1
|
Aoki A, Sugi T, Kawana K, Sugi T, Sakai R. Cross-reactivity of antigenic binding sites of antiphosphatidylserine/prothrombin antibodies in patients with pregnancy loss and epidermal growth factor. J Reprod Immunol 2025; 167:104399. [PMID: 39631139 DOI: 10.1016/j.jri.2024.104399] [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: 11/13/2024] [Accepted: 11/18/2024] [Indexed: 12/07/2024]
Abstract
BACKGROUND Epidermal growth factor (EGF) protein family is essential for implantation and maintenance of normal pregnancy. Results of our previous study on the high incidence of autoantibodies to EGF in patients with pregnancy loss indicated a strong association between EGF autoantibodies and antiphosphatidylserine/prothrombin antibodies (aPS/PT), which are observed in the plasma of patients with thrombosis and adverse pregnancy outcomes. OBJECTIVES To investigate the association between EGF autoantibodies and aPS/PT in patients with pregnancy loss. PATIENTS AND METHODS Plasma specimens were analyzed from patients who experienced pregnancy loss. Direct binding studies using recombinant proteins of the fragments were conducted to determine the antigenic binding sites of aPS/PT, before examining the cross-reactivity of EGF and aPS/PT binding sites. RESULTS Of the 219 patients with pregnancy loss, 26 (11.9 %) were positive for aPS/PT. Moreover, incidence of antihuman EGF autoantibodies (anti-hEGF) was significantly higher in aPS/PT-positive patients than in aPS/PT-negative patients (18/26, 69.2 % and 58/193, 30.1 %, respectively; p = 0.0003). Of the aPS/PT-positive patients, 42.3 % recognized fragment 1 + 2 (F1 + 2), and 61.5 % recognized α-thrombin. Presence of anti-hEGF was correlated with recognition of α-thrombin but not of F1 + 2. Moreover, polyclonal antibodies against EGF recognized α-thrombin, and those against α- thrombin recognized hEGF. CONCLUSIONS In patients with pregnancy loss, aPS/PT recognize F1 + 2 and α-thrombin. Furthermore, α-thrombin and hEGF are immunologically cross-reactive. Therefore, autoantibody-associated disruption of the EGF system may be a cause of pregnancy loss.
Collapse
Affiliation(s)
- Aiko Aoki
- Laboratory for Recurrent Pregnancy Loss, Sugi Women's Clinic, 2-12-1-7 F Shinyokohama, Kohoku, Yokohama, Kanagawa 222-0033, Japan; Department of Obstetrics and Gynecology, Nihon University School of Medicine, 30-1 Oyaguchikamicho, Itabashi-ku, Tokyo 173-0032, Japan
| | - Toshitaka Sugi
- Laboratory for Recurrent Pregnancy Loss, Sugi Women's Clinic, 2-12-1-7 F Shinyokohama, Kohoku, Yokohama, Kanagawa 222-0033, Japan.
| | - Kei Kawana
- Department of Obstetrics and Gynecology, Nihon University School of Medicine, 30-1 Oyaguchikamicho, Itabashi-ku, Tokyo 173-0032, Japan
| | - Toshihiro Sugi
- Laboratory for Recurrent Pregnancy Loss, Sugi Women's Clinic, 2-12-1-7 F Shinyokohama, Kohoku, Yokohama, Kanagawa 222-0033, Japan; Department of Obstetrics and Gynecology, Nihon University School of Medicine, 30-1 Oyaguchikamicho, Itabashi-ku, Tokyo 173-0032, Japan
| | - Rie Sakai
- Laboratory for Recurrent Pregnancy Loss, Sugi Women's Clinic, 2-12-1-7 F Shinyokohama, Kohoku, Yokohama, Kanagawa 222-0033, Japan
| |
Collapse
|
2
|
Takeuchi S, Yamazaki T, Yamaguchi K, Komura F, Tabata T, Nishi H, Azumai S, Miura K, Hirokawa M, Ikemoto K, Kawakami K. Toward the Establishment of a Harmonized Physicochemical Profiling Platform for Therapeutic Oligonucleotides: A Case Study for Aptamers Where the Higher-Order Structure Influences Physical Properties. Mol Pharm 2024; 21:3471-3484. [PMID: 38872243 DOI: 10.1021/acs.molpharmaceut.4c00177] [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] [Indexed: 06/15/2024]
Abstract
Oligonucleotides are short nucleic acids that serve as one of the most promising classes of drug modality. However, attempts to establish a physicochemical evaluation platform of oligonucleotides for acquiring a comprehensive view of their properties have been limited. As the chemical stability and the efficacy as well as the solution properties at a high concentration should be related to their higher-order structure and intra-/intermolecular interactions, their detailed understanding enables effective formulation development. Here, the higher-order structure and the thermodynamic stability of the thrombin-binding aptamer (TBA) and four modified TBAs, which have similar sequences but were expected to have different higher-order structures, were evaluated using ultraviolet spectroscopy (UV), circular dichroism (CD), differential scanning calorimetry (DSC), and nuclear magnetic resonance (NMR). Then, the relationship between the higher-order structure and the solution properties including solubility, viscosity, and stability was investigated. The impact of the higher-order structure on the antithrombin activity was also confirmed. The higher-order structure and intra-/intermolecular interactions of the oligonucleotides were affected by types of buffers because of different potassium concentrations, which are crucial for the formation of the G-quadruplex structure. Consequently, solution properties, such as solubility and viscosity, chemical stability, and antithrombin activity, were also influenced. Each instrumental analysis had a complemental role in investigating the higher-order structure of TBA and modified TBAs. The utility of each physicochemical characterization method during the preclinical developmental stages is also discussed.
Collapse
Affiliation(s)
- Shoko Takeuchi
- Analytical Development, Pharmaceutical Sciences, Takeda Pharmaceutical Co., Ltd., 26-1 Muraoka Higashi 2-Chome, Fujisawa, Kanagawa 2518555, Japan
| | - Tomohiko Yamazaki
- Medical Soft Matter Group, Research Center for Macromolecules and Biomaterials, National Institute for Materials Science, 1-1 Namiki, Tsukuba, Ibaraki 3050044, Japan
| | - Katsutoshi Yamaguchi
- Pharmaceutical Developability, CMC Research, Astellas Pharma Inc., 21 Miyukigaoka, Tsukuba, Ibaraki 3058585, Japan
| | - Fusae Komura
- Analytical Research, Pharmaceutical Science & Technology Unit, Pharmaceutical Profiling & Development Function, Deep Human Biology Learning, Eisai Co., Ltd., 5-1-3 Tokodai, Tsukuba-shi, Ibaraki 3002635, Japan
| | - Takahiro Tabata
- Pharmacokinetics Group, Biological Research Development, Sawai Pharmaceutical Co., Ltd., 5-2-30 Miyahara, Yodogawa-ku, Osaka 5320003, Japan
| | - Hirotaka Nishi
- Formulation Technology Research Laboratories, Pharmaceutical Technology Division, Daiichi Sankyo Co., Ltd., 1-2-58 Hiromachi, Shinagawa-ku, Tokyo 1408710, Japan
| | - Satomi Azumai
- Formulation Technology Research Laboratories, Pharmaceutical Technology Division, Daiichi Sankyo Co., Ltd., 1-2-58 Hiromachi, Shinagawa-ku, Tokyo 1408710, Japan
| | - Kanako Miura
- Formulation Technology Research Laboratories, Pharmaceutical Technology Division, Daiichi Sankyo Co., Ltd., 1-2-58 Hiromachi, Shinagawa-ku, Tokyo 1408710, Japan
| | - Mai Hirokawa
- Modality Pharmaceutical Research Group, CMC Modality Technology Laboratories, Production Technology & Supply Chain Management Division, Mitsubishi Tanabe Pharma Corporation, 7473-2, Onoda, Sanyo-Onoda, Yamaguchi 7560054, Japan
| | - Keisuke Ikemoto
- Modality Pharmaceutical Research Group, CMC Modality Technology Laboratories, Production Technology & Supply Chain Management Division, Mitsubishi Tanabe Pharma Corporation, 7473-2, Onoda, Sanyo-Onoda, Yamaguchi 7560054, Japan
| | - Kohsaku Kawakami
- Medical Soft Matter Group, Research Center for Macromolecules and Biomaterials, National Institute for Materials Science, 1-1 Namiki, Tsukuba, Ibaraki 3050044, Japan
- Graduate School of Pure and Applied Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 3058577, Japan
| |
Collapse
|
3
|
Saladini LY, Magalhães-Junior MJ, da Silva CCF, Oliveira PGC, Kodama RT, Gomes L, Nishiyama-Jr MY, Spencer PJ, da Silva WD, Portaro FCV. Evaluation of the Inhibitory Potential of Synthetic Peptides Homologous to CDR3 Regions of a Monoclonal Antibody against Bothropic Venom Serine Proteases. Int J Mol Sci 2024; 25:5181. [PMID: 38791221 PMCID: PMC11121450 DOI: 10.3390/ijms25105181] [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: 04/01/2024] [Revised: 04/29/2024] [Accepted: 05/01/2024] [Indexed: 05/26/2024] Open
Abstract
Snakebite accidents, neglected tropical diseases per the WHO, pose a significant public health threat due to their severity and frequency. Envenomation by Bothrops genus snakes leads to severe manifestations due to proteolytic enzymes. While the antibothropic serum produced by the Butantan Institute saves lives, its efficacy is limited as it fails to neutralize certain serine proteases. Hence, developing new-generation antivenoms, like monoclonal antibodies, is crucial. This study aimed to explore the inhibitory potential of synthetic peptides homologous to the CDR3 regions of a monoclonal antibody targeting a snake venom thrombin-like enzyme (SVTLE) from B. atrox venom. Five synthetic peptides were studied, all stable against hydrolysis by venoms and serine proteases. Impressively, four peptides demonstrated uncompetitive SVTLE inhibition, with Ki values ranging from 10-6 to 10-7 M. These findings underscore the potential of short peptides homologous to CDR3 regions in blocking snake venom toxins, suggesting their promise as the basis for new-generation antivenoms. Thus, this study offers potential advancements in combatting snakebites, addressing a critical public health challenge in tropical and subtropical regions.
Collapse
Affiliation(s)
- Lucas Yuri Saladini
- Laboratory of Structure and Function of Biomolecules, Butantan Institute, São Paulo 05503-900, Brazil (C.C.F.d.S.); (P.G.C.O.); (R.T.K.); (L.G.)
| | | | | | - Priscila Gonçalves Coutinho Oliveira
- Laboratory of Structure and Function of Biomolecules, Butantan Institute, São Paulo 05503-900, Brazil (C.C.F.d.S.); (P.G.C.O.); (R.T.K.); (L.G.)
| | - Roberto Tadashi Kodama
- Laboratory of Structure and Function of Biomolecules, Butantan Institute, São Paulo 05503-900, Brazil (C.C.F.d.S.); (P.G.C.O.); (R.T.K.); (L.G.)
| | - Lais Gomes
- Laboratory of Structure and Function of Biomolecules, Butantan Institute, São Paulo 05503-900, Brazil (C.C.F.d.S.); (P.G.C.O.); (R.T.K.); (L.G.)
| | - Milton Yutaka Nishiyama-Jr
- Laboratory of Applied Toxinology, Center of Toxins, Immune-Response and Cell Signaling (CeTICS), Instituto Butantan, São Paulo 05503-900, Brazil;
| | - Patrick Jack Spencer
- Biotechnology Center, Nuclear and Energy Research Institute (IPEN/CNEN/SP), São Paulo 05503-900, Brazil;
| | | | - Fernanda Calheta Vieira Portaro
- Laboratory of Structure and Function of Biomolecules, Butantan Institute, São Paulo 05503-900, Brazil (C.C.F.d.S.); (P.G.C.O.); (R.T.K.); (L.G.)
| |
Collapse
|
4
|
Fevraleva I, Mamchich D, Vinogradov D, Chabaeva Y, Kulikov S, Makarik T, Margaryan V, Manasyan G, Novikova V, Rachina S, Melkonyan G, Lytkina K. Role of Genetic Thrombophilia Markers in Thrombosis Events in Elderly Patients with COVID-19. Genes (Basel) 2023; 14:genes14030644. [PMID: 36980916 PMCID: PMC10048352 DOI: 10.3390/genes14030644] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 02/20/2023] [Accepted: 03/02/2023] [Indexed: 03/08/2023] Open
Abstract
Thrombosis is an extremely dangerous complication in elderly patients with COVID-19. Since the first months of the pandemic, anticoagulants have been mandatory in treatment protocols for patients with COVID-19, unless there are serious contraindications. We set out to discover if genetic thrombophilia factors continue to play a triggering role in the occurrence of thrombosis in patients with COVID-19 with prophylactic or therapeutic anticoagulants. We considered the following genetic markers as risk factors for thrombophilia: G1691A in the FV gene, C677T and A1298C in the MTHFR gene, G20210A and C494T in the FII gene, and (−675) 4G/5G in the PAI-I gene. In a cohort of 176 patients, we did not obtain a reliable result indicating a higher risk of thrombotic complications when taking therapeutic doses of anticoagulants in carriers of genetic markers for thrombophilia except the C494T mutation in the FII gene. However, there was still a pronounced tendency to a higher incidence of thrombosis in patients with markers of hereditary thrombophilia, such as FV G1691A and FII G20210A mutations. The presence of the C494T (Thr165Met) allele in the FII gene in this group of patients showed a statistically significant effect of the mutation on the risk of thrombotic complications despite anticoagulant therapy.
Collapse
Affiliation(s)
- Irina Fevraleva
- National Medical Research Center for Hematology, Novy Zykovski Lane 4a, 125167 Moscow, Russia
- Correspondence:
| | - Daria Mamchich
- Hospital Therapy Department No. 2, I.M. Sechenov First Moscow State Medical University, RF Health Ministry, Bolshaya Pirogovskaya St. 2, Bld. 4, 119435 Moscow, Russia
| | - Dmitriy Vinogradov
- Hospital Therapy Department No. 2, I.M. Sechenov First Moscow State Medical University, RF Health Ministry, Bolshaya Pirogovskaya St. 2, Bld. 4, 119435 Moscow, Russia
| | - Yulia Chabaeva
- National Medical Research Center for Hematology, Novy Zykovski Lane 4a, 125167 Moscow, Russia
| | - Sergey Kulikov
- National Medical Research Center for Hematology, Novy Zykovski Lane 4a, 125167 Moscow, Russia
| | - Tatiana Makarik
- National Medical Research Center for Hematology, Novy Zykovski Lane 4a, 125167 Moscow, Russia
| | - Vahe Margaryan
- Hospital Therapy Department No. 2, I.M. Sechenov First Moscow State Medical University, RF Health Ministry, Bolshaya Pirogovskaya St. 2, Bld. 4, 119435 Moscow, Russia
| | - Georgiy Manasyan
- Hospital Therapy Department No. 2, I.M. Sechenov First Moscow State Medical University, RF Health Ministry, Bolshaya Pirogovskaya St. 2, Bld. 4, 119435 Moscow, Russia
| | - Veronika Novikova
- Hospital Therapy Department No. 2, I.M. Sechenov First Moscow State Medical University, RF Health Ministry, Bolshaya Pirogovskaya St. 2, Bld. 4, 119435 Moscow, Russia
| | - Svetlana Rachina
- Hospital Therapy Department No. 2, I.M. Sechenov First Moscow State Medical University, RF Health Ministry, Bolshaya Pirogovskaya St. 2, Bld. 4, 119435 Moscow, Russia
| | | | - Karine Lytkina
- War Veterans Hospital N3, Startovaya St. 4, 129336 Moscow, Russia
| |
Collapse
|
5
|
Mo Z, Xiao Z, He C. Functional expression of a thrombin exosite I inhibitor triabin in Escherichia coli and elucidation of the role of key residues in its inhibitory activity. Biochimie 2022; 208:13-19. [PMID: 36580989 DOI: 10.1016/j.biochi.2022.12.013] [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: 09/16/2022] [Revised: 12/03/2022] [Accepted: 12/22/2022] [Indexed: 12/28/2022]
Abstract
Triabin, a lipocalin-like thrombin inhibitor from the saliva of the blood-sucking triatomine bug Triatoma pallidipennis, exhibits effective inhibition comparable to hirudin despite binding exclusively at exosite I. Interestingly, it was reported that higher triabin doses would not inhibit thrombin completely, which makes it a promising antithrombotic candidate agent with a larger therapeutic window. However, few structural and functional studies about triabin have been reported in the past three decades, mostly due to the lack of a reliable and practicable recombinant expression technology for this seemingly small protein. In this work, we have adopted the SUMO fusion technology for the expression of triabin in E. coli cells-with facile refolding and purification procedures-and the bioactive triabin was produced in ∼12 mg/L culture medium. Subsequently, the structure-function studies through extensive site-directed mutagenesis reveal that triabin's Phe-106 involved in the hydrophobic contacts plays a surprisingly important role in the thrombin inhibition, in contrast to the negatively charged residues Asp-135 or Glu-128 involved in the salt-bridge interaction. As such, this study complements our understanding of the interaction mechanism of natural thrombin inhibitors, which should facilitate the development of anticoagulant drugs with a novel mode of action against thrombin.
Collapse
Affiliation(s)
- Zeyuan Mo
- School of Chemistry and Chemical Engineering, South China University of Technology, Guangzhou, 510640, PR China
| | - Zhenbang Xiao
- School of Chemistry and Chemical Engineering, South China University of Technology, Guangzhou, 510640, PR China
| | - Chunmao He
- School of Chemistry and Chemical Engineering, South China University of Technology, Guangzhou, 510640, PR China.
| |
Collapse
|
6
|
Exosite Binding in Thrombin: A Global Structural/Dynamic Overview of Complexes with Aptamers and Other Ligands. Int J Mol Sci 2021; 22:ijms221910803. [PMID: 34639143 PMCID: PMC8509272 DOI: 10.3390/ijms221910803] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 09/24/2021] [Accepted: 10/01/2021] [Indexed: 12/13/2022] Open
Abstract
Thrombin is the key enzyme of the entire hemostatic process since it is able to exert both procoagulant and anticoagulant functions; therefore, it represents an attractive target for the developments of biomolecules with therapeutic potential. Thrombin can perform its many functional activities because of its ability to recognize a wide variety of substrates, inhibitors, and cofactors. These molecules frequently are bound to positively charged regions on the surface of protein called exosites. In this review, we carried out extensive analyses of the structural determinants of thrombin partnerships by surveying literature data as well as the structural content of the Protein Data Bank (PDB). In particular, we used the information collected on functional, natural, and synthetic molecular ligands to define the anatomy of the exosites and to quantify the interface area between thrombin and exosite ligands. In this framework, we reviewed in detail the specificity of thrombin binding to aptamers, a class of compounds with intriguing pharmaceutical properties. Although these compounds anchor to protein using conservative patterns on its surface, the present analysis highlights some interesting peculiarities. Moreover, the impact of thrombin binding aptamers in the elucidation of the cross-talk between the two distant exosites is illustrated. Collectively, the data and the work here reviewed may provide insights into the design of novel thrombin inhibitors.
Collapse
|
7
|
Vadivel K, Schmidt AE, Cascio D, Padmanabhan K, Krishnaswamy S, Brandstetter H, Bajaj SP. Structure of human factor VIIa-soluble tissue factor with calcium, magnesium and rubidium. Acta Crystallogr D Struct Biol 2021; 77:809-819. [PMID: 34076594 PMCID: PMC8171065 DOI: 10.1107/s2059798321003922] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Accepted: 04/12/2021] [Indexed: 11/10/2022] Open
Abstract
Coagulation factor VIIa (FVIIa) consists of a γ-carboxyglutamic acid (GLA) domain, two epidermal growth factor-like (EGF) domains and a protease domain. FVIIa binds three Mg2+ ions and four Ca2+ ions in the GLA domain, one Ca2+ ion in the EGF1 domain and one Ca2+ ion in the protease domain. Further, FVIIa contains an Na+ site in the protease domain. Since Na+ and water share the same number of electrons, Na+ sites in proteins are difficult to distinguish from waters in X-ray structures. Here, to verify the Na+ site in FVIIa, the structure of the FVIIa-soluble tissue factor (TF) complex was solved at 1.8 Å resolution containing Mg2+, Ca2+ and Rb+ ions. In this structure, Rb+ replaced two Ca2+ sites in the GLA domain and occupied three non-metal sites in the protease domain. However, Rb+ was not detected at the expected Na+ site. In kinetic experiments, Na+ increased the amidolytic activity of FVIIa towards the synthetic substrate S-2288 (H-D-Ile-Pro-Arg-p-nitroanilide) by ∼20-fold; however, in the presence of Ca2+, Na+ had a negligible effect. Ca2+ increased the hydrolytic activity of FVIIa towards S-2288 by ∼60-fold in the absence of Na+ and by ∼82-fold in the presence of Na+. In molecular-dynamics simulations, Na+ stabilized the two Na+-binding loops (the 184-loop and 220-loop) and the TF-binding region spanning residues 163-180. Ca2+ stabilized the Ca2+-binding loop (the 70-loop) and Na+-binding loops but not the TF-binding region. Na+ and Ca2+ together stabilized both the Na+-binding and Ca2+-binding loops and the TF-binding region. Previously, Rb+ has been used to define the Na+ site in thrombin; however, it was unsuccessful in detecting the Na+ site in FVIIa. A conceivable explanation for this observation is provided.
Collapse
Affiliation(s)
- Kanagasabai Vadivel
- Department of Orthopaedic Surgery, University of California, Los Angeles, CA 90095, USA
| | - Amy E. Schmidt
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Duilio Cascio
- DOE Institute for Genomics and Proteomics, University of California, Los Angeles, CA 90095, USA
| | | | - Sriram Krishnaswamy
- Division of Hematology, The Children’s Hospital of Philadelphia University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Hans Brandstetter
- Department of Biosciences, University of Salzburg, 5020 Salzburg, Austria
| | - S. Paul Bajaj
- Department of Orthopaedic Surgery, University of California, Los Angeles, CA 90095, USA
- Molecular Biology Institute, University of California, Los Angeles, CA 90095, USA
| |
Collapse
|
8
|
Liu L, Shan LP, Zhou Y, Chen J. Small Molecule Inhibitors of White Spot Syndrome Virus: Promise in Shrimp Seedling Culture. Int J Mol Sci 2021; 22:ijms22073450. [PMID: 33810591 PMCID: PMC8036603 DOI: 10.3390/ijms22073450] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 03/22/2021] [Accepted: 03/23/2021] [Indexed: 12/29/2022] Open
Abstract
Rapid production of prawn (Litopenaeus vannamei) under artificial pressure can result in a series of obvious challenges and is vulnerable to serious losses related to aquatic environmental issues and the unrestrained outbreak of white spot syndrome (WSS). However, to date, there are no therapeutic strategies to contain the spread of the virus. Here, we synthesized 27 coumarin derivatives and evaluated their anti-white spot syndrome virus (WSSV) activity in L. vannamei larvae. We demonstrated that electron-withdrawing and electron-giving substituent groups play an important role in reducing toxicity and WSSV replication, respectively. Two coumarin C2 (2-amino-5-oxo-4-(p-tolyl)-4H,5H-pyrano[3,2-c]chromene-3-carbonitrile) and C7 (2-amino-4-(4-chlorophenyl)-5-oxo-4H,5H-pyrano[3,2-c]chromene-3-carbonitrile) were regarded as the most promising anti-WSSV compounds with maximum antiviral response <5% and median effective concentration <10 mg/L. The mortality of WSSV-infected larvae decreased by more than 60% after exposure to C2 and C7. With continuous immersion of C2 and C7 exchange, the mortality further decreased to 40% at 120 h. Additionally, C2 and C7 are the relatively stable in aquacultural water, making these agents suitable for use in inhibiting WSSV horizontal transmission in static aquaculture systems. These results showed the marked advantages of using C2 and C7 in the shrimp industry, and suggest that they hold potential for the treatment and prevention of WSSV infection in shrimp seedling culture.
Collapse
Affiliation(s)
- Lei Liu
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo 315211, China; (L.L.); (L.-P.S.); (Y.Z.)
- Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Meishan Campus, Ningbo University, Ningbo 315832, China
- Key Laboratory of Applied Marine Biotechnology of Ministry of Education, Meishan Campus, Ningbo University, Ningbo 315832, China
| | - Li-Peng Shan
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo 315211, China; (L.L.); (L.-P.S.); (Y.Z.)
- Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Meishan Campus, Ningbo University, Ningbo 315832, China
- Key Laboratory of Applied Marine Biotechnology of Ministry of Education, Meishan Campus, Ningbo University, Ningbo 315832, China
| | - Yan Zhou
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo 315211, China; (L.L.); (L.-P.S.); (Y.Z.)
- Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Meishan Campus, Ningbo University, Ningbo 315832, China
- Key Laboratory of Applied Marine Biotechnology of Ministry of Education, Meishan Campus, Ningbo University, Ningbo 315832, China
| | - Jiong Chen
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo 315211, China; (L.L.); (L.-P.S.); (Y.Z.)
- Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Meishan Campus, Ningbo University, Ningbo 315832, China
- Key Laboratory of Applied Marine Biotechnology of Ministry of Education, Meishan Campus, Ningbo University, Ningbo 315832, China
- Correspondence:
| |
Collapse
|
9
|
Mapping specificity, cleavage entropy, allosteric changes and substrates of blood proteases in a high-throughput screen. Nat Commun 2021; 12:1693. [PMID: 33727531 PMCID: PMC7966775 DOI: 10.1038/s41467-021-21754-8] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Accepted: 02/10/2021] [Indexed: 02/06/2023] Open
Abstract
Proteases are among the largest protein families and critical regulators of biochemical processes like apoptosis and blood coagulation. Knowledge of proteases has been expanded by the development of proteomic approaches, however, technology for multiplexed screening of proteases within native environments is currently lacking behind. Here we introduce a simple method to profile protease activity based on isolation of protease products from native lysates using a 96FASP filter, their analysis in a mass spectrometer and a custom data analysis pipeline. The method is significantly faster, cheaper, technically less demanding, easy to multiplex and produces accurate protease fingerprints. Using the blood cascade proteases as a case study, we obtain protease substrate profiles that can be used to map specificity, cleavage entropy and allosteric effects and to design protease probes. The data further show that protease substrate predictions enable the selection of potential physiological substrates for targeted validation in biochemical assays. Characterizing proteases in their native environment is still challenging. Here, the authors develop a proteomics workflow for analyzing protease-specific peptides from cell lysates in 96-well format, providing mechanistic insights into blood proteases and enabling the prediction of protease substrates.
Collapse
|
10
|
Agten SM, Watson EE, Ripoll‐Rozada J, Dowman LJ, Wu MCL, Alwis I, Jackson SP, Pereira PJB, Payne RJ. Potent Trivalent Inhibitors of Thrombin through Hybridization of Salivary Sulfopeptides from Hematophagous Arthropods. Angew Chem Int Ed Engl 2021. [DOI: 10.1002/ange.202015127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Stijn M. Agten
- School of Chemistry and ARC Centre of Excellence for Innovations in Peptide and Protein Science The University of Sydney Sydney NSW 2006 Australia
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM) Maastricht University Universiteitssingel 50 6229 ER Maastricht The Netherlands
| | - Emma E. Watson
- School of Chemistry and ARC Centre of Excellence for Innovations in Peptide and Protein Science The University of Sydney Sydney NSW 2006 Australia
| | - Jorge Ripoll‐Rozada
- IBMC—Instituto de Biologia Molecular e Celular and Instituto de Investigação e Inovação em Saúde Universidade do Porto 4200-135 Porto Portugal
| | - Luke J. Dowman
- School of Chemistry and ARC Centre of Excellence for Innovations in Peptide and Protein Science The University of Sydney Sydney NSW 2006 Australia
| | - Mike C. L. Wu
- Charles Perkins Centre The University of Sydney Sydney NSW 2006 Australia
- Heart Research Institute Sydney NSW 2042 Australia
| | - Imala Alwis
- Charles Perkins Centre The University of Sydney Sydney NSW 2006 Australia
- Heart Research Institute Sydney NSW 2042 Australia
| | - Shaun P. Jackson
- Charles Perkins Centre The University of Sydney Sydney NSW 2006 Australia
- Heart Research Institute Sydney NSW 2042 Australia
| | - Pedro José Barbosa Pereira
- IBMC—Instituto de Biologia Molecular e Celular and Instituto de Investigação e Inovação em Saúde Universidade do Porto 4200-135 Porto Portugal
| | - Richard J. Payne
- School of Chemistry and ARC Centre of Excellence for Innovations in Peptide and Protein Science The University of Sydney Sydney NSW 2006 Australia
| |
Collapse
|
11
|
Agten SM, Watson EE, Ripoll-Rozada J, Dowman LJ, Wu MCL, Alwis I, Jackson SP, Pereira PJB, Payne RJ. Potent Trivalent Inhibitors of Thrombin through Hybridization of Salivary Sulfopeptides from Hematophagous Arthropods. Angew Chem Int Ed Engl 2021; 60:5348-5356. [PMID: 33345438 DOI: 10.1002/anie.202015127] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 12/14/2020] [Indexed: 12/20/2022]
Abstract
Blood feeding arthropods, such as leeches, ticks, flies and mosquitoes, provide a privileged source of peptidic anticoagulant molecules. These primarily operate through inhibition of the central coagulation protease thrombin by binding to the active site and either exosite I or exosite II. Herein, we describe the rational design of a novel class of trivalent thrombin inhibitors that simultaneously block both exosites as well as the active site. These engineered hybrids were synthesized using tandem diselenide-selenoester ligation (DSL) and native chemical ligation (NCL) reactions in one-pot. The most potent trivalent inhibitors possessed femtomolar inhibition constants against α-thrombin and were selective over related coagulation proteases. A lead hybrid inhibitor possessed potent anticoagulant activity, blockade of both thrombin generation and platelet aggregation in vitro and efficacy in a murine thrombosis model at 1 mg kg-1 . The rational engineering approach described here lays the foundation for the development of potent and selective inhibitors for a range of other enzymatic targets that possess multiple sites for the disruption of protein-protein interactions, in addition to an active site.
Collapse
Affiliation(s)
- Stijn M Agten
- School of Chemistry and ARC Centre of Excellence for Innovations in Peptide and Protein Science, The University of Sydney, Sydney, 2006, NSW, Australia
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Universiteitssingel 50, 6229 ER, Maastricht, The Netherlands
| | - Emma E Watson
- School of Chemistry and ARC Centre of Excellence for Innovations in Peptide and Protein Science, The University of Sydney, Sydney, 2006, NSW, Australia
| | - Jorge Ripoll-Rozada
- IBMC-Instituto de Biologia Molecular e Celular and Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135, Porto, Portugal
| | - Luke J Dowman
- School of Chemistry and ARC Centre of Excellence for Innovations in Peptide and Protein Science, The University of Sydney, Sydney, 2006, NSW, Australia
| | - Mike C L Wu
- Charles Perkins Centre, The University of Sydney, Sydney, NSW, 2006, Australia
- Heart Research Institute, Sydney, NSW, 2042, Australia
| | - Imala Alwis
- Charles Perkins Centre, The University of Sydney, Sydney, NSW, 2006, Australia
- Heart Research Institute, Sydney, NSW, 2042, Australia
| | - Shaun P Jackson
- Charles Perkins Centre, The University of Sydney, Sydney, NSW, 2006, Australia
- Heart Research Institute, Sydney, NSW, 2042, Australia
| | - Pedro José Barbosa Pereira
- IBMC-Instituto de Biologia Molecular e Celular and Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135, Porto, Portugal
| | - Richard J Payne
- School of Chemistry and ARC Centre of Excellence for Innovations in Peptide and Protein Science, The University of Sydney, Sydney, 2006, NSW, Australia
| |
Collapse
|
12
|
Rudzińska M, Daglioglu C, Savvateeva LV, Kaci FN, Antoine R, Zamyatnin AA. Current Status and Perspectives of Protease Inhibitors and Their Combination with Nanosized Drug Delivery Systems for Targeted Cancer Therapy. DRUG DESIGN DEVELOPMENT AND THERAPY 2021; 15:9-20. [PMID: 33442233 PMCID: PMC7797289 DOI: 10.2147/dddt.s285852] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Accepted: 12/15/2020] [Indexed: 12/12/2022]
Abstract
In cancer treatments, many natural and synthetic products have been examined; among them, protease inhibitors are promising candidates for anti-cancer agents. Since dysregulated proteolytic activities can contribute to tumor development and metastasis, antagonization of proteases with tailored inhibitors is an encouraging approach. Although adverse effects of early designs of these inhibitors disappeared after the introduction of next-generation agents, most of the proposed inhibitors did not pass the early stages of clinical trials due to their nonspecific toxicity and lack of pharmacological effects. Therefore, new applications that modulate proteases more specifically and serve their programmed way of administration are highly appreciated. In this context, nanosized drug delivery systems have attracted much attention because preliminary studies have demonstrated that the therapeutic capacity of inhibitors has been improved significantly with encapsulated formulation as compared to their free forms. Here, we address this issue and discuss the current application and future clinical prospects of this potential combination towards targeted protease-based cancer therapy.
Collapse
Affiliation(s)
- Magdalena Rudzińska
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, Moscow 119991, Russia
| | - Cenk Daglioglu
- Biotechnology and Bioengineering Application and Research Center, Integrated Research Centers, Izmir Institute of Technology, Urla, Izmir 35430, Turkey
| | - Lyudmila V Savvateeva
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, Moscow 119991, Russia
| | - Fatma Necmiye Kaci
- Department of Molecular Biology and Genetics, Faculty of Science, Erzurum Technical University, Yakutiye, Erzurum 25050, Turkey
| | - Rodolphe Antoine
- CNRS, Institut Lumière Matière, Univ Lyon, Université Claude Bernard Lyon 1, Lyon F-69622, France
| | - Andrey A Zamyatnin
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, Moscow 119991, Russia.,Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow 119991, Russia.,Department of Biotechnology, Sirius University of Science and Technology, Sochi 354340, Russia
| |
Collapse
|
13
|
Liu L, Song DW, Liu GL, Shan LP, Qiu TX, Chen J. Hydroxycoumarin efficiently inhibits spring viraemia of carp virus infection in vitro and in vivo. Zool Res 2020; 41:395-409. [PMID: 32390373 PMCID: PMC7340527 DOI: 10.24272/j.issn.2095-8137.2020.037] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Spring viremia of carp virus (SVCV) causes devastating losses in aquaculture. Coumarin has an advantageous structure for the design of novel antiviral agents with high affinity and specificity. In this study, we evaluated a hydroxycoumarin medicine, i.e., 7-(6-benzimidazole) coumarin (C10), regarding its anti-SVCV effects in vitro and in vivo. Results showed that up to 12.5 mg/L C10 significantly inhibited SVCV replication in the epithelioma papulosum cyprini (EPC) cell line, with a maximum inhibitory rate of >97%. Furthermore, C10 significantly reduced cell death and relieved cellular morphological damage in SVCV-infected cells. Decreased mitochondrial membrane potential (ΔΨm) also suggested that C10 not only protected mitochondria, but also reduced apoptosis in SVCV-infected cells. For in vivo studies, intraperitoneal injection of C10 resulted in an anti-SVCV effect and substantially enhanced the survival rate of virus-infected zebrafish. Furthermore, C10 significantly enhanced antioxidant enzyme activities and decreased reactive oxygen species (ROS) to maintain antioxidant-oxidant balance within the host, thereby contributing to inhibition of SVCV replication. The up-regulation of six interferon (IFN)-related genes also demonstrated that C10 indirectly activated IFNs for the clearance of SVCV in zebrafish. This was beneficial for the continuous maintenance of antiviral effects because of the low viral loads in fish. Thus, C10 is suggested as a therapeutic agent with great potential against SVCV infection in aquaculture.
Collapse
Affiliation(s)
- Lei Liu
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, Zhejiang 315211, China.,Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Meishan Campus, Ningbo University, Ningbo, Zhejiang 315832, China.,Key Laboratory of Applied Marine Biotechnology of Ministry of Education, Meishan Campus, Ningbo University, Ningbo, Zhejiang 315832, China
| | - Da-Wei Song
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, Zhejiang 315211, China.,Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Meishan Campus, Ningbo University, Ningbo, Zhejiang 315832, China.,Key Laboratory of Applied Marine Biotechnology of Ministry of Education, Meishan Campus, Ningbo University, Ningbo, Zhejiang 315832, China
| | - Guang-Lu Liu
- School of Chemistry & Chemical Engineering, Zhoukou Normal University, Zhoukou, Henan 466001, China
| | - Li-Peng Shan
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, Zhejiang 315211, China.,Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Meishan Campus, Ningbo University, Ningbo, Zhejiang 315832, China.,Key Laboratory of Applied Marine Biotechnology of Ministry of Education, Meishan Campus, Ningbo University, Ningbo, Zhejiang 315832, China
| | - Tian-Xiu Qiu
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, Zhejiang 315211, China.,Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Meishan Campus, Ningbo University, Ningbo, Zhejiang 315832, China.,Key Laboratory of Applied Marine Biotechnology of Ministry of Education, Meishan Campus, Ningbo University, Ningbo, Zhejiang 315832, China
| | - Jiong Chen
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, Zhejiang 315211, China.,Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Meishan Campus, Ningbo University, Ningbo, Zhejiang 315832, China.,Key Laboratory of Applied Marine Biotechnology of Ministry of Education, Meishan Campus, Ningbo University, Ningbo, Zhejiang 315832, China. E-mail:
| |
Collapse
|
14
|
Jackson CM, Esnouf P, Duewer DL. Thrombin: An Approach to Developing a Higher-Order Reference Material and Reference Measurement Procedure for Substance Identity, Amount, and Biological Activities. JOURNAL OF RESEARCH OF THE NATIONAL INSTITUTE OF STANDARDS AND TECHNOLOGY 2020; 125:125021. [PMID: 39035347 PMCID: PMC10871826 DOI: 10.6028/jres.125.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Accepted: 06/30/2020] [Indexed: 07/23/2024]
Abstract
Thrombin, the proteolytic enzyme that catalyzes the transformation of soluble fibrinogen to the polymerized fibrin clot, participates in multiple reactions in blood coagulation in addition to the clotting reaction. Although reference materials have existed for many years, structural characterization and measurement of biological activity have never been sufficient to permit claims of clear metrological traceability for the thrombin preparations. Our current state-of-the-art methods for protein characterization and determination of the catalytic properties of thrombin now make it practical to develop and characterize a metrologically acceptable reference material and reference measurement procedure for thrombin. Specifically, α-thrombin, the biologically produced protease formed during prothrombin activation, is readily available and has been extensively characterized. Dependences of thrombin proteolytic and peptide hydrolytic activities on a variety of substrates, pH, specific ions, and temperature are established, although variability remains for the kinetic parameters that describe thrombin enzymatic action. The roles of specific areas on the surface of the thrombin molecule (exosites) in substrate recognition and catalytic efficiency are described and characterized. It is opportune to develop reference materials of high metrological order and technical feasibility. In this article, we review the properties of α-thrombin important for its preparation and suggest an approach suitable for producing a reference material and a reference measurement procedure that is sensitive to thrombin’s catalytic competency on a variety of substrates.
Collapse
Affiliation(s)
| | | | - David L. Duewer
- National Institute of Standards and Technology, Gaithersburg, MD 20899, USA
| |
Collapse
|
15
|
Khan NI, Mousazadehkasin M, Ghosh S, Tsavalas JG, Song E. An integrated microfluidic platform for selective and real-time detection of thrombin biomarkers using a graphene FET. Analyst 2020; 145:4494-4503. [PMID: 32400815 PMCID: PMC7478360 DOI: 10.1039/d0an00251h] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Lab-on-a-chip technology offers an ideal platform for low-cost, reliable, and easy-to-use diagnostics of key biomarkers needed for early screening of diseases and other health concerns. In this work, a graphene field-effect transistor (GFET) functionalized with target-binding aptamers is used as a biosensor for the detection of thrombin protein biomarker. Furthermore, this GFET is integrated with a microfluidic device for enhanced sensing performances in terms of detection limit, sensitivity, and continuous monitoring. Under this platform, a picomolar limit of detection was achieved for measuring thrombin; in our experiment measured as low as 2.6 pM. FTIR, Raman and UV-Vis spectroscopy measurements were performed to confirm the device functionalization steps. Based on the concentration-dependent calibration curve, a dissociation constant of KD = 375.8 pM was obtained. Continuous real-time measurements were also conducted under a constant gate voltage (VGS) to observe the transient response of the sensor when analyte was introduced to the device. The target selectivity of the sensor platform was evaluated and confirmed by challenging the GFET biosensor with various concentrations of lysozyme protein. The results suggest that this device technology has the potential to be used as a general diagnostic platform for measuring clinically relevant biomarkers for point-of-care applications.
Collapse
Affiliation(s)
- Niazul I Khan
- Department of Electrical and Computer Engineering, University of New Hampshire, Durham, NH 03824, USA.
| | | | | | | | | |
Collapse
|
16
|
Abstract
![]()
Accurate determination
of the binding affinity of the ligand to
the receptor remains a difficult problem in computer-aided drug design.
Here, we study and compare the efficiency of Jarzynski’s equality
(JE) combined with steered molecular dynamics and the linear interaction
energy (LIE) method by assessing the binding affinity of 23 small
compounds to six receptors, including β-lactamase, thrombin,
factor Xa, HIV-1 protease (HIV), myeloid cell leukemia-1, and cyclin-dependent
kinase 2 proteins. It was shown that Jarzynski’s nonequilibrium
binding free energy ΔGneqJar correlates with the available
experimental data with the correlation levels R =
0.89, 0.86, 0.83, 0.80, 0.83, and 0.81 for six data sets, while for
the binding free energy ΔGLIE obtained
by the LIE method, we have R = 0.73, 0.80, 0.42,
0.23, 0.85, and 0.01. Therefore, JE is recommended to be used for
ranking binding affinities as it provides accurate and robust results.
In contrast, LIE is not as reliable as JE, and it should be used with
caution, especially when it comes to new systems.
Collapse
Affiliation(s)
- Kiet Ho
- Institute for Computational Sciences and Technology, Quang Trung Software City, SBI Building, Tan Chanh Hiep Ward, District 12, Ho Chi Minh City, Vietnam
| | - Duc Toan Truong
- Institute for Computational Sciences and Technology, Quang Trung Software City, SBI Building, Tan Chanh Hiep Ward, District 12, Ho Chi Minh City, Vietnam.,Department of Theoretical Physics, Faculty of Physics and Engineering Physics, Ho Chi Minh University of Science, Ho Chi Minh City, Vietnam
| | - Mai Suan Li
- Institute of Physics, Polish Academy of Sciences, Al. Lotnikow 32/46, 02-668 Warsaw, Poland
| |
Collapse
|
17
|
Xu S, Fan F, Liu H, Cheng S, Tu M, Du M. Novel Anticoagulant Peptide from Lactoferrin Binding Thrombin at the Active Site and Exosite-I. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2020; 68:3132-3139. [PMID: 32064873 DOI: 10.1021/acs.jafc.9b08094] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Thrombin is currently one of the important targets for the treatment and prevention of thrombosis. At present, there are few reports on the application of lactoferrin peptides in anticoagulation. In this study, a peptide with the amino acid sequence of LRPVAAEIY (LF-LR) derived from lactoferrin was shown to possess antithrombotic activity. LF-LR (5 mM) significantly prolonged activated partial thromboplastin time, prothrombin time, and thrombin time for 13.4, 1.7, and 5.1 s, respectively. It prolonged the coagulation time of fibrinogen from 15.3 ± 0.4 to 20.2 ± 0.5 s by affecting the conformation of thrombin. Using circular dichroism analysis, LF-LR can increase the α-helix content of thrombin from 25.6 to 56.7% and made the β-sheet disappear. In addition, LF-LR also quenched fluorescence of thrombin at about 346 nm (λEx = 280 nm). By means of molecular docking, it was found that LF-LR could bind to both the active site and the exosite-I of thrombin, and the combined LYS60F, TRP60D, ASP189, LYS36, and ARG77A are typical amino acids in the two domains, respectively.
Collapse
Affiliation(s)
- Shiqi Xu
- School of Food Science and Technology, National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian Liaoning, 116034, China
| | - Fengjiao Fan
- College of Food Science and Engineering, Nanjing University of Finance and Economics, Nanjing 210046, China
| | - Hanxiong Liu
- School of Food Science and Technology, National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian Liaoning, 116034, China
| | - Shuzhen Cheng
- School of Food Science and Technology, National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian Liaoning, 116034, China
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China
| | - Maolin Tu
- School of Food Science and Technology, National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian Liaoning, 116034, China
| | - Ming Du
- School of Food Science and Technology, National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian Liaoning, 116034, China
| |
Collapse
|
18
|
Liu H, Tu M, Cheng S, Xu Z, Xu X, Du M. Anticoagulant Decapeptide Interacts with Thrombin at the Active Site and Exosite-I. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2020; 68:176-184. [PMID: 31850760 DOI: 10.1021/acs.jafc.9b06450] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Thrombin can be used as a target for its inhibitors to prevent blood coagulation. A novel peptide (TKLTEEEKNR, PfCN) identified from αS2-casein (fragments 211-220) with high anticoagulant activity was screened and prepared. The activated partial thromboplastin time, prothrombin time, and thrombin time, at the concentration of 4 mM, prolonged about 19, 2.5 and 5.5 s, respectively. At the same concentration, the fibrinogen clotting time prolonged from 25.5 ± 0.7 to 38.3 ± 1.3 s. The thrombin inhibitory efficiency in vitro (IC50 value of 29.27 mM) and antithrombosis effect in vivo were determined. The secondary structure of thrombin, which was influenced by PfCN, indicates that PfCN can bind to thrombin. Isothermal titration calorimetry and the chromogenic substrate test showed that PfCN belongs to the bivalent thrombin inhibitor like bivalirudin. Although the effect was not as good as bivalirudin, in the animal experiment, bleeding occurred in the bivalirudin group but not in the PfCN group. Moreover, molecular docking illustrates the mechanism for the antithrombin activity of PfCN. These results indicated that PfCN could be used as an effective thrombin inhibitor with broad potential for the prevention of thrombotic acute pulmonary embolism and other thrombotic events.
Collapse
Affiliation(s)
- Hanxiong Liu
- School of Food Science and Technology, National Engineering Research Center of Seafood, Collaborative Innovation Center of Seafood Deep Processing , Dalian Polytechnic University , Dalian 116034 , China
| | - Maolin Tu
- School of Food Science and Technology, National Engineering Research Center of Seafood, Collaborative Innovation Center of Seafood Deep Processing , Dalian Polytechnic University , Dalian 116034 , China
| | - ShuZhen Cheng
- School of Food Science and Technology, National Engineering Research Center of Seafood, Collaborative Innovation Center of Seafood Deep Processing , Dalian Polytechnic University , Dalian 116034 , China
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science and Nutritional Engineering , China Agricultural University , Beijing 100083 , China
| | - Zhe Xu
- School of Food Science and Technology, National Engineering Research Center of Seafood, Collaborative Innovation Center of Seafood Deep Processing , Dalian Polytechnic University , Dalian 116034 , China
| | - Xianbing Xu
- School of Food Science and Technology, National Engineering Research Center of Seafood, Collaborative Innovation Center of Seafood Deep Processing , Dalian Polytechnic University , Dalian 116034 , China
| | - Ming Du
- School of Food Science and Technology, National Engineering Research Center of Seafood, Collaborative Innovation Center of Seafood Deep Processing , Dalian Polytechnic University , Dalian 116034 , China
| |
Collapse
|
19
|
Dahms SO, Demir F, Huesgen PF, Thorn K, Brandstetter H. Sirtilins - the new old members of the vitamin K-dependent coagulation factor family. J Thromb Haemost 2019; 17:470-481. [PMID: 30644641 PMCID: PMC6850207 DOI: 10.1111/jth.14384] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Indexed: 12/04/2022]
Abstract
Essentials Blood coagulation is driven by vitamin K (VK)-dependent proteases. We have identified and characterized 'sirtilin' as an additional VK-dependent protease. Sirtilins emerged early in the evolution of the coagulation system of vertebrates. Ubiquitous occurrence might indicate an important functional role of sirtilins. SUMMARY: Background Vitamin K (VK)-dependent proteases are major players in blood coagulation, including both the initiation and the regulation of the cascade. Five different members of this protease family have been described, comprising the following coagulation factors: factor VII, FIX, FX, protein C (PC), and prothrombin (FII). FVII, FIX, FX and PC share a typical domain architecture, with an N-terminal γ-carboxyglutamate (Gla) domain, two epidermal growth factor-like (EGF) domains, and a C-terminal trypsin-like serine protease (SP) domain. Objectives We have identified uncharacterized proteins in snake genomes showing the typical Gla-EGF1-EGF2-SP domain architecture but relatively low sequence conservation compared to known VK-dependent proteases. On the basis of sequence analysis, we hypothesized that these proteins are functional members of the VK-dependent protease family. Methods/results Using phylogenetic analyses, we confirmed the so-called 'sirtilins' as an additional VK-dependent protease class. These proteases were found in several vertebrates, including jawless fish, cartilaginous fish, bony fish, reptiles, birds, and marsupials, but not in other mammals. The recombinant zymogen form of Thamnophis sirtalis sirtilin was produced by in vitro renaturation, and was activated with human activated FXI. The activated form of sirtilin proteolytically cleaved peptide and protein substrates, including prothrombin. Mass spectrometry-based substrate profiling of sirtilin revealed a narrower sequence specificity than those of FIX and FX. Conclusions The ubiquitous occurrence of sirtilins in many vertebrate classes might indicate an important functional role. Understanding the detailed functions of sirtilins might contribute to a deeper understanding of the evolution and function of the vertebrate coagulation system.
Collapse
Affiliation(s)
- Sven O. Dahms
- Department of BiosciencesUniversity of SalzburgSalzburgAustria
| | - Fatih Demir
- ZEA‐3 AnalyticsCentral Institute for Engineering, Electronics and AnalyticsForschungszentrum JülichJülichGermany
| | - Pitter F. Huesgen
- ZEA‐3 AnalyticsCentral Institute for Engineering, Electronics and AnalyticsForschungszentrum JülichJülichGermany
| | - Karina Thorn
- Haemophilia ResearchNovo Nordisk A/SMåløvDenmark
| | | |
Collapse
|
20
|
Gogoi D, Pal A, Chattopadhyay P, Paul S, Deka RC, Mukherjee AK. First Report of Plant-Derived β-Sitosterol with Antithrombotic, in Vivo Anticoagulant, and Thrombus-Preventing Activities in a Mouse Model. JOURNAL OF NATURAL PRODUCTS 2018; 81:2521-2530. [PMID: 30406661 DOI: 10.1021/acs.jnatprod.8b00574] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Inhibitors of thrombin, a key enzyme in the blood coagulation cascade, are of great interest because of their selective specificity and effectiveness in anticoagulation therapy against cardiovascular disorders. The natural soybean phytosterol, β-sitosterol (BSS) demonstrated anticoagulant activity by dose-dependent inhibition of thrombin in an uncompetitive manner with a Ki value of 0.267 μM as well as by partial inhibition of thrombin-catalyzed platelet aggregation with a half-maximal inhibitory concentration (IC50) value of 10.45 ± 2.88 μM against platelet-rich plasma and 9.2 ± 1.2 μM against washed platelets. An in silico study indicated binding of BSS to thrombin, which was experimentally verified by spectrofluorometric and isothermal calorimetric analyses. Under in vitro conditions, BSS demonstrated thrombolytic activity by activating plasminogen, albeit it is devoid of protease (fibrinogenolytic) activity. BSS was noncytotoxic to mammalian cells, nonhemolytic, demonstrated its in vivo anticoagulant activity when administered orally, and inhibited k-carrageen-induced thrombus formation in the tails of mice. Our results suggest that dietary supplementation of BSS may help to prevent thrombosis-associated cardiovascular disorders.
Collapse
Affiliation(s)
| | - Anirban Pal
- In-vivo Testing Laboratory, Molecular Bioprospection Department , Central Institute of Medicinal and Aromatic Plants , Lucknow , India
| | - Pronobesh Chattopadhyay
- Division of Pharmaceutical Technology , Defense Research Laboratory , Tezpur , Assam , India
| | | | | | | |
Collapse
|
21
|
Kunalan S, Othman I, Syed Hassan S, Hodgson WC. Proteomic Characterization of Two Medically Important Malaysian Snake Venoms, Calloselasma rhodostoma (Malayan Pit Viper) and Ophiophagus hannah (King Cobra). Toxins (Basel) 2018; 10:toxins10110434. [PMID: 30373186 PMCID: PMC6266455 DOI: 10.3390/toxins10110434] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Revised: 10/23/2018] [Accepted: 10/23/2018] [Indexed: 02/07/2023] Open
Abstract
Calloselasma rhodostoma (CR) and Ophiophagus hannah (OH) are two medically important snakes found in Malaysia. While some studies have described the biological properties of these venoms, feeding and environmental conditions also influence the concentration and distribution of snake venom toxins, resulting in variations in venom composition. Therefore, a combined proteomic approach using shotgun and gel filtration chromatography, analyzed by tandem mass spectrometry, was used to examine the composition of venoms from these Malaysian snakes. The analysis revealed 114 proteins (15 toxin families) and 176 proteins (20 toxin families) in Malaysian Calloselasma rhodostoma and Ophiophagus hannah species, respectively. Flavin monoamine oxidase, phospholipase A2, phosphodiesterase, snake venom metalloproteinase, and serine protease toxin families were identified in both venoms. Aminopeptidase, glutaminyl-peptide cyclotransferase along with ankyrin repeats were identified for the first time in CR venom, and insulin, c-type lectins/snaclecs, hepatocyte growth factor, and macrophage colony-stimulating factor together with tumor necrosis factor were identified in OH venom for the first time. Our combined proteomic approach has identified a comprehensive arsenal of toxins in CR and OH venoms. These data may be utilized for improved antivenom production, understanding pathological effects of envenoming, and the discovery of biologically active peptides with medical and/or biotechnological value.
Collapse
Affiliation(s)
- Sugita Kunalan
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway, Selangor Darul Ehsan 47500, Malaysia.
| | - Iekhsan Othman
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway, Selangor Darul Ehsan 47500, Malaysia.
| | - Sharifah Syed Hassan
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway, Selangor Darul Ehsan 47500, Malaysia.
| | - Wayne C Hodgson
- Monash Venom Group, Department of Pharmacology, Faculty of Medicine, Nursing and Health Sciences, Monash University, Victoria 3800, Australia.
| |
Collapse
|
22
|
Kim H, An Z, Jang CH. Label-free optical detection of thrombin using a liquid crystal-based aptasensor. Microchem J 2018. [DOI: 10.1016/j.microc.2018.05.010] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
23
|
Chérifi F, Saoud S, Laraba-Djebari F. Molecular modeling, biochemical characterization, and pharmacological properties of Cc 3 -SPase: A platelet-aggregating thrombin-like enzyme purified from Cerastes cerastes venom. J Biochem Mol Toxicol 2018; 32:e22165. [PMID: 29979475 DOI: 10.1002/jbt.22165] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2018] [Revised: 05/23/2018] [Accepted: 06/15/2018] [Indexed: 01/25/2023]
Abstract
Cc3 -SPase (30 kDa-proteinase; pI 5.98) was isolated from Cerastes cerastes venom. Its sequence of 271 residues yielded from LC-MALDI-TOF showed high degrees of homology when aligned with other proteinases. Cc3 -SPase cleaved natural and synthetic proteins such as casein and fibrinogen leaving fibrin clots unaffected. Cc3 -SPase was fully abolished by ion chelators, whereas aprotinin, antithrombin III (Sigma Aldrich, Saint-Louis, Missouri, USA), and heparin were ineffective. Affinity of Cc3 -SPase to benzamidine indicated the presence of an aspartate residue in the catalytic site as confirmed by three-dimensional structure consisting of 14 β-strands and four α-helices. Molecular mechanisms revealed that Cc3 -SPase is capable of promoting dysfunctional platelet aggregation via two signaling pathways mediated by the G-coupled protein receptors and αIIbβ3 integrin. Cc3 -SPase is involved in both extrinsic/intrinsic coagulation pathways in deficient plasmas by replacing defective/lacking factors FII, FVII, and FVIII but not FX. Cc3 -SPase could substitute missing factors in blood diseases related to plasma factor deficiencies.
Collapse
Affiliation(s)
- Fatah Chérifi
- USTHB, Faculty of Biological Sciences, Laboratory of Cellular and Molecular Biology, Bab Ezzouar, Algiers, Algeria
| | - Samah Saoud
- USTHB, Faculty of Biological Sciences, Laboratory of Cellular and Molecular Biology, Bab Ezzouar, Algiers, Algeria
| | - Fatima Laraba-Djebari
- USTHB, Faculty of Biological Sciences, Laboratory of Cellular and Molecular Biology, Bab Ezzouar, Algiers, Algeria
| |
Collapse
|
24
|
Dolot R, Lam CH, Sierant M, Zhao Q, Liu FW, Nawrot B, Egli M, Yang X. Crystal structures of thrombin in complex with chemically modified thrombin DNA aptamers reveal the origins of enhanced affinity. Nucleic Acids Res 2018; 46:4819-4830. [PMID: 29684204 PMCID: PMC5961234 DOI: 10.1093/nar/gky268] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Revised: 03/26/2018] [Accepted: 04/15/2018] [Indexed: 01/11/2023] Open
Abstract
Thrombin-binding aptamer (TBA) is a DNA 15-mer of sequence 5'-GGT TGG TGT GGT TGG-3' that folds into a G-quadruplex structure linked by two T-T loops located on one side and a T-G-T loop on the other. These loops are critical for post-SELEX modification to improve TBA target affinity. With this goal in mind we synthesized a T analog, 5-(indolyl-3-acetyl-3-amino-1-propenyl)-2'-deoxyuridine (W) to substitute one T or a pair of Ts. Subsequently, the affinity for each analog was determined by biolayer interferometry. An aptamer with W at position 4 exhibited about 3-fold increased binding affinity, and replacing both T4 and T12 with W afforded an almost 10-fold enhancement compared to native TBA. To better understand the role of the substituent's aromatic moiety, an aptamer with 5-(methyl-3-acetyl-3-amino-1-propenyl)-2'-deoxyuridine (K; W without the indole moiety) in place of T4 was also synthesized. This K4 aptamer was found to improve affinity 7-fold relative to native TBA. Crystal structures of aptamers with T4 replaced by either W or K bound to thrombin provide insight into the origins of the increased affinities. Our work demonstrates that facile chemical modification of a simple DNA aptamer can be used to significantly improve its binding affinity for a well-established pharmacological target protein.
Collapse
Affiliation(s)
- Rafal Dolot
- Centre of Molecular and Macromolecular Studies, Polish Academy of Sciences, 90–363 Lodz, Sienkiewicza 112, Poland
| | - Curtis H Lam
- AM Biotechnologies, LLC, 12521 Gulf Freeway, Houston, TX 77034, USA
| | - Malgorzata Sierant
- Centre of Molecular and Macromolecular Studies, Polish Academy of Sciences, 90–363 Lodz, Sienkiewicza 112, Poland
| | - Qiang Zhao
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
| | - Feng-Wu Liu
- School of Pharmaceutical Sciences, Zhengzhou University, Science Avenue 100, Zhengzhou 450001, Henan, China
| | - Barbara Nawrot
- Centre of Molecular and Macromolecular Studies, Polish Academy of Sciences, 90–363 Lodz, Sienkiewicza 112, Poland
| | - Martin Egli
- Department of Biochemistry, Vanderbilt University, School of Medicine, Nashville, TN 37232, USA
| | - Xianbin Yang
- AM Biotechnologies, LLC, 12521 Gulf Freeway, Houston, TX 77034, USA
| |
Collapse
|
25
|
Wang A, Ma X, Ye Y, Luo F, Guo L, Qiu B, Lin Z, Chen G. A Simple and Convenient Aptasensor for Protein Using an Electronic Balance as a Readout. Anal Chem 2017; 90:1087-1091. [PMID: 29231705 DOI: 10.1021/acs.analchem.7b03823] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The electronic balance, one of the most common pieces of equipment in the laboratory, is normally used to directly measure the weight of a target with high accuracy. However, little attention has been paid to the extension of its applications. In this study, an electronic balance was used as a readout to develop a novel aptasensor for protein quantification for the first time. Thrombin was selected as a model target, and its two aptamers recognizing different sites of the protein were used (one aptamer was immobilized on the surface of magnetic microparticles and the other aptamer was functionalized with platinum nanoparticles). The two aptamers were specifically bound with the thrombin to form a sandwich structure; thus, the platinum nanoparticles were linked to the magnetic microparticles, and they were separated by a magnet easily. The captured platinum nanoparticles effectively catalyzed the decomposition of H2O2, generating a large volume of O2 to discharge a certain amount of water in a drainage device, because the pressure in the vial is higher than that outside of the vial. The weight of water was accurately measured by an electronic balance. The weight of water increased with the increasing of the thrombin concentration in the range of 0 to 100 nM with a detection limit of 2.8 nM. This is the first time the use of an electronic balance as a signal readout for biomolecule quantitation in bioassay has been reported.
Collapse
Affiliation(s)
- Alian Wang
- Department of Science Research and Training, Fujian Institute of Education , Fuzhou, Fujian 350001, China
| | | | - Yanzhu Ye
- Department of Science Research and Training, Fujian Institute of Education , Fuzhou, Fujian 350001, China
| | | | | | | | | | | |
Collapse
|
26
|
Brahma RK, Blanchet G, Kaur S, Manjunatha Kini R, Doley R. Expression and characterization of haemathrins, madanin-like thrombin inhibitors, isolated from the salivary gland of tick Haemaphysalis bispinosa (Acari: Ixodidae). Thromb Res 2017; 152:20-29. [DOI: 10.1016/j.thromres.2017.01.012] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Revised: 01/26/2017] [Accepted: 01/27/2017] [Indexed: 11/28/2022]
|
27
|
Iyer JK, Koh CY, Kazimirova M, Roller L, Jobichen C, Swaminathan K, Mizuguchi J, Iwanaga S, Nuttall PA, Chan MY, Kini RM. Avathrin: a novel thrombin inhibitor derived from a multicopy precursor in the salivary glands of the ixodid tick,
Amblyomma variegatum. FASEB J 2017; 31:2981-2995. [DOI: 10.1096/fj.201601216r] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Accepted: 03/13/2017] [Indexed: 12/23/2022]
Affiliation(s)
- Janaki Krishnamoorthy Iyer
- Protein Science LaboratoryDepartment of Biological SciencesNational University of Singapore Singapore
- Department of Pathology and Molecular MedicineMcMaster UniversityHamilton Ontario Canada
| | - Cho Yeow Koh
- Protein Science LaboratoryDepartment of Biological SciencesNational University of Singapore Singapore
| | - Maria Kazimirova
- Institute of ZoologySlovak Academy of SciencesBratislava Slovakia
| | - Ladislav Roller
- Institute of ZoologySlovak Academy of SciencesBratislava Slovakia
| | - Chacko Jobichen
- Protein Science LaboratoryDepartment of Biological SciencesNational University of Singapore Singapore
| | | | - Jun Mizuguchi
- The Chemo‐Sero‐Therapeutic Research InstituteKumamoto Japan
| | | | | | - Mark Y. Chan
- Yong Loo Lin School of MedicineNational University of Singapore Singapore
- Department of CardiologyNational University Heart Centre Singapore
| | - R. Manjunatha Kini
- Protein Science LaboratoryDepartment of Biological SciencesNational University of Singapore Singapore
- Singapore Eye Research Institute Singapore
| |
Collapse
|
28
|
Phipps MJS, Fox T, Tautermann CS, Skylaris CK. Intuitive Density Functional Theory-Based Energy Decomposition Analysis for Protein–Ligand Interactions. J Chem Theory Comput 2017; 13:1837-1850. [DOI: 10.1021/acs.jctc.6b01230] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- M. J. S. Phipps
- School of Chemistry, University of Southampton, Highfield, Southampton SO17 1BJ, U.K
| | - T. Fox
- Department of Medicinal Chemistry, Boehringer Ingelheim Pharma GmbH & Co. KG, 88397 Biberach, Germany
| | - C. S. Tautermann
- Department of Medicinal Chemistry, Boehringer Ingelheim Pharma GmbH & Co. KG, 88397 Biberach, Germany
| | - C.-K. Skylaris
- School of Chemistry, University of Southampton, Highfield, Southampton SO17 1BJ, U.K
| |
Collapse
|
29
|
Hassan MZ, Osman H, Ali MA, Ahsan MJ. Therapeutic potential of coumarins as antiviral agents. Eur J Med Chem 2016; 123:236-255. [PMID: 27484512 PMCID: PMC7115672 DOI: 10.1016/j.ejmech.2016.07.056] [Citation(s) in RCA: 217] [Impact Index Per Article: 24.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Revised: 07/21/2016] [Accepted: 07/23/2016] [Indexed: 12/24/2022]
Abstract
Coumarins have received a considerable attention in the last three decades as a lead structures for the discovery of orally bioavailable non-peptidic antiviral agents. A lot of structurally diverse coumarins analogues were found to display remarkable array of affinity with the different molecular targets for antiviral agents and slight modifications around the central motif result in pronounced changes in its antiviral spectrum. This manuscript thoroughly reviews the design, discovery and structure-activity relationship studies of the coumarin analogues as antiviral agents focusing mainly on lead optimization and its development into clinical candidates.
Collapse
Affiliation(s)
- Mohd Zaheen Hassan
- School of Chemical Sciences, Universiti Sains Malaysia, Minden, 11800 Penang, Malaysia; Department of Pharmaceutical Chemistry, Alwar Pharmacy College, M.I.A., Alwar, Rajasthan 301030, India.
| | - Hasnah Osman
- School of Chemical Sciences, Universiti Sains Malaysia, Minden, 11800 Penang, Malaysia.
| | - Mohamed Ashraf Ali
- Institute for Research in Molecular Medicine, Universiti Sains Malaysia, Minden, 11800 Penang, Malaysia
| | | |
Collapse
|
30
|
Design and characterization of an APC-specific serpin for the treatment of hemophilia. Blood 2016; 129:105-113. [PMID: 27789479 DOI: 10.1182/blood-2016-05-718635] [Citation(s) in RCA: 115] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Accepted: 08/06/2016] [Indexed: 01/08/2023] Open
Abstract
Hemophilia is a bleeding disorder caused by deficiency in factors VIII or IX, the two components of the intrinsic Xase complex. Treatment with replacement factor can lead to the development of inhibitory antibodies, requiring the use of bypassing agents such as factor VIIa and factor concentrates. An alternative approach to bypass the Xase complex is to inhibit endogenous anticoagulant activities. Activated protein C (APC) breaks down the complex that produces thrombin by proteolytically inactivating factor Va. Defects in this mechanism (eg, factor V Leiden) are associated with thrombosis but result in less severe bleeding when co-inherited with hemophilia. Selective inhibition of APC might therefore be effective for the treatment of hemophilia. The endogenous inhibitors of APC are members of the serpin family: protein C inhibitor (PCI) and α1-antitrypsin (α1AT); however, both exhibit poor reactivity and selectivity for APC. We mutated residues in and around the scissile P1-P1' bond in PCI and α1AT, resulting in serpins with the desired specificity profile. The lead candidate was shown to promote thrombin generation in vitro and to restore fibrin and platelet deposition in an intravital laser injury model in hemophilia B mice. The power of targeting APC was further demonstrated by the complete normalization of bleeding after a severe tail clip injury in these mice. These results demonstrate that the protein C anticoagulant system can be successfully targeted by engineered serpins and that administration of such agents is effective at restoring hemostasis in vivo.
Collapse
|
31
|
Jackson CV, Satterwhite J, Roberts E. Preclinical and Clinical Pharmacology of Efegatran (LY294468) : A Novel Antithrombin for the Treatment of Acute Coronary Syndromes. Clin Appl Thromb Hemost 2016. [DOI: 10.1177/107602969600200406] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Efegatran (LY294468), a tripeptide arginal in hibitor of the catalytic site of thrombin, is being devel oped as a parenteral anticoagulant for the treatment of acute coronary syndromes. Efegatran was studied in a canine model of coronary artery thrombosis to determine its ability to prevent thrombus formation and as an ad junctive anticoagulant to thrombolysis, in phase I clinical studies, and phase II clinical studies in unstable angina. In the preclinical in vivo studies in dogs, efegatran pro duced a dose-dependent increase in clotting times and demonstrated a selectivity for thrombin time (TT) changes. The activated partial thromboplastin time (APTT)-TT ratio (that is, based on the dose to double each clotting time) determined in dogs from ex vivo blood samples was 8: 1. This observation was similar to that obtained during the phase I studies in normal volunteers where the APTT-TT ratio was 12:1. The canine and hu man clotting systems responded similarly at doses of efe gatran where comparisons could be made (0.25-1.0 mg/ kg/h). The kinetics of the anticoagulant activity of efega tran in dogs and humans were linear and nonsaturable over the dose ranges studied. Efegatran was also found to be an effective adjunctive anticoagulant during streptoki nase-induced thrombolysis in dogs, preventing reocclu sion without increasing bleeding risk. The novel an tithrombin, efegatran, has demonstrated dose-dependent and safe anticoagulation in animal and human studies. Efegatran is presently undergoing phase II clinical studies in unstable angina and acute myocardial infarction pa tients.
Collapse
Affiliation(s)
| | | | - Eiry Roberts
- Lilly Research Laboratories, Indianapolis, Indiana, U.S.A
| |
Collapse
|
32
|
Bowerman S, Wereszczynski J. Detecting Allosteric Networks Using Molecular Dynamics Simulation. Methods Enzymol 2016; 578:429-47. [PMID: 27497176 DOI: 10.1016/bs.mie.2016.05.027] [Citation(s) in RCA: 69] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Abstract
Allosteric networks allow enzymes to transmit information and regulate their catalytic activities over vast distances. In principle, molecular dynamics (MD) simulations can be used to reveal the mechanisms that underlie this phenomenon; in practice, it can be difficult to discern allosteric signals from MD trajectories. Here, we describe how MD simulations can be analyzed to reveal correlated motions and allosteric networks, and provide an example of their use on the coagulation enzyme thrombin. Methods are discussed for calculating residue-pair correlations from atomic fluctuations and mutual information, which can be combined with contact information to identify allosteric networks and to dynamically cluster a system into highly correlated communities. In the case of thrombin, these methods show that binding of the antagonist hirugen significantly alters the enzyme's correlation landscape through a series of pathways between Exosite I and the catalytic core. Results suggest that hirugen binding curtails dynamic diversity and enforces stricter venues of influence, thus reducing the accessibility of thrombin to other molecules.
Collapse
Affiliation(s)
- S Bowerman
- Center for Molecular Study of Condensed Soft Matter, Illinois Institute of Technology, Chicago, IL, United States
| | - J Wereszczynski
- Center for Molecular Study of Condensed Soft Matter, Illinois Institute of Technology, Chicago, IL, United States.
| |
Collapse
|
33
|
Phipps MJS, Fox T, Tautermann CS, Skylaris CK. Energy Decomposition Analysis Based on Absolutely Localized Molecular Orbitals for Large-Scale Density Functional Theory Calculations in Drug Design. J Chem Theory Comput 2016; 12:3135-48. [PMID: 27248370 DOI: 10.1021/acs.jctc.6b00272] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
We report the development and implementation of an energy decomposition analysis (EDA) scheme in the ONETEP linear-scaling electronic structure package. Our approach is hybrid as it combines the localized molecular orbital EDA (Su, P.; Li, H. J. Chem. Phys., 2009, 131, 014102) and the absolutely localized molecular orbital EDA (Khaliullin, R. Z.; et al. J. Phys. Chem. A, 2007, 111, 8753-8765) to partition the intermolecular interaction energy into chemically distinct components (electrostatic, exchange, correlation, Pauli repulsion, polarization, and charge transfer). Limitations shared in EDA approaches such as the issue of basis set dependence in polarization and charge transfer are discussed, and a remedy to this problem is proposed that exploits the strictly localized property of the ONETEP orbitals. Our method is validated on a range of complexes with interactions relevant to drug design. We demonstrate the capabilities for large-scale calculations with our approach on complexes of thrombin with an inhibitor comprised of up to 4975 atoms. Given the capability of ONETEP for large-scale calculations, such as on entire proteins, we expect that our EDA scheme can be applied in a large range of biomolecular problems, especially in the context of drug design.
Collapse
Affiliation(s)
- M J S Phipps
- School of Chemistry, University of Southampton , Highfield, Southampton SO17 1BJ, United Kingdom
| | - T Fox
- Lead Identification and Optimization Support, Boehringer Ingelheim Pharma GmbH & Co. KG , 88397 Biberach, Germany
| | - C S Tautermann
- Lead Identification and Optimization Support, Boehringer Ingelheim Pharma GmbH & Co. KG , 88397 Biberach, Germany
| | - C-K Skylaris
- School of Chemistry, University of Southampton , Highfield, Southampton SO17 1BJ, United Kingdom
| |
Collapse
|
34
|
Colorimetric thrombin assay using aptamer-functionalized gold nanoparticles acting as a peroxidase mimetic. Mikrochim Acta 2015. [DOI: 10.1007/s00604-015-1674-6] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
35
|
Adams TE, Huntington JA. Structural transitions during prothrombin activation: On the importance of fragment 2. Biochimie 2015; 122:235-42. [PMID: 26365066 PMCID: PMC4756804 DOI: 10.1016/j.biochi.2015.09.013] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2015] [Accepted: 09/08/2015] [Indexed: 01/01/2023]
Abstract
Prothrombin is activated to thrombin by the prothrombinase complex through sequential cleavage at two distinct sites. This occurs at sites of vascular injury in a highly regulated cascade of serine protease and cofactor activation, where activated platelets provide a suitable surface for protease/cofactor/substrate assembly. The precise structural and conformational changes undergone during the transition from prothrombin to thrombin have been studied for decades, and several structures of prothrombin fragments along the activation pathway have been solved. Here we present a new structure analyzed in context of other recent structures and biochemical studies. What emerges is an unexpected mechanism that involves a change in the mode of binding of the F2 domain (fragment 2) on the catalytic domain after cleavage at Arg320, and a subsequent reorientation of the linker between the F2 and catalytic domain to present the Arg271 site for cleavage. The catalytic domain of thrombin precursors binds to its F2 domain by two distinct modes. Cleavage of prothrombin at either Arg271 or Arg320 results in shift from mode 2 to mode 1. After cleavage at Arg320, movement of F2 helps to present the second cleavage site at Arg271.
Collapse
Affiliation(s)
- Ty E Adams
- Cambridge Institute for Medical Research, Department of Haematology, University of Cambridge, Wellcome Trust/MRC Building, Hills Road, Cambridge, CB2 0XY, United Kingdom
| | - James A Huntington
- Cambridge Institute for Medical Research, Department of Haematology, University of Cambridge, Wellcome Trust/MRC Building, Hills Road, Cambridge, CB2 0XY, United Kingdom.
| |
Collapse
|
36
|
Boldrini-França J, Santos Rodrigues R, Santos-Silva LK, de Souza DLN, Gomes MSR, Cologna CT, de Pauw E, Quinton L, Henrique-Silva F, de Melo Rodrigues V, Arantes EC. Expression of a new serine protease from Crotalus durissus collilineatus venom in Pichia pastoris and functional comparison with the native enzyme. Appl Microbiol Biotechnol 2015; 99:9971-86. [PMID: 26227411 DOI: 10.1007/s00253-015-6836-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2015] [Revised: 07/07/2015] [Accepted: 07/11/2015] [Indexed: 11/28/2022]
Abstract
Snake venom serine proteases (SVSPs) act primarily on plasma proteins related to blood clotting and are considered promising for the treatment of several hemostatic disorders. We report the heterologous expression of a serine protease from Crotalus durissus collilineatus, named collinein-1, in Pichia pastoris, as well as the enzymatic comparative characterization of the toxin in native and recombinant forms. The complementary DNA (cDNA) encoding collinein-1 was amplified from cDNA library of C. d. collilineatus venom gland and cloned into the pPICZαA vector. The recombinant plasmid was used to transform cells of KM71H P. pastoris. Heterologous expression was induced by methanol and yielded 56 mg of recombinant collinein-1 (rCollinein-1) per liter of culture. The native collinein-1 was purified from C. d. collilineatus venom, and its identity was confirmed by amino acid sequencing. The native and recombinant enzymes showed similar effects upon bovine fibrinogen by releasing preferentially fibrinopeptide A. Although both enzymes have induced plasma coagulation, native Colinein-1 has shown higher coagulant activity. The serine proteases were able to hydrolyze the chromogenic substrates S-2222, S-2238, and S2302. Both enzymes showed high stability on different pH and temperature, and their esterase activities were inhibited in the presence of Zn2+ and Cu2+. The serine proteases showed similar k cat/K m values in enzyme kinetics assays, suggesting no significant differences in efficiency of these proteins to hydrolyze the substrate. These results demonstrated that rCollinein-1 was expressed with functional integrity on the evaluated parameters. The success in producing a functionally active recombinant SVSP may generate perspectives to their future therapeutic applications.
Collapse
Affiliation(s)
- Johara Boldrini-França
- Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Departamento de Física e Química, Universidade de São Paulo, Av. do Café s/n, Monte Alegre, Ribeirão Preto, SP, 14040-903, Brazil
| | - Renata Santos Rodrigues
- Instituto de Genética e Bioquímica, Universidade Federal de Uberlândia, Uberlândia, Brazil.,INCT, Instituto Nacional de Ciência e Tecnologia em Nano-Biofarmacêutica, Belo Horizonte, MG, Brazil
| | | | - Dayane Lorena Naves de Souza
- Instituto de Genética e Bioquímica, Universidade Federal de Uberlândia, Uberlândia, Brazil.,INCT, Instituto Nacional de Ciência e Tecnologia em Nano-Biofarmacêutica, Belo Horizonte, MG, Brazil
| | - Mário Sérgio Rocha Gomes
- Instituto de Genética e Bioquímica, Universidade Federal de Uberlândia, Uberlândia, Brazil.,INCT, Instituto Nacional de Ciência e Tecnologia em Nano-Biofarmacêutica, Belo Horizonte, MG, Brazil
| | | | - Edwin de Pauw
- Department of Chemistry, University of Liège, Liège, Belgium
| | - Loïc Quinton
- Department of Chemistry, University of Liège, Liège, Belgium
| | - Flávio Henrique-Silva
- Departamento de Genética e Evolução, Universidade Federal de São Carlos, São Carlos, Brazil
| | - Veridiana de Melo Rodrigues
- Instituto de Genética e Bioquímica, Universidade Federal de Uberlândia, Uberlândia, Brazil.,INCT, Instituto Nacional de Ciência e Tecnologia em Nano-Biofarmacêutica, Belo Horizonte, MG, Brazil
| | - Eliane Candiani Arantes
- Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Departamento de Física e Química, Universidade de São Paulo, Av. do Café s/n, Monte Alegre, Ribeirão Preto, SP, 14040-903, Brazil.
| |
Collapse
|
37
|
Ma N, Liu XW, Yang YJ, Li JY, Mohamed I, Liu GR, Zhang JY. Preventive Effect of Aspirin Eugenol Ester on Thrombosis in κ-Carrageenan-Induced Rat Tail Thrombosis Model. PLoS One 2015; 10:e0133125. [PMID: 26193677 PMCID: PMC4507943 DOI: 10.1371/journal.pone.0133125] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Accepted: 06/23/2015] [Indexed: 12/28/2022] Open
Abstract
Based on the prodrug principle, aspirin eugenol ester (AEE) was synthesized, which can reduce the side effects of aspirin and eugenol. As a good candidate for new antithrombotic and anti-inflammatory medicine, it is essential to evaluate its preventive effect on thrombosis. Preventive effect of AEE was investigated in κ-carrageenan-induced rat tail thrombosis model. AEE suspension liquids were prepared in 0.5% sodium carboxymethyl cellulose (CMC-Na). AEE was administrated at the dosage of 18, 36 and 72 mg/kg. Aspirin (20 mg/kg), eugenol (18 mg/kg) and 0.5% CMC-Na (30 mg/kg) were used as control drug. In order to compare the effects between AEE and its precursor, integration of aspirin and eugenol group (molar ratio 1:1) was also designed in the experiment. After drugs were administrated intragastrically for seven days, each rat was injected intraperitoneally with 20 mg/kg BW κ-carrageen dissolved in physiological saline to induce thrombosis. The length of tail-thrombosis was measured at 24 and 48 hours. The blank group just was given physiological saline for seven days without κ-carrageenan administrated. The results indicated that AEE significantly not only reduced the average length of thrombus, PT values and FIB concentration, but also reduced the red blood cell (RBC), hemoglobin (HGB), hematocrit (HCT) and platelet (PLT). The effects of AEE on platelet aggregation and anticoagulant in vitro showed that AEE could inhibit adenosine diphosphate (ADP)-induced platelet aggregation as dose-dependence but no notable effect on blood clotting. From these results, it was concluded that AEE possessed positive effect on thrombosis prevention in vivo through the reduction of FIB, PLT, inhibition of platelet aggregation and the change of TT and PT values.
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 CAAS, Lanzhou, 730050, P. R. China
| | - Xi-Wang 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 CAAS, Lanzhou, 730050, P. R. China
| | - Ya-Jun 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 CAAS, Lanzhou, 730050, P. R. China
| | - Jian-Yong 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 CAAS, Lanzhou, 730050, P. R. China
- * E-mail:
| | - Isam Mohamed
- 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 CAAS, Lanzhou, 730050, P. R. China
| | - Guang-Rong 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 CAAS, Lanzhou, 730050, P. R. China
| | - Ji-Yu Zhang
- 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 CAAS, Lanzhou, 730050, P. R. China
| |
Collapse
|
38
|
Abstract
Thrombin (FIIa) is the key enzyme in haemostasis and acts on several substrates involved in clot formation, platelet activation and feed-back regulation of its own formation. During activation of blood coagulation, FIIa is formed by proteolytic cleavage of prothrombin (FII). In the production of recombinant human FII (rhFII), a key question is whether the thrombin formed has the same properties as endogenous thrombin. We have investigated whether FIIa formed from rhFII and plasma-derived human FII (pdhFII) have the same enzymatic and haemostatic properties against a number of substrates and the same haemostatic capacity in plasma, whole blood and on platelets. Pure FIIa was isolated from rhFII and pdhFII cleaved by recombinant ecarin, and analytical methods were developed to compare the activity of FIIa against different substrates. FIIa derived from rhFII and pdhFII were found to have very similar properties in activating FVIII, FXIII, protein C, platelet aggregation and plasma or whole blood coagulation. Further, the same turnover for S-2366 was found with similar KM. However, activation of FV with rhFIIa was approximately 25% more effective than with pdhFIIa and heparin-enhanced inhibition of rhFIIa by antithrombin was significantly more efficient compared with pdhFIIa with 10% higher inhibition both at steady state and at initial rate conditions. Although differences between the two FIIa preparations using ecarin cleavage were observed, FIIa derived from rhFII administered to human would likely be very similar in activity and function as FIIa formed from endogenous FII.
Collapse
|
39
|
A new peptide (Ruviprase) purified from the venom of Daboia russelii russelii shows potent anticoagulant activity via non-enzymatic inhibition of thrombin and factor Xa. Biochimie 2014; 105:149-58. [PMID: 25038567 DOI: 10.1016/j.biochi.2014.07.006] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2014] [Accepted: 07/07/2014] [Indexed: 12/24/2022]
Abstract
Compounds showing dual inhibition of thrombin and factor Xa (FXa) are the subject of great interest owing to their broader specificity for effective anticoagulation therapy against cardiovascular disorders. This is the first report on the functional characterization and assessment of therapeutic potential of a 4423.6 Da inhibitory peptide (Ruviprase) purified from Daboia russelii russelii venom. The secondary structure of Ruviprase is composed of α-helices (61.9%) and random coils (38.1%). The partial N-terminal sequence (E(1)-V(2)-X(3)-W(4)-W(5)-W(6)-A(7)-Q(8)-L(9)-S(10)) of Ruviprase demonstrated significant similarity (80.0%) with an internal sequence of apoptosis-stimulating protein reported from the venom of Ophiophagus hannah and Python bivittatus; albeit Ruviprase did not show sequence similarity with existing thrombin/FXa inhibitors, suggesting its uniqueness. Ruviprase demonstrated a potent in vitro anticoagulant property and inhibited both thrombin and FXa following slow binding kinetics. Ruviprase inhibited thrombin by binding to its active site via an uncompetitive mechanism with a Ki value and dissociation constant (KD) of 0.42 μM and 0.46 μM, respectively. Conversely, Ruviprase demonstrated mixed inhibition (Ki = 0.16 μM) of FXa towards its physiological substrate prothrombin. Furthermore, the biological properties of Ruviprase could not be neutralized by commercial polyvalent or monovalent antivenom. Ruviprase at a dose of 2.0 mg/kg was non-toxic and showed potent in vivo anticoagulant activity after 6 h of intraperitoneal treatment in mice. Because of the potent anticoagulant property as well as non-toxic nature of Ruviprase, the possible application of the peptide as an antithrombotic agent for combating thrombosis-associated ailments appears promising.
Collapse
|
40
|
Wilson R, Bourne C, Chaudhuri RR, Gregory R, Kenny J, Cossins A. Single-step selection of bivalent aptamers validated by comparison with SELEX using high-throughput sequencing. PLoS One 2014; 9:e100572. [PMID: 24963654 PMCID: PMC4070925 DOI: 10.1371/journal.pone.0100572] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2014] [Accepted: 05/26/2014] [Indexed: 01/23/2023] Open
Abstract
The identification of nucleic acid aptamers would be advanced if they could be obtained after fewer rounds of selection and amplification. In this paper the identification of bivalent aptamers for thrombin by SELEX and single-step selection are compared using next generation sequencing and motif finding informatics. Results show that similar aptamers are identified by both methods. This is significant because it shows that next generation sequencing and motif finding informatics have the potential to simplify the selection of aptamers by avoiding multiple rounds of enzymatic transcription and amplification.
Collapse
Affiliation(s)
- Robert Wilson
- Centre for Genomic Research at the Institute of Integrative Biology, University Of Liverpool, Liverpool, United Kingdom
| | - Christian Bourne
- Centre for Genomic Research at the Institute of Integrative Biology, University Of Liverpool, Liverpool, United Kingdom
| | - Roy R. Chaudhuri
- Centre for Genomic Research at the Institute of Integrative Biology, University Of Liverpool, Liverpool, United Kingdom
| | - Richard Gregory
- Centre for Genomic Research at the Institute of Integrative Biology, University Of Liverpool, Liverpool, United Kingdom
| | - John Kenny
- Centre for Genomic Research at the Institute of Integrative Biology, University Of Liverpool, Liverpool, United Kingdom
| | - Andrew Cossins
- Centre for Genomic Research at the Institute of Integrative Biology, University Of Liverpool, Liverpool, United Kingdom
| |
Collapse
|
41
|
Doneux T, De Rache A, Triffaux E, Meunier A, Steichen M, Buess-Herman C. Optimization of the Probe Coverage in DNA Biosensors by a One-Step Coadsorption Procedure. ChemElectroChem 2013. [DOI: 10.1002/celc.201300145] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
|
42
|
Römhildt L, Pahlke C, Zörgiebel F, Braun HG, Opitz J, Baraban L, Cuniberti G. Patterned biochemical functionalization improves aptamer-based detection of unlabeled thrombin in a sandwich assay. ACS APPLIED MATERIALS & INTERFACES 2013; 5:12029-35. [PMID: 24171544 DOI: 10.1021/am4038245] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2023]
Abstract
Here we propose a platform for the detection of unlabeled human α-thrombin down to the picomolar range in a fluorescence-based aptamer assay. In this concept, thrombin is captured between two different thrombin binding aptamers, TBA1 (15mer) and TBA2 (29mer), each labeled with a specific fluorescent dye. One aptamer is attached to the surface, the second one is in solution and recognizes surface-captured thrombin. To improve the limit of detection and the comparability of measurements, we employed and compared two approaches to pattern the chip substrate-microcontact printing of organosilanes onto bare glass slides, and controlled printing of the capture aptamer TBA1 in arrays onto functionalized glass substrates using a nanoplotter device. The parallel presence of functionalized and control areas acts as an internal reference. We demonstrate that both techniques enable the detection of thrombin concentrations in a wide range from 0.02 to 200 nM with a detection limit at 20 pM. Finally, the developed method could be transferred to any substrate to probe different targets that have two distinct possible receptors without the need for direct target labeling.
Collapse
Affiliation(s)
- Lotta Römhildt
- Institute for Materials Science and Max Bergmann Center of Biomaterials and §Center for Advancing Electronics Dresden, TU Dresden , 01062 Dresden, Germany
| | | | | | | | | | | | | |
Collapse
|
43
|
Figueiredo AC, de Sanctis D, Pereira PJB. The tick-derived anticoagulant madanin is processed by thrombin and factor Xa. PLoS One 2013; 8:e71866. [PMID: 23951260 PMCID: PMC3741208 DOI: 10.1371/journal.pone.0071866] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2013] [Accepted: 07/10/2013] [Indexed: 01/20/2023] Open
Abstract
The cysteine-less peptidic anticoagulants madanin-1 and madanin-2 from the bush tick Haemaphysalis longicornis are the founding members of the MEROPS inhibitor family I53. It has been previously suggested that madanins exert their functional activity by competing with physiological substrates for binding to the positively charged exosite I (fibrinogen-binding exosite) of α-thrombin. We hereby demonstrate that competitive inhibition of α-thrombin by madanin-1 or madanin-2 involves binding to the enzyme's active site. Moreover, the blood coagulation factors IIa and Xa are shown to hydrolyze both inhibitors at different, although partially overlapping cleavage sites. Finally, the three-dimensional structure of the complex formed between human α-thrombin and a proteolytic fragment of madanin-1, determined by X-ray crystallography, elucidates the molecular details of madanin-1 recognition and processing by the proteinase. Taken together, the current findings establish the mechanism of action of madanins, natural anticoagulants that behave as cleavable competitive inhibitors of thrombin.
Collapse
Affiliation(s)
- Ana C. Figueiredo
- IBMC – Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
| | - Daniele de Sanctis
- Structural Biology Group, European Synchrotron Radiation Facility (ESRF), Grenoble, France
| | | |
Collapse
|
44
|
Bai L, Chai Y, Yuan R, Yuan Y, Xie S, Jiang L. Amperometric aptasensor for thrombin detection using enzyme-mediated direct electrochemistry and DNA-based signal amplification strategy. Biosens Bioelectron 2013; 50:325-30. [PMID: 23880107 DOI: 10.1016/j.bios.2013.06.050] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2013] [Revised: 06/13/2013] [Accepted: 06/24/2013] [Indexed: 10/26/2022]
Abstract
In this work, a new electrochemical aptasensor based on direct electron transfer and electrocatalysis of horseradish peroxidase (HRP) using exonuclease-catalyzed target recycling and hybridization chain reaction (HCR) for signal amplification was developed for highly sensitive detection of thrombin. The electrochemical signal was originated from HRP without the addition or labeling of redox probes. To construct the aptasensor, the capture probe was immobilized on gold nanoparticles (AuNPs) modified electrode for the following hybridization with the complementary thrombin binding aptamer. In the presence of thrombin, the formation of aptamer-thrombin complex would result in the dissociation of aptamer from the double-strand DNA (dsDNA). Subsequently, with the employment of exonuclease, aptamer was selectively digested and thrombin could be released for analyte recycling. The capture probe and two hairpin helper DNAs lead to the formation of extended dsDNA polymers through HCR on the electrode surface. Then the biotin-labeled dsDNA polymers could introduce numerous avidin-labeled HRP, resulting in significantly amplified electrochemical signal through the direct electrochemistry and electrocatalysis of HRP. The proposed strategy combined the amplification of analyte recycling and HCR, as well as the inherent electroactivity and catalytic activity of HRP, which exhibited high sensitivity for thrombin determination with an ultra-low detection limit of 1.2×10(-13) M. Moreover, the detection scheme could be easily extended to the detection of other biomolecules.
Collapse
Affiliation(s)
- Lijuan Bai
- Education Ministry Key Laboratory on Luminescence and Real-Time Analysis, College of Chemistry and Chemical Engineering, Southwest University, Chongqing 400715, People's Republic of China
| | | | | | | | | | | |
Collapse
|
45
|
Wang G, He X, Zhu Y, Chen L, Wang L, Zhang X. G-Quadruplex-Linked Supersandwich DNA Structure for Electrochemical Amplified Detection of Thrombin. ELECTROANAL 2013. [DOI: 10.1002/elan.201300184] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
46
|
Loo AH, Bonanni A, Pumera M. Thrombin aptasensing with inherently electroactive graphene oxide nanoplatelets as labels. NANOSCALE 2013; 5:4758-4762. [PMID: 23604556 DOI: 10.1039/c3nr00511a] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
Graphene and its associated materials are commonly used as the transducing platform in biosensing. We propose a different approach for the application of graphene in biosensing. Here, we utilized graphene oxide nanoplatelets as the inherently electroactive labels for the aptasensing of thrombin. The basis of detection lies in the ability of graphene oxide to be electrochemically reduced, thereby providing a well-defined reduction wave; one graphene oxide nanoplatelet of dimension 50 × 50 nm can provide a reduction signal by accepting ~22,000 electrons. We demonstrate that by using graphene oxide nanoplatelets as an inherently electroactive label, we can detect thrombin in the concentration range of 3 pM-0.3 μM, with good selectivity of the aptamer towards interferences by bovine serum albumin, immunoglobulin G and avidin. Therefore, the inherently electroactive graphene oxide nanoplatelets are a material which can serve as an electroactive label, in a manner similar to metallic nanoparticles.
Collapse
Affiliation(s)
- Adeline Huiling Loo
- Division of Chemistry & Biological Chemistry, School of Physical and Mathematical Sciences, Nanyang Technological University, Singapore, 637371, Singapore
| | | | | |
Collapse
|
47
|
Oroval M, Climent E, Coll C, Eritja R, Aviñó A, Marcos MD, Sancenón F, Martínez-Máñez R, Amorós P. An aptamer-gated silica mesoporous material for thrombin detection. Chem Commun (Camb) 2013; 49:5480-2. [PMID: 23660687 DOI: 10.1039/c3cc42157k] [Citation(s) in RCA: 81] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
An aptamer-capped mesoporous material for the selective and sensitive detection of α-thrombin in human plasma and serum has been prepared and characterised.
Collapse
Affiliation(s)
- Mar Oroval
- Centro de Reconocimiento Molecular y Desarrollo Tecnológico (IDM), Unidad Mixta Universidad Politécnica de Valencia - Universidad de Valencia, Spain
| | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Winquist J, Geschwindner S, Xue Y, Gustavsson L, Musil D, Deinum J, Danielson UH. Identification of structural-kinetic and structural-thermodynamic relationships for thrombin inhibitors. Biochemistry 2013; 52:613-26. [PMID: 23290007 DOI: 10.1021/bi301333z] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
To improve our understanding of drug-target interactions, we explored the effect of introducing substituted amine residues with increased chain length in the P3 residue of the thrombin inhibitor melagatran. Inhibition, kinetic, and thermodynamic data obtained via stopped-flow spectroscopy (SF), isothermal microcalorimetry (ITC), and surface plasmon resonance (SPR) biosensor analysis were interpreted with the help of X-ray crystal structures of the enzyme-inhibitor complexes. The association rate became faster when the lipophilicity of the inhibitors was increased. This was coupled to an increased enthalpic component and a corresponding decreased entropic component. The dissociation rates were reduced with an increase in chain length, with only a smaller increase and a decrease in the enthalpic and entropic components, respectively. Overall, the affinity increased with an increase in chain length, with similar changes in the enthalpic and entropic components. ITC analysis confirmed the equilibrium data from SPR analysis, showing that the interaction of melagatran was the most enthalpy-driven interaction. Structural analysis of the thrombin-inhibitor complex showed that the orientation of the P1 and P2 parts of the molecules was very similar, but that there were significant differences in the interaction between the terminal part of the P3 side chain and the binding pocket. A combination of charge repulsion, H-bonds, and hydrophobic interactions could be used to explain the observed kinetic and thermodynamic profiles for the ligands. In conclusion, changes in the structure of a lead compound can have significant effects on its interaction with the target that translate directly into kinetic and thermodynamic effects. In contrast to what may be intuitively expected, hydrogen bond formation and breakage are not necessarily reflected in enthalpy gains and losses, respectively.
Collapse
Affiliation(s)
- Johan Winquist
- Department of Chemistry-BMC, Uppsala University, SE-751 23 Uppsala, Sweden
| | | | | | | | | | | | | |
Collapse
|
49
|
Unique thrombin inhibition mechanism by anophelin, an anticoagulant from the malaria vector. Proc Natl Acad Sci U S A 2012; 109:E3649-58. [PMID: 23223529 DOI: 10.1073/pnas.1211614109] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Anopheles mosquitoes are vectors of malaria, a potentially fatal blood disease affecting half a billion humans worldwide. These blood-feeding insects include in their antihemostatic arsenal a potent thrombin inhibitor, the flexible and cysteine-less anophelin. Here, we present a thorough structure-and-function analysis of thrombin inhibition by anophelin, including the 2.3-Å crystal structure of the human thrombin·anophelin complex. Anophelin residues 32-61 are well-defined by electron density, completely occupying the long cleft between the active site and exosite I. However, in striking contrast to substrates, the D50-R53 anophelin tetrapeptide occupies the active site cleft of the enzyme, whereas the upstream residues A35-P45 shield the regulatory exosite I, defining a unique reverse-binding mode of an inhibitor to the target proteinase. The extensive interactions established, the disruption of thrombin's active site charge-relay system, and the insertion of residue R53 into the proteinase S(1) pocket in an orientation opposed to productive substrates explain anophelin's remarkable specificity and resistance to proteolysis by thrombin. Complementary biophysical and functional characterization of point mutants and truncated versions of anophelin unambiguously establish the molecular mechanism of action of this family of serine proteinase inhibitors (I77). These findings have implications for the design of novel antithrombotics.
Collapse
|
50
|
Loo AH, Bonanni A, Pumera M. Biorecognition on graphene: physical, covalent, and affinity immobilization methods exhibiting dramatic differences. Chem Asian J 2012; 8:198-203. [PMID: 23090869 DOI: 10.1002/asia.201200756] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2012] [Indexed: 01/09/2023]
Abstract
The preparation of biorecognition layers on the surface of a sensing platform is a very crucial step for the development of sensitive and selective biosensors. Different protocols have been used thus far for the immobilization of biomolecules onto various electrode surfaces. In this work, we investigate how the protocol followed for the immobilization of a DNA aptamer affects the performance of the fabricated thrombin aptasensor. Specifically, the differences in selectivity and optimum amount of immobilized aptamer of the fabricated aptasensors adopting either physical, covalent, or affinity immobilization were compared. It was discovered that while all three methods of immobilization uniformly show a similar optimum amount of immobilized aptamer, physical, and covalent immobilization methods exhibit higher selectivity than affinity immobilization. Hence, it is believed that our findings are very important in order to optimize and improve the performance of graphene-based aptasensors.
Collapse
Affiliation(s)
- Adeline Huiling Loo
- Division of Chemistry & Biological Chemistry, School of Physical and Mathematical Sciences, Nanyang Technological University, Singapore 637371, Singapore
| | | | | |
Collapse
|