ArticlePDF Available

The antithrombotic activity of corilagin purified from korean herb-Phyllanthus ussuriensis

Authors:
  • university of science, Democratic People’s Republic of Korea

Abstract

Plasminogen activator inhibitor-1(PAI-1) is a main negative regulator of the fibrinolytic system. In animal studies, inhibition of PAI-1 activity prevents arterial and venous thrombosis, indicating that PAI-1 inhibitors may be used as a new class of antithrombotics. In this study, we revealed that corilagin which purified from Korean herb, Phyllanthus ussuriensis, inhibited plasma PAI-1 and increased the activity of t-PA in vitro and in vivo. We then observed that when injected more than 6mg/kg of corilain into the rat electrically stimulated carotid artery thrombosis rat, the rethrombosis ratio was lower than 28.6%, and it was much lower than when 20000IU/kg of urokinase was injected. At the end we found out that corilagin remarkably decreased the content of plasma fibrin in the thrombosis rat and the decrease at dose of more than 6mg/kg was about 4.4 fold than the control. Our data show that corilagin is a new class of small molecule PAI-1 inhibitors with anti-thrombotic potential and has great prospect as thrombosis therapeutic drug.
The antithrombotic activity of corilagin purified from korean
herb-Phyllanthus ussuriensis
Kum Chol Ri1,*, Chol Ho Ju1
Abstract: Plasminogen activator inhibitor-1(PAI-1) is a main negative regulator of the
fibrinolytic system. In animal studies, inhibition of PAI-1 activity prevents arterial and venous
thrombosis, indicating that PAI-1 inhibitors may be used as a new class of antithrombotics. In
this study, we revealed that corilagin which purified from Korean herb, Phyllanthus
ussuriensis, inhibited plasma PAI-1 and increased the activity of t-PA in vitro and in vivo. We
then observed that when injected more than 6mg/kg of corilain into the rat electrically
stimulated carotid artery thrombosis rat, the rethrombosis ratio was lower than 28.6%, and it
was much lower than when 20000IU/kg of urokinase was injected. At the end we found out
that corilagin remarkably decreased the content of plasma fibrin in the thrombosis rat and the
decrease at dose of more than 6mg/kg was about 4.4 fold than the control. Our data show that
corilagin is a new class of small molecule PAI-1 inhibitors with anti-thrombotic potential and
has great prospect as thrombosis therapeutic drug.
Key Words:Plasminogen activator, Plasminogen activator inhibitor-1 (PAI-1), corilagin, thrombosis
Introduction
Fibrinolysis is an important physiologic mechanism
that removes intravascular thrombi to maintain vascular
patency in the setting of thrombosis. Plasminogen
activator inhibitor-1(PAI-1) is an immediate inhibitor of
tissue-type plasminogen activator (t-PA) and urokinse-
type plasminogen activator (u-PA), and therefore is a
negative regulator of the fibrinolytic system.
Recent studies suggest that PAI-1 also contributes
directly to the complications of obesity, including type 2
diabetes and coronary arterial thrombi, and may even
influence the accumulation of visceral fat [1]. Other
studies substantiate PAI-1 plays an important role in
cellular motility and tumor angiogenesis, and an orally
active PAI-1 inhibitor prevents angiogenesis in a
Matrigel implant [2].
It was found out that high levels of PAI-1 reduce
fibrinolytic potential and contribute to the development
of thrombosis. In the clinic, elevated levels of PAI-1
antigen and activity have been reported in patients with a
variety of thrombotic diseases including deep vein
thrombosis, disseminated intravascular coagulation,
unstable angina, myocardial infarction and coronary
artery disease [4, 5]. In animal studies, transgenic mice
expressing high levels of PAI-1 develop spontaneous
thrombosis, whereas PAI-1-deficient mice are more
resistant to venous or arterial thrombosis induced by
either endotoxin or chemical and electric injury [6].
1 Life Science Department, Univercity of Science, Pyongyang 999093,
Democratic People’s Republic of Korea .* address for correspondence:
15124579835 @163.com..Kum Chol Ri
Inhibition of PAI-1 activity by monoclonal or polyclonal
antibodies prevents thrombus formation in animal models,
indicating that inhibition of PAI-1 is a potential strategy
to prevent thrombosis. Mechanistically, the anti-
thrombotic effects of PAI-1 inhibition are achieved by
enhancing endogenous fibrinolytic potential without
directly affecting blood coagulation or platelet function.
In PAI-1-deficient mice, for example, no significant
defects were found in coagulation assays. PAI-1-deficient
mice showed no significant abnormalities in bleeding
tests and exhibited no spontaneous bleeding [7, 17].
These data suggest that an antithrombotic agent based on
PAI-1 inhibition may have a lower risk of bleeding than
that of conventional antiplatelet and anticoagulant drugs,
and therefore have a better therapeutic index.
Therefor, some studies to find out small molecule
compounds which inhibit either PAI-1 production or
activity have been performed [3]. Recently, we have
identified a small molecule PAI-1 inhibitor, corilagin,
among different compounds purified from korean herbs
by a high-throughput screening. We now report
antithrombotic characteristic of corilagin.
Materials and methods
The corilagin was prepared by filtering the extract of
Korean herb Phyllanthus ussuriensis at the gel
chromatography, and its chemical structure was verified
by nuclear magnetic resonance spectro-scopy and mass
spectrometry. To study the effects of corilagin on the
activity of PAI-1 in vitro, we collected blood from a
healthy rat, prevented coagulation by using 3.8% citric
Fig. 1 The structure of corilagin
sodium solution, and obtained a platelet-free plasma by
centrifugating for 30 minutes in 4, 2500rpm. The
plasma was mixed with different concentrations of
corilagin and kept for 45 minutes. The activities of t-PA
and PAI-1 were measured in the 96-well microtiter plate
according to the chromogenic substrate analysis kits
(Chromogenix) [3]. ThermoMax plate reader(Molecular
Devices, Sunnyvale, CA) was used for the chromogenic
analysis.
We also studied the effects of corilagin on the activities
of PAI-1 and tPA of the rat plasma in vivo. We divided
rats into two groups and injected 0.9% saline solution
and corilagin respectively for 20 minutes at the rate of
0.5mg/kg/min. We gathered blood before injection and
10 minutes, 30 minutes, 60 minutes, 90 minutes,
120minutes after injection respectively, and measured the
the activities of PAI-1 and tPA in the plasma.
To examine the thrombolysis effect of corilagin in the
thrombosis animals, electrically stimulated carotid artery
thrombosis rats [9] were used. In the electrically
stimulated carotid artery thrombosis rats, we divided rats
into 5 groups and 0.9% saline solution, 20000 IU/kg of
urokinase(Sigma, St.Louis, MO), 3mg/kg of corilagin, 6
mg/kg of corilagin, 9 mg/kg of corilagin were injected
respectively. At one hour after injection, the hematocele
was measured with supersonic tachometer (DVM-4200,
Hayashi Denki). We evaluated the vessels to be opened if
the hematocele is over half of the original hematocele
(the hematocele measured before the thrombus was
formed) and to fail to be opened if the hematocele is less
than half of the original hematocele. The vessels which
was opened at first but decreased to less than 25% at 1
hour after successful reperfusion were evaluated as re-
occlusion.
In order to study the effect of corilagin on the content
of the plasma fibrin, we divided thrombosis rats into 4
groups and 0.9% saline solution, 3 mg/kg of corilagin, 6
mg/kg of corilagin and 9 mg/kg of corilagin were
injected respectively. At 1 hour after that, we gathered
blood from each rat, prevented coagulation by using
citric sodium solution, and obtained a platelet-free
plasma by centrifuging for 15 minutes in 2500rpm. 1ml
of 0.025M CaCl2solution was added to each of the 1.9ml
of plasma from test groups in order to form the fibrin.
After centrifuging the fibrin fibers, the wet and dried
weight was measured with an electronic balance.
The MDA content of the brain was measured using the
TBA method [15].
Results and Discussion
- The chemical structure of corilagin
The chemical structure of corilagin is shown in Fig. 1.
- The effects of corilagin on the activities of PAI-1
and tPA of the rat plasma in vitro
We examined the effects of corilagin on the activities
of PAI-1 and tPA of the rat plasma, and the results were
shown in Table 1.
Table 1. The effect of corilagin on rat plasma PAI-1 and
tPA activities in vitro
( * P<0.05, ** P<0.01(comparison with saline solution)
As Table 1 shows, corilagin markedly inhibits the
activity of PAI-1 of the rat plasma and raises that of tPA
in vitro. The IC50 on the PAI-1 of the rat plasma
calculated from Table 1 was 18mg /L.
- The effects of corilagin on the activities of PAI-1
and tPA of the rat plasma in vivo
Table 2 and Table 3 show the effects of corilagin on
the activities of PAI-1 and tPA of the rat plasma in vivo,
respectively .
As Table 2 and Table 3 show, corilagin decreased
remarkably the activity of PAI-1 of the plama and
increased that of tPA of the plasma. At 90 minutes after
injection, the rat plasma PAI-1 activity was decreased to
the minimum value of 16.1±3.7 IU/ml and the activity of
the tPA of the plasma was mostly increased.
corilagin
mg/L
PAI-1 activity
IU/mL
tPA activity
IU/mL
0
10
20
40
80
160
19.2±2.5
16.1±2.4
11.5±2.2**
7.4±3.5**
4.5±1.6**
3.7±1.4**
3.3±1.4
4.7±1.6
5.2±1.8
5.7±1.6**
6.5±1.7**
7.0±1.8**
Table 2. The change in the activity of the rat plasma PAI-1 according to the time intervals
since injection of corilagin
The unit of PAI-1 activity is IU/ml.
*P<0.05, **P<0.01(comparison with the same time of saline solution), #P<0.01(comparison with 0 min)
Table 3. The change in the activity of the rat plasma tPA according to the time intervals
since injection of corilagin
The unit of t-PA activity is IU/ml.
**P<0.01(comparison with the same time of saline solution), #P<0.01(comparison with 0 min)
- The thrombolysis effect of the corilagin on the
thrombus in the carotid artery of electrically stimulated
carotid artery thrombosis rats
We examined the thrombolysis effect of the corilagin
on the thrombus in the carotid artery of electrically
stimulated carotid artery thrombosis rat, and the results
were shown in Table 4.
Table 4. The thrombolysis effect of the corilagin on the
carotid artery thrombus in rats
Medicine
reperfusion
/total
reocclusion
/reperfusion
saline
corilagin
urokinase
0/10
2/10
7/10
7/10
7/10
0/0
2/2
2/7
2/7
3/7
* P<0.05(comparison with saline)
As Table 4 shows, when injected 6mg/kg of corilagin,
reperfusion ratio was 70%, and reocclusion ratio was
28.6%, which is better than the injection of 20000IU/kg
of urokinase (reocclusion ratio was 42.9% in this case).
- The change in the content of the plasma fibrin
according to the variation in the corilagin content in the
thrombosis rats
The change in the content of the plasma fibrin
according to the variation in the corilagin dose in the
thrombosis rats was shown in Table 5.
Table 5 shows that the plasma fibrin content was
markedly reduced in thrombosis rats injected with
corilagin in comparison with the control. When more
Table 5. The change in the content of the plasma fibrin
according to the variation in the corilagin dose in the
thrombosis rats
dose,
mg/kg
the content of the plasma fibrin
wet weight,
mg/ml
dried weight,
mg/ml
saline
3
6
9
26.9±1.9
17.8±1.2
6.5±0.7**
6.4±0.9**
9.3±0.8
5.1±0.5
2.1±0.3**
2.0±0.2**
*P<0.05, **P<0.01(comparison with saline solution)
than 6mg/kg of corilagin was injected, the fibrin content
was decreased by 4.4 times in comparison with the
control.
- The effect of corilagin on MDA content of the brain
tissue in thrombosis rats
Table 6 shows the effect of corilagin on MDA content
of the brain tissue in thrombosis rats.
Table 6. The effect of corilagin on MDA content of the
brain tissue
corilagin dose,
mg/kg
MDAcontent,
nmol/g
saline
3
6
9
555.6±14.4
513.9±13.7
395.7±15.8
392.6±14.2
*P<0.05(comparison with the same time of saline solution)
As Table 6 shows, the MDA content of the brain tissue
in the rats was decreased according to the increase in the
corilagin dose.
time, min
medicine
0
30
60
90
120
saline
corilagin
26.4±4.8
26.7±5.2
25.9±5.0
24.1±4.6
27.4±4.3
20.3±5.4
26.9±5.7
16.1±3.7**
27.4±4.9
18.8±3.5
time, min
medicine
0
30
60
90
120
saline
corilagin
2.4±0.6
2.3±0.7
1.9±0.3
3.6±1.3**
2.0±0.5
7.3±1.4**
1.9±0.2
8.3±1.7**
2.1±0.4
5.8±1.5**
Conclusions
Through our experiments we revealed that corilagin
inhibited plasma PAI-1 and increased the activity of t-PA
in vitro and in vivo. We then observed that when injected
more than 6mg/kg of corilagin into the electrically
stimulated carotid artery thrombosis rats, the
rethrombosis ratio was lower than 28.6%, and it was
much lower than when 20000IU/kg of urokinase was
injected. At the end we found out that corilagin
remarkably decreased the content of plasma fibrin in the
thrombosis rats and the decrease at dose of more than
6mg/kg was about 4.4 fold than the control.
Thrombosis is a major cardiovascular disease,
afflicting millions people in the world. Currently used
antithrombotic drugs are inhibitors of either platelets or
blood clotting factors. More recently, several small
molecule PAI-1 inhibitors have been identified [9]. Most
of them have been reported that inhibit PAI-1 activity in
vitro [8, 10–12]. One compound series among them,
represented by XR5118, was shown to have
antithrombotic efficacy in the electrically stimulated
arterial thrombosis rats [13]. Another PAI-1 inhibitor,
WAY-140312, was shown to enhance arterial blood flow
and reduce venous thrombus weight when it was
administered orally in the thrombosis rats [14]. These
studies have demonstrated the feasibility of developing
nonpeptic small molecule PAI-1 inhibitors as a new class
of antithrombotic agents[16, 18, 19, 20]. Our data show
that corilagin is a new class of small molecule PAI-1
inhibitors with anti-thrombotic potential and has great
prospect as thrombosis therapeutic drug.
References
1. B De-Taeye, et al., Plasminogen activator
inhibitor-1: a common denominator in obesity, diabetes
and cardiovascular disease, Curr-Opin-Pharmacol, Vol
5(2005), 149-154pp
2. C. E. Leik, et al., Effect of pharmacologic
plasminogen activator inhibitor-1 inhibition on cell
motility and tumor angiogenesis, J-Thromb-Haemost,
Vol 4(2006), 2710-2715pp
3. A. Liang, et al., Characterization of a small
molecule PAI-1 inhibitor, ZK4044, Thromb-Res, Vol
115(2005), 341-350pp
4. B. Wiman, Plasminogen activator inhibitor-
1(PAI-1) in plasma: its role in thrombotic disease,
Thromb Haemost, Vol74(1995), 71-76pp
5. H. M. Hoffmeister, et al., Correlation between
coronary morphology and molecular markers of
fibrinolysis in unstable angina pectoris, Atherosclerosis,
Vol 144(1999), 151-157pp
6. M. Eren, et al., Age-dependent spontaneous
coronary arterial thrombosis in transgenic mice that
express a stable form of human plasminogen activator
inhibitor-1, Circulation, Vol 106(2002), 491-496pp
7. T. Abrahamsson, et al., Anti-thrombotic effect of
a PAI-1 inhibitor in rats given endotoxin, Thromb
Haemost, Vol 75(1996), 118-126pp
8. P. A. Chariton, et al., Evaluation of a low
molecular weight modulator of human plasminogen
activator inhibitor-1 activity, Thrombosis and
Haemostasis, Vol 75(1996), 808-815pp
9. Q. Wu, Z. Zhao, Inhibition of PAI-1: a new anti-
thrombotic approach, Curr Drug Targets Cardiovasc
Haematol Disord, Vol 2(2002), 27–42pp
10. P. Bjorquist, et al., Identification of the binding
site for a low-molecular-weight inhibitor of plasminogen
activator inhibitor type 1 by site-directed mutagenesis,
Biochemistry Vol 37(1998), 1227–1234pp
11. H. Elokdah, et al., Tiplaxtinin, a novel, orally
efficacious inhibitor of plasminogen activator inhib-itor-1:
design, synthesis, and preclinical characterization, J Med
Chem Vol 47(2004), 3491–3494pp
12. B. Ye, et al., Synthesis and biological evaluation
of piperazine-based derivatives as inhibitors of plasmino-
gen activator inhibitor-1 (PAI-1), Bioorg Med Chem Lett
Vol 14(2004), 761–765pp
13. P. A. Charlton, et al., XR5118, a novel
modulator of plasminogen activator inhibitor-1 (PAI-1),
increases endogenous tPA activity in the rat, Fibrinolysis
Proteol-ysis Vol 11(1997) 51–56pp
14. Kumchol Ri, Yanqing Wang, Xiran Zhang.
Innovator’s Innovative Genetic Model: From Biological
to Social Perspective, Science Journal of Business and
Management, 2018; 6(2): 38-44
15. D. L., Crandall, et al., Characterization and
comparative evaluation of a structurally unique PAI-1
inhibitor exhibiting oral in vivo efficacy, J Thromb
Haemost Vol 2(2004) 1422–1428pp
16. Bing jing zheng et al., development and
validation of an UPLC-PDA method for the
determination of corilagin in rat plasma and its
application to pharmacokinetic study, journal of
chromatography B vol 1031(2016) 76-79pp
17. Kum Chol Ri, Jong Su Kim, Chol Kim,.
Identification ofKlf6-Related Super Enhancer in Human
Hepatoma (HepG2) Cells by CRISPR Technique.
(2017).Genetics and Molecular Research 16(4):
gmr16039841.
18. F tong et al., corilagin attenuates radiation
induced brain injury in mice, molecular neurobiology
vol53(10),2016,6982-6996
19. Chen qq et al., determination of corilagin in rat
plasma using a liquid chromatography-electrospray
ionization tandem mass spectrometric method, biomed
chromatogr, vol29(10),2015,1553-8
20. KumChol Ri, KumChol Kim, SunHyok Kong, JuHua
Ri,. The Disruption of Klf6-Related Super- Enhancer
Induces Growth Inhibition and Apoptosis in Human HepG2
Cells. (2018). Genetics and Molecular Research 17 (1):
gmr16039888.
... Herbs of the formulations such as Yashtimadhu, Pathyadi Kwath, Sanjeevani Vati, Samshamni Vati were reported to have antithrombotic activity. Additionally, compounds such as Glycyrrhizin, Corilagin, Withanoferin A also reported for antithrombotic activity (Ku & Bae, 2014;Lugun, 2018;Mendes-Silva, 2003;Ri & Ho Ju, 2018;Saleem, 2019). ...
Article
Full-text available
Herbal formulations mentioned in traditional medicinal texts were investigated for in silico effect against SARS-COV-2 proteins involved in various functions of a virus such as attachment, entry, replication, transcription, etc. To repurpose and validate polyherbal formulations, molecular docking was performed to study the interactions of more than 150 compounds from various formulations against the SARS-CoV-2 proteins. Molecular dynamics (MD) simulation was performed to evaluate the interaction of top scored ligands with the various receptor proteins. The docking results showed that Liquiritic acid, Liquorice acid, Terchebulin, Glabrolide, Casuarinin, Corilagin, Chebulagic acid, Neochebulinic acid, Daturataturin A, and Taraxerol were effective against SARS-COV-2 proteins with higher binding affinities with different proteins. Results of MD simulations validated the stability of ligands from potent formulations with various receptors of SARS-CoV-2. Binding free energy analysis suggested the favourable interactions of phytocompounds with the recpetors. Besides, in silico comparison of the various formulations determined that Pathyadi kwath, Sanjeevani vati, Yashtimadhu, Tribhuvan Keeratiras, and Septillin were more effective than Samshamni vati, AYUSH-64, and Trikatu. Polyherbal formulations having anti-COVID-19 potential can be used for the treatment with adequate monitoring. New formulations may also be developed for systematic trials based on ranking from these studies. Communicated by Ramaswamy H. Sarma.
Article
Full-text available
Klf6 is a member of the Krüppel-like factor family, which is a group of zinc finger transcription factors involved in differentiation and development. In general, Klf6 gene plays an important role in growth-related signal transduction pathways, cell proliferation, apoptosis, and angiogenesis. Recently, the Klf6 gene is considered as one of tumor suppressor genes in several kinds of tumours, but there is also a conflict of experimental data. In this study, we investigated the influence of Klf6-related super-enhancer (SE) on tumor cell growth in human hepatoma (HepG2) cells. As a result, SE-deleted clones were obtained via the CRISPR/Cas9-mediated gene-editing technique. Moreover, it was found that some crucial properties associated with malignancy in those clones were equally affected by the disruption of Klf6-related super-enhancer: proliferation, migration, clonogenic potential and drug resistance. These results contribute to the deeper understanding on the potential roles of the Klf6 related super enhancer in human hepatoma (HepG2) cells.
Article
Full-text available
The Klf6 gene is a tumor suppressor gene belonging to the family of the Klf gene and closely associated with tumor formation. Recently, super enhancers(SEs) have been shown to play a particularly important role in regulating cell identity and are attempting to evaluate the in vivo function of SEs. But direct functional evidence of SE associated with Klf6 is lacking. Using genomic editing technology, we have attempted to identify super enhancer associated with the expression of Klf6 and clarify its function in human hepatoma (HepG2) cells. As a result, it identified the Klf6 related SE and demonstrated that the Klf6-related SE is responsible for more than 80% of Klf6 gene expression. It also revealed the hierarchical structure of the Klf-6 related SE and the function of individual enhancers. Our results provide the functional significance of the super enhancer in understanding the transcriptional regulation mechanism of Klf6.
Data
Full-text available
Another clone has an extra g before the polyA tract.
Article
Full-text available
Cranial irradiation-induced inflammation plays a critical role in the initiation and progression of radiation-induced brain injury (RIBI). Anti-inflammation treatment may provide therapeutic benefits. Corilagin (beta-1-O-galloyl-3, 6-(R)-hexahydroxydiphenoyl-D-glucose, C27H22O18) was a novel member of the tannin family with anti-inflammatory properties and is isolated from some medicinal plants, such as Phyllanthus amarus and Caesalpinia coriaria. In this study, the effect of Corilagin on RIBI was investigated and the underlying mechanisms were explored. Spatial learning and memory ability of mice were investigated by the Morris water maze test. Evans blue leakage and electron microscopy were used to assess the integrity of blood-brain barrier (BBB). The mRNA and protein expressions of inflammatory cytokines, TNF-α and IL-1β, were measured by using real-time PCR and Western blotting. The activation of microglial cells and expression of TNF-α were examined by immunofluorescence staining. Phosphorylated signal transducers and activators of transcription 3 (p-STAT3) and IκBα, and the translocation of p65 from cytoplasm to nucleus were detected by using Western blotting. Morris water maze test showed that Corilagin ameliorated the neurocognitive deficits in RIBI mice. Evans blue leakage and electron microscopy exhibited that Corilagin partially protected the BBB integrity from cranial irradiation-caused damage; immunofluorescence staining showed that Corilagin could inhibit microglial activation and TNF-α expression. Real-time PCR and Western blotting revealed that Corilagin downregulated the expression of TNF-α and IL-1β and inhibited the irradiation-induced activation of NF-κB pathways by upregulating p-STAT3 expression. In conclusion, Corilagin could attenuate RIBI through inhibiting microglial activation and the expressions of inflammatory cytokines. Corilagin might inhibit the activation of NF-κB pathway in a STAT3-associated manner, thereby downregulating the inflammatory cytokine expressions.
Article
Full-text available
A novel low-molecular-weight inhibitor, AR-H029953XX, was developed from a known fibrinolytic compound, flufenamic acid, which prevented complex formation of human plasminogen activator inhibitor type 1 (PAI-1) with tissue plasminogen activator (tPA) by inhibition of PAI-1. To explore the binding site for AR-H029953XX, mutants of human PAI-1 were constructed by site-directed mutagenesis and were then expressed in CHO cells, purified, activated, and characterized. (1) PAI-1 with mutations in the reactive center loop: L1-PAI-1 (P10, Ser337Glu) had stability and activity similar to those of wild-type PAI-1 (wt-PAI-1), and L2-PAI-1 (P12, Ala335Glu) was highly stable but was a substrate for tPA. (2) PAI-1 with mutations near the binding epitope for the strongly inhibiting monoclonal antibody CLB-2C8: C1-PAI-1 (Phe114Glu), C2-PAI-1 (Val121Phe), C3-PAI-1 (Arg76Glu/Arg115Glu/Arg118Glu), and C4-PAI-1 (Arg115Glu) were all comparable in activity and stability to wt-PAI-1. AR-H029953XX (Ki = 25 microM) prevented complex formation between tPA and active wt-PAI-1 as well as that with mutants L1-, L2-, C1-, C2-, and C4-PAI-1. AR-H029953XX also inhibited binding of these PAI-1 variants to the antibody CLB-2C8, as measured by surface plasmon resonance. In contrast, AR-H029953XX had almost no inhibitory effect on the complex formation of tPA with C3-PAI-1. Moreover, AR-H029953XX had no effect on the binding rate of CLB-2C8 to C3-PAI-1, or on the binding to latent PAI-1 or to cleaved L2-PAI-1. The binding site of AR-H029953XX thus appears to be located in the neighborhood of the postulated epitope for CLB-2C8, near residues Arg76 and/or Arg118. This specific domain of the PAI-1 molecule might thus also be important for the mechanism of inhibitory activity toward tPA. Moreover, the structure of this region in active PAI-1 has to be different from the corresponding regions in latent and cleaved PAI-1.
Article
A sensitive and simple liquid chromatography-tandem mass spectrometric (HPLC-MS/MS) method for the determination of corilagin in rat plasma has been developed. Samples were prepared with protein precipitation method and analyzed with a triple quadrupole tandem mass spectrometer. We employed negative electrospray ionization as the ionization source and the analytes were detected in multiple reaction monitoring mode. Separation was achieved on a C8 column eluted with mobile phase consisting of methanol-0.1% formic acid in a gradient mode at the flow rate of 0.3 mL/min. The total run time was 7.0 min.This method was proved to have good linearity in the concentration range of 2.5-1000.0 ng/mL. The lower limit of quantification of corilagin was 2.5 ng/mL. The intra- and inter-day relative standard deviationa across three validation runs for four concentration levels were both <9.8%. The relative error was within ±6.0%. This assay offers advantages in terms of expediency and suitability for the analysis of corilagin in rat plasma. The practical utility of this new HPLC-MS/MS method was confirmed in pilot plasma concentration studies in rats following oral administration. Copyright © 2015 John Wiley & Sons, Ltd. Copyright © 2015 John Wiley & Sons, Ltd.
Article
The aim of this study was to investigate the anti-thrombotic effects of an inhibitor of the plasminogen activator inhibitor-1 (PAI-1) in rats given endotoxin. In studies in vitro, PRAP-1, a Fab-fragment of a polyclonal antibody against human PAI-1, was shown to inhibit PAI-1 activity in rat plasma as well as to stimulate clot-lysis of the euglobulin fraction derived from rat plasma. Endotoxin administered to anaesthetised rats produced a marked increase in plasma PAI-1 activity. To study fibrin formation and lysis in vivo after intravenous (i.v.) injection of the coagulant enzyme batroxobin, 125I-fibrinogen was administered to the animals. The thrombi formed by batroxobin were rapidly lysed in control animals, while the rate of lysis was markedly attenuated in rats given endotoxin. PRAP-1 was administered i.v. (bolus+infusion) to rats given endotoxin and batroxobin and the PAI-1 inhibitor caused a dose-dependent decrease in the 125I-fibrin deposition in the lungs. An immunohistochemical technique was used to confirm this decrease in density of fibrin clots in the tissue. Furthermore, PRAP-1 decreased plasma PAI-1 activity in the rats and this reduction was correlated to the decrease in lung 125I-fibrin deposition at the corresponding time point. It is concluded that in this experimental model the PAI-1 antibody PRAP-1 may indeed inhibit thrombosis in animals exposed to endotoxin.
Article
A critical component in the regulation of thrombus formation and clearance is the balance between tissue plasminogen activator (tPA) and plasminogen activator inhibitor type-1 (PAI-1). An increase in the plasma concentration of PAI-1 has been proposed as a risk factor in thrombotic disease. Inhibition of PAI-1 activity may have utility in the treatment of thromboembolic disease. We report here the evaluation of three diketopiperazine-based low molecular weight inhibitors of PAI-1 activity (XR334, XR1853 and XR5082). In vitro these compounds reversed the inhibitory effects of PAI-1 against both tPA and urokinase (UK) (IC50: 5 to 80 muM). In contrast, other serpin-serine protease interactions, including alpha 1-antitrypsin-trypsin, alpha 2-antiplasmin- plasmin and antithrombin-thrombin, were not affected, neither did these inhibitors affect global tests of haemostasis. In the light of this promising in vitro profile these compounds were evaluated in a standard radioisotopic assay of clot lysis in whole rat blood following intravenous administration. In this assay these compounds dose-dependently enhanced fibrinolysis ex vivo. After intravenous bolus administration XR334, XR1853 and XR5082 at 5 mg/kg increased clot lysis by 32.0 +/- 5.1% SEM (n = 25, p < 0.01), 36.7 +/- 3.5% SEM (n = 36, p < 0.01) and 60.0 +/- 2.8% SEM (n = 17, p < 0.01) respectively compared to vehicle. Intravenous infusion of these compounds (1 mg/kg/min for 20 min) significantly prolonged (approximately twofold) the time to blood vessel occlusion in the rat electrically-stimulated carotid artery thrombosis model. Thus, these low molecular weight inhibitors of PAI-1 activity enhanced fibrinolysis ex vivo and protected against thrombus formation in the rat.
Article
Impaired fibrinolytic function, mainly due to an elevation of plasma plasminogen activator inhibitor 1 concentration, is a common finding in patients with thrombotic disease. In patients subjected to hip surgery, preoperatively increased levels of plasminogen activator inhibitor 1 seem to be predictive of postoperative deep venous thrombosis. Several prospective studies of patients with angina pectoris or a past myocardial infarction have shown strong correlation between plasmatic plasminogen activator inhibitor 1 elevation and future cardiovascular events. However, before plasminogen activator inhibitor 1 can be regarded as a risk factor in the conventional epidemiologic sense, its relationship to myocardial infarction must be demonstrated in prospective studies of healthy populations. The regulation of plasminogen activator inhibitor 1 concentration in plasma is complex and at present not well understood. Multiple interactions with disturbances of both carbohydrate and lipoprotein metabolism are evident. Studies performed in cultured cells, transformed or natural, can be translated into human pathophysiology only with great caution. Both environmental and genetic factors seem to be of importance for the plasma plasminogen activator inhibitor 1 concentration. Platelet plasminogen activator inhibitor 1 seems to play a role in the resistance of platelet-rich thrombi to thrombolytic treatment.
Article
In acute coronary syndromes, marked alterations of coagulation and fibrinolysis have been observed, but no data are available concerning a possible relation to coronary stenosis morphology. Thirty one patients with unstable angina pectoris were included. Culprit stenosis morphology judged from coronary angiography was graded using the modified ACC/AHA classification. Molecular and functional markers of hemostasis and fibrinolysis were determined from venous plasma samples obtained at admission. Patients with unstable angina pectoris had a moderate procoagulant state, especially a contact phase activation compared with age-matched controls (factor XII 93.9 +/- 5.6 vs 112.8 +/- 5.4%; P < 0.05; high molecular weight kininogen 55.3 +/- 5.4 vs 86.1 +/- 6.5%; P < 0.01). Thrombin-antithrombin (TAT) was not significantly elevated (7.6 +/- 1.9 vs 4.0 +/- 0.5 microg/l). Elevated plasminogen activator mass concentration (16.6 +/- 2.1 vs 5.4 +/- 0.6 ng/ml; P < 0.01) and plasminogen activator inhibitor (PAI) activity (9.9 +/- 3.0 vs 5.6 +/- 3.0 AU/ml; P < 0.05) indicated an alteration of the fibrinolysis. Complexity of coronary stenosis was positively correlated with tissue-type plasminogen activator (TPA) mass concentration (P < 0.01) and PAI activity (P < 0.05). No association was found to markers of a hypercoagulative state. These findings indicate a relation between alterations of the fibrinolytic system and coronary morphology, whereas the acute changes of coagulation occur independently of culprit stenosis complexity.