1
|
Liu Y, Li R, Song L, Li K, Yu H, Xing R, Liu S, Li P. Intermediate molecular weight-fucosylated chondroitin sulfate from sea cucumber Cucumaria frondosa is a promising anticoagulant targeting intrinsic factor IXa. Int J Biol Macromol 2024; 269:131952. [PMID: 38692541 DOI: 10.1016/j.ijbiomac.2024.131952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 04/25/2024] [Accepted: 04/26/2024] [Indexed: 05/03/2024]
Abstract
Thromboembolic diseases pose a serious risk to human health worldwide. Fucosylated chondroitin sulfate (FCS) is reported to have good anticoagulant activity with a low bleeding risk. Molecular weight plays a significant role in the anticoagulant activity of FCS, and FCS smaller than octasaccharide in size has no anticoagulant activity. Therefore, identifying the best candidate for developing novel anticoagulant FCS drugs is crucial. Herein, native FCS was isolated from sea cucumber Cucumaria frondosa (FCScf) and depolymerized into a series of lower molecular weights (FCScfs). A comprehensive assessment of the in vitro anticoagulant activity and in vivo bleeding risk of FCScfs with different molecule weights demonstrated that 10 kDa FCScf (FCScf-10 K) had a greater intrinsic anticoagulant activity than low molecular weight heparin (LMWH) without any bleeding risk. Using molecular modeling combined with experimental validation, we revealed that FCScf-10 K can specifically inhibit the formation of the Xase complex by binding the negatively charged sulfate group of FCScf-10 K to the positively charged side chain of arginine residues on the specific surface of factor IXa. Thus, these data demonstrate that the intermediate molecular weight FCScf-10 K is a promising candidate for the development of novel anticoagulant drugs.
Collapse
Affiliation(s)
- Yuanjie Liu
- College of Chemical and Molecular Engineering, Qingdao University of Science and Technology, Qingdao 266042, China
| | - Rongfeng Li
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Center for Ocean Mega-Science, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China; Laboratory for Marine Drugs and Bioproducts, Qingdao National Laboratory for Marine Science and Technology, No. 1 Wenhai Road, Qingdao 266237, China.
| | - Lin Song
- College of Marine Science and Biological Engineering, Qingdao University of Science and Technology, Qingdao 266042, China.
| | - Kecheng Li
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Center for Ocean Mega-Science, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China; Laboratory for Marine Drugs and Bioproducts, Qingdao National Laboratory for Marine Science and Technology, No. 1 Wenhai Road, Qingdao 266237, China
| | - Huahua Yu
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Center for Ocean Mega-Science, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China; Laboratory for Marine Drugs and Bioproducts, Qingdao National Laboratory for Marine Science and Technology, No. 1 Wenhai Road, Qingdao 266237, China
| | - Ronge Xing
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Center for Ocean Mega-Science, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China; Laboratory for Marine Drugs and Bioproducts, Qingdao National Laboratory for Marine Science and Technology, No. 1 Wenhai Road, Qingdao 266237, China
| | - Song Liu
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Center for Ocean Mega-Science, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China; Laboratory for Marine Drugs and Bioproducts, Qingdao National Laboratory for Marine Science and Technology, No. 1 Wenhai Road, Qingdao 266237, China
| | - Pengcheng Li
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Center for Ocean Mega-Science, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China; Laboratory for Marine Drugs and Bioproducts, Qingdao National Laboratory for Marine Science and Technology, No. 1 Wenhai Road, Qingdao 266237, China
| |
Collapse
|
2
|
Huang J, Liang X, Zhao M, Zhang Y, Chen Z. Metabolomics and network pharmacology reveal the mechanism of antithrombotic effect of Asperosaponin VI. Biomed Pharmacother 2024; 173:116355. [PMID: 38493592 DOI: 10.1016/j.biopha.2024.116355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2023] [Revised: 02/23/2024] [Accepted: 02/26/2024] [Indexed: 03/19/2024] Open
Abstract
Dipsaci Radix may possess antithrombotic properties, and one of its primary active ingredients is Asperosaponin VI. However, the antithrombotic effects and pharmacological mechanisms of Asperosaponin VI remain unclear. An in vivo experimental study has demonstrated the antithrombotic activity of Asperosaponin VI. Asperosaponin VI also exhibits anticoagulant properties. Asperosaponin VI significantly hindered collagen adrenergic-induced acute pulmonary thrombosis in mice and enhanced their survival rate. This hinders the formation of acute pulmonary embolisms induced by adenosine diphosphate (ADP) and decreases recovery time. A comprehensive strategy that combines metabolomics, network pharmacology, molecular docking, and experimental validation has the potential to reveal the antithrombotic mechanisms of Asperosaponin VI. Metabolomic evidence suggests that Asperosaponin VI may influence platelet aggregation and the production of anti-inflammatory metabolites through the regulation of pathways such as phenylalanine and arachidonic acid metabolism, thereby inhibiting thrombosis. Network pharmacology identified the pharmacological targets of Asperosaponin VI and indicated that it treats thrombi by partially regulating the signaling pathways related to inflammation and platelet aggregation. Asperosaponin VI showed strong binding affinity for F2, PTPRC, JUN, STAT3, SRC, AKT1. The antiplatelet aggregation activity of Asperosaponin VI was validated based on the metabolomic and network pharmacology results. Asperosaponin VI inhibits platelet aggregation induced by ADP, AA, and collagen. Therefore, Asperosaponin VI exerts antithrombotic effects through antiplatelet aggregation. Therefore, Asperosaponin VI is a promising antithrombotic agent.
Collapse
Affiliation(s)
- Jin Huang
- Shenzhen Pingle Orthopedic Hospital (Shenzhen Pingshan Traditional Chinese Medicine Hospital), Shenzhen 518001, China
| | - Xuewen Liang
- Shenzhen Pingle Orthopedic Hospital (Shenzhen Pingshan Traditional Chinese Medicine Hospital), Shenzhen 518001, China
| | - Minrui Zhao
- Shenzhen Pingle Orthopedic Hospital (Shenzhen Pingshan Traditional Chinese Medicine Hospital), Shenzhen 518001, China
| | - Yue Zhang
- Shenzhen Pingle Orthopedic Hospital (Shenzhen Pingshan Traditional Chinese Medicine Hospital), Shenzhen 518001, China.
| | - Ziyang Chen
- Huizhou first Maternal and Child Health Care Hospital, Huizhou 516000, China.
| |
Collapse
|
3
|
Lee SH, Song SY, Choi JH, Kim S, Lee HJ, Park JW, Park DH, Bae CS, Cho SS. Partial Purification and Biochemical Evaluation of Protease Fraction (MA-1) from Mycoleptodonoides aitchisonii and Its Fibrinolytic Effect. Antioxidants (Basel) 2023; 12:1558. [PMID: 37627553 PMCID: PMC10451839 DOI: 10.3390/antiox12081558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 08/01/2023] [Accepted: 08/02/2023] [Indexed: 08/27/2023] Open
Abstract
The antioxidative proteolytic fraction, MA-1, was partially purified from Mycoleptodonoides aitchisonii. MA-1 was purified to homogeneity using a two-step procedure, which resulted in an 89-fold increase in specific activity and 42.5% recovery. SDS-PAGE revealed two proteins with a molecular weight of 48 kDa. The zymography results revealed proteolytic activity based on the MA-1 band. MA-1 was found to be stable in the presence of Na+, Ca2+, Fe3+, K+, and Mg2+. MA-1 was also stable in methanol, ethanol, and acetone, and its enzyme activity increased by 15% in SDS. MA-1 was inhibited by ethylenediaminetetra-acetic acid or ethylene glycol tetraacetic acid and exerted the highest specificity for the substrate, MeO-Suc-Arg-Pro-Tyr-pNA, for chymotrypsin. Accordingly, MA-1 belongs to the family of chymotrypsin-like metalloproteins. The optimum temperature was 40 °C and stability was stable in the range of 20 to 35 °C. The optimum pH and stability were pH 5.5 and pH 4-11. MA-1 exhibited stronger fibrinolytic activity than plasmin. MA-1 hydrolyzed the Aα, Bβ, and γ chains of fibrinogen within 2 h. MA-1 exhibited an antithrombotic effect in animal models. MA-1 was devoid of hemorrhagic activity at a dose of 80,000 U/kg. Overall, our results show that M. aitchisonii produces an acid-tolerant and antioxidative chymotrypsin-like fibrinolytic enzyme, and M. aitchisonii containing MA-1 could be a beneficial functional material for the prevention of cardiovascular diseases and possible complications.
Collapse
Affiliation(s)
- Sung-Ho Lee
- Department of Pharmacy, College of Pharmacy, Mokpo National University, Muan 58554, Republic of Korea; (S.-H.L.); (S.-Y.S.); (J.W.P.)
- Department of Biomedicine, Health & Life Convergence Sciences, BK21 Four, Biomedical and Healthcare Research Institute, Mokpo National University, Mokpo 58554, Republic of Korea
| | - Seung-Yub Song
- Department of Pharmacy, College of Pharmacy, Mokpo National University, Muan 58554, Republic of Korea; (S.-H.L.); (S.-Y.S.); (J.W.P.)
- Department of Biomedicine, Health & Life Convergence Sciences, BK21 Four, Biomedical and Healthcare Research Institute, Mokpo National University, Mokpo 58554, Republic of Korea
| | - Jun-Hui Choi
- Department of Food Science and Biotechnology, Gwangju University, Gwangju 61743, Republic of Korea; (J.-H.C.); (S.K.); (H.-J.L.)
| | - Seung Kim
- Department of Food Science and Biotechnology, Gwangju University, Gwangju 61743, Republic of Korea; (J.-H.C.); (S.K.); (H.-J.L.)
| | - Hyo-Jeong Lee
- Department of Food Science and Biotechnology, Gwangju University, Gwangju 61743, Republic of Korea; (J.-H.C.); (S.K.); (H.-J.L.)
| | - Jin Woo Park
- Department of Pharmacy, College of Pharmacy, Mokpo National University, Muan 58554, Republic of Korea; (S.-H.L.); (S.-Y.S.); (J.W.P.)
- Department of Biomedicine, Health & Life Convergence Sciences, BK21 Four, Biomedical and Healthcare Research Institute, Mokpo National University, Mokpo 58554, Republic of Korea
| | - Dae-Hun Park
- College of Oriental Medicine, Dongshin University, Naju-si 58245, Republic of Korea;
| | - Chun-Sik Bae
- College of Veterinary Medicine, Chonnam National University, 77 Yongbong-ro, Buk-gu, Gwangju 61186, Republic of Korea;
| | - Seung-Sik Cho
- Department of Pharmacy, College of Pharmacy, Mokpo National University, Muan 58554, Republic of Korea; (S.-H.L.); (S.-Y.S.); (J.W.P.)
- Department of Biomedicine, Health & Life Convergence Sciences, BK21 Four, Biomedical and Healthcare Research Institute, Mokpo National University, Mokpo 58554, Republic of Korea
| |
Collapse
|
4
|
Song Y, Liang Y, Zeng R, Li R, Zhou Y, Huang S, Li X, Zhang N, Xu M, Xiong K, Fu K, Ye H, Wu L, Yu S, Chen W, Tang C, Jiang M, Wang Z. Qualitative and quantitative analyses of chemical constituents in vitro and in vivo and systematic evaluation of the pharmacological effects of Tibetan medicine Zhixue Zhentong capsules. Front Pharmacol 2023; 14:1204947. [PMID: 37529700 PMCID: PMC10389267 DOI: 10.3389/fphar.2023.1204947] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Accepted: 06/12/2023] [Indexed: 08/03/2023] Open
Abstract
Introduction: Zhixue Zhentong capsules (ZXZTCs) are a Tibetan medicine preparation solely composed of Lamiophlomis rotata (Benth.) Kudo. L. rotata is the only species of the genus Laniophlomis (family Lamiaceae) that has medicinal constituents derived from the grass or root and rhizome. L. rotata is one of the most extensively used folk medicines by Tibetan, Mongolian, Naxi, and other ethnic groups in China and has been listed as a first-class endangered Tibetan medicine. The biological effects of the plant include hemostasis, analgesia, and the removal of blood stasis and swelling. Purpose: This study aimed to profile the overall metabolites of ZXZTCs and those entering the blood. Moreover, the contents of six metabolites were measured and the hemostatic, analgesic, and anti-inflammatory effects of ZXZTCs were explored. Methods: Ultra-performance liquid chromatography-tandem quadrupole time-of-flight high-resolution mass spectrometry (UPLC-Q-TOF-MS) was employed for qualitative analysis of the metabolites of ZXZTCs and those entering the blood. Six metabolites of ZXZTCs were quantitatively determined via high-performance liquid chromatography The hemostatic, analgesic, and anti-inflammatory effects of ZXZTCs were evaluated in various animal models. Results: A total of 36 metabolites of ZXZTCs were identified, including 13 iridoid glycosides, 9 flavonoids, 9 phenylethanol glycosides, 4 phenylpropanoids, and 1 other metabolite. Overall, 11 metabolites of ZXZTCs entered the blood of normal rats. Quantitative analysis of the six main metabolites, shanzhiside methyl ester, chlorogenic acid, 8-O-acetyl shanzhiside methyl ester, forsythin B, luteoloside, and verbascoside, was extensively performed. ZXZTCs exerted hemostatic effects by reducing platelet aggregation and thrombosis and shortening bleeding time. Additionally, ZXZTCs clearly had an analgesic effect, as observed through the prolongation of the latency of writhing, reduction in writhing, and increase in the pain threshold of experimental rats. Furthermore, significant anti-inflammatory effects of ZXZTCs were observed, including a reduction in capillary permeability, the inhibition of foot swelling, and a reduction in the proliferation of granulation tissue. Conclusion: Speculative identification of the overall metabolites of ZXZTCs and those entering the blood can provide a foundation for determining its biologically active constituents. The established method is simple and reproducible and can help improve the quality control level of ZXZTCs as a medicinal product. Evaluating the hemostatic, analgesic, and anti-inflammatory activities of ZXZTCs can help reveal its mechanism.
Collapse
Affiliation(s)
- Yinglian Song
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yan Liang
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Rong Zeng
- Chengdu Jiuzhitang Jinding Pharmaceutical Company Limited, Chengdu, China
| | - Ran Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- College of Ethnomedicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - You Zhou
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | | | - Xiaoli Li
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Ning Zhang
- Chengdu Jiuzhitang Jinding Pharmaceutical Company Limited, Chengdu, China
| | - Min Xu
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Kaipeng Xiong
- Chengdu Jiuzhitang Jinding Pharmaceutical Company Limited, Chengdu, China
| | - Ke Fu
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Huixuan Ye
- Jiuzhitang Company Limited, Changsha, China
| | - Lei Wu
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Shaopeng Yu
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Wanyue Chen
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Ce Tang
- College of Ethnomedicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Miao Jiang
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Zhang Wang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- College of Ethnomedicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
5
|
Liu R, Xu B, Ma Z, Ye H, Guan X, Ke Y, Xiang Z, Shi Q. Controlled release of nitric oxide for enhanced tumor drug delivery and reduction of thrombosis risk. RSC Adv 2022; 12:32355-32364. [PMID: 36425712 PMCID: PMC9650582 DOI: 10.1039/d2ra05438h] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 10/31/2022] [Indexed: 08/28/2023] Open
Abstract
Platelets activation and hypercoagulation induced by tumor cell-specific thrombotic secretions such as tissue factor (TF) and cancer procoagulant (CP), microparticles (MPs), and cytokines not only increase cancer-associated thrombosis but also accelerate cancer progress. In addition, the tumor heterogeneity such avascular areas, vascular occlusion and interstitial fluid pressure still challenges efficient drug delivery into tumor tissue. To overcome these adversities, we herein present an antiplatelet strategy based on a proteinic nanoparticles co-assembly of l-arginine (LA) and photosensitizer IR783 for local NO release to inhibit the activation of tumor-associated platelets and normalize angiogenesis, suppressing thrombosis and increasing tumoral accumulation of the nanoagent. In addition, NIR-controlled release localizes the NO spatiotemporally to tumor-associated platelets and prevents undesirable systemic bleeding substantially. Moreover, NO can transform to more cytotoxic peroxynitrite to destroy cancer cells. Our study describes an antiplatelet-directed cancer treatment, which represents a promising area of targeted cancer therapy.
Collapse
Affiliation(s)
- Rui Liu
- Department of VIP Unit, China-Japan Union Hospital of Jilin University Changchun Jilin 130022 China
| | - Baofeng Xu
- Stroke Center and Department of Neurology, First Hospital of Jilin University Changchun Jilin 130022 China
| | - Zhifang Ma
- State Key Laboratory of Polymer Physics and Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences Changchun Jilin 130022 China
| | - Hongbo Ye
- State Key Laboratory of Polymer Physics and Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences Changchun Jilin 130022 China
| | - Xinghua Guan
- State Key Laboratory of Polymer Physics and Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences Changchun Jilin 130022 China
| | - Yue Ke
- State Key Laboratory of Polymer Physics and Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences Changchun Jilin 130022 China
- University of Science and Technology of China Hefei Anhui 230026 China
| | - Zehong Xiang
- State Key Laboratory of Polymer Physics and Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences Changchun Jilin 130022 China
- University of Science and Technology of China Hefei Anhui 230026 China
| | - Qiang Shi
- State Key Laboratory of Polymer Physics and Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences Changchun Jilin 130022 China
- University of Science and Technology of China Hefei Anhui 230026 China
| |
Collapse
|
6
|
Sharma C, Osmolovskiy A, Singh R. Microbial Fibrinolytic Enzymes as Anti-Thrombotics: Production, Characterisation and Prodigious Biopharmaceutical Applications. Pharmaceutics 2021; 13:1880. [PMID: 34834294 PMCID: PMC8625737 DOI: 10.3390/pharmaceutics13111880] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 10/23/2021] [Accepted: 10/29/2021] [Indexed: 12/19/2022] Open
Abstract
Cardiac disorders such as acute myocardial infarction, embolism and stroke are primarily attributed to excessive fibrin accumulation in the blood vessels, usually consequential in thrombosis. Numerous methodologies including the use of anti-coagulants, anti-platelet drugs, surgical operations and fibrinolytic enzymes are employed for the dissolution of fibrin clots and hence ameliorate thrombosis. Microbial fibrinolytic enzymes have attracted much more attention in the management of cardiovascular disorders than typical anti-thrombotic strategies because of the undesirable after-effects and high expense of the latter. Fibrinolytic enzymes such as plasminogen activators and plasmin-like proteins hydrolyse thrombi with high efficacy with no significant after-effects and can be cost effectively produced on a large scale with a short generation time. However, the hunt for novel fibrinolytic enzymes necessitates complex purification stages, physiochemical and structural-functional attributes, which provide an insight into their mechanism of action. Besides, strain improvement and molecular technologies such as cloning, overexpression and the construction of genetically modified strains for the enhanced production of fibrinolytic enzymes significantly improve their thrombolytic potential. In addition, the unconventional applicability of some fibrinolytic enzymes paves their way for protein hydrolysis in addition to fibrin/thrombi, blood pressure regulation, anti-microbials, detergent additives for blood stain removal, preventing dental caries, anti-inflammatory and mucolytic expectorant agents. Therefore, this review article encompasses the production, biochemical/structure-function properties, thrombolytic potential and other surplus applications of microbial fibrinolytic enzymes.
Collapse
Affiliation(s)
- Chhavi Sharma
- Amity Institute of Microbial Technology, Amity University Uttar Pradesh, Noida 201313, India;
| | - Alexander Osmolovskiy
- Department of Microbiology, Faculty of Biology, Lomonosov Moscow State University, 119991 Moscow, Russia
| | - Rajni Singh
- Amity Institute of Microbial Technology, Amity University Uttar Pradesh, Noida 201313, India;
| |
Collapse
|
7
|
Diwan D, Usmani Z, Sharma M, Nelson JW, Thakur VK, Christie G, Molina G, Gupta VK. Thrombolytic Enzymes of Microbial Origin: A Review. Int J Mol Sci 2021; 22:10468. [PMID: 34638809 PMCID: PMC8508633 DOI: 10.3390/ijms221910468] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 09/23/2021] [Accepted: 09/24/2021] [Indexed: 01/10/2023] Open
Abstract
Enzyme therapies are attracting significant attention as thrombolytic drugs during the current scenario owing to their great affinity, specificity, catalytic activity, and stability. Among various sources, the application of microbial-derived thrombolytic and fibrinolytic enzymes to prevent and treat vascular occlusion is promising due to their advantageous cost-benefit ratio and large-scale production. Thrombotic complications such as stroke, myocardial infarction, pulmonary embolism, deep venous thrombosis, and peripheral occlusive diseases resulting from blood vessel blockage are the major cause of poor prognosis and mortality. Given the ability of microbial thrombolytic enzymes to dissolve blood clots and prevent any adverse effects, their use as a potential thrombolytic therapy has attracted great interest. A better understanding of the hemostasis and fibrinolytic system may aid in improving the efficacy and safety of this treatment approach over classical thrombolytic agents. Here, we concisely discuss the physiological mechanism of thrombus formation, thrombo-, and fibrinolysis, thrombolytic and fibrinolytic agents isolated from bacteria, fungi, and algae along with their mode of action and the potential application of microbial enzymes in thrombosis therapy.
Collapse
Affiliation(s)
- Deepti Diwan
- Department of Neurosurgery, Washington University School of Medicine, Saint Louis, MO 63110, USA; (D.D.); (J.W.N.)
| | - Zeba Usmani
- Department of Applied Biology, University of Science & Technology, Techno City, Killing Road, Baridua 9th Mile 793101, Meghalaya, India; (Z.U.); (M.S.)
| | - Minaxi Sharma
- Department of Applied Biology, University of Science & Technology, Techno City, Killing Road, Baridua 9th Mile 793101, Meghalaya, India; (Z.U.); (M.S.)
| | - James W. Nelson
- Department of Neurosurgery, Washington University School of Medicine, Saint Louis, MO 63110, USA; (D.D.); (J.W.N.)
| | - Vijay Kumar Thakur
- Biorefining and Advanced Materials Research Center, SRUC, Edinburgh EH9 3JG, UK;
- School of Engineering, University of Petroleum & Energy Studies (UPES), Dehradun 248007, Uttarakhand, India
| | - Graham Christie
- Department of Chemical Engineering & Biotechnology, University of Cambridge, Cambridge CB2 1TN, UK;
| | - Gustavo Molina
- Laboratory of Bioflavors and Bioactive Compounds, Department of Food Science, Faculty of Food Engineering, State University of Campinas, R. Monteiro Lobato, 80, Campinas, São Paulo 13083-862, Brazil;
| | - Vijai Kumar Gupta
- Biorefining and Advanced Materials Research Center, SRUC, Edinburgh EH9 3JG, UK;
- Centre for Safe and Improved Food, SRUC, Edinburgh EH9 3JG, UK
| |
Collapse
|
8
|
Huang J, Song W, Hua H, Yin X, Huang F, Alolga RN. Antithrombotic and anticoagulant effects of a novel protein isolated from the venom of the Deinagkistrodon acutus snake. Biomed Pharmacother 2021; 138:111527. [PMID: 33773469 DOI: 10.1016/j.biopha.2021.111527] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2021] [Revised: 03/16/2021] [Accepted: 03/17/2021] [Indexed: 12/19/2022] Open
Abstract
The venom of the Deinagkistrodon acutus snake is composed of numerous bioactive proteins and peptides. In this study, we report the antithrombotic and anticoagulant activities of one of such proteins, herein known as SLPC. This novel protein was isolated and purified via multi-gel chromatography. Its amino acid sequence, structure and function were then determined. This protein was found to exhibit defibration, anticoagulation and general antithrombotic effects based on the results of both in vitro and in vivo studies. Based on same studies, it was found to cleave the α, β, γ chains of fibrinogen and generally improved antiplatelet aggregation and blood rheology. A metabolomic insight of the antithrombotic effects of SLPC was found to be mainly linked to perturbations in the synthesis of unsaturated fatty acids, glycerophospholipid metabolism, arachidonic acid metabolism and other metabolic pathways. In summary, the novel protein SLPC, elicits its antithrombotic effects via degradation of fibrinogen and regulation of various thrombogenic factors in multiple metabolic pathways.
Collapse
Affiliation(s)
- Jin Huang
- Shenzhen Pingle Orthopedic Hospital (Shenzhen Pingshan Traditional Chinese Medicine Hospital), Shenzhen 518001, China
| | - Wei Song
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Haibing Hua
- The Affiliated Jiangyin Hospital of Nanjing University of Chinese Medicine, Jiangyin 214400, China
| | - Xiaojian Yin
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China.
| | - Fang Huang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China.
| | - Raphael N Alolga
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China.
| |
Collapse
|
9
|
Zaman KAU, Park JH, DeVine L, Hu Z, Wu X, Kim HS, Cao S. Secondary Metabolites from the Leather Coral-Derived Fungal Strain Xylaria sp. FM1005 and Their Glycoprotein IIb/IIIa Inhibitory Activity. JOURNAL OF NATURAL PRODUCTS 2021; 84:466-473. [PMID: 33491454 DOI: 10.1021/acs.jnatprod.0c01330] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Five new tyrosine derivatives (1-5), one new phenylacetic acid derivative (6), two new quinazolinone analogues (7 and 8), one new naphthalenedicarboxylic acid (9), and one new 3,4-dihydroisocoumarin derivative (10), together with seven known compounds, were isolated from the fungus Xylaria sp. FM1005, which was isolated from Sinularia densa (leather coral) collected in the offshore region of the Big Island, Hawaii. The structures of compounds 1-10 were elucidated by extensive analysis of NMR spectroscopy, HRESIMS, and ECD data. Due to their structure similarity to the antiplatelet drug tirofiban, compounds 1-5 together with 6 were investigated for their antithrombotic activities. Compounds 1 and 2 strongly inhibited the binding of fibrinogen to purified integrin IIIb/IIa in a dose-dependent manner with the IC50 values of 0.89 and 0.61 μM, respectively, and compounds 1 and 2 did not show any cytotoxicity against A2780 and HEK 293 at 40 μM.
Collapse
Affiliation(s)
- Kh Ahammad Uz Zaman
- Department of Pharmaceutical Sciences, Daniel K. Inouye College of Pharmacy, University of Hawai'i at Hilo, Hilo, Hawaii 96720, United States
| | - Jae Hyeon Park
- School of Pharmacy, Sungkyunkwan University, Gyeonggi-do 16419, Republic of Korea
| | - Lela DeVine
- Department of Pharmaceutical Sciences, Daniel K. Inouye College of Pharmacy, University of Hawai'i at Hilo, Hilo, Hawaii 96720, United States
| | - Zhenquan Hu
- Warshel Institute for Computational Biology, The Chinese University of Hong Kong, Shenzhen 518172, People's Republic of China
- School of Chemistry and Materials Science, University of Science and Technology of China, Hefei, Anhui 230052, People's Republic of China
| | - Xiaohua Wu
- Department of Pharmaceutical Sciences, Daniel K. Inouye College of Pharmacy, University of Hawai'i at Hilo, Hilo, Hawaii 96720, United States
| | - Hyung Sik Kim
- School of Pharmacy, Sungkyunkwan University, Gyeonggi-do 16419, Republic of Korea
| | - Shugeng Cao
- Department of Pharmaceutical Sciences, Daniel K. Inouye College of Pharmacy, University of Hawai'i at Hilo, Hilo, Hawaii 96720, United States
| |
Collapse
|
10
|
Kim ES, Lee JS, Lee HG. Improvement of antithrombotic activity of red ginseng extract by nanoencapsulation using chitosan and antithrombotic cross-linkers: polyglutamic acid and fucodian. J Ginseng Res 2020; 45:236-245. [PMID: 33841004 PMCID: PMC8020352 DOI: 10.1016/j.jgr.2020.04.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2019] [Revised: 04/01/2020] [Accepted: 04/03/2020] [Indexed: 10/25/2022] Open
Abstract
Background Red ginseng (RG) extract, especially ginsenoside Rg1 and Rb1 fractions has been reported to have antithrombotic activities. However, gastric instability and low intestinal permeability are considered to be obstacles to its oral administration. We hypothesized that stability, permeability, and activities of RG might be improved by encapsulation within nanoparticles (NPs) prepared with antithrombotic coating materials. Methods RG-loaded chitosan (CS) NPs (PF-NPs) were prepared by complex ionic gelation with the antithrombotic wall materials, polyglutamic acid (PGA), and fucoidan (Fu). The concentrations of PGA (mg/mL, X1) and Fu (mg/mL, X2) were optimized for the smallest particle size by response surface methodology. Antithrombotic activities of RG and PF-NPs were analyzed using ex vivo and in vivo antiplatelet activities, in vivo carrageenan-induced mouse tail, and arteriovenous shunt rat thrombosis models. Results In accordance with a quadratic regression model, the smallest PF-NPs (286 ± 36.6 nm) were fabricated at 0.628 mg/mL PGA and 0.081 mg/mL Fu. The inhibitory activities of RG on ex vivo and in vivo platelet aggregation and thrombosis in in vivo arteriovenous shunt significantly (p < 0.05) increased to approximately 66.82%, 35.42%, and 38.95%, respectively, by encapsulation within PF-NPs. For an in vivo carrageenan-induced mouse tail thrombosis model, though RG had a weaker inhibitory effect, PF-NPs reduced thrombus significantly due to the presence of PGA and Fu. Conclusion PF-NPs contributed to improve the activities of RG not only by nanoencapsulation but also by antithrombotic coating materials. Therefore, PG-NPs can be suggested as an efficient delivery system for oral administration of RG.
Collapse
Affiliation(s)
- Eun Suh Kim
- Department of Food and Nutrition, Hanyang University, 222, Wangsimni-ro, Seongdong-gu, Seoul 04763, Republic of Korea
| | - Ji-Soo Lee
- Department of Food and Nutrition, Hanyang University, 222, Wangsimni-ro, Seongdong-gu, Seoul 04763, Republic of Korea
| | - Hyeon Gyu Lee
- Department of Food and Nutrition, Hanyang University, 222, Wangsimni-ro, Seongdong-gu, Seoul 04763, Republic of Korea
| |
Collapse
|
11
|
Ma N, Yang Y, Liu X, Li S, Qin Z, Li J. Plasma metabonomics and proteomics studies on the anti-thrombosis mechanism of aspirin eugenol ester in rat tail thrombosis model. J Proteomics 2019; 215:103631. [PMID: 31891783 DOI: 10.1016/j.jprot.2019.103631] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Revised: 12/10/2019] [Accepted: 12/27/2019] [Indexed: 01/09/2023]
Abstract
Aspirin eugenol eater (AEE), a new drug compound, was synthesized through the combination of aspirin and eugenol. Antithrombotic effects of AEE have been confirmed in carrageenan-induced rat tail thrombosis model. However, its mechanism is unclear. With the application of integrated approach combining proteomics and metabolomics, the profilings of protein and metabolite in plasma were examined in thrombosis rat pretreated with AEE, aspirin and eugenol, respectively. A clear separation of the plasma metabolic profiles from different groups was found in score plots. 15 metabolites related with the metabolism of fatty acid, energy and amino acid were found. A total of 144, 38, 41 and 54 differentially abundant proteins (DAPs) were identified in control, AEE, aspirin and eugenol group, respectively. Proteomic results showed that aspirin modulated 7 proteins in amino acid metabolism and 4 proteins in complement system; eugenol regulated the 8 proteins related with coagulation cascades and fibrinogen; AEE improved 3 proteins in TCA cycle and 3 in lipid metabolism. Integrated analysis suggested that AEE improved fatty acid, energy and lipid metabolism to against thrombosis. Results of this study indicated AEE had different action mechanism on thrombosis from aspirin and eugenol, and contribute to understanding the mechanisms of AEE on thrombosis. SIGNIFICANCE: Thrombosis is a threat to human health, and there is an urgent need for new drug. In this study, compared with the model group, plasma metabolic profiles in AEE-treated rats were clearly separated; 15 metabolites and 38 proteins were picked out. These metabolites and proteins may assist in understanding the action mechanism of AEE on thrombosis. The results of plasma metabonomics and proteomics also revealed the different action mechanism among AEE, aspirin and eugenol on thrombosis. This study established the foundation to further evaluate the druggability of AEE on thrombosis treatment.
Collapse
Affiliation(s)
- Ning Ma
- Key Lab of New Animal Drug Project of Gansu Province, Key Lab of Veterinary Pharmaceutical Development, Ministry of Agriculture, Lanzhou Institute of Husbandry and Pharmaceutical Science of Chinese Academy of Agricultural Sciences, Lanzhou 730050, PR China; College of Veterinary Medicine, Agricultural University of Hebei, Baoding, Hebei 071000, PR China
| | - Yajun Yang
- Key Lab of New Animal Drug Project of Gansu Province, Key Lab of Veterinary Pharmaceutical Development, Ministry of Agriculture, Lanzhou Institute of Husbandry and Pharmaceutical Science of Chinese Academy of Agricultural Sciences, Lanzhou 730050, PR China
| | - Xiwang Liu
- Key Lab of New Animal Drug Project of Gansu Province, Key Lab of Veterinary Pharmaceutical Development, Ministry of Agriculture, Lanzhou Institute of Husbandry and Pharmaceutical Science of Chinese Academy of Agricultural Sciences, Lanzhou 730050, PR China
| | - Shihong Li
- Key Lab of New Animal Drug Project of Gansu Province, Key Lab of Veterinary Pharmaceutical Development, Ministry of Agriculture, Lanzhou Institute of Husbandry and Pharmaceutical Science of Chinese Academy of Agricultural Sciences, Lanzhou 730050, PR China
| | - Zhe Qin
- Key Lab of New Animal Drug Project of Gansu Province, Key Lab of Veterinary Pharmaceutical Development, Ministry of Agriculture, Lanzhou Institute of Husbandry and Pharmaceutical Science of Chinese Academy of Agricultural Sciences, Lanzhou 730050, PR China
| | - Jianyong Li
- Key Lab of New Animal Drug Project of Gansu Province, Key Lab of Veterinary Pharmaceutical Development, Ministry of Agriculture, Lanzhou Institute of Husbandry and Pharmaceutical Science of Chinese Academy of Agricultural Sciences, Lanzhou 730050, PR China.
| |
Collapse
|
12
|
Gogoi D, Ramani S, Bhartari S, Chattopadhyay P, Mukherjee AK. Characterization of active anticoagulant fraction and a fibrin(ogen)olytic serine protease from leaves of Clerodendrum colebrookianum, a traditional ethno-medicinal plant used to reduce hypertension. JOURNAL OF ETHNOPHARMACOLOGY 2019; 243:112099. [PMID: 31326559 DOI: 10.1016/j.jep.2019.112099] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Revised: 05/21/2019] [Accepted: 07/16/2019] [Indexed: 06/10/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Cardiovascular diseases are the major cause of mortality and morbidity, causing over 17.9 million deaths a year worldwide. Currently used therapy is often having side effects and expensive, dietary interventions and alternative medicines are required. Clerodendrum colebrookianum has been used to treat cardiac hypertension but anticoagulant potency was not evaluated. AIM OF THE STUDY To characterize an active anticoagulant fraction (AAFCC) and a 30 kDa fibrin(ogen)olytic serine protease (clerofibrase) isolated from aqueous leave extract of C. colebrookianum. MATERIALS AND METHODS AAFCC/clerofibrase was subjected to extensive biochemical and pharmacological characterization including LC-MS/MS, amino acid compositional and GC-MS analyses. Interaction between clerofibrase with fibrinogen was studied by spectrofluorometric analysis. In vitro thrombolytic, antiplatelet and cytotoxicity assay were performed. In vivo toxicity, anticoagulant, defibrinogen and antithrombotic activities were determined on Swiss albino mice. RESULTS The in vitro anticoagulant activity of AAFCC was found to be superior to heparin and clerofibrase and comparable to Nattokinase and warfarin. The proteomics and amino acid composition analyses suggest that clerofibrase is a previously uncharacterized novel plant protease capable of degrading the -αβ chains of fibrinogen/fibrin. AAFCC/clerofibrase exerts their anticoagulant action via fibrinogenolytic activity and partially by antiplatelet activity albeit they have no effect on thrombin and FXa inhibition. The spectrofluorometric analysis revealed the binding of clerofibrase to fibrinogen but not to thrombin and FXa. The phytochemical constituents and bioactive components of AAFCC were characterized by biochemical, and GC-MS analyses. The AAFCC and clerofibrase inhibited collagen/ADP-induced mammalian platelet aggregation, showed in vitro thrombolytic activity, and non-cytotoxic to mammalian cells. The AAFCC showed and dose-dependent in vivo plasma defibrinogenating and anticoagulant activities and inhibited k-carrageen-induced thrombus formation in the tails of mice. CONCLUSION The potent in vivo anticoagulant and antithrombotic effects of AAFCC suggests its pharmacological significance as herbal anticoagulant drug for the prevention and/or treatment of hyperfibrinogenemia- and thrombosis associated cardiovascular disorders.
Collapse
Affiliation(s)
- Debananda Gogoi
- Microbial Biotechnology and Protein Research Laboratory, Department of Molecular Biology and Biotechnology, School of Sciences, Tezpur University, Tezpur, 784028, Assam, India
| | - Sheetal Ramani
- Microbial Biotechnology and Protein Research Laboratory, Department of Molecular Biology and Biotechnology, School of Sciences, Tezpur University, Tezpur, 784028, Assam, India
| | - Surbhi Bhartari
- Microbial Biotechnology and Protein Research Laboratory, Department of Molecular Biology and Biotechnology, School of Sciences, Tezpur University, Tezpur, 784028, Assam, India
| | - Pronobesh Chattopadhyay
- Division of Pharmaceutical Technology, Defense Research Laboratory, Tezpur, 784501, Assam, India
| | - Ashis K Mukherjee
- Microbial Biotechnology and Protein Research Laboratory, Department of Molecular Biology and Biotechnology, School of Sciences, Tezpur University, Tezpur, 784028, Assam, India.
| |
Collapse
|
13
|
Huang J, Fan H, Yin X, Huang F. Isolation of a Novel Metalloproteinase from Agkistrodon Venom and Its Antithrombotic Activity Analysis. Int J Mol Sci 2019; 20:E4088. [PMID: 31438579 PMCID: PMC6747553 DOI: 10.3390/ijms20174088] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 08/15/2019] [Accepted: 08/18/2019] [Indexed: 12/12/2022] Open
Abstract
Snake venom contains large amounts of active proteins and peptides. In this study, a novel snake protein, metalloproteinase SP, was successfully isolated from the venom of Agkistrodon acutus by multi-gel chromatography. The isolated protein exhibits anti-platelet aggregation activity. Animal experiments showed that it exhibited defibration, anticoagulation, and antithrombotic effects and contributes to improved blood rheology and antiplatelet aggregation. In vivo experiments demonstrated that it prolonged clotting time, partial thromboplastin time, prothrombin time, thrombin time, fibrinogen time and reduced fibrinogen content of mice. Also, metalloproteinase SP inhibited carrageenan-induced tail thrombosis, ADP-induced acute pulmonary embolism, and ADP, Arachidonic acid (AA), or collagen-induced platelet aggregation. In vitro experiments showed that the protein cleaved the α, β, and γ chains of fibrinogen. Metabolomic analysis upon metalloproteinase SP treatment revealed that 14 metabolites, which are mainly involved in phenylalanine, tyrosine, and tryptophan biosynthesis, responded to metalloproteinase SP treatment. In summary, the isolated snake venom protein inhibits formation of acute pulmonary embolism probably through regulating and restoring perturbed energy, lipid, and amino acid metabolism.
Collapse
Affiliation(s)
- Jin Huang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Hui Fan
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Xiaojian Yin
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China.
- Chinese Medicine College, China Pharmaceutical University, Nanjing 210009, China.
| | - Fang Huang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China.
- Chinese Medicine College, China Pharmaceutical University, Nanjing 210009, China.
| |
Collapse
|
14
|
Li H, Liu B, Wu J, Yu H, Huang H, Chen X, Chen B, Wu S, Ma J, Liu W, Chen X, Lan L, He Z, Zhang H. The inhibitory effect of tachyplesin I on thrombosis and its mechanisms. Chem Biol Drug Des 2019; 94:1672-1679. [PMID: 31108023 DOI: 10.1111/cbdd.13570] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Revised: 05/07/2019] [Accepted: 05/13/2019] [Indexed: 12/31/2022]
Abstract
Thrombotic diseases are major cause of cardiovascular diseases. This study was designed to investigate the effect of tachyplesin I on platelet aggregation and thrombosis. Platelet aggregation was analysed with a whole blood aggregometer. The mice were employed to investigate the effect of tachyplesin I on thrombosis in vivo. Tachyplesin I inhibited thrombin-induced platelet aggregation in a dose-dependent manner. Furthermore, tachyplesin I significantly reduced thrombosis in carrageenan-induced tail thrombosis model by intraperitoneal injection (0.1, 0.2 or 0.4 mg/kg) or intragastric administration (15, 30 or 60 mg/kg). Tachyplesin I also prolonged the bleeding time (BT) and clotting time (CT). The results revealed that tachyplesin I inhibited platelet aggregation and thrombosis by interfering the PI3K/AKT pathway. Tachyplesin I did not show significantly toxicity to mice under 300 mg/kg via intravenous injection. The results show that tachyplesin I inhibits thrombosis and has low toxicity. It is suggested that tachyplesin I has the potential to develop a new anti-thrombotic drug.
Collapse
Affiliation(s)
- Huimin Li
- Department of Biochemistry and Molecular Biology, Guangdong Medical University, Zhanjiang, China.,Guangdong Key Laboratory for Research and Development of Natural Drugs, Guangdong Medical University, Zhanjiang, China
| | - Bin Liu
- Department of Biochemistry and Molecular Biology, Guangdong Medical University, Zhanjiang, China
| | - Jun Wu
- Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Huajun Yu
- Department of Biochemistry and Molecular Biology, Guangdong Medical University, Zhanjiang, China.,Guangdong Key Laboratory for Research and Development of Natural Drugs, Guangdong Medical University, Zhanjiang, China
| | - Hui Huang
- Department of Biochemistry and Molecular Biology, Guangdong Medical University, Zhanjiang, China
| | - Xi Chen
- Department of Biochemistry and Molecular Biology, Guangdong Medical University, Zhanjiang, China
| | - Baoan Chen
- Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Shang Wu
- Department of Biochemistry and Molecular Biology, Guangdong Medical University, Zhanjiang, China
| | - Jingyao Ma
- Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Wen Liu
- Department of Biochemistry and Molecular Biology, Guangdong Medical University, Zhanjiang, China
| | - Xiaoyi Chen
- Department of Biochemistry and Molecular Biology, Guangdong Medical University, Zhanjiang, China
| | - Liubo Lan
- Department of Biochemistry and Molecular Biology, Guangdong Medical University, Zhanjiang, China
| | - Zhan He
- Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Haitao Zhang
- Department of Biochemistry and Molecular Biology, Guangdong Medical University, Zhanjiang, China.,Guangdong Key Laboratory for Research and Development of Natural Drugs, Guangdong Medical University, Zhanjiang, China
| |
Collapse
|
15
|
Methods available to assess therapeutic potential of fibrinolytic enzymes of microbial origin: a review. J Anal Sci Technol 2018. [DOI: 10.1186/s40543-018-0143-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
|
16
|
Production of Fibrinolytic Enzyme by the Marine Isolate Serratia marcescens subsp. sakuensis and its In-vitro Anticoagulant and Thrombolytic Potential. JOURNAL OF PURE AND APPLIED MICROBIOLOGY 2017. [DOI: 10.22207/jpam.11.4.41] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|
17
|
Choi JH, Kim JE, Kim S, Yoon J, Park DH, Shin HJ, Lee HJ, Cho SS. Purification and partial characterization of a low molecular fibrinolytic serine metalloprotease C142 from the culture supernatant of Bacillus subtilis C142. Int J Biol Macromol 2017; 104:724-731. [PMID: 28600208 DOI: 10.1016/j.ijbiomac.2017.06.025] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Revised: 06/01/2017] [Accepted: 06/05/2017] [Indexed: 11/20/2022]
Abstract
Novel serine metalloprotease-like enzyme, C142 was purified from the culture supernatant of Bacillus subtilis C142. The C142 was purified to homogeneity by a two-step procedure with a 20.7-fold increase in specific activity and 0.9% recovery. The molecular mass of C142 was approximately 23.5kDa based on SDS-PAGE. The N-terminal amino acid sequence of the first 21 amino acids of C142 was AQSVPYGISQIKAPALHSQGY. Its optimum pH, optimum temperature, pH stability, and thermal stability were pH 6, 40°C, pH 6-8, and 20-35°C, respectively. C142 was strongly inhibited by PMSF and EGTA, suggesting that C142 was a serine metalloprotease-like enzyme. C142 showed the highest specificity toward the substrate for t-PA. The apparent Km, Vmax, and Kcat values of C142 toward H-d-Ile-Pro-Arg-pNA were determined as 0.34mM, 0.25mmolmg-1min-1, and 46.83s-1. C142 exhibited fibrinolytic activity, which is stronger than that of plasmin. C142 hydrolyzed Aα, and Bβ-chains of fibrinogen, but did not cleave γ-chains. C142 had antithrombotic effect in three animal models. C142 was devoid of hemorrhagic activity at a dose of 20,000FU/kg. Taken together, our results indicate that B. subtilis C142 produces a serine metalloprotease-like enzyme/fibrinolytic enzyme and this enzyme might be used as a new thrombolytic agent.
Collapse
Affiliation(s)
- Jun-Hui Choi
- Department of Food Science and Biotechnology, Gwangju University, Gwangju, 61743, Republic of Korea
| | - Jung-Eun Kim
- Department of Pharmacy, College of Pharmacy, Mokpo National University, Muan, Jeonnam 58554, Republic of Korea
| | - Seung Kim
- Department of Food Science and Biotechnology, Gwangju University, Gwangju, 61743, Republic of Korea
| | - Jaewoo Yoon
- College of Pharmacy, Keimyung University, 1095 Dalgubeoldaero, Dalseo-Gu, Daegu, 42601, Republic of Korea
| | - Dae-Hun Park
- Department of Nursing, Dongshin University, Naju, Jeonnam 58245, Republic of Korea
| | - Hyun-Jae Shin
- Department of Biochemical and Polymer Engineering, Chosun University, Gwangju, 61452, Republic of Korea
| | - Hyo-Jeong Lee
- Department of Food Science and Biotechnology, Gwangju University, Gwangju, 61743, Republic of Korea.
| | - Seung-Sik Cho
- Department of Pharmacy, College of Pharmacy, Mokpo National University, Muan, Jeonnam 58554, Republic of Korea.
| |
Collapse
|
18
|
Lee IJ, Yang YC, Hsu JW, Chang WT, Chuang YJ, Liau I. Zebrafish model of photochemical thrombosis for translational research and thrombolytic screening in vivo. JOURNAL OF BIOPHOTONICS 2017; 10:494-502. [PMID: 27174426 DOI: 10.1002/jbio.201500287] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/05/2015] [Revised: 04/22/2016] [Accepted: 04/22/2016] [Indexed: 06/05/2023]
Abstract
Acute thromboembolic diseases remain the major global cause of death or disability. Although an array of thrombolytic and antithrombotic drugs has been approved to treat or prevent thromboembolic diseases, many more drugs that target specific clotting mechanisms are under development. Here a novel zebrafish model of photochemical thrombosis is reported and its prospective application for the screening and preclinical testing of thrombolytic agents in vivo is demonstrated. Through photochemical excitation, a thrombus was induced to form at a selected section of the dorsal aorta of larval zebrafish, which had been injected with photosensitizers. Such photochemical thrombosis can be consistently controlled to occlude partially or completely the targeted blood vessel. Detailed mechanistic tests indicate that the zebrafish model of photochemical thrombosis exhibits essential features of classical coagulation and a thrombolytic pathway. For demonstration, tissue plasminogen activator (tPA), a clinically feasible thrombolytic agent, was shown to effectively dissolve photochemically induced blood clots. In light of the numerous unique advantages of zebrafish as a model organism, our approach is expected to benefit not only the development of novel thrombolytic and antithrombotic strategies but also the fundamental or translational research targeting hereditary thrombotic or coagulation disorders.
Collapse
Affiliation(s)
- I-Ju Lee
- Department of Applied Chemistry and Institute of Molecular Science, National Chiao Tung University, Hsinchu, 300, Taiwan
| | - Yi-Cyun Yang
- Department of Applied Chemistry and Institute of Molecular Science, National Chiao Tung University, Hsinchu, 300, Taiwan
| | - Jia-Wen Hsu
- Department of Applied Chemistry and Institute of Molecular Science, National Chiao Tung University, Hsinchu, 300, Taiwan
| | - Wei-Tien Chang
- Department of Emergency Medicine and Cardiovascular Center, National Taiwan University Hospital and College of Medicine, Taipei, 100, Taiwan
| | - Yung-Jen Chuang
- Department of Medical Science and Institute of Bioinformatics and Structural Biology, National Tsing Hua University, Hsinchu, 300, Taiwan
| | - Ian Liau
- Department of Applied Chemistry and Institute of Molecular Science, National Chiao Tung University, Hsinchu, 300, Taiwan
| |
Collapse
|
19
|
Hu M, Zhang J, Ding R, Fu Y, Gong T, Zhang Z. Improved oral bioavailability and therapeutic efficacy of dabigatran etexilate via Soluplus-TPGS binary mixed micelles system. Drug Dev Ind Pharm 2017; 43:687-697. [PMID: 28032534 DOI: 10.1080/03639045.2016.1278015] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
The clinical use of dabigatran etexilate (DABE) is limited by its poor absorption and relatively low bioavailability. Our study aimed to explore the potential of a mixed micelle system composed of Soluplus® and D-alpha tocopheryl polyethylene glycol 1000 succinate (TPGS) to improve the oral absorption and bioavailability of DBAE. DBAE was first encapsulated into Soluplus/TPGS mixed micelles by a simple thin film hydration method. The DBAE loaded micelles displayed an average size distribution of around 83.13 nm. The cellular uptake of DBAE loaded micelles in Caco-2 cell monolayer was significantly enhanced by 2-2.6 fold over time as compared with DBAE suspension. Both lipid raft/caveolae and macropinocytosis-mediated the cell uptake of DBAE loaded micelles through P-glycoprotein (P-gp)-independent pathway. Compared with the DBAE suspension, the intestinal absorption of DBAE from DBAE mixed micelles in rats was significantly improved by 8 and 5-fold in ileum at 2 h and 4 h, respectively. Moreover, DBAE mixed micelles were absorbed into systemic circulation via both portal vein and lymphatic pathway. The oral bioavailability of DBAE mixed micelles in rats was 3.37 fold higher than that of DBAE suspension. DBAE mixed micelles exhibited a comparable anti-thrombolytic activity with a thrombosis inhibition rate of 63.18% compared with DBAE suspension in vivo. Thus, our study provides a promising drug delivery system to enhance the oral bioavailability and therapeutic efficacy of DBAE.
Collapse
Affiliation(s)
- Mei Hu
- a Key Laboratory of Drug Targeting and Drug Delivery Systems , Ministry of Education, Sichuan University , Chengdu , China
| | - Jinjie Zhang
- a Key Laboratory of Drug Targeting and Drug Delivery Systems , Ministry of Education, Sichuan University , Chengdu , China
| | - Rui Ding
- b Beijing Institute for Drug Control , Beijing , China
| | - Yao Fu
- a Key Laboratory of Drug Targeting and Drug Delivery Systems , Ministry of Education, Sichuan University , Chengdu , China
| | - Tao Gong
- a Key Laboratory of Drug Targeting and Drug Delivery Systems , Ministry of Education, Sichuan University , Chengdu , China
| | - Zhirong Zhang
- a Key Laboratory of Drug Targeting and Drug Delivery Systems , Ministry of Education, Sichuan University , Chengdu , China
| |
Collapse
|
20
|
Kim ES, Lee JS, Lee HG. Nanoencapsulation of Red Ginseng Extracts Using Chitosan with Polyglutamic Acid or Fucoidan for Improving Antithrombotic Activities. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2016; 64:4765-4771. [PMID: 27181678 DOI: 10.1021/acs.jafc.6b00911] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
The potential of nanoencapsulation using bioactive coating materials for improving antithrombotic activities of red ginseng extract (RG) was examined. RG-loaded chitosan (CS) nanoparticles were prepared using antithrombotic materials, polyglutamic acid (PGA) or fucoidan (Fu). Both CS-PGA (P-NPs, 360 ± 67 nm) and CS-Fu nanoparticles (F-NPs, 440 ± 44 nm) showed sustained ginsenoside release in an acidic environment and improved ginsenoside solubility by approximately 122.8%. Both in vitro rabbit and ex vivo rat platelet aggregation of RG (22.3 and 41.5%) were significantly (p < 0.05) decreased within P-NPs (14.4 and 30.0%) and F-NPs (12.3 and 30.3%), respectively. Although RG exhibited no effect on in vivo carrageenan-induced mouse tail thrombosis, P-NPs and F-NPs demonstrated significant effects, likely the anticoagulation activity of PGA and Fu. Moreover, in the in vivo rat arteriovenous shunt model, P-NPs (156 ± 6.8 mg) and F-NPs (160 ± 3.2 mg) groups showed significantly lower thrombus formation than that of RG (190 ± 5.5 mg). Therefore, nanoencapsulation using CS, PGA, and Fu is a potential for improving the antithrombotic activity of RG.
Collapse
Affiliation(s)
- Eun Suh Kim
- Department of Food and Nutrition, Hanyang University , 222 Wangsimni-ro, Seongdong-gu, Seoul 04763, Republic of Korea
| | - Ji-Soo Lee
- Department of Food and Nutrition, Hanyang University , 222 Wangsimni-ro, Seongdong-gu, Seoul 04763, Republic of Korea
| | - Hyeon Gyu Lee
- Department of Food and Nutrition, Hanyang University , 222 Wangsimni-ro, Seongdong-gu, Seoul 04763, Republic of Korea
| |
Collapse
|
21
|
Ma N, Liu XW, Yang YJ, Shen DS, Zhao XL, Mohamed I, Kong XJ, Li JY. Evaluation on antithrombotic effect of aspirin eugenol ester from the view of platelet aggregation, hemorheology, TXB2/6-keto-PGF1α and blood biochemistry in rat model. BMC Vet Res 2016; 12:108. [PMID: 27296110 PMCID: PMC4907079 DOI: 10.1186/s12917-016-0738-0] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Accepted: 06/07/2016] [Indexed: 12/28/2022] Open
Abstract
Background Based on the prodrug principle, aspirin and eugenol, as starting precursors, were esterified to synthesize aspirin eugenol ester (AEE). The aim of the present study was to evaluate the antithrombotic effect of AEE in an animal disease model. In order to compare the therapeutic effects of AEE and its precursors, aspirin, eugenol and a combination of aspirin and eugenol were designed at the same molar quantities as the AEE medium dose in the control group. Methods After oral administration of AEE (dosed at 18, 36 and 72 mg/kg) for seven days, rats were treated with k-carrageenan to induce tail thrombosis. Following the same method, aspirin (20 mg/kg), eugenol (18 mg/kg) and 0.5 % CMC-Na (30 mg/kg) were administered as control drug. Different drug effects on platelet aggregation, hemorheology, TXB2/6-keto-PGF1α ratio and blood biochemistry were studied. Results AEE significantly inhibited ADP and AA-induced platelet aggregation in vivo. AEE also significantly reduced blood and plasma viscosity. Moreover, AEE down-regulated TXB2 and up-regulated 6-keto-PGF1α, normalizing the TXB2/6-keto-PGF1α ratio and blood biochemical profile. In comparison with aspirin and eugenol, AEE produced more positive therapeutic effects than its precursors under the same molar quantity. Conclusion It may be concluded that AEE was a good candidate for new antithrombotic and antiplatelet medicine. Additionally, this study may help to understand how AEE works on antithrombosis in different ways.
Collapse
Affiliation(s)
- Ning Ma
- Key Lab of New Animal Drug Project, Gansu Province, Lanzhou, 730050, People's Republic of China.,Key Lab of Veterinary Pharmaceutical Development, Ministry of Agriculture, Lanzhou, 730050, People's Republic of China.,Lanzhou Institute of Husbandry and Pharmaceutical Science of CAAS, Lanzhou, 730050, People's Republic of China
| | - Xi-Wang Liu
- Key Lab of New Animal Drug Project, Gansu Province, Lanzhou, 730050, People's Republic of China.,Key Lab of Veterinary Pharmaceutical Development, Ministry of Agriculture, Lanzhou, 730050, People's Republic of China.,Lanzhou Institute of Husbandry and Pharmaceutical Science of CAAS, Lanzhou, 730050, People's Republic of China
| | - Ya-Jun Yang
- Key Lab of New Animal Drug Project, Gansu Province, Lanzhou, 730050, People's Republic of China.,Key Lab of Veterinary Pharmaceutical Development, Ministry of Agriculture, Lanzhou, 730050, People's Republic of China.,Lanzhou Institute of Husbandry and Pharmaceutical Science of CAAS, Lanzhou, 730050, People's Republic of China
| | - Dong-Shuai Shen
- Key Lab of New Animal Drug Project, Gansu Province, Lanzhou, 730050, People's Republic of China.,Key Lab of Veterinary Pharmaceutical Development, Ministry of Agriculture, Lanzhou, 730050, People's Republic of China.,Lanzhou Institute of Husbandry and Pharmaceutical Science of CAAS, Lanzhou, 730050, People's Republic of China
| | - Xiao-Le Zhao
- Key Lab of New Animal Drug Project, Gansu Province, Lanzhou, 730050, People's Republic of China.,Key Lab of Veterinary Pharmaceutical Development, Ministry of Agriculture, Lanzhou, 730050, People's Republic of China.,Lanzhou Institute of Husbandry and Pharmaceutical Science of CAAS, Lanzhou, 730050, People's Republic of China
| | - Isam Mohamed
- Key Lab of New Animal Drug Project, Gansu Province, Lanzhou, 730050, People's Republic of China.,Key Lab of Veterinary Pharmaceutical Development, Ministry of Agriculture, Lanzhou, 730050, People's Republic of China.,Lanzhou Institute of Husbandry and Pharmaceutical Science of CAAS, Lanzhou, 730050, People's Republic of China
| | - Xiao-Jun Kong
- Key Lab of New Animal Drug Project, Gansu Province, Lanzhou, 730050, People's Republic of China.,Key Lab of Veterinary Pharmaceutical Development, Ministry of Agriculture, Lanzhou, 730050, People's Republic of China.,Lanzhou Institute of Husbandry and Pharmaceutical Science of CAAS, Lanzhou, 730050, People's Republic of China
| | - Jian-Yong Li
- Key Lab of New Animal Drug Project, Gansu Province, Lanzhou, 730050, People's Republic of China. .,Key Lab of Veterinary Pharmaceutical Development, Ministry of Agriculture, Lanzhou, 730050, People's Republic of China. .,Lanzhou Institute of Husbandry and Pharmaceutical Science of CAAS, Lanzhou, 730050, People's Republic of China. .,No.335, Jiangouyan, Qilihe District, Lanzhou, 730050, China.
| |
Collapse
|
22
|
Meshram V, Saxena S, Paul K. Xylarinase: a novel clot busting enzyme from an endophytic fungus Xylaria curta. J Enzyme Inhib Med Chem 2016; 31:1502-11. [PMID: 27033431 DOI: 10.3109/14756366.2016.1151013] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Xylarinase is a bi-functional fibrinolytic metalloprotease isolated from the culture filtrate of endophytic fungus Xylaria curta which is monomeric with a molecular mass of ∼33.76 kDa. The enzyme displayed both plasmin and tissue plasminogen activator like activity under in vitro conditions. It hydrolyses Aα and Bβ chains of the fibrinogen. Optimal fibrinolytic activity of xylarinase is observed at 35 °C, pH 8. Ca(2+) stimulated the fibrinolytic activity of xylarinase while Fe(2+) and Zn(2+) inhibited suggesting it to be a metalloprotease. The Km and Vmax values of xylarinase were 240.9 μM and 1.10 U/ml for fibrinogen and 246 μM and 1.22 U/ml for fibrin, respectively. Xylarinase was found to prolong the activated partial thromboplastin time and prothrombin time. The N-terminal sequence of xylarinase (SNGPLPGGVVWAG) did not show any homology with previously known fibrinolytic enzymes. Thus xylarinase is a novel fibrinolytic metalloprotease which could be possibly used as a new clot busting enzyme.
Collapse
Affiliation(s)
- Vineet Meshram
- a Department of Biotechnology , Thapar University , Patiala , India and
| | - Sanjai Saxena
- a Department of Biotechnology , Thapar University , Patiala , India and
| | - Karan Paul
- b Department of Biochemistry , DAV University , Jalandhar , Punjab , India
| |
Collapse
|
23
|
Sun Z, Liu P, Cheng G, Zhang B, Dong W, Su X, Huang Y, Cui Z, Kong Y. A fibrinolytic protease AfeE from Streptomyces sp. CC5, with potent thrombolytic activity in a mouse model. Int J Biol Macromol 2015; 85:346-54. [PMID: 26721382 DOI: 10.1016/j.ijbiomac.2015.12.059] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2015] [Revised: 12/16/2015] [Accepted: 12/17/2015] [Indexed: 12/31/2022]
Abstract
Fibrinolytic proteases have potential applications in cardiovascular disease therapy. A novel fibrinolytic protease, AfeE, with strong thrombolytic activity was purified from Streptomyces sp. CC5. AfeE displayed maximum activity at 40°C in the pH range of 7.0-12.0. It was strongly inhibited by serine protease inhibitor phenylmethanesulfonylfluoride, soybean trypsin inhibitor, tosyl-l-lysine chloromethyl ketone and tosyl-l-phenylalanine chloromethyl ketone. The activity of the enzyme was partially inhibited by Cu(2+), Co(2+) and Zn(2+). AfeE exhibited higher substrate specificity for fibrin than fibrinogen, which has rarely been reported in fibrinolytic enzymes. AfeE also showed high thrombolytic activity in a carrageenan-induced mouse tail thrombosis model. AfeE prolonged prothrombin time, activated partial thromboplastin time, and thrombin time in rat blood. A bleeding time assay revealed that AfeE did not prolong bleeding time in mice at a dose of 1mg/kg. No acute cytotoxicity was observed for AfeE at 320μg/well in human umbilical vein endothelial cells. The afeE gene was cloned from the genome of Streptomyces sp. CC5. Full-length AFE-CC5E contained 434 amino acids and was processed into a mature form consisting 284 amino acids by posttranslational modification, as revealed by high-resolution mass spectrometry analysis. These results indicate that AfeE is a prospective candidate for antithrombotic drug development.
Collapse
Affiliation(s)
- Zhibin Sun
- Key Laboratory of Environmental Microbiology of Ministry of Agriculture, Nanjing Agricultural University, Nanjing 210095, People's Republic of China
| | - Pingping Liu
- Key Laboratory of Environmental Microbiology of Ministry of Agriculture, Nanjing Agricultural University, Nanjing 210095, People's Republic of China
| | - Guangyan Cheng
- College of Life Sciences and Technology, China Pharmaceutical University, Nanjing 210009, People's Republic of China
| | - Biying Zhang
- Key Laboratory of Environmental Microbiology of Ministry of Agriculture, Nanjing Agricultural University, Nanjing 210095, People's Republic of China
| | - Weiliang Dong
- Key Laboratory of Environmental Microbiology of Ministry of Agriculture, Nanjing Agricultural University, Nanjing 210095, People's Republic of China
| | - Xingli Su
- College of Life Sciences and Technology, China Pharmaceutical University, Nanjing 210009, People's Republic of China
| | - Yan Huang
- Key Laboratory of Environmental Microbiology of Ministry of Agriculture, Nanjing Agricultural University, Nanjing 210095, People's Republic of China
| | - Zhongli Cui
- Key Laboratory of Environmental Microbiology of Ministry of Agriculture, Nanjing Agricultural University, Nanjing 210095, People's Republic of China.
| | - Yi Kong
- College of Life Sciences and Technology, China Pharmaceutical University, Nanjing 210009, People's Republic of China.
| |
Collapse
|
24
|
Wang X, Jiang S, Wang X, Liao J, Yin Z. Preparation and evaluation of nattokinase-loaded self-double-emulsifying drug delivery system. Asian J Pharm Sci 2015. [DOI: 10.1016/j.ajps.2015.04.005] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
|
25
|
Chen C, Yang FQ, Zhang Q, Wang FQ, Hu YJ, Xia ZN. Natural Products for Antithrombosis. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2015; 2015:876426. [PMID: 26075003 PMCID: PMC4449941 DOI: 10.1155/2015/876426] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/24/2014] [Revised: 03/22/2015] [Accepted: 03/24/2015] [Indexed: 12/25/2022]
Abstract
Thrombosis is considered to be closely related to several diseases such as atherosclerosis, ischemic heart disease and stroke, as well as rheumatoid arthritis, hyperuricemia, and various inflammatory conditions. More and more studies have been focused on understanding the mechanism of molecular and cellular basis of thrombus formation as well as preventing thrombosis for the treatment of thrombotic diseases. In reality, there is considerable interest in the role of natural products and their bioactive components in the prevention and treatment of thrombosis related disorders. This paper briefly describes the mechanisms of thrombus formation on three aspects, including coagulation system, platelet activation, and aggregation, and change of blood flow conditions. Furthermore, the natural products for antithrombosis by anticoagulation, antiplatelet aggregation, and fibrinolysis were summarized, respectively.
Collapse
Affiliation(s)
- Cen Chen
- School of Chemistry and Chemical Engineering, Chongqing University, Chongqing 400030, China
| | - Feng-Qing Yang
- School of Chemistry and Chemical Engineering, Chongqing University, Chongqing 400030, China
| | - Qian Zhang
- School of Chemistry and Chemical Engineering, Chongqing University, Chongqing 400030, China
| | - Feng-Qin Wang
- School of Chemistry and Chemical Engineering, Chongqing University, Chongqing 400030, China
| | - Yuan-Jia Hu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau
| | - Zhi-Ning Xia
- School of Chemistry and Chemical Engineering, Chongqing University, Chongqing 400030, China
| |
Collapse
|
26
|
Cho G, Han K, Yoon J. Stability Test and Quantitative and Qualitative Analyses of the Amino Acids in Pharmacopuncture Extracted from Scolopendra subspinipes mutilans. J Pharmacopuncture 2015; 18:44-55. [PMID: 25830058 PMCID: PMC4379475 DOI: 10.3831/kpi.2015.18.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2014] [Accepted: 12/31/2014] [Indexed: 11/13/2022] Open
Abstract
Objectives: Scolopendra subspinipes mutilans (S. subspinipes mutilans) is known as a traditional medicine and includes various amino acids, peptides and proteins. The amino acids in the pharmacopuncture extracted from S. subspinipes mutilans by using derivatization methods were analyzed quantitatively and qualitatively by using high performance liquid chromatography (HPLC) over a 12 month period to confirm its stability. Methods: Amino acids of pharmacopuncture extracted from S. subspinipes mutilans were derived by using O-phthaldialdehyde (OPA) & 9-fluorenyl methoxy carbonyl chloride (FMOC) reagent and were analyzed using HPLC. The amino acids were detected by using a diode array detector (DAD) and a fluorescence detector (FLD) to compare a mixed amino acid standard (STD) to the pharmacopuncture from centipedes. The stability tests on the pharmacopuncture from centipedes were done using HPLC for three conditions: a room temperature test chamber, an acceleration test chamber, and a cold test chamber. Results: The pharmacopuncture from centipedes was prepared by using the method of the Korean Pharmacopuncture Institute (KPI) and through quantitative analyses was shown to contain 9 amino acids of the 16 amino acids in the mixed amino acid STD. The amounts of the amino acids in the pharmacopuncture from centipedes were 34.37 ppm of aspartate, 123.72 ppm of arginine, 170.63 ppm of alanine, 59.55 ppm of leucine and 57 ppm of lysine. The relative standard deviation (RSD %) results for the pharmacopuncture from centipedes had a maximum value of 14.95% and minimum value of 1.795% on the room temperature test chamber, the acceleration test chamber and the cold test chamber stability tests. Conclusion: Stability tests on and quantitative and qualitative analyses of the amino acids in the pharmacopuncture extracted from centipedes by using derivatization methods were performed by using HPLC. Through research, we hope to determine the relationship between time and the concentrations of the amino acids in the pharmacopuncture extracted from centipedes.
Collapse
Affiliation(s)
- GyeYoon Cho
- Department of Research & Development, Pharmacopuncture Research Laboratory, Korean Pharmacopuncture Institute, Seoul, Korea
| | - KyuChul Han
- Department of Research & Development, Pharmacopuncture Research Laboratory, Korean Pharmacopuncture Institute, Seoul, Korea
| | - JinYoung Yoon
- Department of Research & Development, Pharmacopuncture Research Laboratory, Korean Pharmacopuncture Institute, Seoul, Korea
| |
Collapse
|
27
|
Majumdar S, Chattopadhyay P, Mukherjee AK. In Vivo Anticoagulant and Thrombolytic Activities of a Fibrinolytic Serine Protease (Brevithrombolase) With the k-Carrageenan-Induced Rat Tail Thrombosis Model. Clin Appl Thromb Hemost 2015; 22:594-8. [PMID: 25657326 DOI: 10.1177/1076029615569567] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
In the present study, in vivo thrombolysis efficiency of Brevithrombolase, a nontoxic fibrinolytic enzyme purified from Brevibacillus brevis strain FF02B, was affirmed by significant inhibition of thrombus formation in the k-carrageenan-induced rat tail, in a dose-dependent manner. Brevithrombolase at a dose of 600 µg/kg showed an efficacy that was comparable to streptokinase and plasmin, in dissolving in vivo thrombus of k-carrageenan-treated rats under identical conditions. The in vivo anticoagulant property of Brevithrombolase was demonstrated by its prolongation of activated partial thromboplastin time, prothrombin time, and thrombin time in Wistar rats. However, the Brevithrombolase-treated rats demonstrated an insignificant decrease in fibrinogen (Fg) level of plasma compared with Fg level of control group of rats corroborating in vivo as well as in vitro anticoagulant activity of Brevithrombolase is due to its hydrolytic action on thrombin. These findings unequivocally suggest that Brevithrombolase may serve a promising alternative to the commercial thrombolytic drugs.
Collapse
Affiliation(s)
- Sourav Majumdar
- Department of Molecular Biology and Biotechnology, Microbial Biotechnology and Protein Research Laboratory, School of Science, Tezpur University, Tezpur, Assam, India
| | | | - Ashis K Mukherjee
- Department of Molecular Biology and Biotechnology, Microbial Biotechnology and Protein Research Laboratory, School of Science, Tezpur University, Tezpur, Assam, India
| |
Collapse
|
28
|
Kim DW, Choi JH, Park SE, Kim S, Sapkota K, Kim SJ. Purification and characterization of a fibrinolytic enzyme from Petasites japonicus. Int J Biol Macromol 2015; 72:1159-67. [DOI: 10.1016/j.ijbiomac.2014.09.046] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2014] [Revised: 08/06/2014] [Accepted: 09/27/2014] [Indexed: 12/28/2022]
|
29
|
Choi JH, Sapkota K, Kim MK, Kim S, Kim SJ. Undariase, a direct-acting fibrin(ogen)olytic enzyme from Undaria pinnatifida, inhibits thrombosis in vivo and exhibits in vitro thrombolytic properties. Appl Biochem Biotechnol 2014; 173:1985-2004. [PMID: 24938821 DOI: 10.1007/s12010-014-0981-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2014] [Accepted: 05/19/2014] [Indexed: 12/14/2022]
Abstract
A direct-acting fibrinolytic serine protease named undariase possessing anticoagulant and antiplatelet properties was purified from Undaria pinnatifida. Undariase showed a molecular weight of 50 kDa by sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE) and mass spectrometry. It displayed a strong fibrin zymogram lysis band corresponding to the same molecular mass. The N-terminal sequence of undariase, LTATTCEELAAAPTD, does not match with any known fibrinolytic enzyme. The enzyme was stable and active at high temperatures (35-70 °C). The fibrinolytic activity of undariase was strongly inhibited by phenylmethylsulfonyl fluoride (PMSF) and 4-(amidinophenyl) methanesulfonyl fluoride (APMSF). The K m and V max values for substrate S-2251 were determined as 6.15 mM and 90.91 mM/min/ml, respectively. Undariase resulted in clot lysis by directly cleaving α and β chains of fibrin. Similarly, it preferentially acted on the Aα chain of fibrinogen followed by cleavage of the Bβ chain. It significantly prolonged the PFA-100 closure times of citrated whole human blood. In addition, undariase delayed the coagulation time and increased activated partial thromboplastin time (APTT), prothrombin time (PT), and thrombin time (TT). Undariase exerted a significant protective effect against collagen plus epinephrine-induced pulmonary thromboembolism in mice. It prevented carrageenan-induced thrombus formation in the tail of mice. It also resulted in prolongation of APTT ex vivo. In conclusion, these results suggested a therapeutic potential of undariase for thrombosis.
Collapse
Affiliation(s)
- Jun-Hui Choi
- Department of Life Science & BK21-Plus Research Team for Bioactive Control Technology, Chosun University, 375 Seosuk-dong, Dong-gu, Gwangju, 501-759, Republic of Korea
| | | | | | | | | |
Collapse
|
30
|
Bi Q, Han B, Feng Y, Jiang Z, Yang Y, Liu W. Antithrombotic effects of a newly purified fibrinolytic protease from Urechis unicinctus. Thromb Res 2013; 132:e135-44. [DOI: 10.1016/j.thromres.2013.07.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2013] [Revised: 06/21/2013] [Accepted: 07/04/2013] [Indexed: 11/26/2022]
|
31
|
Kong Y, Shao Y, Chen H, Ming X, Wang JB, Li ZY, Wei JF. A Novel Factor Xa-Inhibiting Peptide from Centipedes Venom. Int J Pept Res Ther 2013; 19:303-311. [PMID: 24273471 PMCID: PMC3824214 DOI: 10.1007/s10989-013-9353-0] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/05/2013] [Indexed: 11/26/2022]
Abstract
Centipedes have been used as traditional medicine for thousands of years in China. Centipede venoms consist of many biochemical peptides and proteins. Factor Xa (FXa) is a serine endopeptidase that plays the key role in blood coagulation, and has been used as a new target for anti-thrombotic drug development. A novel FXa inhibitor, a natural peptide with the sequence of Thr-Asn-Gly-Tyr-Thr (TNGYT), was isolated from the venom of Scolopendra subspinipesmutilans using a combination of size-exclusion and reverse-phase chromatography. The molecular weight of the TNGYT peptide was 554.3 Da measured by electrospray ionization mass spectrometry. The amino acid sequence of TNGYT was determined by Edman degradation. TNGYT inhibited the activity of FXa in a dose-dependent manner with an IC50 value of 41.14 mg/ml. It prolonged the partial thromboplastin time and prothrombin time in both in vitro and ex vivo assays. It also significantly prolonged whole blood clotting time and bleeding time in mice. This is the first report that an FXa inhibiting peptide was isolated from centipedes venom.
Collapse
Affiliation(s)
- Yi Kong
- School of Life Science & Technology, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing, 210009 People’s Republic of China
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009 People’s Republic of China
| | - Yu Shao
- School of Life Science & Technology, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing, 210009 People’s Republic of China
| | - Hao Chen
- School of Life Science & Technology, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing, 210009 People’s Republic of China
| | - Xin Ming
- Division of Molecular Pharmaceutics, UNC Eshelman School of Pharmacy, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599 USA
| | - Jin-Bin Wang
- School of Pharmacy, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing, 210009 People’s Republic of China
| | - Zhi-Yu Li
- School of Pharmacy, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing, 210009 People’s Republic of China
| | - Ji-Fu Wei
- Research Division of Clinical Pharmacology, The First Affiliated Hospital with Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029 China
| |
Collapse
|
32
|
Carrageenan-induced colonic inflammation is reduced in Bcl10 null mice and increased in IL-10-deficient mice. Mediators Inflamm 2013; 2013:397642. [PMID: 23766559 PMCID: PMC3677668 DOI: 10.1155/2013/397642] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2013] [Revised: 04/29/2013] [Accepted: 05/08/2013] [Indexed: 12/19/2022] Open
Abstract
The common food additive carrageenan is a known activator of inflammation in mammalian tissues and stimulates both the canonical and noncanonical pathways of NF-κB activation. Exposure to low concentrations of carrageenan (10 μg/mL in the water supply) has produced glucose intolerance, insulin resistance, and impaired insulin signaling in C57BL/6 mice. B-cell leukemia/lymphoma 10 (Bcl10) is a mediator of inflammatory signals from Toll-like receptor (TLR) 4 in myeloid and epithelial cells. Since the TLR4 signaling pathway is activated in diabetes and by carrageenan, we addressed systemic and intestinal inflammatory responses following carrageenan exposure in Bcl10 wild type, heterozygous, and null mice. Fecal calprotectin and circulating keratinocyte chemokine (KC), nuclear RelA and RelB, phospho(Thr559)-NF-κB-inducing kinase (NIK), and phospho(Ser36)-IκBα in the colonic epithelial cells were significantly less (P < 0.001) in the carrageenan-treated Bcl10 null mice than in controls. IL-10-deficient mice exposed to carrageenan in a germ-free environment showed an increase in activation of the canonical pathway of NF-κB (RelA) activation, but without increase in RelB or phospho-Bcl10, and exogenous IL-10 inhibited only the canonical pathway of NF-κB activation in cultured colonic cells. These findings demonstrate a Bcl10 requirement for maximum development of carrageenan-induced inflammation and lack of complete suppression by IL-10 of carrageenan-induced inflammation.
Collapse
|
33
|
Purification and Characterization of a New Serine Protease with Fibrinolytic Activity from the Marine Invertebrate, Urechis unicinctus. Appl Biochem Biotechnol 2013; 170:525-40. [DOI: 10.1007/s12010-013-0168-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2012] [Accepted: 02/26/2013] [Indexed: 12/24/2022]
|