Next Article in Journal
Commercially Available Viola odorata Oil, Chemical Variability and Antimicrobial Activity
Next Article in Special Issue
Protective Effects of Hemp (Cannabis sativa) Root Extracts against Insulin-Deficient Diabetes Mellitus In Mice
Previous Article in Journal
Photodynamics of the Molecular Ruby [Cr(ddpd)2]3+
Previous Article in Special Issue
Current Landscape of Methods to Evaluate Antimicrobial Activity of Natural Extracts
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Plant-Derived Compounds and Extracts as Modulators of Plasmin Activity—A Review

by
Joanna Kolodziejczyk-Czepas
1,* and
Jan Czepas
2
1
Department of General Biochemistry, Faculty of Biology and Environmental Protection, University of Lodz, 90-236 Lodz, Poland
2
Department of Oncobiology and Epigenetics, Faculty of Biology and Environmental Protection, University of Lodz, 90-236 Lodz, Poland
*
Author to whom correspondence should be addressed.
Molecules 2023, 28(4), 1677; https://doi.org/10.3390/molecules28041677
Submission received: 30 December 2022 / Revised: 3 February 2023 / Accepted: 8 February 2023 / Published: 9 February 2023

Abstract

:
Functionality of the fibrinolytic system is based on activity of its central enzyme, plasmin, responsible for the removal of fibrin clots. Besides the hemostasis, fibrinolytic proteins are also involved in many other physiological and pathological processes, including immune response, extracellular matrix degradation, cell migration, and tissue remodeling. Both the impaired and enhanced activity of fibrinolytic proteins may result in serious physiological consequences: prothrombotic state or excessive bleeding, respectively. However, current medicine offers very few options for treating fibrinolytic disorders, particularly in the case of plasmin inhibition. Although numerous attempts have been undertaken to identify natural or to develop engineered fibrinolytic system modulators, structural similarities within serine proteases of the hemostatic system and pleiotropic activity of fibrinolytic proteins constitute a serious problem in discovering anti- or profibrinolytic agents that could precisely affect the target molecules and reduce the risk of side effects. Therefore, this review aims to present a current knowledge of various classes of natural inhibitors and stimulators of the fibrinolytic system being well-defined low-molecular plant secondary metabolites or constituents of plant extracts as well as plant peptides. This work also discusses obstacles caused by low specificity of most of natural compounds and, hence, outlines recent trends in studies aimed at finding more efficient modulators of plasmin activity, including investigation of modifications of natural pharmacophore templates.

1. Introduction

Among complex mechanisms of hemostasis, the fibrinolytic system has been primarily considered a natural counterbalance of the blood coagulation cascade, responsible for degradation of fibrin clots and maintaining blood fluidity. Physiology of the fibrinolytic system is based on proteolytic activity of the plasmin enzyme and different regulatory mechanisms preserving the hemostatic balance and preventing an excessive proteolysis within blood vessels and tissues surrounding the vasculature. However, removal of fibrin clots is only a part of pleiotropic activity, both of plasmin itself and other fibrinolytic proteins. Functions of the fibrinolytic system extend beyond blood physiology and vascular hemostasis and include participation in extracellular matrix (ECM) degradation, tissue remodeling, wound healing, immune response, and other physiological and pathological processes [1,2,3] (Figure 1). Besides degradation of fibrinogen and fibrin, plasmin hydrolyzes many other protein substrates, including blood coagulation factors (V, VIII, IX, X, von Willebrand factor, FXII, and pre-kallikrein), vascular endothelial growth factor, C3 and C5 complement components, protease-activated receptor-1 (PAR-1), matrix metalloproteinase zymogenes (proMMPs), as well as ECM proteins, e.g., fibronectin, tenascin, collagen, and laminin [4].
Due to an increased risk of thrombo-embolic complications in diseases of civilization, including atherosclerosis, metabolic syndrome, obesity, and cancer, numerous studies have been undertaken to improve diagnostics of coagulation cascade abnormalities and current anticoagulant therapies, as well as to develop novel therapeutic strategies. Many of the aforementioned studies have been mostly focused on inhibition of key proteinases of the blood coagulation cascade and reduction in platelet activity [5,6,7,8,9]. This imbalance between the blood coagulation-centered approach and fibrinolysis-targeting studies has partly contributed to less advanced development of fibrinolysis-targeting therapy. Current medicine offers only three inhibitors to reduce fibrinolysis and few activators of fibrinolysis [10].
Although plasminogen deficiency is very rare (1.6 per million) [11], diverse types of fibrinolysis disorders occur in many other diseases. Under pathological conditions associated with a pro-coagulant state, impaired fibrinolysis becomes an additional risk factor of thrombosis [12,13]. On the other hand, an increased activity of the fibrinolytic system is a risk of uncontrolled bleedings associated with different surgical procedures [14,15]. Antifibrinolytics are used in patients with von Willebrand disease [16] and to treat thrombolytic-induced bleedings [17]. Although antifibrinolytics may be helpful in the treatment of mucosal bleedings, they should be administered with caution to patients with congenital fibrinogen disorders with a personal or family history of thrombotic episode [18].
Fibrinolysis is a tightly regulated process, involving interactions of the fibrin surface, plasminogen, and its main activator, i.e., the tissue-type plasminogen activator (t-PA). It begins with formation of the fibrin clot and the activation of t-PA. Release of t-PA from endothelial cells is a critical step in intravascular fibrinolysis, enabling the conversion of the plasminogen zymogen into the plasmin enzyme [19]. Fibrinogen mediates both the coagulation and fibrinolysis processes through thrombin binding and limiting its activity as well as through interactions with the coagulation factor FXIII, plasminogen, t-PA, and antiplasmin [20]. Preliminary efforts to recognize possibility of fibrinolysis modulation by natural compounds or extracts originated from plants were undertaken as early as the 1970s and 1980s [21]. From that time, many interesting results have appeared. This review aims to summarize a current state of the art in the field of plant-derived modulators of fibrinolysis, in the context of a search for inhibitors of fibrinolytic proteins and substances displaying thrombolytic or fibrinolysis-stimulatory properties.

2. Plasminogen Structure and Main Components of the Fibrinolytic System

The functional core of the fibrinolytic system is the plasminogen (zymogene)-plasmin (the active enzyme) axis, controlled by activators and inhibitors (Figure 2). The structure of native plasminogen, named the Glu-plasminogen (Glu-Plg, 92 kDa), comprises 791 amino acids (aa) and is organized into seven domains, as follows: N-terminal Pan-apple (PAp) domain, five homologous kringle domains (K1-K5), and a serine protease (SP) domain. Plasminogen (Plg) is mainly synthesized in the liver and circulates in blood flow at a concentration of about 2 μM (0.18 mg/mL) in two single-chain glycoforms, i.e., glycoform I and II. Hydrolytic removal of the N-terminal peptide (77 aa) from the Glu-Plg molecule by plasmin leads to the formation of a slightly shorter form of the zymogene, i.e., Lys-Plg, which is more prone to the proteolytic activation by plasmin, compared to the Glu-Plg. The kringle domains in the plasminogen/plasmin molecule (about 80 aa) are triple-looped structures, stabilized by three intrachain disulfide bonds and contain the lysine binding sites (LBS). Among five kringle domains, four contain the functional LBS. In the K1, K2, K4, and K5 domains, DXD/E motifs enabling their interaction with lysine ligands are present. The K3 domain contains a DXK sequence instead, which results in the lack of ability to bind lysine. This dysfunctional LBS can be activated as a result of substitution of Lys311 to Asp [22,23,24]. A general scheme of kringle sequences is based on anionic and cationic centers separated by a hydrophobic region. While the anionic centers of K1, K2, K4, and K5 are very similar (including two acidic residues at identical positions), the cationic centers display higher variability. The cationic center of K1 contains two positively charged amino acid residues (i.e., R117 and R153), with a hydrophobic residue of Y146. The K2 cationic center contains only R234. In the cationic center of the K4 domain, K392 and R426 are present. The K5 domain lacks the cationic center [25]. Structural differences between the kringles (and LBS) influence their ligand binding capacity. The K1 and K4 domains were found to display a preference for lysine-like dipolar molecules, and K2 and K5 have a marginal affinity for these types of ligands [26].
In blood flow, plasminogen circulates in a closed conformation, maintained by various intramolecular interactions (for details see: [4,22]). The critical step in plasminogen activation is its LBS-mediated interaction with the target protein or receptors on the cell surface. This interaction initiates conformational changes in the plasminogen molecule, converting its closed structure into open conformation to expose the activation loop. During the immobilization, K2, K4, and K5 domains (bound to the self-lysines/arginines in the closed conformation of plasminogen) are switched onto lysine residues of the target surface. The LBS are responsible for interactions both with fibrinogen/fibrin and targets such as α2-antiplasmin, plasminogen receptors on the cell surface, as well as other ligands [22,23,24,27].
The activation of plasminogen to plasmin occurs through a cleavage of the Arg561-Val562 peptide bond. The plasmin (EC 3.4.21.7) is a two-chain enzyme with an active center (Ser741, His603, and Asp646) located in the B (light) chain (25 kDa). The heavy A chain (60 kDa) contains the kringle structures. Plasminogen conversion to plasmin is primarily mediated by the tissue-type plasminogen activator (t-PA, EC 3.4.21.68) and the urokinase-type plasminogen activator (u-PA/urokinase, EC 3.4.21.73). The t-PA-mediated plasminogen conversion to plasmin is the main mechanism of thrombolysis activation in blood vessels, whereas the u-PA and its receptor are mainly involved in the extravascular proteolysis, including the tissue remodeling. The plasminogen proenzyme can be also proteolytically activated by small amounts of already generated plasmin as well as by interactions with other proteases, including the blood coagulation cascade enzymes such as factors XIa and XIIa and kallikrein. Plasmin is able to amplify fibrinolysis efficiency by a direct activation of plasminogen molecules or by proteolysis of the single chain t-PA and u-PA to their more active, double-chain forms [28].
During the phase of plasminogen conversion to plasmin, activity of the fibrinolytic system is regulated by the plasminogen activator inhibitors (PAIs), members of the SERPINs family. The plasminogen activator inhibitor-1 (PAI-1), able to inhibit both t-PA and u-PA, is the main inhibitor of plasmin generation. The plasminogen activator inhibitor-2 (PAI-2) selectively inactivates u-PA and is considered an additional regulator of plasmin formation. Plasmin activity is inhibited via the formation of irreversible complexes with α2-antiplasmin (the main plasmin inhibitor) and, to a minor extent, by α2-macroglobulin [29]. Indirectly, plasminogen activation and plasmin generation are controlled by the thrombin-activatable fibrinolysis inhibitor (TAFI). This regulator of the fibrinolytic rate removes C-terminal lysine residues from the partially degraded fibrin clot, and therefore it limits plasminogen binding and activation [30].
Biochemical characteristics of main components of the fibrinolytic system are briefly presented in Table 1.

3. Current Pro- and Anti-Fibrinolytic Therapies

The number of pharmacological regimens available for patients requiring thrombolytic or antifibrinolytic therapy is limited [31,32,33,34] (Table 2). Currently used drugs enable modulation of the fibrinolytic system activity by regulation (activation or inhibition) of plasminogen conversion to plasmin or by inhibition of plasmin activity. Maintaining the fibrinolytic potential of blood plasma is an important element of anti-thrombotic therapy. Thrombolytic therapy is crucial for dissolving intravascular clots and thus for maintaining blood flow and preventing ischemic damage in patients with acute myocardial infarction, acute ischemic stroke, deep vein thrombosis, pulmonary embolism, and other types of thrombo-embolic complications. It is based on non-fibrin-specific and fibrin-specific plasminogen activators, including exogenous activators such as streptokinase and variants of t-PA, i.e., tenecteplase (TNK-tPA), and reteplase (Figure 3). Adverse effects of this type of treatment are bleeding, hypotension, and allergic reactions (most frequently, after use of streptokinase) [35,36,37].
Possibility of extensive postoperative bleeding or other risks of blood loss are reduced by the antifibrinolytic treatment. At the level of plasminogen activation, fibrinolysis is inhibited by administration of lysine analogues, impairing plasminogen activation to plasmin [38,39,40]. Clinically used drugs belonging to this group (i.e., tranexamic acid (TXA) and aminocaproic acid (EACA)) are capable of blocking the LBS of the kringle domains in the plasminogen molecule, which are crucial for interactions of this protein with fibrin and other target surfaces. The amino group of TXA or EACA is bound to the DXD motif of the LBS anionic centers, and the carboxyl group interacts with the cationic centers [26]. Among four kringle domains containing functional LBS, the K1 displays the highest affinity for both TXA and EACA, followed by K4, K5, and K2 [22,41].
The only currently approved direct inhibitor of plasmin is aprotinin (Trasylol), belonging to the Kunitz-type inhibitors of serine proteases. However, its use in clinical routine has been widely discussed due to adverse effects, including an increased risk of anaphylaxis, renal failure, and mortality. Based on an increasing number of data on serious adverse effects, aprotinin was withdrawn from the European market in 2008. Later, however, clinical use of aprotinin was reacknowledged, but its administration requires careful risk–benefit assessment [42].

4. Plant-Derived Inhibitors of the Fibrinolytic System

4.1. General Insight into Phytochemicals-Plasmin(ogen) Interactions and the Problem of Selectivity of Compounds

Analogously to physiological mechanisms of fibrinolytic activity control, the possibility of fibrinolysis modulation by plant-derived substances at different molecular levels can also be taken into consideration. Existing reports provide data from tests of different substances, both synthetic and natural, with anti-plasmin activity and emphasize the potential of small molecules. The trypsin-like proteases, including enzymes of the hemostatic system, seem to be prone to action of plant-derived substances, based on different carbon backbones and representing diverse classes of low-molecular phytochemicals, e.g., flavonoids, alcoholic β-glucosides, coumarin derivatives, anthrones, and dianthrones and even the steroid-type compounds such as bufadienolides [43,44,45,46,47,48,49,50,51,52,53,54,55] (Table 3). However, promising outcomes obtained from studies on native or modified peptides encourage further research on plant polypeptides as well. Studies on peptide-based plant inhibitors of serine proteases evidently indicate their higher efficiency when compared to (poly)phenolic compounds, for example, AVPI-12, a protease inhibitor from Aloe vera L. leaves with a unique RDWAEPNDGY motif starting its N-terminal region inhibited the plasmin-mediated hydrolysis of human fibrin(ogen) in vitro with efficiency comparable to α2-macroglobulin [56]. Both the Kunitz and other types of peptide inhibitors of different plant origin act mostly at nanomolar and even sub-nanomolar concentrations [57,58,59,60,61,62,63,64,65,66,67,68] (Table 4). In contrast, the most active plant polyphenolics are able to inhibit serine proteases or other enzymes at micromolar concentrations (Table 3).
The central enzyme of fibrinolysis, plasmin, is an obvious target in the search for new regulators of the activity of the fibrinolytic system. However, as a member of the trypsin-like protease group (family S1 (trypsin-like fold), the PA clan), plasmin shares many structural and functional features with other serine proteases, including two β-barrel-like regions in the catalytic domain (described as the “Greek key”), the catalytic triad (composed of His, Asp and Ser) and the nucleophilic attack of the hydroxyl group of the active serine on the carbonyl of the peptide substrate and preferential cleaving on the arginine or lysine [69]. Similarities of the trypsin-like enzymes significantly hinder identification of natural compounds or development of new drugs capable of precisely targeting the activity of plasmin. Even aprotinin, a clinically approved Kunitz-type inhibitor of serine proteases, may interact not only with the active center of plasmin but also with other enzymes, including trypsin, kallikrein, urokinase, elastase, and thrombin. Moreover, the inhibitory efficiency of aprotinin towards plasmin is markedly lower than its ability to block the trypsin enzyme (Ki of 0.01 and 0.00006 nM for plasmin and trypsin, respectively) [70]. Therefore, development of an effective and specific plasmin inhibitor formulation is a demanding challenge, but the plant-derived substances are considered as promising pharmacophores, also in the context of selectivity. One previous study screened 55 synthetic, sulfated glycosaminoglycan mimetics, based on nine distinct scaffolds. Among the examined chalcones, flavonoids, sucrose octasulfate, quinazolinones, tetrahydroisoquinolines, flavonoid-quinazolinone heterodimers, bis-quinazolinone homodimers, and bis-flavonoid homodimers, a pentasulfated flavonoid-quinazolinone dimer was the most effective inhibitor of plasmin (IC50 = 45 μM). Furthermore, the flavonoid-quinazolinone dimer was selective for plasmin over thrombin and the coagulation factor Xa [71].

4.2. Plant-Derived Polypeptide Inhibitors of Fibrinolysis

Plants may be a source of a wide range of peptide-based protease inhibitors such as SERPINs, Kunitz-type inhibitors, Bowman-Birk inhibitors, Kazal-domain inhibitors, cyclic cysteine-rich peptides, phytocystatins, metallocarboxypeptidase inhibitors, potato-type inhibitors, and mustard-type trypsin inhibitors [72,73]. Many of the identified serine protease inhibitors have been isolated from the Leguminosae (Fabaceae) family. The best-known is, of course, the soybean trypsin inhibitor (STI), isolated and characterized by Kunitz [74,75], but plants synthesize a variety of Kunitz-type inhibitors with significant diversity in cysteine content, arrangement of disulfide bonds, as well as other structural and functional differences [76]. For example, Bauhinia rufa (Bong.) Steud. seeds contain BrTI, a Kunitz proteinase inhibitor containing the RGD sequence [77], which could be useful in modulation of biological processes which are unrelated to removal of the fibrin clot. The RGD sequence has been found in many integrin ligands (e.g., fibronectin, vitronectin, and fibrinogen) and acts as one of the most important cell adhesion motifs, crucial for the ECM-cell interactions, platelet aggregation, and other protein-cell membrane interactions [78,79]. Incorporation of these three amino acid motifs is considered an important modification in development of new biomaterials and drugs. In the past few years, a formula of cyclic RGD-functionalized liposomes with encapsulated urokinase was proposed for enhancement of thrombolytic efficacy [80].
However, low selectivity still remains one of the major bottlenecks to overcome. While modulation of the hemostatic system requires a precise impact on the target enzymes to recover the fibrinolysis-coagulation balance, plant-derived inhibitors are mostly capable of interacting with both the fibrinolytic enzymes and the coagulation cascade factors (Table 4). Moreover, their inhibitory efficiency towards the coagulation cascade and fibrinolytic proteins may be similar. Although the Bauhinia genus has been considered a rich source of natural inhibitors of cysteine and serine proteinases, so far, inhibitors isolated from these plants have been shown to influence the activities of plasmin and coagulation factors with comparable efficiency. For BuXI, one of the Bauhinia-derived inhibitors, the plasmin Ki value was established to be 76 nM, and the factor XIIa Ki attained 74 nM [61]. Another Kunitz-type inhibitor, CeKI isolated from Caesalpinia echinata Lam., was found to inhibit plasmin and the coagulation factor XIIa activities with the same efficiency, i.e., with an Ki value of 0.18 nM [62]. Some selectivity has been observed for AaTI, a serine protease inhibitor isolated from Araucaria angustifolia (Bertol.) Kuntze seeds. The inhibitor did not influence activities of chymotrypsin, human plasma kallikrein, or other coagulation enzymes; however, AaTI was more active towards trypsin (Ki = 85 nM) than plasmin (Ki = 7.0 μM) [59].
Among plant-derived peptide inhibitors, a 14-amino acid backbone-cyclic sunflower trypsin inhibitor-1 (SFTI-1) seems to be particularly promising. It is the smallest known Bowman-Birk peptide inhibitor, and its structure is stabilized by a single disulfide bond and an intramolecular hydrogen bond network formed between the antiparallel β-strands. The SFTI-1 template, modified by amino acid sequences derived from serpin reactive center loops, has been used to develop selective inhibitors of kallikrein-related peptidases 5 and 7 [81]. The SFTI-1 scaffold was also used as a base for development of a new and effective plasmin inhibitor, called SFTIv3 [82]. Unlike the aprotinin, the designed inhibitor displayed a higher potency towards plasmin (Ki = 0.051 nM) than trypsin (Ki = 160 nM) and a million-fold selectivity over the S1 family of serine proteases of blood. The inhibitor was characterized by no detectable inhibition of thrombin, FIXa, FXa, FXIa, FXIIa, t-PA, u-PA, plasma kallikrein, or matriptase. Moreover, further studies resulted in development of its rapid synthesis method, based on transient expression of the peptide in Nicotiana benthamiana Domin [83].

4.3. (Poly)Phenolic Compounds and Other Secondary Metabolites as Inhibitors of Fibrinolysis

Many aspects of plasmin structure and specificity are strictly related to its interactions with (poly)peptide substrates (e.g., preference for Lys over Arg at the P1 position, a slight preference for Tyr over Phe or Trp at the P2 site, and more restricted preferences at other subsites, with reduced levels of lysis of sequences with Trp at the P2 site when Arg was located at the P1 site, were found [84]). These features provide new opportunities for further research and may be useful in development of peptidomimetics of various types (reviewed in [85]) but cannot be directly transferred into the polyphenolic compounds-plasmin interactions. On the other hand, these findings may be helpful in the design of new, combined inhibitors. It has been emphasized that some structural features of both plant-derived and synthetic substances may be essential for their inhibitory action on serine proteases. Three-dimensional QSAR comparative molecular field analysis (CoMFA), employing u-PA, t-PA, plasmin, the coagulation factor Xa, thrombin, and trypsin, suggested the selectivity of molecules with indole/benzoimidazole-5-carboxamidine component towards serine proteases [86]. Some structural similarities of these substances with the quercetin molecule, such as the sterically favorable presence of a phenyl ring and electrostatically interacting hydroxyl group, have been also described [53].
Phytochemicals with two adjacent phenolic hydroxyl groups (or catechol moiety) may be inhibitors of various trypsin-like proteases due to highly conserved structural analogies in enzymes from the S1 family. The X-ray crystallography analyses of the quercetin-u-PA complex structure demonstrated that this flavonoid interacted with the active site of u-PA. The phenyl ring bearing a catechol group (commonly referred also as the B ring) of quercetin was found to insert into the specific substrate binding pocket (S1 pocket), and two hydroxyl groups of the benzene ring (A ring) of the benzopyran part of quercetin molecule interacted with the S2 pocket of u-PA [87]. The role of hydroxyl groups was also confirmed in study on anthocyanidins, which demonstrated the ability of delphinidin, cyanidin, and petunidin to inhibit the u-PA-dependent activation of plasminogen to plasmin. Delphinidin with three hydroxyl groups in the B-ring was the most potent inhibitor, followed by compounds containing two hydroxyl groups in the B-ring, i.e., cyanidin and petunidin. Furthermore, analyses of inhibitory effects of anthocyanidins on the migration of glioblastoma cells involving plasmin suggested an important role of a free hydroxyl group at position 3. Delphinidin was the most effective inhibitor of cell migration, but the presence of sugar residue in position 3 (delphinidin 3-glucoside) abolished the inhibitory action [88]. Recently, pharmacophore modeling, in silico docking, and molecular dynamics simulation of the flavonoid-enzyme interactions demonstrated four features (signed as F1-F4) that may be essential for selective modulation of urokinase. These critical features included the presence of aromatic rings (F1 and F2), connected to Cys191, Gln192, Ser195, and Trp215 residues; a cationic H-bond donor (F3) corresponding to Asp189, Ser190, and Gly219; and a metal ligator (F4) corresponding to Gly216. Based on above features, isorhamnetin, rhamnetin, quercetin, and kaempferol were indicated as the most effective ligands for u-PA [89].
Experiments on plasmin-inhibitory effects of flavonoids isolated from Blumea balsamifera DC. demonstrated that two the most effective compounds (IC50 = 1.5 and 2.3 µM) had hydroxyl groups at 3′ and 4′ positions. These most efficient inhibitors were characterized by the presence of hydroxyl or methoxyl groups in position 3 and 7, and their efficiency was comparable to leupeptin (IC50 = 1.2 µM), used as a reference inhibitor of serine proteases [90]. However, studies on the complement cascade, the activation of which is partly associated with plasmin activity, revealed no inhibition for epicatechin, myricitrin, myricetin, or quercetin, with only weak inhibitory effects of afzelin, quercitrin, kaempferol, and tiliroside (IC50 values of 258, 440, 730, and 101 µM, respectively). Anti-complement efficiency of the tested compounds increased in inverse proportion to a number of free hydroxyl groups in the B-ring of a flavonoid structure [91].

4.4. Plasmin-Targeting Low-Molecular Plant Metabolites and Their Derivatives

Among natural, non-modified phytochemicals, quercetin is the best-described compound with the serine protease-inhibitory properties. Interactions between this flavonoid and plasmin active center have been visualized in silico by Cuccioloni et al. [92]. Quercetin has been reported to inhibit plasmin, urokinase, thrombin, trypsin, and elastase. Part of the available data indicated competitive characteristics of plasmin inhibition by quercetin and its glycoside derivative, rutin. Molecular docking revealed interactions between the catalytic triad of plasmin (Asp646, His603, Ser741) and quercetin and rutoside (rutin). Other amino acid residues participating in these interactions were Glu606, Arg644, Trp731 for the quercetin- and the Arg644 residue for the rutin-induced inhibition, respectively. Based on the equilibrium dissociation constants, over 10-fold higher affinity of quercetin to plasmin when compared to rutin has been established. The dissociation constants (Kd) for plasmin-quercetin and plasmin-rutin complexes were 0.715 ± 0.096 and 12.98 ± 0.76 μM, respectively [86]. However, a competitive action of quercetin seems to be one of the possible mechanisms of interaction of this compound with plasmin, and the final effect may be related to the experimental settings, including the used substrate. Comparative studies on inhibitory effects of the plant-derived bufadienolides and quercetin revealed the uncompetitive characteristics of bufadienolide-triggered plasmin inhibition, while the quercetin acted as a non-competitive inhibitor of this enzyme. In silico analyses revealed that quercetin was bound in proximity but not exactly in the active site, independently of the substrate presence or absence in the active site [43]. The aforementioned differences in mechanisms of plasmin inhibition displayed by quercetin, revealed by various research groups, may indicate a mixed type of plasmin inhibition by quercetin as well. Interestingly, different mechanisms of the inhibitory actions were also observed for TXA, a lysine analogue, which typically blocks the LBS. However, under certain experimental conditions, i.e., its high concentrations (∼25 mM), TXA was able to directly inhibit plasmin through binding to the primary S1 pocket of this protease [93]. Diverse effects on plasmin were also found in studies on the bufadienolide rich-fraction isolated from Kalanchoe daigremontiana Raym.-Hamet & H. Perrier. This fraction was capable of stimulating or inhibiting plasmin activity, dependently on the applied concentration. While at lower concentrations (i.e., 0.05–2.5 μg/mL) a significant profibrinolytic effect was found, at a higher concentration (i.e., 50 μg/mL), the bufadienolide fraction inhibited the activity of plasmin [43].
The hemostatic-modulatory properties, including inhibition of platelets, changes in erythrocyte membrane fluidity, as well as stimulation or inhibition of coagulation cascade and fibrinolytic proteins, have been also demonstrated for tannins [94]. Browplasminin, a condensed tannin based on heteroflavan-3-ols of catechin with B-type linkages, isolated from flowers of Brownea grandiceps Jacq., was identified as an inhibitor of plasmin (IC50 = 47.80 μg/mL, Ki = 0.76 μg/mL). This tannin did not influence the activity of t-PA or u-PA but slightly inhibited FXa (IC50 = 237.08 μg/mL, Ki = 61.61 μg/mL) [95]. Animal studies demonstrated diverse effects of tannins on the physiology of the fibrinolytic system. Oral administration of the extract from Potentilla erecta (L.) Raeusch. rhizome increased the activity of t-PA (200 mg/kg, for 14 days) in streptozotocin-induced diabetic rats. However, the euglobulin clot lysis time was prolonged [96]. In normoglycemic rats, administration of the extract (400 mg/kg, for 14 days) decreased the activity of t-PA with no effects on PAI-1 [97]. Corilagin, an ellagitannin isolated from Phyllanthus urinaria L. aerial parts increased activity of t-PA and reduced the activity of PAI-1 in blood plasma of healthy rats [98].

4.5. Natural and Modified Plant Based Inhibitors of Other Components of the Fibrinolytic System

An excessive inhibition of plasmin, a downstream factor involved in multiple critical biological processes, may result in serious physiological consequences, similar to those observed in plasminogen deficiency. Plasminogen-deficient animals have been shown to suffer from growth disorders, impaired fertility, and low survival [99]. For that reason, the upstream factors such as uPAR, u-PA, t-PA, and PAI-1 have been suggested as more specific and safer targets in the context of clinical treatment [100]. Accordingly, both natural (tannic acid (TA), epigallocatechin monogallate (EGCG), epigallocatechin-3,5-digallate (EGCDG), hexachlorophene, quinalizarin, gallic acid, and sennoside A) and synthetic polyphenolic inhibitors have been investigated. Sennoside A, representing dianthrone compounds, was the weakest inhibitor of PAI-1 activity (IC50 = 51.0 µM). TA, EGCG, EGCDG, and gallic acid backbones contain galloyl or gallo-galloyl moieties. However, their inhibitory efficiency towards PAI-1 was only partly related to the number of these galloyl units, suggesting that the orientation of units and other factors are also important. While a monomeric compound, i.e., gallic acid (IC50 = 6.6 µM), was 1000-fold less active compared to oligomeric TA (IC50 = 0.007 µM), EGCG (IC50 = 0.091 µM) was only about 10-fold weaker than TA [101].
The u-PA-inhibitory effects were found for crude methanol extracts from six medicinal plant species, i.e., Croton lucidus Gage (the most effective one, IC50 = 13.52 µg/mL), Erythroxylum aerolatum L., Tabebuia heterophylla Britton, Lantana camara L., Cananga odorata (Lam.) Hook.f. & Thomson, and Amyris elemifera Kuntze. The chloroform fraction from C. lucidus leaves was the most effective u-PA inhibitor (IC50 = 3.52 µg/mL) [102]. Quercetin was reported to inhibit the activity of u-PA with an IC50 of 12.1 [50] or 20 μM [49]. This plasminogen activator was also inhibited by esculin, rutin [46], silybin, hypericin, and hyperoside [50]. Furthermore, available studies indicate a possibility of urokinase and its receptor control by plant substances at the level of protein expression, especially in the context of anti-cancer therapy [103,104] and modulation of other processes associated with cell migration and ECM degradation [105].
It has been also suggested that selective modifications of polyphenols, such as lipophilization with fatty acids, may represent a new approach in development of inhibitors of serine proteases, including the urokinase. Screening of the protease-inhibitory efficiency of rutin, phloridzin, and esculin esters revealed that the most potent serine protease inhibitors were rutin derivatives, containing medium to long as well as mono- and polyunsaturated fatty acids. However, in the case of inhibition of urokinase, none of the examined esters was more efficient than rutin. The established IC50 values for rutin and its most efficient derivatives, i.e., rutin butyrate and rutin arachidonate, were as follows: 6, 8, and 9 μM, respectively [49].

5. Pro-Fibrinolytic Effects of Natural, Plant-Derived Substances

Numerous reports devoted to pro-fibrinolytic or thrombolytic activity of different plant extracts or isolated compounds have been published so far [106,107,108,109,110,111,112,113,114,115,116,117,118,119,120,121]. Using plants as sources of fibrinolytic substances seems to be an attractive prospect for future research, but as yet, most of the available data derive from basic in vitro screening tests employing whole blood or plasma fibrin clots (Table 5). Unfortunately, very few in vivo studies are devoted to pro-fibrinolytic action of plant extracts. However, in mice, administration of 200 mg/mL and 400 mg/kg of body weight doses of ethanolic extract of Ocimum basilicum L. shoots has been found to stimulate the fibrinolytic system, resulting in a higher content of fibrin degradation products (FDPs) [122]. In humans, supplementation with a dried garlic powder (900 mg/day; for 14 days; placebo-controlled study) increased the t-PA activity but did not influence PAI-1 or fibrinogen levels [123]. Favorable changes in plasma clotting, clot formation, and lysis were found in middle-aged patients with metabolic syndrome taking 100 mg of Aronia melanocarpa (Michx.) Elliott extract (three times a day, for 2 months) [124]. It is assumed that in simple experimental systems, the observed effects may be a result of the presence of plant proteases or modulation of the fibrinolytic enzymes by members of various classes of phytochemicals that are present in plant material. At the level of the whole organism, molecular mechanisms of pro-fibrinolytic or anti-thrombotic actions are much more complicated. The plant-derived substances are able to affect diverse components of the hemostatic system [125,126], and their final effects may be a result of stimulation of the fibrinolytic system, modulation of the coagulation cascade and platelet activity, or other activities.
However, in some cases, substances that the most likely display the fibrinolytic activity have been at least preliminarily identified. For instance, an edible and medicinal plant Aster yomena (Kitam.) Honda has been found to synthesize a protease (named kitamase) with thrombolytic (plasmin-like) and anticoagulant properties. Kitamase preferentially digested the Aα- and γ-γ chains of fibrin(ogen). It also acted as an anticoagulant, i.e., prolonged the coagulation time, the activated partial thromboplastin time, and prothrombin time [127]. Experiments on thrombolytic potential of Gastrodia elata Blume demonstrated that ethyl acetate and water extracts enhanced plasmin activity. More in-depth analyses revealed three the most active compounds from these extracts, i.e., 4-hydroxybenzyl alcohol, 4-hydroxybenzaldehyde, and 4-ethoxymethylphenol, that significantly stimulated plasmin activity [128]. In studies on Lagenaria siceraria (Molina) Standley fruit extract, kaempferol was found to be a key ingredient of this plant preparation, responsible for stimulating fibrinolytic activity in blood clots [129]. However, in other studies, kaempferol was found to inhibit serine proteases and impair the activity of complement proteins [91].
Thrombolytic activity was also demonstrated for plant latex proteases. Interestingly, most of the identified plant latex proteases display both thrombin-like (pro-coagulant) and plasmin-like activity. The thrombin-like activity of plant proteases has been found in in vitro and in vivo investigations. Plant latex proteases can hydrolyze (but with different efficiency) all fibrinogen subunits (i.e., Aα, Bβ, and γ chains) leading to the clot degradation; however, they can also stimulate fibrin formation by partial degradation of fibrinogen, involving removal of fibrinopeptides FpA and FpB from N-terminal regions of the fibrinogen Aα and Bβ chains, respectively [130,131]. During the blood coagulation cascade, hydrolytic release of these peptides from fibrinogen by thrombin is critical for starting its polymerization and formation of the fibrin network. Comparative analyses of hydrolytic activities of Calotropis gigantea R. Br., Synadenium grantii Hook. f., and Wrightia tinctoria R. Br. latex extracts demonstrated that Aα chains of fibrin(ogen) are the most susceptible to degradation by the examined latex proteases, followed by Bβ and γ chains. The clot dissolving properties were as follows: C. gigantea > S. grantii > W. tinctoria. The same pattern of efficiency of the examined extracts was observed during the fibrin clot formation tests, after recalcification of blood plasma [132]. Pergularain e I, a cysteine protease (23.3 kDa, N-terminal sequence: LPHDVE) from Pergularia extensa (Jacq.) N.E.Br. latex displayed both thrombin-like and thrombolytic properties [133]. Hirtin, a 34 kDa serine protease with the N-terminal sequence YAVYIGLILETAA/NNE was identified in the latex of a medicinal herb Euphorbia hirta L. The enzyme had a broad range of hydrolytic activity, including azocaseinolytic, gelatinolytic, fibrinogenolytic, and fibrinolytic properties (with the following preference: Aα > Bβ > γ units) [134]. Another serine protease (AMP48) displaying plasmin-like activity was isolated from Artocarpus heterophyllus Lam. latex. A 48-kDa enzyme, characterized by the N-terminal aa sequence AQEGGKDDDGG, was able to effectively hydrolyze Aα chains and partly degrade Bβ and γ subunits of human fibrinogen [135].

6. Concluding Remarks and Perspectives

Diverse classes of plant-derived substances have been evidenced to inhibit fibrinolytic proteins or to possess fibrinolytic activity. Both (poly)phenols and other low molecular secondary metabolites may be considered as promising templates for fibrinolysis inhibitors or modulators; however, given the ample evidence, the peptide-type substances displayed higher inhibitory efficiency. The Laskowski folds may be used for designing new structures or combinations with other substances to obtain plasmin inhibitors that could be useful in different diseases. However, low specificity of most of naturally occurring substances limits the possibility of undertaking studies in animals and is still a significant obstacle to developing effective fibrinolysis-targeting drugs.
SFTI-1 may be a template for different modifications and a base for development of new formulae of delivery of this potential drug. Both the STFI-1 structural scaffold and other scaffolds that are present in different plant-derived peptide-type inhibitors may be modified to obtain structures (e.g., by optimization of the sequence in the canonical loop), providing both increased inhibitory efficiency and selectivity. Some attempts to modify the Kunitz-type inhibitors have been also undertaken. The genetic engineering techniques enable modification of natural substances and their production systems. For example, a recombinant kallikrein inhibitor (rBbKI) and a recombinant cruzipain inhibitor from B. bauhinioides (rBbCI) were obtained by heterologous expression and production in the form of functional proteins in Escherichia coli [136]. Native BbCI and BbKI share over 80% sequence similarity; however, some important divergences have been found in their reactive sites. This was the context for the development of recombinant BbKI with a single mutation replacing the Arg64 residue with Ala. The R64A mutation led to significant modification of BbKI specificity to a weaker inhibitor of plasma kallikrein than the wild-type rBbKI (Ki changed from 2 to 98 nM) and a strong inhibitory action towards plasmin (Ki changed from 33 to 2.6 nM) [137].
Furthermore, the latest interesting trends in research on fibrinolysis modulators include other ways of exploiting the potential of plant-derived substances, e.g., the use of modified nanoparticles (NPs) as drug carriers. Recently, Yu and co-authors [138] described the polyphenol-based nanocarriers of thrombolytic drugs. The nanoparticle concept was used to improve drug delivery system using TA, which is capable of interacting with diverse surfaces and target molecules through a wide spectrum of mechanisms, including hydrophobic, hydrogen bond, covalent bond, and electrostatic interactions; metal coordination; and π interactions. TA was used to cross-link u-PA-loaded sacrificial mesoporous silica template and a thrombin-cleavable peptide to obtain the bioresponsive NP carrier of u-PA. Preliminary in vitro assessments of the nanocarrier action demonstrated an increase in Plg activation in the presence of thrombin (1.14-fold) and lower association with macrophages and monocytes. Further, an animal study revealed delayed blood clearance of the modified nanocarriers (90% clearance at 60 min) compared to the mesoporous silica template NPs (90% clearance at 5 min).
Therefore, the latest approach based on the use of natural pharmacophore templates and on investigation of their modifications could offer a promising way to overcome the difficulties related to low specificity of natural compounds.

Author Contributions

Conceptualization: J.K.-C.; writing, review and editing: J.K.-C. and J.C. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Kwaan, H.C. The Role of Fibrinolytic System in Health and Disease. Int. J. Mol. Sci. 2022, 23, 5262. [Google Scholar] [CrossRef] [PubMed]
  2. Miles, L.; Ny, L.; Wilczynska, M.; Shen, Y.; Ny, T.; Parmer, R. Plasminogen Receptors and Fibrinolysis. Int. J. Mol. Sci. 2021, 22, 1712. [Google Scholar] [CrossRef] [PubMed]
  3. Keragala, C.B.; Medcalf, R.L. Plasminogen: An enigmatic zymogen. Blood 2021, 137, 2881–2889. [Google Scholar] [CrossRef] [PubMed]
  4. Wu, G.; Quek, A.J.; Caradoc-Davies, T.T.; Ekkel, S.M.; Mazzitelli, B.; Whisstock, J.C.; Law, R.H. Structural studies of plasmin inhibition. Biochem. Soc. Trans. 2019, 47, 541–557. [Google Scholar] [CrossRef]
  5. Hogg, K.; Carrier, M. Prevention and treatment of venous thromboembolism in patients with cancer. Ther. Adv. Hematol. 2011, 3, 45–58. [Google Scholar] [CrossRef]
  6. Kapil, N.; Datta, Y.H.; Alakbarova, N.; Bershad, E.; Selim, M.; Liebeskind, D.S.; Bachour, O.; Rao, G.H.R.; Divani, A.A. Antiplatelet and Anticoagulant Therapies for Prevention of Ischemic Stroke. Clin. Appl. Thromb. 2016, 23, 301–318. [Google Scholar] [CrossRef]
  7. Mackman, N.; Spronk, H.M.; Stouffer, G.A.; Cate, H.T. Dual Anticoagulant and Antiplatelet Therapy for Coronary Artery Disease and Peripheral Artery Disease Patients. Arter. Thromb. Vasc. Biol. 2018, 38, 726–732. [Google Scholar] [CrossRef]
  8. Larson, E.A.; German, D.M.; Shatzel, J.; Deloughery, T.G. Anticoagulation in the cardiac patient: A concise review. Eur. J. Haematol. 2018, 102, 3–19. [Google Scholar] [CrossRef]
  9. Lamponi, S. Bioactive Natural Compounds with Antiplatelet and Anticoagulant Activity and Their Potential Role in the Treatment of Thrombotic Disorders. Life 2021, 11, 1095. [Google Scholar] [CrossRef]
  10. Ali, R.; Hossain, M.S.; Islam, A.; Arman, S.I.; Raju, G.S.; Dasgupta, P.; Noshin, T.F. Aspect of Thrombolytic Therapy: A Review. Sci. World J. 2014, 2014, 1–8. [Google Scholar] [CrossRef]
  11. Shapiro, A.D.; Menegatti, M.; Palla, R.; Boscarino, M.; Roberson, C.; Lanzi, P.; Bowen, J.; Nakar, C.; Janson, I.A.; Peyvandi, F. An international registry of patients with plasminogen deficiency (HISTORY). Haematologica 2020, 105, 554–561. [Google Scholar] [CrossRef]
  12. Okafor, O.N.; Gorog, D.A. Endogenous fibrinolysis: An important mediator of thrombus formation and cardiovascular risk. J. Am. Coll. Cardiol. 2015, 65, 1683–1699. [Google Scholar] [CrossRef] [PubMed]
  13. Kanji, R.; Kubica, J.; Navarese, E.P.; Gorog, D.A. Endogenous fibrinolysis—Relevance to clinical thrombosis risk assessment. Eur. J. Clin. Investig. 2020, 51, e13471. [Google Scholar] [CrossRef] [PubMed]
  14. Saes, J.; Schols, S.; van Heerde, W.; Nijziel, M. Hemorrhagic disorders of fibrinolysis: A clinical review. J. Thromb. Haemost. 2018, 16, 1498–1509. [Google Scholar] [CrossRef] [PubMed]
  15. Marinho, D.S. Perioperative hyperfibrinolysis—Physiology and pathophysiology. Braz. J. Anesthesiol. 2020, 71, 65–75. [Google Scholar] [CrossRef]
  16. Lavin, M.; O’Donnell, J.S. New treatment approaches to von Willebrand disease. Hematology 2016, 2016, 683–689. [Google Scholar] [CrossRef]
  17. O’Carroll, C.B.; Aguilar, M.I. Management of Postthrombolysis Hemorrhagic and Orolingual Angioedema Complications. Neurohospitalist 2015, 5, 133–141. [Google Scholar] [CrossRef]
  18. Simurda, T.; Asselta, R.; Zolkova, J.; Brunclikova, M.; Dobrotova, M.; Kolkova, Z.; Loderer, D.; Skornova, I.; Hudecek, J.; Lasabova, Z.; et al. Congenital Afibrinogenemia and Hypofibrinogenemia: Laboratory and Genetic Testing in Rare Bleeding Disorders with Life-Threatening Clinical Manifestations and Challenging Management. Diagnostics 2021, 11, 2140. [Google Scholar] [CrossRef]
  19. Simurda, T.; Vilar, R.; Zolkova, J.; Ceznerova, E.; Kolkova, Z.; Loderer, D.; Neerman-Arbez, M.; Casini, A.; Brunclikova, M.; Skornova, I.; et al. A Novel Nonsense Mutation in FGB (c.1421G>A; p.Trp474Ter) in the Beta Chain of Fibrinogen Causing Hypofibrinogenemia with Bleeding Phenotype. Biomedicines 2020, 8, 605. [Google Scholar] [CrossRef]
  20. Brunclikova, M.; Simurda, T.; Zolkova, J.; Sterankova, M.; Skornova, I.; Dobrotova, M.; Kolkova, Z.; Loderer, D.; Grendar, M.; Hudecek, J.; et al. Heterogeneity of Genotype–Phenotype in Congenital Hypofibrinogenemia—A Review of Case Reports Associated with Bleeding and Thrombosis. J. Clin. Med. 2022, 11, 1083. [Google Scholar] [CrossRef]
  21. Collen, D. Natural inhibitors of fibrinolysis. J. Clin. Pathol. Suppl. R. Coll. Pathol. 1980, 14, 24–30. [Google Scholar] [CrossRef] [PubMed]
  22. Law, R.H.; Caradoc-Davies, T.; Cowieson, N.; Horvath, A.J.; Quek, A.J.; Encarnacao, J.A.; Steer, D.; Cowan, A.; Zhang, Q.; Lu, B.G.; et al. The X-ray Crystal Structure of Full-length Human Plasminogen. Cell Rep. 2012, 1, 185–190. [Google Scholar] [CrossRef] [PubMed]
  23. Schaller, J.; Gerber, S.S. The plasmin–antiplasmin system: Structural and functional aspects. Cell. Mol. Life Sci. 2010, 68, 785–801. [Google Scholar] [CrossRef]
  24. Medcalf, R.L.; Keragala, C.B. The Fibrinolytic System: Mysteries and Opportunities. Hemasphere 2021, 5, e570. [Google Scholar] [CrossRef] [PubMed]
  25. Steinmetzer, T.; Pilgram, O.; Wenzel, B.M.; Wiedemeyer, S.J.A. Fibrinolysis Inhibitors: Potential Drugs for the Treatment and Prevention of Bleeding. J. Med. Chem. 2019, 63, 1445–1472. [Google Scholar] [CrossRef] [PubMed]
  26. Battistel, M.D.; Grishaev, A.; An, S.S.A.; Castellino, F.J.; Llinas, M. Solution structure and functional characterization of human plasminogen kringle 5. Biochemistry 2009, 48, 10208–10219. [Google Scholar] [CrossRef] [PubMed]
  27. Law, R.H.; Abu-Ssaydeh, D.; Whisstock, J.C. New insights into the structure and function of the plasminogen/plasmin system. Curr. Opin. Struct. Biol. 2013, 23, 836–841. [Google Scholar] [CrossRef]
  28. Longstaff, C.; Kolev, K. Basic mechanisms and regulation of fibrinolysis. J. Thromb. Haemost. 2015, 13, S98–S105. [Google Scholar] [CrossRef]
  29. Rehman, A.A.; Ahsan, H.; Khan, F.H. α-2-Macroglobulin: A physiological guardian. J. Cell Physiol. 2013, 228, 1665–1675. [Google Scholar] [CrossRef]
  30. Sillen, M.; Declerck, P. Thrombin Activatable Fibrinolysis Inhibitor (TAFI): An Updated Narrative Review. Int. J. Mol. Sci. 2021, 22, 3670. [Google Scholar] [CrossRef]
  31. European Medicine Agency. Antifibrinolytics Containing Aprotinin, Aminocaproic Acid and Tranexamic Acid. Aminocaproic acid. EMA/680964/Assessment Report. Available online: https://www.ema.europa.eu/en/documents/referral/assessment-report-antifibrinolytic-medicines-aminocaproic-acid_en.pdf (accessed on 10 October 2012).
  32. Ockerman, A.; Vanassche, T.; Garip, M.; Vandenbriele, C.; Engelen, M.M.; Martens, J.; Politis, C.; Jacobs, R.; Verhamme, P. Tranexamic acid for the prevention and treatment of bleeding in surgery, trauma and bleeding disorders: A narrative review. Thromb. J. 2021, 19, 54. [Google Scholar] [CrossRef] [PubMed]
  33. Mangano, D.T.; Tudor, I.C.; Dietzel, C. The Risk Associated with Aprotinin in Cardiac Surgery. N. Engl. J. Med. 2006, 354, 353–365. [Google Scholar] [CrossRef] [PubMed]
  34. Karkouti, K.; Beattie, W.S.; Dattilo, K.M.; McCluskey, S.A.; Ghannam, M.; Hamdy, A.; Wijeysundera, D.N.; Fedorko, L.; Yau, T.M. A propensity score case-control comparison of aprotinin and tranexamic acid in high-transfusion-risk cardiac surgery. Transfusion 2006, 46, 327–338. [Google Scholar] [CrossRef] [PubMed]
  35. Ucar, E.Y. Update on Thrombolytic Therapy in Acute Pulmonary Thromboembolism. Eurasian J. Med. 2019, 51, 185–189. [Google Scholar] [CrossRef]
  36. Abraham, P.; Arroyo, D.A.; Giraud, R.; Bounameaux, H.; Bendjelid, K. Understanding haemorrhagic risk following thrombolytic therapy in patients with intermediate-risk and high-risk pulmonary embolism: A hypothesis paper. Open Heart 2018, 5, e000735. [Google Scholar] [CrossRef]
  37. Do Amaral Baruzzi, A.C.; Stefanini, E.; Manzo, G. Fibrinolytics: Indications and treatment of hemorrhagic complications. Rev. Soc. Cardiol. Estado São Paulo 2018, 28, 421–427. [Google Scholar] [CrossRef]
  38. Ozier, Y.; Bellamy, L. Pharmacological agents: Antifibrinolytics and desmopressin. Best Pract. Res. Clin. Anaesthesiol. 2010, 24, 107–119. [Google Scholar] [CrossRef]
  39. Levy, J.H.; Koster, A.; Quinones, Q.J.; Milling, T.J.; Key, N.S. Antifibrinolytic Therapy and Perioperative Considerations. Anesthesiology 2018, 128, 657–670. [Google Scholar] [CrossRef]
  40. Colomina, M.J.; Contreras, L.; Guilabert, P.; Koo, M.; Ndez, E.M.; Sabate, A. Clinical use of tranexamic acid: Evidences and controversies. Braz. J. Anesthesiol. 2021, 72, 795–812. [Google Scholar] [CrossRef]
  41. Marti, D.N.; Hu, C.-K.; An, S.S.A.; von Haller, P.; Schaller, J.; Llinás, M. Ligand Preferences of Kringle 2 and Homologous Domains of Human Plasminogen: Canvassing Weak, Intermediate, and High-Affinity Binding Sites by 1H-NMR. Biochemistry 1997, 36, 11591–11604. [Google Scholar] [CrossRef]
  42. The European Society of Anaesthesiology task force reports on the place of aprotinin in clinical anaesthesia. Aprotinin: Is it time to reconsider? Eur. J. Anaesthesiol. 2015, 32, 591–595. [Google Scholar] [CrossRef] [PubMed]
  43. Kolodziejczyk-Czepas, J.; Pasiński, B.; Ponczek, M.B.; Moniuszko-Szajwaj, B.; Kowalczyk, M.; Pecio, Ł.; Nowak, P.; Stochmal, A. Bufadienolides from Kalanchoe daigremontiana modulate the enzymatic activity of plasmin—In vitro and in silico analyses. Int. J. Biol. Macromol. 2018, 120, 1591–1600. [Google Scholar] [CrossRef] [PubMed]
  44. Kolodziejczyk-Czepas, J.; Sieradzka, M.; Moniuszko-Szajwaj, B.; Pecio, Ł.; Ponczek, M.B.; Nowak, P.; Stochmal, A. Bufadienolides from Kalanchoe daigremontiana as thrombin inhibitors—In vitro and in silico study. Int. J. Biol. Macromol. 2017, 99, 141–150. [Google Scholar] [CrossRef] [PubMed]
  45. Bijak, M.; Ziewiecki, R.; Saluk, J.; Ponczek, M.; Pawlaczyk, I.; Krotkiewski, H.; Wachowicz, B.; Nowak, P. Thrombin inhibitory activity of some polyphenolic compounds. Med. Chem. Res. 2013, 23, 2324–2337. [Google Scholar] [CrossRef] [PubMed]
  46. Bijak, M.; Ponczek, M.B.; Nowak, P. Polyphenol compounds belonging to flavonoids inhibit activity of coagulation factor X. Int. J. Biol. Macromol. 2014, 65, 129–135. [Google Scholar] [CrossRef]
  47. Sartor, L.; Pezzato, E.; Dell’Aica, I.; Caniato, R.; Biggin, S.; Garbisa, S. Inhibition of matrix-proteases by polyphenols: Chemical insights for anti-inflammatory and anti-invasion drug design. Biochem. Pharmacol. 2002, 64, 229–237. [Google Scholar] [CrossRef]
  48. Mozzicafreddo, M.; Cuccioloni, M.; Eleuteri, A.M.; Fioretti, E.; Angeletti, M. Flavonoids inhibit the amidolytic activity of human thrombin. Biochimie 2006, 88, 1297–1306. [Google Scholar] [CrossRef]
  49. Viskupicova, J.; Danihelova, M.; Majekova, M.; Liptaj, T.; Sturdik, E. Polyphenol fatty acid esters as serine protease inhibitors: A quantum-chemical QSAR analysis. J. Enzym. Inhib. Med. Chem. 2011, 27, 800–809. [Google Scholar] [CrossRef]
  50. Jedinák, A.; Maliar, T.; Grancai, D.; Nagy, M. Inhibition activities of natural products on serine proteases. Phytother. Res. 2006, 20, 214–217. [Google Scholar] [CrossRef]
  51. Melzig, M.F.; Löser, B.; Ciesielski, S. Inhibition of neutrophil elastase activity by phenolic compounds from plants. Die Pharm. 2001, 56, 967–970. [Google Scholar]
  52. Liu, L.; Ma, H.; Yang, N.; Tang, Y.; Guo, J.; Tao, W.; Duan, J. A Series of Natural Flavonoids as Thrombin Inhibitors: Structure-activity relationships. Thromb. Res. 2010, 126, e365–e378. [Google Scholar] [CrossRef] [PubMed]
  53. Mozzicafreddo, M.; Cuccioloni, M.; Bonfili, L.; Eleuteri, A.M.; Fioretti, E.; Angeletti, M. Antiplasmin activity of natural occurring polyphenols. Biochim. Biophys. Acta BBA Proteins Proteom. 2008, 1784, 995–1001. [Google Scholar] [CrossRef] [PubMed]
  54. Cuccioloni, M.; Bonfili, L.; Mozzicafreddo, M.; Cecarini, V.; Eleuteri, A.M.; Angeletti, M. Sanguisorba minor extract suppresses plasmin-mediated mechanisms of cancer cell migration. Biochim. et Biophys. Acta BBA Gen. Subj. 2012, 1820, 1027–1034. [Google Scholar] [CrossRef] [PubMed]
  55. Weir, M.P.; Bethell, S.S.; Cleasby, A.; Campbell, C.J.; Dennis, R.J.; Dix, C.J.; Finch, H.; Jhoti, H.; Mooney, C.J.; Patel, S.; et al. Novel natural product 5,5-trans-lactone inhibitors of human alpha-thrombin: Mechanism of action and structural studies. Biochemistry 1998, 37, 6645–6657. [Google Scholar] [CrossRef]
  56. Siritapetawee, J.; Sojikul, P.; Soontaranon, S.; Limphirat, W.; Thammasirirak, S. A Protein from Aloe vera that Inhibits the Cleavage of Human Fibrin(ogen) by Plasmin. Appl. Biochem. Biotechnol. 2013, 170, 2034–2045. [Google Scholar] [CrossRef]
  57. Tanaka, A.S.; Sampaio, M.U.; Marangoni, S.; de Oliveira, B.; Novelle, J.C.; Oliva, M.L.V.; Fink, E.; Sampaio, C.A.M. Purification and Primary Structure Determination of a Bowman-Birk Trypsin Inhibitor from Torresea cearensis Seeds. Biol. Chem. 1997, 378, 273–282. [Google Scholar] [CrossRef]
  58. Lopes, J.; Valadares, N.; Moraes, D.; Rosa, J.; Araújo, H.; Beltramini, L. Physico-chemical and antifungal properties of protease inhibitors from Acacia plumosa. Phytochemistry 2009, 70, 871–879. [Google Scholar] [CrossRef]
  59. Alves, F.L.; Sallai, R.C.; Salu, B.R.; Miranda, A.; Oliva, M.L.V. Identification and characterisation of serine protease inhibitors from Araucaria angustifolia seeds. Nat. Prod. Res. 2016, 30, 2712–2715. [Google Scholar] [CrossRef]
  60. Oliva, M.; Mendes, C.; Santomauro-Vaz, E.; Juliano, M.; Mentele, R.; Auerswald, E.; Sampaio, M.; Sampaio, C. Bauhinia bauhinioides Plasma Kallikrein Inhibitor Interaction with Synthetic Peptides and Fluorogenic Peptide Substrates Related to the Reactive Site Sequence. Curr. Med. Chem. 2001, 8, 977–984. [Google Scholar] [CrossRef]
  61. Oliva, M.L.; Sampaio, U.M. Bauhinia Kunitz-type proteinase inhibitors: Structural characteristics and biological properties. Biol. Chem. 2008, 389, 1007–1013. [Google Scholar] [CrossRef]
  62. Cruz-Silva, I.; Gozzo, A.J.; Nunes, V.A.; Carmona, A.K.; Faljoni-Alario, A.; Oliva, M.L.V.; Sampaio, M.U.; Sampaio, C.A.M.; Araujo, M.S. A proteinase inhibitor from Caesalpinia echinata (pau-brasil) seeds for plasma kallikrein, plasmin and factor XIIa. Biol. Chem. 2004, 385, 1083–1086. [Google Scholar] [CrossRef] [PubMed]
  63. Chong, G.L.; Reeck, G.R. Interaction of trypsin, β-factor XIIa, and plasma kallikrein with a trypsin inhibitor isolated from barley seeds: A comparison with the corn inhibitor of activated hageman factor. Thromb. Res. 1987, 48, 211–221. [Google Scholar] [CrossRef] [PubMed]
  64. Zhou, D.; Lobo, Y.A.; Batista, I.F.C.; Marques-Porto, R.; Gustchina, A.; Oliva, M.L.V.; Wlodawer, A. Crystal Structures of a Plant Trypsin Inhibitor from Enterolobium contortisiliquum (EcTI) and of Its Complex with Bovine Trypsin. PLoS ONE 2013, 8, e62252. [Google Scholar] [CrossRef] [PubMed]
  65. Do Socorro, M.; Cavalcanti, M.; Oliva, M.L.; Fritz, H.; Jochum, M.; Mentele, R.; Sampaio, M.; Coelho, L.C.; Batista, I.F.; Sampaio, C.A. Characterization of a Kunitz trypsin inhibitor with one disulfide bridge purified from Swartzia pickellii. Biochem. Biophys. Res. Commun. 2002, 291, 635–639. [Google Scholar] [CrossRef] [PubMed]
  66. Patil, D.N.; Chaudhary, A.; Sharma, A.K.; Tomar, S.; Kumar, P. Structural basis for dual inhibitory role of tamarind Kunitz inhibitor (TKI) against factor Xa and trypsin. FEBS J. 2012, 279, 4547–4564. [Google Scholar] [CrossRef]
  67. Krishnamoorthi, R.; Gong, Y.; Richardson, M. A new protein inhibitor of trypsin and activated hageman factor from pumpkin (Cucurbita maxima) seeds. FEBS Lett. 1990, 273, 163–167. [Google Scholar] [CrossRef]
  68. Hayashi, K.; Takehisa, T.; Hamato, N.; Takano, R.; Hara, S.; Miyata, T.; Kato, H. Inhibition of Serine Proteases of the Blood Coagulation System by Squash Family Protease Inhibitors. J. Biochem. 1994, 116, 1013–1018. [Google Scholar] [CrossRef]
  69. Laskar, A.; Rodger, E.J.; Chatterjee, A.; Mandal, C. Modeling and structural analysis of PA clan serine proteases. BMC Res. Notes 2012, 5, 256. [Google Scholar] [CrossRef]
  70. Millers, E.-K.I.; Johnson, L.A.; Birrell, G.W.; Masci, P.P.; Lavin, M.; De Jersey, J.; Guddat, L.W. The structure of Human Microplasmin in Complex with Textilinin-1, an Aprotinin-like Inhibitor from the Australian Brown Snake. PLoS ONE 2013, 8, e54104. [Google Scholar] [CrossRef]
  71. Al-Horani, R.A.; Karuturi, R.; White, D.T.; Desai, U.R. Plasmin Regulation through Allosteric, Sulfated, Small Molecules. Molecules 2015, 20, 608–624. [Google Scholar] [CrossRef]
  72. Swedberg, J.E.; De Veer, S.J.; Harris, J.M. Natural and engineered kallikrein inhibitors: An emerging pharmacopoeia. Biol. Chem. 2010, 391, 357–374. [Google Scholar] [CrossRef] [PubMed]
  73. Hellinger, R.; Gruber, C.W. Peptide-based protease inhibitors from plants. Drug Discov. Today 2019, 24, 1877–1889. [Google Scholar] [CrossRef] [PubMed]
  74. Oliva, M.L.V.; Ferreira, R.D.S.; Ferreira, J.G.; De Paula, C.A.A.; Salas, C.E.; Sampaio, M.U. Structural and functional properties of kunitz proteinase inhibitors from leguminosae: A mini review. Curr. Protein Pept. Sci. 2011, 12, 348–357. [Google Scholar] [CrossRef] [PubMed]
  75. Bendre, A.D.; Ramasamy, S.; Suresh, C. Analysis of Kunitz inhibitors from plants for comprehensive structural and functional insights. Int. J. Biol. Macromol. 2018, 113, 933–943. [Google Scholar] [CrossRef] [PubMed]
  76. Oliva, M.L.V.; Silva, M.C.; Sallai, R.C.; Brito, M.V.; Sampaio, M.U. A novel subclassification for Kunitz proteinase inhibitors from leguminous seeds. Biochimie 2010, 92, 1667–1673. [Google Scholar] [CrossRef] [PubMed]
  77. Nakahata, A.M.; Bueno, N.R.; Rocha, H.A.; Franco, C.R.; Chammas, R.; Nakaie, C.R.; Jasiulionis, M.; Nader, H.B.; Santana, L.A.; Sampaio, M.U.; et al. Structural and inhibitory properties of a plant proteinase inhibitor containing the RGD motif. Int. J. Biol. Macromol. 2006, 40, 22–29. [Google Scholar] [CrossRef]
  78. Ruoslahti, E. Rgd and other recognition sequences for integrins. Annu. Rev. Cell Dev. Biol. 1996, 12, 697–715. [Google Scholar] [CrossRef]
  79. Bachmann, M.; Kukkurainen, S.; Hytönen, V.P.; Wehrle-Haller, B. Cell Adhesion by Integrins. Physiol. Rev. 2019, 99, 1655–1699. [Google Scholar] [CrossRef]
  80. Zhang, N.; Li, C.; Zhou, D.; Ding, C.; Jin, Y.; Tian, Q.; Meng, X.; Pu, K.; Zhu, Y. Cyclic RGD functionalized liposomes encapsulating urokinase for thrombolysis. Acta Biomater. 2018, 70, 227–236. [Google Scholar] [CrossRef]
  81. Jendrny, C.; Beck-Sickinger, A.G. Inhibition of Kallikrein-Related Peptidases 7 and 5 by Grafting Serpin Reactive-Center Loop Sequences onto Sunflower Trypsin Inhibitor-1 (SFTI-1). Chembiochem 2015, 17, 719–726. [Google Scholar] [CrossRef]
  82. Swedberg, J.E.; Wu, G.; Mahatmanto, T.; Durek, T.; Caradoc-Davies, T.T.; Whisstock, J.C.; Law, R.H.P.; Craik, D.J. Highly Potent and Selective Plasmin Inhibitors Based on the Sunflower Trypsin Inhibitor-1 Scaffold Attenuate Fibrinolysis in Plasma. J. Med. Chem. 2018, 62, 552–560. [Google Scholar] [CrossRef] [PubMed]
  83. Jackson, M.; Yap, K.; Poth, A.G.; Gilding, E.; Swedberg, J.E.; Poon, S.; Qu, H.; Durek, T.; Harris, K.; Anderson, M.; et al. Rapid and Scalable Plant-Based Production of a Potent Plasmin Inhibitor Peptide. Front. Plant Sci. 2019, 10, 602. [Google Scholar] [CrossRef] [PubMed]
  84. Swedberg, J.E.; Harris, J.M. Plasmin Substrate Binding Site Cooperativity Guides the Design of Potent Peptide Aldehyde Inhibitors. Biochemistry 2011, 50, 8454–8462. [Google Scholar] [CrossRef] [PubMed]
  85. Al-Horani, R.A.; Desai, U.R. Recent Advances on Plasmin Inhibitors for the Treatment of Fibrinolysis-Related Disorders. Med. Res. Rev. 2014, 34, 1168–1216. [Google Scholar] [CrossRef] [PubMed]
  86. Bhongade, B.A.; Gouripur, V.V.; Gadad, A.K. 3D-QSAR CoMFA studies on trypsin-like serine protease inhibitors: A comparative selectivity analysis. Bioorganic Med. Chem. 2005, 13, 2773–2782. [Google Scholar] [CrossRef]
  87. Xue, G.; Gong, L.; Yuan, C.; Xu, M.; Wang, X.; Jiang, L.; Huang, M. A structural mechanism of flavonoids in inhibiting serine proteases. Food Funct. 2017, 8, 2437–2443. [Google Scholar] [CrossRef]
  88. Lamy, S.; Lafleur, R.; Bédard, V.; Moghrabi, A.; Barrette, S.; Gingras, D.; Béliveau, R. Anthocyanidins inhibit migration of glioblastoma cells: Structure-activity relationship and involvement of the plasminolytic system. J. Cell. Biochem. 2006, 100, 100–111. [Google Scholar] [CrossRef]
  89. Tjahjono, D.H.; Sari, B.L.; Ibrahim, S. Pharmacophore Modeling, Docking, and Molecular Dynamics Simulation of Flavonoids as Inhibitors of Urokinase-type Plasminogen Activator. J. Math. Fundam. Sci. 2021, 53, 451–465. [Google Scholar] [CrossRef]
  90. Osaki, N.; Koyano, T.; Kowithayakorn, T.; Hayashi, M.; Komiyama, K.; Ishibashi, M. Sesquiterpenoids and plasmin-inhibitory flavonoids from Blumea balsamifera. J. Nat. Prod. 2005, 68, 447–449. [Google Scholar] [CrossRef]
  91. Lee, S.-Y.; Min, B.-S.; Kim, J.-H.; Lee, J.; Kim, T.-J.; Kim, C.-S.; Kim, Y.-H.; Lee, H.-K. Flavonoids from the leaves ofLitsea japonica and their anti-complement activity. Phytother. Res. 2005, 19, 273–276. [Google Scholar] [CrossRef]
  92. Cuccioloni, M.; Mozzicafreddo, M.; Bonfili, L.; Cecarini, V.; Eleuteri, A.M.; Angeletti, M. Natural Occurring Polyphenols as Template for Drug Design. Focus on Serine Proteases. Chem. Biol. Drug Des. 2009, 74, 1–15. [Google Scholar] [CrossRef] [PubMed]
  93. Law, R.H.; Wu, G.; Leung, E.W.W.; Hidaka, K.; Quek, A.J.; Caradoc-Davies, T.T.; Jeevarajah, D.; Conroy, P.J.; Kirby, N.M.; Norton, R.S.; et al. X-ray crystal structure of plasmin with tranexamic acid-derived active site inhibitors. Blood Adv. 2017, 1, 766–771. [Google Scholar] [CrossRef] [PubMed]
  94. Marcińczyk, N.; Gromotowicz-Popławska, A.; Tomczyk, M.; Chabielska, E. Tannins as Hemostasis Modulators. Front. Pharmacol. 2022, 12, 4055. [Google Scholar] [CrossRef] [PubMed]
  95. Pereira, B.; Brazón, J.; Rincón, M.; Vonasek, E. Browplasminin, a condensed tannin with anti-plasmin activity isolated from an aqueous extract of Brownea grandiceps Jacq. flowers. J. Ethnopharmacol. 2017, 198, 282–290. [Google Scholar] [CrossRef] [PubMed]
  96. Marcinczyk, N.; Gołaszewska, A.; Gromotowicz-Poplawska, A.; Misztal, T.; Strawa, J.; Tomczyk, M.; Kasacka, I.; Chabielska, E. Multidirectional Effects of Tormentil Extract on Hemostasis in Experimental Diabetes. Front. Pharmacol. 2021, 12, 682987. [Google Scholar] [CrossRef]
  97. Marcinczyk, N.; Jarmoc, D.; Leszczynska, A.; Zakrzeska, A.; Kramkowski, K.; Strawa, J.; Gromotowicz-Poplawska, A.; Chabielska, E.; Tomczyk, M. Antithrombotic Potential of Tormentil Extract in Animal Models. Front. Pharmacol. 2017, 8, 534. [Google Scholar] [CrossRef]
  98. Shen, Z.-Q.; Dong, Z.-J.; Peng, H.; Liu, J.-K. Modulation of PAI-1 and tPA Activity and Thrombolytic Effects of Corilagin. Planta Med. 2003, 69, 1109–1112. [Google Scholar] [CrossRef]
  99. Ploplis, V.A.; Carmeliet, P.; Vazirzadeh, S.; Van Vlaenderen, I.; Moons, L.; Plow, E.F.; Collen, D. Effects of Disruption of The Plasminogen Gene on Thrombosis, Growth, and Health in Mice. Circulation 1995, 92, 2585–2593. [Google Scholar] [CrossRef]
  100. Lin, H.; Xu, L.; Yu, S.; Hong, W.; Huang, M.; Xu, P. Therapeutics targeting the fibrinolytic system. Exp. Mol. Med. 2020, 52, 367–379. [Google Scholar] [CrossRef]
  101. Cale, J.M.; Li, S.-H.; Warnock, M.; Su, E.J.; North, P.R.; Sanders, K.L.; Puscau, M.M.; Emal, C.D.; Lawrence, D. Characterization of a Novel Class of Polyphenolic Inhibitors of Plasminogen Activator Inhibitor-1. J. Biol. Chem. 2010, 285, 7892–7902. [Google Scholar] [CrossRef]
  102. Zha, X.; Diaz, R.; Franco, J.J.R.; Sanchez, V.F.; Fasoli, E.; Barletta, G.; Carvajal, A.; Bansal, V. Inhibitors of Urokinase Type Plasminogen Activator and Cytostatic Activity from Crude Plants Extracts. Molecules 2013, 18, 8945–8958. [Google Scholar] [CrossRef] [PubMed]
  103. Uzawa, K.; Amelio, A.L.; Kasamatsu, A.; Saito, T.; Kita, A.; Fukamachi, M.; Sawai, Y.; Toeda, Y.; Eizuka, K.; Hayashi, F.; et al. Resveratrol Targets Urokinase-Type Plasminogen Activator Receptor Expression to Overcome Cetuximab-Resistance in Oral Squamous Cell Carcinoma. Sci. Rep. 2019, 9, 12179. [Google Scholar] [CrossRef] [PubMed]
  104. Xia, Y.; Yuan, M.; Li, S.; Thuan, U.T.; Nguyen, T.T.; Kang, T.W.; Liao, W.; Lian, S.; Jung, Y.D. Apigenin suppresses the IL-1β-induced expression of the urokinase-type plasminogen activator receptor by inhibiting MAPK-mediated AP-1 and NF-κB signaling in human bladder cancer T24 cells. J. Agric. Food Chem. 2018, 66, 7663–7673. [Google Scholar] [CrossRef] [PubMed]
  105. Sandra, D.; Radha, M.; Harishkumar, M.; Yuichi, N.; Sayuri, O.; Masugi, M. Downregulation of urokinase-type plasminogen activator and plasminogen activator inhibitor-1 by grape seed proanthocyanidin extract. Phytomedicine 2010, 17, 42–46. [Google Scholar] [CrossRef] [PubMed]
  106. Sadeghi, M.; Safaeian, L.; Ghazvini, M.A.; Ramezani, M. Evaluation of fibrinolytic and antioxidant effects of Allium affine hydroalcoholic extract. Res. Pharm. Sci. 2017, 12, 299–306. [Google Scholar] [CrossRef]
  107. Safaeian, L.; Zolfaghari, B.; Aghaye-Ghazvini, M.; Behnampour, M. Evaluation of fibrinolytic and antioxidant effects of Allium elburzense bulb extracts. Avicenna J. Phytomedicine 2017, 7, 223–231. [Google Scholar] [CrossRef]
  108. Omotoso Abayomi, E.; Ezealisiji, K.; Bagbi, B.M.; Maduakor Ogochukwu, H. Fibrinolytic activity of some Nigerian medicinal plants. J. Pharm. Pharmacol. 2014, 2, 177–184. [Google Scholar]
  109. Ibrahim, M.Y.; Ansari, P.; Riasat-ul-Islam, A.K.M.; Sultana, M.; Akter Zhumur, N.; Shafi, S.M. Evaluation of thrombolytic and cytotoxic activities of an ornamental medicinal plant: Byttneria pilosa. Am. J. Biomed. Res. 2015, 3, 35–39. [Google Scholar]
  110. Klafke, J.Z.; da Silva, M.A.; Rossato, M.F.; Trevisan, G.; Walker, C.I.B.; Leal, C.A.M.; Borges, D.O.; Schetinger, M.R.C.; Moresco, R.N.; Duarte, M.M.M.F.; et al. Antiplatelet, Antithrombotic, and Fibrinolytic Activities of Campomanesia xanthocarpa. Evid. Based Complement. Altern. Med. 2011, 2012, 954748. [Google Scholar] [CrossRef]
  111. Quintal-Martínez, J.P.; Quintal-Ortiz, I.G.; Alonzo-Salomón, L.G.; Muñoz-Rodríguez, D.; Segura-Campos, M.R. Antithrombotic Study and Identification of Metabolites in Leaf Extracts of Chaya [Cnidoscolus aconitifolius (Mill.) I.M. Johnst.]. J. Med. Food 2021, 24, 1304–1312. [Google Scholar] [CrossRef]
  112. Godatwar, P.K.; Chaudhary, S.; Sharma, R. In vitro thrombolytic activity of Dhamasa (Fagonia arabica Linn.), Kushta (Saussurea lappa Decne.), and Guduchi (Tinospora cordifolia Thunb.). Ayu 2015, 36, 421–424. [Google Scholar] [CrossRef]
  113. Chowdhury, T.A.; Hasanat, A.; Jakaria; Kamal, A.M.; Kabir, M.S.H.; Hossain, S.; Mamur, A.; Hossain, M.M. Thrombolytic and cytotoxic activity of methanolic extract of Commelina benghalensis (Family: Commelinaceae) Leaves. J. Sci. Innov. Res. 2015, 4, 100–104. [Google Scholar] [CrossRef]
  114. Madhu, C.; Sharada, A. Fibrinogenolytic activity of serine proteases(s) from Cucumis dipsaceus. Biocatal. Agric. Biotechnol. 2019, 17, 685–689. [Google Scholar] [CrossRef]
  115. Kabir, M.S.H.; Mahamoud, S.; Chakrabarty, N.; Ahmad, S.; Al Masum, A.; Hoque, A.; Hossain, M.M.; Rahman, M.; Uddin, M.M.N. Antithrombotic and cytotoxic activities of four Bangladeshi plants and PASS prediction of their isolated compounds. J. Basic Clin. Physiol. Pharmacol. 2016, 27, 659–666. [Google Scholar] [CrossRef] [PubMed]
  116. Kim, H.K.; Sung, Y.-Y.; Yang, W.-K. Antiplatelet, anticoagulant and fibrinolytic effects of Litchi chinensis Sonn. extract. Mol. Med. Rep. 2011, 5, 721–724. [Google Scholar] [CrossRef]
  117. Srinivasa Reddy, C.; Ammani, K.; Rose Mary, T. In vitro evaluation of fibrinolytic and antioxidant activities of Maba buxifolia (Rottb.) Juss. stem. J. Pharmacogn. Phytochem. 2015, 3, 148–151. [Google Scholar]
  118. Shahik, S.; Sikder, M.O.F.; Patwary, N.I.A.; Sohel; Islam, S.; Nishi, T.F.; Sultana, T.; Barua, R. In vitro thrombolytic and cytotoxic evaluation of Mentha arvensis L., Mentha spicata L. and Mentha viridis L. IOSR J. Pharm. Biol. Sci. 2014, 9, 97–102. [Google Scholar] [CrossRef]
  119. Mahmud, S.; Akhter, S.; Rahman, A.; Aklima, J.; Akhter, S.; Merry, S.R.; Jubair, S.M.R.; Dash, R.; Bin Emran, T. Antithrombotic Effects of Five Organic Extracts of Bangladeshi Plants In Vitro and Mechanisms in In Silico Models. Evid. Based Complement. Altern. Med. 2015, 2015, 782742. [Google Scholar] [CrossRef]
  120. Prasad, S.; Kashyap, R.S.; Deopujari, J.Y.; Purohit, H.J.; Taori, G.M.; Daginawala, H.F. Effect of Fagonia Arabica (Dhamasa) on in vitro thrombolysis. BMC Complement. Altern. Med. 2007, 7, 36. [Google Scholar] [CrossRef]
  121. Bin Emran, T.; Rahman, A.; Uddin, M.M.N.; Rahman, M.; Uddin, Z.; Dash, R.; Layzu, C. Effects of organic extracts and their different fractions of five Bangladeshi plants on in vitro thrombolysis. BMC Complement. Altern. Med. 2015, 15, 128. [Google Scholar] [CrossRef]
  122. Kuerban, A.; Almulaiky, Y.Q.; Sheikh, R.A.; Alzahrani, S.A.; Al-Talhi, H.A.; Nasrullah; Moselhy, S.S.; Abulnaja, K.O. In vivo Antithrombotic Activity of Ethanolic Extract from Ocimum basilicum L. Annu. Res. Rev. Biol. 2017, 20, 1–6. [Google Scholar] [CrossRef]
  123. Legnani, C.; Frascaro, M.; Guazzaloca, G.; Ludovici, S.; Cesarano, G.; Coccheri, S. Effects of a dried garlic preparation on fibrinolysis and platelet aggregation in healthy subjects. Arzneimittelforschung 1993, 43, 119–122. [Google Scholar] [PubMed]
  124. Sikora, J.; Broncel, M.; Markowicz, M.; Chałubiński, M.; Wojdan, K.; Mikiciuk-Olasik, E. Short-term supplementation with Aronia melanocarpa extract improves platelet aggregation, clotting, and fibrinolysis in patients with metabolic syndrome. Eur. J. Nutr. 2011, 51, 549–556. [Google Scholar] [CrossRef] [PubMed]
  125. Ebrahimi, F.; Torbati, M.; Mahmoudi, J.; Valizadeh, H. Medicinal Plants as Potential Hemostatic Agents. J. Pharm. Pharm. Sci. 2020, 23, 10–23. [Google Scholar] [CrossRef]
  126. Subramani, B.; Sathiyarajeswaran, P. Current update on herbal sources of antithrombotic activity—A comprehensive review. Egypt. J. Intern. Med. 2022, 34, 26. [Google Scholar] [CrossRef]
  127. Choi, J.-H.; Kim, D.-W.; Park, S.-E.; Choi, B.-S.; Sapkota, K.; Kim, S.; Kim, S.-J. Novel thrombolytic protease from edible and medicinal plant Aster yomena (Kitam.) Honda with anticoagulant activity: Purification and partial characterization. J. Biosci. Bioeng. 2014, 118, 372–377. [Google Scholar] [CrossRef]
  128. Jeon, J.S.; Kim, J.; Park, S.; Ryou, C.; Kim, C.Y. Preparative purification of plasmin activity stimulating phenolic derivatives from Gastrodia elata using centrifugal partition chromatography. Biomed Chromatogr. 2016, 30, 976–982. [Google Scholar] [CrossRef]
  129. Rajput, M.; Mathur, V.; Agrawal, P.; Chandrawanshi, H.; Pilaniya, U. Fibrinolytic activity of kaempferol isolated from the fruits of Lagenaria siceraria(Molina) Standley. Nat. Prod. Res. 2011, 25, 1870–1875. [Google Scholar] [CrossRef]
  130. Urs, A.P.; Rudresha, G.V.; Manjuprasanna, V.N.; Suvilesh, K.N.; Gowda, M.D.M.; Yariswamy, M.; Hiremath, V.; Ramakrishnan, C.; Savitha, M.N.; Jayachandra, K.; et al. Plant latex thrombin-like cysteine proteases alleviates bleeding by bypassing factor VIII in murine model. J. Cell. Biochem. 2019, 120, 12843–12858. [Google Scholar] [CrossRef]
  131. Urs, A.P.; Manjuprasanna, V.N.; Rudresha, G.V.; Hiremath, V.; Sharanappa, P.; Rajaiah, R.; Vishwanath, B.S. Thrombin-like serine protease, antiquorin from Euphorbia antiquorum latex induces platelet aggregation via PAR1-Akt/p38 signaling axis. Biochim. Biophys. Acta BBA Mol. Cell Res. 2020, 1868, 118925. [Google Scholar] [CrossRef]
  132. Rajesh, R.; Shivaprasad, H.V.; Gowda, C.D.R.; Nataraju, A.; Dhananjaya, B.L.; Vishwanath, B.S. Comparative Study on Plant Latex Proteases and their Involvement in Hemostasis: A Special Emphasis on Clot Inducing and Dissolving Properties. Planta Medica 2007, 73, 1061–1067. [Google Scholar] [CrossRef] [PubMed]
  133. Shivaprasad, H.V.; Rajaiah, R.; Frey, B.M.; Frey, F.J.; Vishwanath, B.S. ‘Pergularain e I’—A plant cysteine protease with thrombin-like activity from Pergularia extensa latex. Thromb. Res. 2010, 125, e100–e105. [Google Scholar] [CrossRef] [PubMed]
  134. Patel, G.K.; Kawale, A.A.; Sharma, A.K. Purification and physicochemical characterization of a serine protease with fibrinolytic activity from latex of a medicinal herb Euphorbia hirta. Plant Physiol. Biochem. 2011, 52, 104–111. [Google Scholar] [CrossRef] [PubMed]
  135. Siritapetawee, J.; Thumanu, K.; Sojikul, P.; Thammasirirak, S. A novel serine protease with human fibrino(geno)lytic activities from Artocarpus heterophyllus latex. Biochim. Biophys. Acta BBA Proteins Proteom. 2012, 1824, 907–912. [Google Scholar] [CrossRef]
  136. Araújo, A.P.U.; Hansen, D.; Vieira, D.F.; de Oliveira, C.; Santana, L.A.; Beltramini, L.M.; Sampaio, C.A.; Sampaio, M.U.; Oliva, M.L.V. Kunitz-type Bauhinia bauhinioides inhibitors devoid of disulfide bridges: Isolation of the cDNAs, heterologous expression and structural studies. Biol. Chem. 2005, 386, 561–568. [Google Scholar] [CrossRef]
  137. Zhou, D.; Hansen, D.; Shabalin, I.; Gustchina, A.; Vieira, D.F.; De Brito, M.V.; Araujo, A.P.U.; Oliva, M.L.; Wlodawer, A. Structure of BbKI, a disulfide-free plasma kallikrein inhibitor. Acta Crystallogr. Sect. F Struct. Biol. Commun. 2015, 71, 1055–1062. [Google Scholar] [CrossRef]
  138. Yu, H.; Palazzolo, J.S.; Zhou, J.; Hu, Y.; Niego, B.; Pan, S.; Ju, Y.; Wang, T.-Y.; Lin, Z.; Hagemeyer, C.E.; et al. Bioresponsive Polyphenol-Based Nanoparticles as Thrombolytic Drug Carriers. ACS Appl. Mater. Interfaces 2022, 14, 3740–3751. [Google Scholar] [CrossRef]
Figure 1. Brief representation of hemostatic and non-hemostatic activity of the fibrinolytic system. ECM—extracellular matrix; proMMPs—matrix metalloproteinase zymogenes.
Figure 1. Brief representation of hemostatic and non-hemostatic activity of the fibrinolytic system. ECM—extracellular matrix; proMMPs—matrix metalloproteinase zymogenes.
Molecules 28 01677 g001
Figure 2. Main components of the fibrinolytic system and their physiological and therapeutical activators and inhibitors. C3, C5—complement components; EACA—aminocaproic acid; ECM—extracellular matrix; PAI-1, PAI-2—plasminogen activator inhibitors 1 and 2; PAR—protease-activated receptor; Plg-SK—plasminogen-streptokinase; proMMPs—matrix metalloproteinase zymogenes; rt-PA—recombinant t-PA; SK—streptokinase; TAFI—thrombin-activatable fibrinolysis inhibitor; t-PA—tissue-type plasminogen activator; TXA—tranexamic acid; u-PA—urokinase-type plasminogen activator.
Figure 2. Main components of the fibrinolytic system and their physiological and therapeutical activators and inhibitors. C3, C5—complement components; EACA—aminocaproic acid; ECM—extracellular matrix; PAI-1, PAI-2—plasminogen activator inhibitors 1 and 2; PAR—protease-activated receptor; Plg-SK—plasminogen-streptokinase; proMMPs—matrix metalloproteinase zymogenes; rt-PA—recombinant t-PA; SK—streptokinase; TAFI—thrombin-activatable fibrinolysis inhibitor; t-PA—tissue-type plasminogen activator; TXA—tranexamic acid; u-PA—urokinase-type plasminogen activator.
Molecules 28 01677 g002
Figure 3. Different generations of thrombolytic drugs.
Figure 3. Different generations of thrombolytic drugs.
Molecules 28 01677 g003
Table 1. General characteristics of main proteins of the fibrinolytic system.
Table 1. General characteristics of main proteins of the fibrinolytic system.
Protein of the Fibrinolytic SystemFunctionMolecular Weight (Da)/Number of Amino AcidsCatalytic triad of
Enzymes/Reactive Site
of Inhibitors
Main OriginsPlasma Concentration: µg/mL (Molar) Plasma Half-Life (T1/2)
Plasminogenproenzyme of plasmin92,000/791 -Liver180 (2 µM)~2.2 days
Plasmin
(EC 3.4.21.7)
serine protease85,000/530His603, Asp646, Ser741Liver180 (2 µM)
t-PA
(EC 3.4.21.68)
serine protease (plasminogen activator)70,000/527His322, Asp371, Ser478endothelial cells0.005 (70 pM)~5–6 min
u-PA
(EC 3.4. 21.73)
serine protease (plasminogen activator)53,000/411His204, Asp255, Ser356kidney, lung, and many other cell types0.002 (40 pM)~8–10 min
uPARurokinase receptor55,000/313-various cell types-
α2-antiplasminserine protease inhibitor (SERPIN)70,000/464Arg376-Met377Liver70 (1 µM)~2.6 days
α2-macroglobulinserine protease and metalloproteinase inhibitor725,000/5804
(tetramer)
the “bait region” and the reactive site formed by thiol esterification of β-SH group of Cys-949 and γ-carbonyl group of Glx-952Liver2000–4000 (2.4–4.8 µM)5–8 days
PAI-1serine protease inhibitor (SERPIN)52,000/379Arg346-Met347liver,
endothelial cells, megakaryocytes, smooth muscle
<0.05 (<1 nM)1–2 h
PAI-2serine protease inhibitor (SERPIN) 47,000 (non-glycosylated form) or 60,000 (glycosylated form)/393Arg358-Thr359placenta, monocytes/macrophages<0.250 (during pregnancy)24 h
TAFI/TAFIa procarboxypeptidase B/carboxypeptidase B—fibrinolysis inhibitor60,000/401 Liver4–15 (73–275 nM)~10 min (TAFIa)
Table 2. Currently used anti-fibrinolytic drugs.
Table 2. Currently used anti-fibrinolytic drugs.
Molecular
Action
NameBiochemical Mechanisms of ActionPharmacokinetics/
Elimination Route
Exemplary Clinical UseSide Effects
Inhibition of plasminogen activation to plasminEpsilon aminocaproic acid (EACA; 6-aminohexanoic acid)synthetic lysine analogues; competitive inhibitors of plasminogen binding to fibrin (and its activation to plasmin)terminal elimination T1/2 of 2 h/renalCardiac, prostate, gynecological and other surgery; bleeding associated with hyperfibrinolysis;
bleeding associated with hematopoietic disorders
Hypotension, nasal and conjunctival congestion, gastrointestinal disturbances (diarrhea, nausea, vomiting, abdominal pain), dizziness, headache, tinnitus, and ejaculation disorders [31]
Tranexamic Acid (TXA; trans-4-(aminomethyl)cyclohexanecarboxylic acid)terminal elimination T1/2 of 2–3 h/renalCardiac, orthopedic, gastrointestinal, and other surgery [32]Hypotension; longer-term administration may induce rashes, nausea, vomiting, weakness, retrograde ejaculation, myopathy, and rhabdomyolysis [32]
Inhibition of plasminAprotininpolypeptide inhibitor of serine proteinases, isolated from bovine lung, forms reversible complexes with the active serine residue in proteases and inhibits not only plasmin but also kallikrein, chymotrypsin, and trypsinterminal elimination T1/2 of 10 h/mainly by proteolysis, <10% renalCardiac surgeryRenal failure, myocardial infarction, heart failure, stroke, encephalopathy, mortality [33], anaphylaxis, graft occlusion, and stroke [34]
Table 3. The plant-derived substances with the highest potency in vitro to inhibit the serine proteases. Compounds with IC50 ≥ 50 µM have been considered weak inhibitors, and therefore they are not listed in this table. Kd—dissociation constants of the serine protease-plant-derived compound complexes; IC50—half-maximal inhibitory concentration.
Table 3. The plant-derived substances with the highest potency in vitro to inhibit the serine proteases. Compounds with IC50 ≥ 50 µM have been considered weak inhibitors, and therefore they are not listed in this table. Kd—dissociation constants of the serine protease-plant-derived compound complexes; IC50—half-maximal inhibitory concentration.
CompoundInhibited EnzymeKdIC50References
BufadienolidesPlasmin 16.70 μg/mL[43]
Thrombin 2.79 µg/mL[44]
CyanidinThrombin 0.25 μM[45]
Factor Xa 3.0 μM[46]
Epigallocatechin gallate (EGCG)Elastase 0.4 μM[47]
Elagic acidThrombin7.6 µM [48]
EsculinUrokinase 25.0 µM[49]
HypericinTrypsin 4.5 μM[50]
Urokinase 30.7 μM
HyperosideElastase 0.3 μM[51]
Trypsin 14.5 μM[50]
Urokinase 8.3 μM
Myricetin Thrombin 6.0 μM[52]
Elastase 4.0 μM[47]
Procyanidin B2Factor Xa 1.2 µM[46]
RutinPlasmin10.7 µM [53]
Thrombin 25.0 µM[49]
Trypsin 40 µM
Urokinase 6.0 µM
SalicinThrombin 11.4 µM[50]
Sennoside ATrypsin 6.1 µM[50]
Sennoside BTrypsin 10.6 µM[50]
SilybinThrombin 20.9 µM[50]
25 µM[45]
Trypsin 3.7 µM[50]
Urokinase 21.0 µM
Factor Xa 35.0 µM[46]
QuercetinPlasmin0.62 µM [53]
Thrombin0.35 µM [48]
30.0 µM[50]
1.5 µM[45]
35.0 µM[49]
Factor Xa 5.5 µM[46]
Trypsin 15.4 µM[50]
10.0 µM[49]
Urokinase 12.1 µM[50]
20.0 µM[49]
Elastase 50.0 µM[50]
Quercetin-3-glucuronidePlasmin0.14 µM [54]
5,5-trans-fused cyclic lactone euphane triterpene (GR133487)Trypsin 0.12 µM[55]
Chymotrypsin 0.01 µM
Thrombin 0.004 µM
Factor Xa > 10 µM
Factor XIa 1.1 µM
Factor XIIa > 10 µM
t-PA > 10 µM
Plasmin 5.6 µM
Elastase > 10 µM
Cathepsin G > 10 µM
5,5-trans-fused cyclic lactone euphane triterpene (GR133686)Trypsin 0.07 µM[55]
Chymotrypsin 0.07 µM
Thrombin 0.04 µM
Factor Xa > 10 µM
Factor XIa 0.7 µM
Factor XIIa > 10 µM
t-PA 1.0 µM
Plasmin 3.5 µM
Elastase > 10 µM
Cathepsin G 2.2 µM
Table 4. Peptide-based plant inhibitors of serine proteases of the blood coagulation cascade and fibrinolytic system.
Table 4. Peptide-based plant inhibitors of serine proteases of the blood coagulation cascade and fibrinolytic system.
Type of InhibitorNamePlant SourceTarget Serine ProteaseKiReferences
Bowman-BirkTcTITorresea cearensis Allemão Plasmin36.0 nM[57]
Factor XIIa1.45 µM
KunitzApTIAAcacia plumosa LoweKallikrein0.55 µM[58]
ApTIB0.58 µM
ApTIC0.65 µM
AaTIAraucaria angustifolia (Bertol.) KuntzePlasmin7.0 µM[59]
BbKIBauhinia bauhinioides (Mart.) J.F.Macbr.Kallikrein0.35 nM[60]
Plasmin33.0 nM[61]
Factor XIIa110.0 nM
BuXIPlasmin76.0 nM
Factor Xa14.0 nM
Factor XIIa74.0 nM
BvTIKallikrein23.0 nM
Factor XIIa21.0 nM
BrTIKallikrein14.0 nM
CeKICaesalpinia echinata Lam.Plasmin0.18 nM[62]
Factor XIIa0.18 nM
Factor Xa490 nM
Kallikrein3.1 nM
Corn Hageman factor inhibitorZea mays L.Factor XIIa2.1 nM[63]
STIGlycine max (L.) Merr.Plasmin192.10 nM[64]
Factor XIIa1367.59 nM
Kallikrein5.70 nM
SWTI Swartzia pickelii KillipPlasmin2.52 nM[65]
Kallikrein20.25 nM
Tamarind Kunitz inhibitorTamarindus indica L.Factor Xa220.0 nM[66]
Potato-type PICMTI-VCucurbita maxima Lam.Factor XIIa41.0 nM[67]
Squash PIMCTI-IMomordica charantia L.Factor XIIa13 nM[68]
Kallikrein110 μM
Factor Xa100 μM
MCTI-IIMomordica charantia L.Factor XIIa56 nM
Kallikrein100 μM
Factor Xa1.4 μM
Factor XIa18 μM
MCTI-IIIMomordica charantia L.Factor XIIa1.6 μM
Kallikrein140 μM
Factor Xa59 μM
CMTI-IIICucurbita maxima Lam.Factor XIIa70 nM
Kallikrein130 μM
Factor Xa23 μM
LLTI-IILagenaria leucantha Rusby var. Gourda MakinoFactor XIIa1.4 μM
Kallikrein27 μM
Factor Xa41 μM
LLTI-IIILagenaria leucantha Rusby var. Gourda MakinoFactor XIIa4.2 μM
Kallikrein200 μM
Factor Xa19 μM
LCTI-IILuffa cylindrica Roem.Factor XIIa75 nM
Kallikrein20 μM
Factor Xa780μM
LCTI-IIILuffa cylindrica Roem.Factor XIIa3.8 nM
Kallikrein38 μM
Factor Xa100 μM
Table 5. Exemplary data from in vitro studies on effects of plant-derived preparations on fibrin clot lysis. SK—streptokinase.
Table 5. Exemplary data from in vitro studies on effects of plant-derived preparations on fibrin clot lysis. SK—streptokinase.
The Examined PlantsBiological Materials and the Used Concentrations of Plant ExtractsReference
Fibrinolytic Drug
Main Findings References
Allium affine Ledeb.Human blood clots; hydro-alcoholic extract from aerial parts of A. affine (70% ethanol); 0.005–50 mg/mLSK (8000 IU)A. affine efficiency—up to ~30% of clot lysis; SK—51.40%[103]
Allium elburzense WendelboHuman blood clots; hydroalcoholic (70% ethanol), aqueous, chloroformic and butanolic extracts from A. elburzense bulb; 5 mg/mLSK (30,000 IU)Clot lysis efficiency: aqueous extract (33.11%) > hydroalcoholic extract (22.40%) > butanolic extract (16.75) > chloroformic (9.77%) extract; SK—60.59%[104]
Anona senegalensis (A.DC.) Pichon, Buchholzia coriacea Engl., Citrullus colocynthis (L.) Schrad., Cnidoscolus aconitifolius (Mill.) I. M. Johnst., Curculigo pilosa (Schumach. & Thonn.) Engl., Nicotiana tabacum L., Parinari curatellifolia Planch. ex Benth., Peperomia pellucida (L.) Kunth, Sida acuta Burm. f., Xylopia aethiopica (Dunal) A.Rich.Human blood clots; the crude methanolic extracts; 100 μg/mLSK (30,000 IU)The highest clot lysis efficiency: P. curatellifolia—56.12%, C. aconitifolius 48.38%, A. senegalensis—46.36%, X. aethiopica —43.20%, B. coriacea—27.06%; SK—60.20%[105]
Byttneria pilosa Roxb.Human blood clots; crude, methanol extract of B. pilosa; 100 µL/clot sampleSK (30,000 IU)Clot lysis in samples treated with B. pilosa 46.20 ± 2.27%; in samples treated with SK: 82.60 ± 2.45%[106]
Campomanesia xanthocarpa (Mart.) O. BergHuman and mice blood clots; water extract from C. xanthocarpa leaves; 1, 3, 10, 30, and 100 μg/mLSK (1, 3, 10, 30, and 100 μg/mL)In mice blood, EC50 for C. xanthocarpa extract and SK: 21 μg/mL and 24 μg/mL, respectively; the Emax (at 100 μg/mL) was 56 ± 9% and 60 ± 14%, respectively. In human blood, the EC50 for C. xanthocarpa extract and SK was 11 and 4 μg/mL, respectively; Emax = 62 ± 7% and 70 ± 6%, respectively[107]
Cnidoscolus aconitifolius (Mill.) I.M. Johnst.Human blood clots; aqueous (Aq), ethanolic (EtOH), acetonic (An), ethyl acetate (AcOEt), diethyl ether (Et2O), and hexane (Hx) extracts from C. aconitifolius leaves; 0.1, 1, and 10 mg/mLSK (30,000 IU)The most effective were AcOEt, An, and Hx extracts (at 10 mg/mL), yielding 18.4—24% lysis; SK—76% lysis[108]
Fagonia arabica L., Saussurea lappa Decne., Tinospora cordifolia Thunb.Human blood clots; aqueous extracts from whole dry plant of F. arabica, dry bark of S. lappa, dry stem of T. cordifolia; 100 µL/clot sampleSK (30,000 IU)Clot lysis efficiency: 68.06%, 14.85%, 25.01%, and 92.54%, for F. arabica, S. lappa, T. cordifolia, and SK, respectively.[109]
Commelina benghalensis Forssk.Human blood clots; methanol extract from C. benghalensis leaves; 10–1000 μg/mL SK (30,000 IU)Average lytic activity of 40.94% for the extract, and 75% for SK[110]
Cucumis dipsaceus Wender. ex Steud.Fibrinogen preparation; 25 and 50 µg/mL-Fibrinogenolytic activity of the aqueous C. dipsaceus dialysate fraction (AqCDF): hydrolysis of Aα and Bβ chains of fibrinogen, partial degradation of γ chain[111]
Curculigo recurvata W.T. Aiton (Satipata), Amorphophallus bulbifer Roxb. (Olkachu), Phyllanthus sikkimensis Muell. Arg., Thunbergia grandiflora Roxb. (Nillata)Human blood clots; methanol extracts from the examined plants; 100 µL/clot sampleSK (30,000 IU)Clot lysis efficiency: C. recurvata 28.10%, A. bulbifer 42.47%, P. sikkimensis—32.86%, T. grandiflora 25.51%, SK—78.23%[112]
Litchi chinensis Sonn.Rat fibrin clots; ethanol (70%) extract from dried fruits; fibrin plate assay, 0.5, 1, 2, and 4 mg/mLPlasmin, 1–2 µg/fibrin clot plateInduction of fibrinolysis at 2 and 4 mg/mL
[113]
Maba buxifolia (Rottb.) Juss.Human blood clots; methanol extract from the plant stem; 1 mg/mLUrokinase (50,000 IU)24.3% of clot lysis for the extract; 100% lysis for urokinase-treated samples[114]
Mentha arvensis L., Mentha spicata L., Mentha viridis L.Human blood clots; Mentha sp. extracts (10 mg/mL): 95% methanol, 99.50% ethanol, 95% chloroform and acetone extracts); 100 µL/clot sampleSK (15,000 IU)32.56%, 30.89%, 30.29% clot lysis for the methanol extracts; 32.04%, 30.37%, 30.02% clot lysis for the ethanol extracts; 31.87%, 29.77%, 29.05% clot lysis efficiency for the chloroform extracts; and 30.29%, 28.45%, 27.55% clot lysis efficiency for the acetone extracts for M. arvensis, M. spicata, and M. viridis, respectively[115]
Ocimum tenuiflorum Burm.f., Andrographis paniculate (Burm.f.) Wall., Adhatoda vasica Nees, Leea macrophylla Roxb., Litsea glutinosa (Lour.) C.B.Rob.Human blood clot; methanol extracts; 100 µL/clot sampleSK (30,000 IU)L. macrophylla extract had the highest activity (47.47% of clot lysis); SK—71.14% of clot lysis
[116]
Tinospora cordifolia (Willd.) Miers, Rubia cordifolia L., Hemidesmus indicus (L.) R. Br. ex Schult., Glycyrrhiza glabra L., Fagonia arabica L., Bacopa monnieri Hayata & Matsum.Human blood clots; aqueous herbal extracts; 100 µL/clot sampleSK (30,000 IU)F. Arabica—75.6% clot lysis, B. monnieri—41.8% clot lysis, other extracts < 20% clot lysis; SK—86% clot lysis[117]
Trema orientalis L., Bacopa monnieri Hayata & Matsum., Capsicum frutescens L., Brassica oleracea L., Urena sinuata L.Human blood clots; methanol crude extracts and fractions (i.e., chloroform, n-hexane, hydro-methanol, and ethyl acetate fractions); 100 µL/clot sampleSK (30,000 IU)The highest clot lysis activity: chloroform fractions from T. orientalis, B. monnieri, C. frutescens, B. oleracea, and U. sinuata, i.e., 46.44 ± 2.44%, 48.39 ± 10.12%, 36.87 ± 1.27%, 30.24 ± 0.95%, and 47.89 ± 6.83%, respectively; SK: 80.77 ± 1.12%[118]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Kolodziejczyk-Czepas, J.; Czepas, J. Plant-Derived Compounds and Extracts as Modulators of Plasmin Activity—A Review. Molecules 2023, 28, 1677. https://doi.org/10.3390/molecules28041677

AMA Style

Kolodziejczyk-Czepas J, Czepas J. Plant-Derived Compounds and Extracts as Modulators of Plasmin Activity—A Review. Molecules. 2023; 28(4):1677. https://doi.org/10.3390/molecules28041677

Chicago/Turabian Style

Kolodziejczyk-Czepas, Joanna, and Jan Czepas. 2023. "Plant-Derived Compounds and Extracts as Modulators of Plasmin Activity—A Review" Molecules 28, no. 4: 1677. https://doi.org/10.3390/molecules28041677

Article Metrics

Back to TopTop