thrombosis through peroxisome proliferator-activated receptor-gamma pathway. Biomed Pharmacother, 2017; 96: 1163–1169.10.1016/j.biopha.2017.11.106Search in Google Scholar PubMed

[121] Zhou Q, Jiang L, Xu C, et al. Ginsenoside Rg1 inhibits platelet activation and arterial thrombosis. Thromb Res, 2014; 133(1): 57–65.10.1016/j.thromres.2013.10.032Search in Google Scholar

[122] Liu Y, Liu T, Ding K, et al. Phospholipase cgamma2 signaling cascade contribute to the antiplatelet effect of notoginsenoside fc. Front Pharmacol, 2018; 9: 1293.10.3389/fphar.2018.01293Search in Google Scholar

[123] Zhang W J, Wojta J, Binder B R. Effect of notoginsenoside R1 on the synthesis of components of the fibrinolytic system in cultured smooth muscle cells of human pulmonary artery. Cell mol biol (Noisy-le-grand), 1997; 43(4): 581–587.Search in Google Scholar

[124] Li Z M, Xu S W, Liu P Q. Salvia miltiorrhizaBurge (Danshen): a golden herbal medicine in cardiovascular therapeutics. Acta Pharmacol Sin, 2018; 39(5): 802–824.10.1038/aps.2017.193Search in Google Scholar

[125] Tang M K, Ren D C, Zhang J T, et al. Effect of salvianolic acids from Radix Salviae miltiorrhizae on regional cerebral blood flow and platelet aggregation in rats. Phytomedicine, 2002; 9(5): 405–409.10.1078/09447110260571634Search in Google Scholar

[126] Fan H Y, Fu F H, Yang M Y, et al. Antiplatelet and antithrombotic activities of salvianolic acid A. Thromb Res, 2010; 126(1): e17–22.10.1016/j.thromres.2010.04.006Search in Google Scholar

[127] Kasimu R, Wang X, Wang X, et al. Antithrombotic effects and related mechanisms of Salvia deserta Schang root EtOAc extracts. Sci Rep, 2018; 8(1): 17753.10.1038/s41598-018-36026-7Search in Google Scholar

[128] Yang Y Y, Wu Z Y, Zhang H, et al. LC-MS-based multivariate statistical analysis for the screening of potential thrombin/factor Xa inhibitors from Radix Salvia Miltiorrhiza. Chin Med, 2020; 15(1): 38.10.1186/s13020-020-00320-2Search in Google Scholar

[129] Wang D, Girard T J, Kasten T P, et al. Inhibitory activity of unsaturated fatty acids and anacardic acids toward soluble tissue factor-factor VIIa complex. J Nat Prod, 1998; 61(11): 1352–1355.10.1021/np980117pSearch in Google Scholar

[130] Wu J, Luo Y, Deng D, et al. Coptisine from Coptis chinensis exerts diverse beneficial properties: A concise review. J Cell Mol Med, 2019; 23: 7946–7960.10.1111/jcmm.14725Search in Google Scholar

[131] Yang J, Ljn J. Advance on study in anti-tumor mechamism of bererine (Ber). Zhongguo Zhong Yao Za Zhi, 2007; 32(10): 881–883, 934.Search in Google Scholar

[132] Huang C G, Chu Z L, Wei S J, et al. Effect of berberine on arachidonic acid metabolism in rabbit platelets and endothelial cells. Thromb Res, 2002; 106(4–5): 223–227.10.1016/S0049-3848(02)00133-0Search in Google Scholar

[133] Hui K K, Yu J L, Chan W F, et al. Interaction of berberine with human platelet alpha 2 adrenoceptors. Life Sci, 1991; 49(4): 315–324.10.1016/0024-3205(91)90019-8Search in Google Scholar

[134] Wang X, Zhang Y, Yang Y, et al. Identification of berberine as a direct thrombin inhibitor from traditional Chinese medicine through structural, functional and binding studies. Sci Rep, 2017; 7(1): 44040.10.1038/srep44040Search in Google Scholar

[135] Ho J W, Jie M. Pharmacological activity of cardiovascular agents from herbal medicine. Cardiovasc Hematol Agents Med Chem, 2007; 5(4): 273–277.10.2174/187152507782109854Search in Google Scholar

[136] Sheu J R, Kan Y C, Hung W C, et al. Mechanisms involved in the antiplatelet activity of tetramethylpyrazine in human platelets. Thromb Res, 1997; 88(3): 259–270.10.1016/S0049-3848(97)00253-3Search in Google Scholar

[137] Liu S Y, Sylvester D M. Antiplatelet activity of tetramethylpyrazine. Thromb Res, 1994; 75: 51–62.10.1016/0049-3848(94)90139-2Search in Google Scholar

[138] Sheu J R, Kan Y C, Hung W C, et al. The antiplatelet activity of tetramethylpyrazine is mediated through activation of NO synthase. Life Sci, 2000; 67: 937–947.10.1016/S0024-3205(00)00686-XSearch in Google Scholar

[139] Li L, Chen H, Shen A, et al. Ligustrazine inhibits platelet activation via suppression of the Akt pathway. Int J Mol Med, 2019; 43(1): 575–582.10.3892/ijmm.2018.3970Search in Google Scholar

[140] Zhang Q, Yang Y X, Li S Y, et al. An ultrafiltration and high performance liquid chromatography coupled with diode array detector and mass spectrometry approach for screening and characterizing thrombin inhibitors from Rhizoma Chuanxiong. J Chromatogr B Analyt Technol Biomed Life Sci, 2017; 1061–1062: 421–429.10.1016/j.jchromb.2017.07.050Search in Google Scholar

[141] Tsui P F, Lin C S, Ho L J, et al. Spices and Atherosclerosis. Nutrients, 2018; 10(11): 1724.10.3390/nu10111724Search in Google Scholar

[142] Srivas K C. Effects of aqueous extracts of onion, garlic and ginger on platelet aggregation and metabolism of arachidonic acid in the blood vascular system: in vitro study. Prostaglandins Leukot Med, 1984; 13(2): 227–235.10.1016/0262-1746(84)90014-3Search in Google Scholar

[143] Srivastava K C. Effect of onion and ginger consumption on platelet thromboxane production in humans. Prostaglandins Leukot Essent Fatty Acids, 1989; 35(3): 183–185.10.1016/0952-3278(89)90122-1Search in Google Scholar

[144] Koo K L, Ammit A J, Tran V H, et al. Gingerols and related analogues inhibit arachidonic acid-induced human platelet serotonin release and aggregation. Thromb Res, 2001; 103(5): 387–397.10.1016/S0049-3848(01)00338-3Search in Google Scholar

[145] Nurtjahja-Tjendraputra E, Ammit A J, Roufogalis B D, et al. Effective anti-platelet and COX-1 enzyme inhibitors from pungent constituents of ginger. Thromb Res, 2003; 111(4–5): 259–265.10.1016/j.thromres.2003.09.009Search in Google Scholar PubMed

[146] Lee W, Ku S K, Kim M A, et al. Anti-factor Xa activities of zingerone with anti-platelet aggregation activity. Food Chem Toxicol, 2017; 105: 186–193.10.1016/j.fct.2017.04.012Search in Google Scholar PubMed

[147] Jiang W, Shan T Z, Xu J J, et al. Cytotoxic abietane and kaurane diterpenoids from Celastrus orbiculatus. J Nat Med, 2019; 73(11): 841–846.10.1007/s11418-019-01326-3Search in Google Scholar PubMed

[148] Guo Y Q, Li X, Xu J, et al. Sesquiterpene esters from the fruits of Celastrus orbiculatus. Chem Pharm Bull (Tokyo), 2004; 52(9): 1134–1136.10.1248/cpb.52.1134Search in Google Scholar PubMed

[149] Zhou J, Zhai J, Zheng W, et al. The antithrombotic activity of the active fractions from the fruits of Celastrus orbiculatus Thunb through the anti-coagulation, anti-platelet activation and anti-fibrinolysis pathways. J Ethnopharmacol, 2019; 241: 111974.10.1016/j.jep.2019.111974Search in Google Scholar PubMed

[150] Han J, Ye M, Xu M, et al. Characterization of flavonoids in the traditional Chinese herbal medicine-Huangqin by liquid chromatography coupled with electrospray ionization mass spectrometry. J Chromatogr B Analyt Technol Biomed Life Sci, 2007; 848(2): 355–362.10.1016/j.jchromb.2006.10.061Search in Google Scholar PubMed

[151] Zhang Y, Wang X, Wang X, et al. Protective effect of flavonoids from Scutellaria baicalensis Georgi on cerebral ischemia injury. J Ethnopharmacol, 2006; 108(3): 355–360.10.1016/j.jep.2006.05.022Search in Google Scholar PubMed

[152] Ku S K, Lee I C, Bae J S. Antithrombotic activities of oroxylin A in vitro and in vivo. Arch Pharm Res, 2014; 37: 679–686.10.1007/s12272-013-0233-0Search in Google Scholar PubMed

[153] Lee W, Ku S K, Bae J S. Antiplatelet, anticoagulant, and profibrinolytic activities of baicalin. Arch Pharm Res, 2015; 38(5): 893–903.10.1007/s12272-014-0410-9Search in Google Scholar PubMed

[154] Ku S K, Bae J S. Antithrombotic activities of wogonin and wogonoside via inhibiting platelet aggregation. Fitoterapia, 2014; 98: 27–35.10.1016/j.fitote.2014.07.006Search in Google Scholar PubMed

[155] Liu X F, Liu M L, Iyanagi T, et al. Inhibition of rat liver NAD(P)H:quinone acceptor oxidoreductase (DT-diaphorase) by flavonoids isolated from the Chinese herb scutellariae radix (Huang Qin). Mol Pharmacol, 1990; 37(6): 911–915.Search in Google Scholar

[156] Wu Y H, Chuang L P, Yu C L, et al. Anticoagulant effect of wogonin against tissue factor expression. Eur J Pharmacol, 2019; 859: 172517.10.1016/j.ejphar.2019.172517Search in Google Scholar PubMed

[157] Zhang X, Jin M, Tadesse N, et al. Dioscorea zingiberensis C. H. Wright: An overview on its traditional use, phytochemistry, pharmacology, clinical applications, quality control, and toxicity. J Ethnopharmacol, 2018; 220: 283–293.10.1016/j.jep.2018.03.017Search in Google Scholar PubMed

[158] Li H, Huang W, Wen Y, et al. Anti-thrombotic activity and chemical characterization of steroidal saponins from dioscorea zingiberensis C.H. Wright. Fitoterapia, 2010; 81(1): 1147–1156.10.1016/j.fitote.2010.07.016Search in Google Scholar PubMed

[159] Gong G, Qin Y, Huang W. Anti-thrombosis effect of diosgenin extract from Dioscorea zingiberensis C.H. Wright in vitro and in vivo. Phytomedicine, 2011; 18(6): 458–463.10.1016/j.phymed.2010.08.015Search in Google Scholar PubMed

[160] Zheng H, Wei Z, Xin G, et al. Preventive effect of a novel diosgenin derivative on arterial and venous thrombosis in vivo. Bioorg Med Chem Lett, 2016; 26(14): 3364–3369.10.1016/j.bmcl.2016.05.032Search in Google Scholar PubMed

[161] Chen Y, Chen P D, Bao B H, et al. Anti-thrombotic and pro-angiogenic effects of Rubia cordifolia extract in zebrafish. J Ethnopharmacol, 2018; 219: 152–160.10.1016/j.jep.2017.11.005Search in Google Scholar PubMed

[162] Shan M, Yu S, Yan H, et al. A review of the botany, phytochemistry, pharmacology and toxicology of rubiae radix et rhizoma. Molecules, 2016; 21(12): 1747.10.3390/molecules21121747Search in Google Scholar PubMed PubMed Central

[163] Nishimura N, Takai M, Yamamoto E, et al. Purpurin as a specific inhibitor of spermidine-induced autoactivation of the protease plasma hyaluronan-binding protein. Biol Pharm Bull, 2010; 33(8): 1430–1433.10.1248/bpb.33.1430Search in Google Scholar PubMed

[164] Auyeung K K, Han Q B, Ko J K. Astragalus membranaceus: A review of its protection against inflammation and gastrointestinal cancers. Am J Chin Med, 2016; 44(1): 1–22.10.1142/S0192415X16500014Search in Google Scholar

[165] Zhao J, Yang P, Li F, et al. Therapeutic effects of astragaloside IV on myocardial injuries: multi-target identification and network analysis. PLoS One, 2012; 7(9): e44938.10.1371/journal.pone.0044938Search in Google Scholar

[166] Zhang W J, Wojta J, Binder B R. Regulation of the fibrinolytic potential of cultured human umbilical vein endothelial cells: astragaloside IV downregulates plasminogen activator inhibitor-1 and upregulates tissue-type plasminogen activator expression. J Vasc Res, 1997; 34(4): 273–280.10.1159/000159234Search in Google Scholar

[167] Koo Y K, Kim J M, Koo J Y, et al. Platelet anti-aggregatory and blood anti-coagulant effects of compounds isolated from Paeonia lactiflora and Paeonia suffruticosa. Pharmazie, 2010; 65(8): 624–628.Search in Google Scholar

[168] Xie P, Cui L, Shan Y, et al. Antithrombotic effect and mechanism of radix paeoniae rubra. Biomed Res Int, 2017; 17: 9475074.10.1155/2017/9475074Search in Google Scholar

[169] Ye J, Duan H, Yang X, et al. Anti-thrombosis effect of paeoniflorin: evaluated in a photochemical reaction thrombosis model in vivo. Planta Med, 2001; 67(8): 766–767.10.1055/s-2001-18364Search in Google Scholar

[170] Ngo T, Kim K, Bian Y, et al. Antithrombotic effects of paeoniflorin from paeonia suffruticosa by selective inhibition on shear stress-induced platelet aggregation. Int J Mol Sci, 2019; 20(20): 5040.10.3390/ijms20205040Search in Google Scholar

[171] Ye S, Mao B, Yang L, et al. Thrombosis recanalization by paeoniflorin through the upregulation of urokinasetype plasminogen activator via the MAPK signaling pathway. Mol Med Rep, 2016; 13(6): 4593–4598.10.3892/mmr.2016.5146Search in Google Scholar

[172] Kocaadam B, Sanlier N. Curcumin, an active component of turmeric (Curcuma longa), and its effects on health. Crit Rev Food Sci Nutr, 2017; 57(13): 2889–2895.10.1080/10408398.2015.1077195Search in Google Scholar

[173] Srivastava K C, Bordia A, Verma S K. Curcumin, a major component of food spice turmeric (Curcuma longa) inhibits aggregation and alters eicosanoid metabolism in human blood platelets. Prostaglandins Leukot Essent Fatty Acids, 1995; 52(4): 223–227.10.1016/0952-3278(95)90040-3Search in Google Scholar

[174] Lee H S. Antiplatelet property of Curcuma longa L. rhizome-derived ar-turmerone. Bioresour Technol, 2006; 97(12): 1372–1376.10.1016/j.biortech.2005.07.006Search in Google Scholar PubMed

[175] Manikandan P, Sumitra M, Aishwarya S, et al. Curcumin modulates free radical quenching in myocardial ischaemia in rats. Int J Biochem Cell Biol, 2004; 36(10): 1967–1980.10.1016/j.biocel.2004.01.030Search in Google Scholar PubMed

[176] Heemskerk J W, Sage S O. Calcium signalling in platelets and other cells. Platelets, 1994; 5(6): 295–316.10.3109/09537109409006439Search in Google Scholar PubMed

[177] Mayanglambam A, Dangelmaier C A, Thomas D, et al. Curcumin inhibits GPVI-mediated platelet activation by interfering with the kinase activity of Syk and the subsequent activation of PLCgamma2. Platelets, 2010; 21(3): 211–220.10.3109/09537100903528269Search in Google Scholar PubMed

[178] Xia Q, Wang X, Xu D J, et al. Inhibition of platelet aggregation by curdione from Curcuma wenyujin essential Oil. Thromb Res, 2012; 130(3): 409–414.10.1016/j.thromres.2012.04.005Search in Google Scholar PubMed

[179] Kim D C, Ku S K, Bae J S. Anticoagulant activities of curcumin and its derivative. BMB Rep, 2012; 45(4): 221–226.10.5483/BMBRep.2012.45.4.221Search in Google Scholar

[180] Koizume S, Yokota N, Miyagi E, et al. Hepatocyte nuclear factor-4-independent synthesis of coagulation factor VII in breast cancer cells and its inhibition by targeting selective histone acetyltransferases. Mol Cancer Res, 2009; 7(12): 1928–1936.10.1158/1541-7786.MCR-09-0372Search in Google Scholar PubMed PubMed Central

[181] Madhyastha R, Madhyastha H, Nakajima Y, et al. Curcumin facilitates fibrinolysis and cellular migration during wound healing by modulating urokinase plasminogen activator expression. Pathophysiol Haemost Thromb, 2010; 37(2–4): 59–66.10.1159/000321375Search in Google Scholar PubMed

[182] Grau T, Vilcinskas A, Joop G. Sustainable farming of the mealworm Tenebrio molitor for the production of food and feed. Z Naturforsch C J Biosci, 2017; 72(9): 337–349.10.1515/znc-2017-0033Search in Google Scholar PubMed

[183] Lee W, Kim M A, Park I, et al. Novel direct factor Xa inhibitory compounds from Tenebrio molitor with anti-platelet aggregation activity. Food Chem Toxicol, 2017; 109(Pt 1): 19–27.10.1016/j.fct.2017.08.026Search in Google Scholar PubMed

Received: 2021-01-20
Accepted: 2021-03-31
Published Online: 2021-09-18

© 2021 Xueqing Tang et al., published by Sciendo

This work is licensed under the Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License.

Downloaded on 30.4.2024 from https://www.degruyter.com/document/doi/10.2478/fzm-2021-0005/html
Scroll to top button