1
|
Liu Y, Zhao Z, Huang X, Xiao Y, Li N, Yang W, Feng R, Feng W, Long T, Wu H, Peng G, Chen S, Chang G, Huang K, Li Z. 2-Methoxybenzoic acid ameliorates arterial thrombosis via inhibiting carbon anhydrase activity in platelet. J Thromb Haemost 2025; 23:1893-1907. [PMID: 40056982 DOI: 10.1016/j.jtha.2025.02.027] [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: 10/14/2024] [Revised: 02/18/2025] [Accepted: 02/26/2025] [Indexed: 03/30/2025]
Abstract
BACKGROUND 2-Methoxybenzoic acid (2MOA) is a natural compound with potential salicylate-like effects; however, its impact on arterial thrombosis remains unclear. OBJECTIVES This study aimed to investigate the effects of 2MOA on thrombogenesis and its underlying mechanisms. METHODS FeCl3-induced carotid artery injury and laser-induced cremaster artery injury thrombosis assays were used to explore the effect of 2MOA on thrombogenesis in vivo. Various ex vivo platelet function assays were conducted to evaluate the impacts of 2MOA on platelet activity. In addition, untargeted metabolomics analysis was performed to identify the alterations in intraplatelet metabolites following 2MOA treatment. RESULTS We found that 2MOA significantly ameliorated thrombosis in a dose-dependent manner, without affecting the normal hemostasis in C57BL/6J mice. 2MOA suppressed platelet reactivity as indicated by decreased spreading, retraction, and aggregation in both mouse and human platelets. Metabolomics analysis revealed significantly alterations in purine metabolism following 2MOA treatment, which increased cyclic guanosine monophosphate production in platelets. Mechanistically, 2MOA inhibited the activity of carbonic anhydrase, leading to elevated intraplatelet cGMP level, and subsequent suppression of cytosolic phospholipase A2 phosphorylation. CONCLUSION Our study illustrates that 2MOA efficaciously inhibits platelet reactivity and alleviates thrombogenesis via suppressing carbonic anhydrase activity, which should be a promising reagent in the prevention and treatment of arterial thrombotic events.
Collapse
Affiliation(s)
- Yunchong Liu
- Division of Vascular Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China; National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Zhengde Zhao
- Division of Vascular Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China; National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Xiuyi Huang
- Division of Vascular Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China; National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Ying Xiao
- Department of Anesthesiology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Na Li
- Division of Vascular Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China; National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Wenchao Yang
- Division of Vascular Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China; National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Ruijia Feng
- Division of Vascular Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China; National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Weiqi Feng
- Division of Vascular Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China; National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Ting Long
- Division of Vascular Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China; National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Haoliang Wu
- Department of Anesthesiology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Guiyan Peng
- Division of Vascular Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China; National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Sifan Chen
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China; Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-sen Memorial Hospital, Foshan, Guangdong, China
| | - Guangqi Chang
- Division of Vascular Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China; National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Kan Huang
- Division of Vascular Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China; National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China.
| | - Zilun Li
- Division of Vascular Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China; National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China.
| |
Collapse
|
2
|
Imberg L, Siutkina AI, Erbacher C, Schmidt J, Broekmans DF, Ovsepyan RA, Daniliuc CG, Gonçalves de Oliveira E, Serafim MSM, O’Donoghue AJ, Pillaiyar T, Panteleev MA, Poso A, Kalinina SA, Bermúdez M, Nekipelov K, Bendas G, Karst U, Kalinin DV. Pyrazinyl-Substituted Aminoazoles as Covalent Inhibitors of Thrombin: Synthesis, Structure, and Anticoagulant Properties. ACS Pharmacol Transl Sci 2025; 8:146-172. [PMID: 39816788 PMCID: PMC11730114 DOI: 10.1021/acsptsci.4c00515] [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: 08/26/2024] [Revised: 11/17/2024] [Accepted: 11/22/2024] [Indexed: 01/18/2025]
Abstract
This study presents a novel series of N-acylated 1,2,4-triazol-5-amines and 1H-pyrazol-5-amines, featuring a pyrazin-2-yl moiety, developed as covalent inhibitors of thrombin. These compounds demonstrated potent inhibitory activity, with derivatives 13a and 13b achieving IC50 values as low as 0.7 and 0.8 nM, respectively. Mass-shift assays confirmed that these inhibitors covalently bind to the catalytic Ser195 of thrombin, leading to temporary inhibition of its activity through specific acylation. The anticoagulant efficacy of these compounds was validated in plasma coagulation assays, with selected derivatives extending coagulation times in both an activated partial thromboplastin time (aPTT) and prothrombin time (PT) test. Thrombin generation assays further demonstrated that compounds of this series effectively reduced thrombin generation without substantially prolonging clotting times, suggesting a lower risk of bleeding. Selected compounds also strongly inhibited cancer cell- and thrombin-induced platelet aggregation. These results indicate that acylated aminoazoles hold a promise as new anticoagulants.
Collapse
Affiliation(s)
- Lukas Imberg
- Institute of Pharmaceutical and Medicinal Chemistry,
University of Münster, 48149 Münster,
Germany
| | - Alena I. Siutkina
- Institute of Pharmaceutical and Medicinal Chemistry,
University of Münster, 48149 Münster,
Germany
| | - Catharina Erbacher
- Institute of Inorganic and Analytical Chemistry,
University of Münster, 48149 Münster,
Germany
| | - Judith Schmidt
- Institute of Pharmaceutical and Medicinal Chemistry,
University of Münster, 48149 Münster,
Germany
| | - Darius F. Broekmans
- Institute of Pharmaceutical and Medicinal Chemistry,
University of Münster, 48149 Münster,
Germany
| | - Ruzanna A. Ovsepyan
- Center for Theoretical Problems of
Physicochemical Pharmacology, Russian Academy of Sciences, 119991 Moscow,
Russia
| | | | - Ellen Gonçalves de Oliveira
- Center for Discovery and Innovation in Parasitic
Diseases, Skaggs School of Pharmacy and Pharmaceutical Sciences, University
of California, La Jolla, San Diego, California 92093, United
States
- Department of Microbiology, Institute of Biological Sciences,
Federal University of Minas Gerais, 31270-901 Belo Horizonte,
Minas Gerais, Brazil
| | - Mateus Sá Magalhães Serafim
- Center for Discovery and Innovation in Parasitic
Diseases, Skaggs School of Pharmacy and Pharmaceutical Sciences, University
of California, La Jolla, San Diego, California 92093, United
States
- Department of Microbiology, Institute of Biological Sciences,
Federal University of Minas Gerais, 31270-901 Belo Horizonte,
Minas Gerais, Brazil
| | - Anthony J. O’Donoghue
- Center for Discovery and Innovation in Parasitic
Diseases, Skaggs School of Pharmacy and Pharmaceutical Sciences, University
of California, La Jolla, San Diego, California 92093, United
States
| | - Thanigaimalai Pillaiyar
- Institute of Pharmacy, Pharmaceutical/Medicinal
Chemistry, Eberhard Karls University Tübingen, 72076
Tübingen, Germany
- Tübingen Center for Academic Drug
Discovery (TüCAD2), 72076 Tübingen,
Germany
| | - Mikhail A. Panteleev
- Center for Theoretical Problems of
Physicochemical Pharmacology, Russian Academy of Sciences, 119991 Moscow,
Russia
- Laboratory of Translational Medicine,
Dmitriy Rogachev National Medical Research Center of Pediatric Hematology, Oncology, and
Immunology, 117997 Moscow, Russia
| | - Antti Poso
- School of Pharmacy, Faculty of Health Sciences,
University of Eastern Finland, 70211 Kuopio,
Finland
- Department of Internal Medicine VIII,
University Hospital Tübingen, 72076 Tübingen,
Germany
| | | | - Marcel Bermúdez
- Institute of Pharmaceutical and Medicinal Chemistry,
University of Münster, 48149 Münster,
Germany
| | - Katrin Nekipelov
- Pharmaceutical Institute, University
of Bonn, 53121 Bonn, Germany
| | - Gerd Bendas
- Pharmaceutical Institute, University
of Bonn, 53121 Bonn, Germany
| | - Uwe Karst
- Institute of Inorganic and Analytical Chemistry,
University of Münster, 48149 Münster,
Germany
| | - Dmitrii V. Kalinin
- Institute of Pharmaceutical and Medicinal Chemistry,
University of Münster, 48149 Münster,
Germany
| |
Collapse
|
3
|
Dai Y, Kretz CA, Kim PY, Gross PL. A specific fluorescence resonance energy quenching-based biosensor for measuring thrombin activity in whole blood. J Thromb Haemost 2024; 22:1627-1639. [PMID: 38382740 DOI: 10.1016/j.jtha.2024.02.007] [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: 10/17/2023] [Revised: 02/06/2024] [Accepted: 02/09/2024] [Indexed: 02/23/2024]
Abstract
BACKGROUND At sites of vessel injury, thrombin acts as the central mediator of coagulation by catalyzing fibrin clot formation and platelet activation. Thrombin generation is most frequently measured in plasma samples using small-molecule substrates; however, these have low specificity for thrombin and limited utility in whole blood. Plasma assays are limited because they ignore the hemostatic contributions of blood cells and require anticoagulation and the addition of supraphysiological concentrations of calcium. OBJECTIVES To overcome these limitations, we designed and characterized a fluorescence resonance energy quenching-based thrombin sensor (FTS) protein. METHODS The fluorescence resonance energy quenching pair of mAmetrine and tTomato, separated by a thrombin recognition sequence, was developed. The protein was expressed using Escherichia coli, and purity was assessed using sodium dodecyl sulfate-polyacrylamide gel electrophoresis. The cleavage of FTS was monitored by fluorescence using excitation at 406 nm and emission at 526 nm and 581 nm. RESULTS Compared with small-molecule substrates, the FTS demonstrated high specificity for thrombin; it is not cleaved by thrombin or inhibited by α2-macroglobulin and interacts with thrombin's anion-binding exosite I. The FTS can effectively measure thrombin generation in plasma and in finger-prick whole blood, which allows it to be developed into a point-of-care test of thrombin generation. The FTS does not inhibit standard thrombin-generation assays. Lastly, FTS-based thrombin generation in nonanticoagulated finger-prick blood was delayed but enhanced compared with that in citrated plasma. CONCLUSION The FTS will broaden our understanding of thrombin generation in ways that are not attainable with current methods.
Collapse
Affiliation(s)
- Ying Dai
- Hamilton Health Sciences and Departments of Medicine and Medical Sciences, Thrombosis and Atherosclerosis Research Institute, McMaster University, Hamilton, Ontario, Canada
| | - Colin A Kretz
- Hamilton Health Sciences and Departments of Medicine and Medical Sciences, Thrombosis and Atherosclerosis Research Institute, McMaster University, Hamilton, Ontario, Canada
| | - Paul Y Kim
- Hamilton Health Sciences and Departments of Medicine and Medical Sciences, Thrombosis and Atherosclerosis Research Institute, McMaster University, Hamilton, Ontario, Canada
| | - Peter L Gross
- Hamilton Health Sciences and Departments of Medicine and Medical Sciences, Thrombosis and Atherosclerosis Research Institute, McMaster University, Hamilton, Ontario, Canada.
| |
Collapse
|
4
|
Erbacher C, Athmer M, Kröger E, Dahrendorf L, Imberg L, Kalinin DV, Karst U. An automated analysis method enabling the screening of covalent thrombin and factor XIIa inhibitors via liquid chromatography-mass spectrometry. Drug Test Anal 2024; 16:314-322. [PMID: 37482900 DOI: 10.1002/dta.3549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2023] [Revised: 07/04/2023] [Accepted: 07/06/2023] [Indexed: 07/25/2023]
Abstract
An automated sample preparation and separation method for the analysis of various enzyme-inhibitor combinations using liquid chromatography (LC) coupled to mass spectrometry (MS) is presented. As conventional anticoagulants have several drawbacks, the most severe being the elevated risk of internal bleedings, it is necessary to develop new-generation anticoagulants with reduced side effects. Therefore, the screening of potential inhibitors against anticoagulation targets like thrombin and FXIIa is important to design a potent and selective inhibitor. To facilitate the analysis of numerous enzyme-inhibitor covalent complexes, automation of the analysis using an LC system with a user-defined injection sequence is helpful. The developed method ensures comparable reaction conditions like reaction time and temperature for all enzyme-inhibitor complexes. Furthermore, it prevents time-consuming manual sample preparation and potential manual errors. To achieve good reproducibility with relative standard deviation of approximately 3% for three-fold determination, multiple cleaning steps were added to the automated sample preparation. Subsequently, this method was applied to screen a variety of 15 aminopyrazole- and aminotriazole-based inhibitors with a covalent mechanism of action against thrombin and to test two covalent inhibitors for FXIIa. Successful complex formation and acylation of the catalytic center of the enzymes was monitored using deconvoluted mass spectra and the matching mass shifts of the acyl moiety of the analyzed inhibitors. The inhibitors' structure directly influenced reaction yields. Sterically demanding aminotriazoles and acyl moieties both affected the product formation negatively. However, the screening yielded several promising candidates for new covalent thrombin inhibitors, which might find their application as prospective anticoagulants.
Collapse
Affiliation(s)
- Catharina Erbacher
- Institute of Inorganic and Analytical Chemistry, University of Münster, Münster, Germany
| | - Mathis Athmer
- Institute of Inorganic and Analytical Chemistry, University of Münster, Münster, Germany
| | - Erik Kröger
- Institute of Inorganic and Analytical Chemistry, University of Münster, Münster, Germany
| | - Laureen Dahrendorf
- Institute of Inorganic and Analytical Chemistry, University of Münster, Münster, Germany
| | - Lukas Imberg
- Institute of Pharmaceutical and Medicinal Chemistry, University of Münster, Münster, Germany
| | - Dmitrii V Kalinin
- Institute of Pharmaceutical and Medicinal Chemistry, University of Münster, Münster, Germany
| | - Uwe Karst
- Institute of Inorganic and Analytical Chemistry, University of Münster, Münster, Germany
| |
Collapse
|
5
|
Short KM, Estiarte MA, Pham SM, Williams DC, Igoudin L, Dash S, Sandoval N, Datta A, Pozzi N, Di Cera E, Kita DB. Discovery of novel N-acylpyrazoles as potent and selective thrombin inhibitors. Eur J Med Chem 2023; 246:114855. [PMID: 36462436 DOI: 10.1016/j.ejmech.2022.114855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 10/08/2022] [Accepted: 10/08/2022] [Indexed: 01/04/2023]
Abstract
Direct oral anticoagulants (DOACs), which includes thrombin and factor Xa inhibitors, have emerged as the preferred therapeutics for thrombotic disorders, penetrating a market previously dominated by warfarin and heparin. This article describes the discovery and profiling of a novel series of N-acylpyrazoles, which act as selective, covalent, reversible, non-competitive inhibitors of thrombin. We describe in vitro stability issues associated with this chemotype and, importantly, demonstrate that N-acylpyrazoles successfully act in vivo as anticoagulants in basic thrombotic animal models. Crucially, this anticoagulant nature is unaccompanied by the higher bleeding risk profile that has become an undesirable characteristic of the DTIs and factor Xa inhibitors. We propose that the N-acylpyrazole chemotype shows intriguing promise as next-generation oral anticoagulants.
Collapse
Affiliation(s)
- Kevin M Short
- Verseon, 47071 Bayside Parkway, Fremont, CA, 94538, USA.
| | | | - Son M Pham
- Verseon, 47071 Bayside Parkway, Fremont, CA, 94538, USA
| | | | - Lev Igoudin
- Verseon, 47071 Bayside Parkway, Fremont, CA, 94538, USA
| | - Subhadra Dash
- Verseon, 47071 Bayside Parkway, Fremont, CA, 94538, USA
| | | | - Anirban Datta
- Verseon, 47071 Bayside Parkway, Fremont, CA, 94538, USA
| | - Nicola Pozzi
- Department of Biochemistry and Molecular Biology, Edward A. Doisy Research Center, Saint Louis University School of Medicine, 1100 South Grand Blvd., St. Louis, MO, 63104, USA
| | - Enrico Di Cera
- Department of Biochemistry and Molecular Biology, Edward A. Doisy Research Center, Saint Louis University School of Medicine, 1100 South Grand Blvd., St. Louis, MO, 63104, USA
| | - David B Kita
- Verseon, 47071 Bayside Parkway, Fremont, CA, 94538, USA
| |
Collapse
|
6
|
Imberg L, Platte S, Erbacher C, Daniliuc CG, Kalinina SA, Dörner W, Poso A, Karst U, Kalinin DV. Amide-functionalized 1,2,4-Triazol-5-amines as Covalent Inhibitors of Blood Coagulation Factor XIIa and Thrombin. ACS Pharmacol Transl Sci 2022; 5:1318-1347. [PMID: 36524012 PMCID: PMC9745896 DOI: 10.1021/acsptsci.2c00204] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Indexed: 12/05/2022]
Abstract
To counteract thrombosis, new safe and efficient antithrombotics are required. We herein report the design, synthesis, and biological activity of a series of amide-functionalized acylated 1,2,4-triazol-5-amines as selective inhibitors of blood coagulation factor XIIa and thrombin. The introduction of an amide moiety into the main scaffold of 3-aryl aminotriazoles added certain three-dimensional properties to synthesized compounds and allowed them to reach binding sites in FXIIa and thrombin previously unaddressed by non-functionalized 1,2,4-triazol-5-amines. Among synthesized compounds, one quinoxaline-derived aminotriazole bearing N-butylamide moiety inhibited FXIIa with the IC50 value of 28 nM, whereas the N-phenylamide-derived aminotriazole inhibited thrombin with the IC50 value of 41 nM. Performed mass-shift experiments and molecular modeling studies proved the covalent mechanism of FXIIa and thrombin inhibition by synthesized compounds. In plasma coagulation tests, developed aminotriazoles showed anticoagulant properties mainly affecting the intrinsic blood coagulation pathway, activation of which is associated with thrombosis but is negligible for hemostasis.
Collapse
Affiliation(s)
- Lukas Imberg
- Institute
of Pharmaceutical and Medicinal Chemistry, University of Münster, Münster 48149, Germany
| | - Simon Platte
- Institute
of Pharmaceutical and Medicinal Chemistry, University of Münster, Münster 48149, Germany
| | - Catharina Erbacher
- Institute
of Inorganic and Analytical Chemistry, University
of Münster, Münster 48149, Germany
| | | | | | - Wolfgang Dörner
- Institute
of Biochemistry, University of Münster, Münster 48149, Germany
| | - Antti Poso
- School
of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, Kuopio 70211, Finland
- Department
of Internal Medicine VIII, University Hospital
Tübingen, Tübingen 72076, Germany
| | - Uwe Karst
- Institute
of Inorganic and Analytical Chemistry, University
of Münster, Münster 48149, Germany
| | - Dmitrii V. Kalinin
- Institute
of Pharmaceutical and Medicinal Chemistry, University of Münster, Münster 48149, Germany
| |
Collapse
|
7
|
Dunker C, Imberg L, Siutkina AI, Erbacher C, Daniliuc CG, Karst U, Kalinin DV. Pyrazole-Based Thrombin Inhibitors with a Serine-Trapping Mechanism of Action: Synthesis and Biological Activity. Pharmaceuticals (Basel) 2022; 15:1340. [PMID: 36355511 PMCID: PMC9696832 DOI: 10.3390/ph15111340] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 10/24/2022] [Accepted: 10/25/2022] [Indexed: 07/25/2023] Open
Abstract
New antithrombotic drugs are needed to combat thrombosis, a dangerous pathology that causes myocardial infarction and ischemic stroke. In this respect, thrombin (FIIa) represents an important drug target. We herein report the synthesis and biological activity of a series of 1H-pyrazol-5-amine-based thrombin inhibitors with a serine-trapping mechanism of action. Among synthesized compounds, flexible acylated 1H-pyrazol-5-amines 24e, 34a, and 34b were identified as potent 16-80 nM thrombin inhibitors, which showed practically no off-targeting effect against other physiologically relevant serine proteases. To prove that synthesized compounds are covalent thrombin inhibitors, the most potent derivative 24e (FIIa IC50 = 16 nM) was studied in a mass-shift assay, where it has been shown that 24e transfers its acyl moiety (pivaloyl) to the catalytic Ser195 of thrombin. Performed herein docking studies also confirmed the covalent mechanism of thrombin inhibition by synthesized compounds. Acylated aminopyrazoles found during this study showed only limited effects on plasma coagulation in activated partial thrombin time (aPTT) and prothrombin time (PT) in vitro assays. However, such thrombin inhibitors are expected to have virtually no effect on bleeding time and can be used as a starting point for developing a safer alternative to traditional non-covalent anticoagulants.
Collapse
Affiliation(s)
- Calvin Dunker
- Institute of Pharmaceutical and Medicinal Chemistry, University of Münster, 48149 Münster, Germany
| | - Lukas Imberg
- Institute of Pharmaceutical and Medicinal Chemistry, University of Münster, 48149 Münster, Germany
| | - Alena I. Siutkina
- Institute of Pharmaceutical and Medicinal Chemistry, University of Münster, 48149 Münster, Germany
| | - Catharina Erbacher
- Institute of Inorganic and Analytical Chemistry, University of Münster, 48149 Münster, Germany
| | | | - Uwe Karst
- Institute of Inorganic and Analytical Chemistry, University of Münster, 48149 Münster, Germany
| | - Dmitrii V. Kalinin
- Institute of Pharmaceutical and Medicinal Chemistry, University of Münster, 48149 Münster, Germany
| |
Collapse
|
8
|
Montinari MR, Minelli S. From ancient leech to direct thrombin inhibitors and beyond: New from old. Biomed Pharmacother 2022; 149:112878. [PMID: 35364378 DOI: 10.1016/j.biopha.2022.112878] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Revised: 03/13/2022] [Accepted: 03/23/2022] [Indexed: 11/30/2022] Open
Abstract
Medicinal leeches have been used in health care since before written history, with widely varying popularity over the centuries. Nowadays, medicinal leech therapy is mainly used in plastic and reconstructive microsurgery, with new interesting potential therapeutic applications in many other diseases. The leech's best-known salivary product, hirudin - one of the most powerful natural anticoagulants - was the only remedy to prevent blood clotting until the discovery of heparin. Starting from hirudin, pharmacological research succeeded in developing new anticoagulants, which represent a cornerstone of prevention and treatment of thromboembolic disease. While we are perhaps on the threshold of a new era of anticoagulation, with the development of FXI and XII inhibitors and direct reversible covalent thrombin inhibitors, which promise to achieve effective anticoagulation without bleeding risk. This review retraces the intriguing journey of these drugs in cardiovascular disease, highlighting the fil rouge that links the ancient leech to the current and oncoming antithrombotic therapy. We think that knowledge of the past is key to understanding and appreciating the present and to seize future opportunities.
Collapse
Affiliation(s)
- Maria Rosa Montinari
- Chair of History of Medicine, Department of Biological and Environmental Sciences and Technologies, University of Salento, Lecce, Italy.
| | | |
Collapse
|
9
|
Mukherjee AK, Chattopadhyay DJ. Potential clinical applications of phytopharmaceuticals for the in-patient management of coagulopathies in COVID-19. Phytother Res 2022; 36:1884-1913. [PMID: 35147268 PMCID: PMC9111032 DOI: 10.1002/ptr.7408] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Revised: 01/21/2022] [Accepted: 01/26/2022] [Indexed: 12/21/2022]
Abstract
Thrombotic complications occur in many cardiovascular pathologies and have been demonstrated in COVID‐19. The currently used antithrombotic drugs are not free of adverse reactions, and COVID‐19 patients in particular, when treated with a therapeutic dose of an anticoagulant do not receive mortality benefits. The clinical management of COVID‐19 is one of the most difficult tasks for clinicians, and the search for safe, potent, and effective antithrombotic drugs may benefit from exploring naturally bioactive molecules from plant sources. This review describes recent advances in understanding the antithrombotic potential of herbal drug prototypes and points to their future clinical use as potent antithrombotic drugs. Although natural products are perceived to be safe, their clinical and therapeutic applications are not always apparent or accepted. More in‐depth studies are necessary to demonstrate the clinical usefulness of plant‐derived, bioactive compounds. In addition, holistic approaches in systematic investigations and the identification of antithrombotic mechanisms of the herbal bioactive molecule(s) need to be conducted in pre‐clinical studies. Moreover, rigorous studies are needed to compare the potency of herbal drugs to that of competitor chemical antithrombotic drugs, and to examine their interactions with Western antithrombotic medicines. We have also proposed a road map to improve the commercialization of phytopharmaceuticals.
Collapse
Affiliation(s)
- Ashis K Mukherjee
- Division of Life Sciences, Institute of Advanced Study in Science and Technology, Guwahati, India.,Department of Molecular Biology and Biotechnology, School of Sciences, Tezpur University, Tezpur, India
| | | |
Collapse
|
10
|
Platte S, Korff M, Imberg L, Balicioglu I, Erbacher C, Will JM, Daniliuc CG, Karst U, Kalinin DV. Microscale Parallel Synthesis of Acylated Aminotriazoles Enabling the Development of Factor XIIa and Thrombin Inhibitors. ChemMedChem 2021; 16:3672-3690. [PMID: 34278727 PMCID: PMC9292294 DOI: 10.1002/cmdc.202100431] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Indexed: 01/12/2023]
Abstract
Herein we report a microscale parallel synthetic approach allowing for rapid access to libraries of N-acylated aminotriazoles and screening of their inhibitory activity against factor XIIa (FXIIa) and thrombin, which are targets for antithrombotic drugs. This approach, in combination with post-screening structure optimization, yielded a potent 7 nM inhibitor of FXIIa and a 25 nM thrombin inhibitor; both compounds showed no inhibition of the other tested serine proteases. Selected N-acylated aminotriazoles exhibited anticoagulant properties in vitro influencing the intrinsic blood coagulation pathway, but not extrinsic coagulation. Mechanistic studies of FXIIa inhibition suggested that synthesized N-acylated aminotriazoles are covalent inhibitors of FXIIa. These synthesized compounds may serve as a promising starting point for the development of novel antithrombotic drugs.
Collapse
Affiliation(s)
- Simon Platte
- Institute of Pharmaceutical and Medicinal ChemistryUniversity of MünsterCorrensstr. 4848149MünsterGermany
| | - Marvin Korff
- Institute of Pharmaceutical and Medicinal ChemistryUniversity of MünsterCorrensstr. 4848149MünsterGermany
| | - Lukas Imberg
- Institute of Pharmaceutical and Medicinal ChemistryUniversity of MünsterCorrensstr. 4848149MünsterGermany
| | - Ilker Balicioglu
- Institute of Pharmaceutical and Medicinal ChemistryUniversity of MünsterCorrensstr. 4848149MünsterGermany
| | - Catharina Erbacher
- Institute of Inorganic and Analytical ChemistryUniversity of MünsterCorrensstr. 3048149MünsterGermany
| | - Jonas M. Will
- Institute of Inorganic and Analytical ChemistryUniversity of MünsterCorrensstr. 3048149MünsterGermany
| | | | - Uwe Karst
- Institute of Inorganic and Analytical ChemistryUniversity of MünsterCorrensstr. 3048149MünsterGermany
| | - Dmitrii V. Kalinin
- Institute of Pharmaceutical and Medicinal ChemistryUniversity of MünsterCorrensstr. 4848149MünsterGermany
| |
Collapse
|
11
|
Abstract
Introduction: Blood coagulation factor XII (FXII) is an emerging and potentially safe drug target, which dysregulation is associated with thrombosis, hereditary angioedema, and (neuro)inflammation. At the same time, FXII-deficiency is practically asymptomatic. Industrial and academic institutions have developed a number of potential therapeutic agents targeting either FXII zymogen or its active form FXIIa for the treatment of thrombotic and inflammatory conditions associated with the activity of this enzyme.Areas covered: A short overview of the FXII(a) structure and function, underlining its suitability as a drug target, is given. The article reviews patents reported over the last three decades on FXII(a)-targeting therapeutic agents. These agents include small molecules, proteins, peptides, oligonucleotides, siRNAs, and monoclonal antibodies.Expert opinion: The performed analysis of patents revealed that many FXII(a) inhibitors are in the early preclinical stage, while several already showed efficacy in vivo animal models of thrombosis, sepsis, hereditary angioedema, and multiple sclerosis. Two anti-FXIIa agents namely tick protein Ir-CPI and monoclonal antibody CSL312 are currently in human clinical trials. The results of these trials and further studies of FXII(a) pathophysiological functions will encourage the development of new FXII(a) inhibitors.
Collapse
Affiliation(s)
- Dmitrii V Kalinin
- Institute of Pharmaceutical and Medicinal Chemistry, University of Münster, Münster, Germany
| |
Collapse
|
12
|
Korff M, Imberg L, Will JM, Bückreiß N, Kalinina SA, Wenzel BM, Kastner GA, Daniliuc CG, Barth M, Ovsepyan RA, Butov KR, Humpf HU, Lehr M, Panteleev MA, Poso A, Karst U, Steinmetzer T, Bendas G, Kalinin DV. Acylated 1H-1,2,4-Triazol-5-amines Targeting Human Coagulation Factor XIIa and Thrombin: Conventional and Microscale Synthesis, Anticoagulant Properties, and Mechanism of Action. J Med Chem 2020; 63:13159-13186. [DOI: 10.1021/acs.jmedchem.0c01635] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Marvin Korff
- Institute of Pharmaceutical and Medicinal Chemistry, University of Münster, Corrensstraße 48, 48149 Münster, Germany
| | - Lukas Imberg
- Institute of Pharmaceutical and Medicinal Chemistry, University of Münster, Corrensstraße 48, 48149 Münster, Germany
| | - Jonas M. Will
- Institute of Inorganic and Analytical Chemistry, University of Münster, Corrensstraße 30, 48149 Münster, Germany
| | - Nico Bückreiß
- Pharmaceutical Institute, University of Bonn, An der Immenburg 4, 53121 Bonn, Germany
| | - Svetlana A. Kalinina
- Institute of Food Chemistry, University of Münster, Corrensstraße 45, 48149 Münster, Germany
| | - Benjamin M. Wenzel
- Department of Pharmacy, Institute of Pharmaceutical Chemistry, Philipps University Marburg, Marbacher Weg 6, 35032 Marburg, Germany
| | - Gregor A. Kastner
- Institute of Pharmaceutical and Medicinal Chemistry, University of Münster, Corrensstraße 48, 48149 Münster, Germany
| | - Constantin G. Daniliuc
- Institute for Organic Chemistry, University of Münster, Corrensstraße 40, 48149 Münster, Germany
| | - Maximilian Barth
- Institute of Pharmaceutical and Medicinal Chemistry, University of Münster, Corrensstraße 48, 48149 Münster, Germany
| | - Ruzanna A. Ovsepyan
- Laboratory of Translational Medicine, Dmitriy Rogachev National Medical Research Center of Pediatric Hematology, Oncology, and Immunology, Samory Mashela str. 1, GSP-7, 117997 Moscow, Russia
- Center for Theoretical Problems of Physicochemical Pharmacology, Russian Academy of Sciences, 4 Kosygina St, 119991 Moscow, Russia
| | - Kirill R. Butov
- Laboratory of Translational Medicine, Dmitriy Rogachev National Medical Research Center of Pediatric Hematology, Oncology, and Immunology, Samory Mashela str. 1, GSP-7, 117997 Moscow, Russia
- Center for Theoretical Problems of Physicochemical Pharmacology, Russian Academy of Sciences, 4 Kosygina St, 119991 Moscow, Russia
| | - Hans-Ulrich Humpf
- Institute of Food Chemistry, University of Münster, Corrensstraße 45, 48149 Münster, Germany
| | - Matthias Lehr
- Institute of Pharmaceutical and Medicinal Chemistry, University of Münster, Corrensstraße 48, 48149 Münster, Germany
| | - Mikhail A. Panteleev
- Laboratory of Translational Medicine, Dmitriy Rogachev National Medical Research Center of Pediatric Hematology, Oncology, and Immunology, Samory Mashela str. 1, GSP-7, 117997 Moscow, Russia
- Faculty of Physics, Lomonosov Moscow State University, 1/2 Leninskie gory, 119991 Moscow, Russia
- Center for Theoretical Problems of Physicochemical Pharmacology, Russian Academy of Sciences, 4 Kosygina St, 119991 Moscow, Russia
- Faculty of Biological and Medical Physics, Moscow Institute of Physics and Technology, 9 Institutskii per., 141700 Dolgoprudnyi, Russia
| | - Antti Poso
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, 70211 Kuopio, Finland
- Department of Internal Medicine VIII, University Hospital Tübingen, 72076 Tübingen, Germany
| | - Uwe Karst
- Institute of Inorganic and Analytical Chemistry, University of Münster, Corrensstraße 30, 48149 Münster, Germany
| | - Torsten Steinmetzer
- Department of Pharmacy, Institute of Pharmaceutical Chemistry, Philipps University Marburg, Marbacher Weg 6, 35032 Marburg, Germany
| | - Gerd Bendas
- Pharmaceutical Institute, University of Bonn, An der Immenburg 4, 53121 Bonn, Germany
| | - Dmitrii V. Kalinin
- Institute of Pharmaceutical and Medicinal Chemistry, University of Münster, Corrensstraße 48, 48149 Münster, Germany
| |
Collapse
|
13
|
Wakui M, Fujimori Y, Nakamura S, Oka S, Ozaki Y, Kondo Y, Nakagawa T, Katagiri H, Murata M. Characterisation of antithrombin-dependent anticoagulants through clot waveform analysis to potentially distinguish them from antithrombin-independent inhibitors targeting activated coagulation factors. J Clin Pathol 2020; 74:251-256. [PMID: 32796051 DOI: 10.1136/jclinpath-2020-206835] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 06/26/2020] [Accepted: 06/30/2020] [Indexed: 11/04/2022]
Abstract
AIMS While antithrombin (AT)-independent inhibitors targeting thrombin or activated factor X have been assessed through clot waveform (CWA), there are no reports on assessment with respect to AT-dependent anticoagulants. The present study aims to characterise AT-dependent anticoagulants through CWA to distinguish them from AT-independent inhibitors. METHODS CWA was applied to the activated partial thromboplastin time (APTT) assay of plasma samples spiked with each of AT-dependent drugs (unfractionated heparin, enoxaparin and fondaparinux) and AT-independent drugs (rivaroxaban, apixaban, edoxaban, dabigatran, argatroban, hirudin and bivalirudin), which was performed using the CS-5100 or CN-6000 (Sysmex). The APTT-CWA data were automatically gained by the analyser program. The positive mode of clotting reaction curves was defined as the direction towards fibrin generation. RESULTS Regarding dose-response curves in AT-dependent anticoagulants, the maximum positive values of the first and secondary derivatives (Max1 and Maxp2, respectively) and the maximum negative values of the secondary derivative (Maxn2) seemed to drop to zero without making an asymptotic line, consistent with the irreversibility. Such a feature was observed also in hirudin, as reported previously. Notably, the symmetric property of Max1 peaks in the waveforms was distorted dose dependently in AT independent but not AT-dependent drugs. A plot of Maxp2 logarithm versus Maxn2 logarithm was linear. The slope was about 1 in AT-dependent drugs while that was more than 1 in AT-independent drugs. These features made it possible to distinguish AT-dependent and AT-independent drugs. CONCLUSIONS The results aid in further understanding of the pharmacological aspects of anticoagulation and in screening of candidates for novel anticoagulants.
Collapse
Affiliation(s)
- Masatoshi Wakui
- Department of Laboratory Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Yuta Fujimori
- Office of Clinical Laboratory Technology, Keio University Hospital, Tokyo, Japan
| | - Shoko Nakamura
- Clinical Laboratory, Keio University Hospital, Tokyo, Japan
| | - Shusaku Oka
- Clinical Laboratory, Keio University Hospital, Tokyo, Japan
| | - Yuko Ozaki
- Clinical Laboratory, Keio University Hospital, Tokyo, Japan
| | - Yoshino Kondo
- Clinical Laboratory, Keio University Hospital, Tokyo, Japan
| | | | | | - Mitsuru Murata
- Department of Laboratory Medicine, Keio University School of Medicine, Tokyo, Japan
| |
Collapse
|
14
|
Sivaraja M, Clemens DM, Sizikov S, Dash S, Xu C, Rienzo M, Yang B, Ryan M, Chattopadhyay M, Igoudin L, Chang SS, Keutzer S, Zalicki P, Estiarte MA, Shiau TP, Short KM, Williams DC, Datta A, Pozzi N, Di Cera E, Gibson CM, Fox KAA, Kita DB. VE-1902-A direct thrombin inhibitor with reversible covalent mechanism of action shows efficacy with reduced bleeding in rodent models of thrombosis. Thromb Res 2020; 190:112-121. [PMID: 32339947 DOI: 10.1016/j.thromres.2020.04.020] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 03/18/2020] [Accepted: 04/15/2020] [Indexed: 12/31/2022]
Abstract
INTRODUCTION High incidence of bleeding events remains a key risk for patients taking anticoagulants, especially those in need of long-term combination therapy with antiplatelet agents. As a consequence, patients may not receive clinically indicated combination antithrombotic therapy. Here, we report on VE-1902, a member of a novel class of precision oral anticoagulants (PROACs) that combines effective anticoagulation with reduced bleeding in preclinical testing. METHODS AND RESULTS Acting through covalent, reversible active-site modification of thrombin similar to a previously described molecule [1], VE-1902 shows potency and selectivity for thrombin inhibition in human plasma comparable to clinically relevant direct thrombin inhibitors (DTI) such as argatroban and dabigatran (thrombin generation assay ETP EC50 = 1.3 μM compared to 0.36 μM and 0.31 μM for argatroban and dabigatran; >100-fold selectivity against related serine proteases). Unlike the current anticoagulants, VE-1902 does not significantly inhibit thrombin-mediated platelet activation in in vivo models of thrombosis. In the thrombin generation assay, the compound inhibits thrombin formation without significantly delaying the initiation phase of the clotting cascade. These features are possibly responsible for the observed reduced bleeding in tail bleeding and saphenous vein bleeding models. Consistent with this novel pharmacological profile, VE-1902 shows efficacious anticoagulation in several fibrin-driven animal models of thrombosis (arteriovenous shunt, venous stasis thrombosis, and thrombin-induced thromboembolism models), whereas it does not significantly prevent arterial occlusion in the platelet dependent FeCl3 model. CONCLUSIONS By leaving platelet activation following vascular injury mostly unaffected, VE-1902, and the PROACs more generally, represent a new generation of precision anticoagulants with reduced bleeding risk.
Collapse
Affiliation(s)
| | | | - Sivan Sizikov
- Verseon Corporation, Fremont, CA, United States of America
| | - Subhadra Dash
- Verseon Corporation, Fremont, CA, United States of America
| | - Chengpei Xu
- Verseon Corporation, Fremont, CA, United States of America
| | - Matthew Rienzo
- Verseon Corporation, Fremont, CA, United States of America
| | - Bo Yang
- Verseon Corporation, Fremont, CA, United States of America
| | - Molly Ryan
- Verseon Corporation, Fremont, CA, United States of America
| | | | - Lev Igoudin
- Verseon Corporation, Fremont, CA, United States of America
| | | | - Samuel Keutzer
- Verseon Corporation, Fremont, CA, United States of America
| | - Piotr Zalicki
- Verseon Corporation, Fremont, CA, United States of America
| | | | | | - Kevin M Short
- Verseon Corporation, Fremont, CA, United States of America
| | | | - Anirban Datta
- Verseon Corporation, Fremont, CA, United States of America
| | - Nicola Pozzi
- Edward A. Doisy Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, St. Louis, MO, United States of America
| | - Enrico Di Cera
- Edward A. Doisy Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, St. Louis, MO, United States of America
| | - C Michael Gibson
- TIMI Study Group, Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, United States of America
| | - Keith A A Fox
- Edinburgh Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, United Kingdom
| | - David B Kita
- Verseon Corporation, Fremont, CA, United States of America
| |
Collapse
|
15
|
Cheng B, Liu F, Guo Q, Lu Y, Shi H, Ding A, Xu C. Identification and characterization of hirudin-HN, a new thrombin inhibitor, from the salivary glands of Hirudo nipponia. PeerJ 2019; 7:e7716. [PMID: 31592161 PMCID: PMC6776071 DOI: 10.7717/peerj.7716] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Accepted: 08/21/2019] [Indexed: 01/18/2023] Open
Abstract
Transcriptome sequencing data (6.5 Gb) of the salivary glands of the haematophagous leech Hirudo nipponia was obtained by using the BGIseq-500 platform. After identification and analysis, one transcript (Unigene5370) was annotated to hirudin HV3 from Hirudo medicinalis with an e-value of 1e-29 and was named hirudin-HN. This transcript was a new thrombin inhibitor gene belonging to the proteinase inhibitor I14 (hirudin) family. Hirudin-HN, with a 270-bp cDNA, encodes an 89-aa protein containing a 20-aa signal peptide. The mature hirudin-HN protein contains the typical structural characteristics of hirudin, e.g., three conserved disulfide bonds and the PKP and DFxxIP motifs. Proteins (Hir and M-Hir) were obtained via prokaryotic expression, and the mature hirudin-HN protein was shown to have anticoagulant activity and thrombin affinity by using the chromogenic substrate S2238 and surface plasmon resonance (SPR) interaction analysis, respectively. The N-terminal structure of the mature hirudin-HN protein was shown to be important for anticoagulant activity by comparing the activity and thrombin affinity of Hir and M-Hir. The abundances of Hirudin-HN mRNA and protein were higher in the salivary glands of starving animals than in those of feeding or fed leeches. These results provided a foundation for further study on the structure-function relationship of hirudin-HN with thrombin.
Collapse
Affiliation(s)
- Boxing Cheng
- Institute of Chinese Medicinal Materials, Nanjing Agricultural University, Nanjing, China.,School of Biological Sciences, Guizhou Education University, Gui Yang, China
| | - Fei Liu
- School of Marine and Bioengineering, Yancheng Institute of Technology, Yan Cheng, China
| | - Qiaosheng Guo
- Institute of Chinese Medicinal Materials, Nanjing Agricultural University, Nanjing, China
| | - Yuxi Lu
- Institute of Chinese Medicinal Materials, Nanjing Agricultural University, Nanjing, China
| | - Hongzhuan Shi
- Institute of Chinese Medicinal Materials, Nanjing Agricultural University, Nanjing, China
| | - Andong Ding
- Institute of Chinese Medicinal Materials, Nanjing Agricultural University, Nanjing, China
| | - Chengfeng Xu
- Institute of Chinese Medicinal Materials, Nanjing Agricultural University, Nanjing, China
| |
Collapse
|
16
|
Ábrányi-Balogh P, Petri L, Imre T, Szijj P, Scarpino A, Hrast M, Mitrović A, Fonovič UP, Németh K, Barreteau H, Roper DI, Horváti K, Ferenczy GG, Kos J, Ilaš J, Gobec S, Keserű GM. A road map for prioritizing warheads for cysteine targeting covalent inhibitors. Eur J Med Chem 2018; 160:94-107. [DOI: 10.1016/j.ejmech.2018.10.010] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Revised: 09/07/2018] [Accepted: 10/03/2018] [Indexed: 01/01/2023]
|