Skip to main content
Log in

Thrombin inhibitory peptides derived from Mytilus edulis proteins: identification, molecular docking and in silico prediction of toxicity

  • Original Paper
  • Published:
European Food Research and Technology Aims and scope Submit manuscript

Abstract

Optimization of the thrombin inhibitory activities of different enzymatic hydrolysates was conducted, and then an optimal hydrolysis condition by trypsin (5000 u/g) was determined as follows, digested at 45 °C and pH 8.5 for 2 h with a protein concentration of 25 mg/mL. Thrombin inhibitory activity was proved to be 76.92 ± 4.66% under this condition. A total of 39 peptides were identified in the hydrolysate by UPLC-Q-TOF–MS/MS, and all the peptides were predicted to be nontoxic by in silico predictive approaches. Twenty-six peptides were predicted to be anticoagulant peptides by molecular docking method, and the peptide 26 (Lys-Asn-Ala-Glu-Asn-Glu-Leu-Gly-Glu-Val-Thr-Val-Arg) was predicted to be a better anticoagulant peptide through both structure–activity relationship and affinity activity to thrombin. The interactional positions between peptide and thrombin were also involved in the interaction site on the S1 pocket of thrombin and strongly promoted its thrombin inhibitory activity. The firmly non-bonded interactions made the bound of peptide and thrombin firmly. Eventually, the chemical identification and activity verification of synthetic peptide 26 were conducted, and the thrombin inhibitory activity was 89.96 ± 5.30% at the concentration of 9 mg/mL. This study optimized an enzymatic hydrolysis and a virtual screening method for predicting and verifying the anticoagulant peptide from Mytilus edulis, respectively, which provided a good theoretical basis and application method for the research and development of the anticoagulant peptides, especially from the seafood products.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2
Fig. 3
Fig. 4
Fig. 5
Fig. 6

Similar content being viewed by others

References

  1. Lazcano-Pérez F, Román-González SA, Sánchez-Puig N, Arreguin-Espinosa R (2012) Bioactive peptides from marine organisms: a short overview [J]. Protein Pept Lett 19:700–707

    Article  Google Scholar 

  2. Kim SK, Mendis E (2006) Bioactive compounds from marine processing byproducts—a review [J]. Food Res Int 39:383–393

    Article  CAS  Google Scholar 

  3. Dutta PK, Tripathi S, Mehrotra GK, Joydeep D (2009) Perspectives for chitosan based antimicrobial films in food applications [J]. Food Chem 114:1173–1182

    Article  CAS  Google Scholar 

  4. Najafian L, Babji AS (2012) A review of fish-derived antioxidant and antimicrobial peptides: their production, assessment, and applications [J]. Peptides 33:178–185

    Article  CAS  Google Scholar 

  5. Betoret E, Betoret N, Vidal D, Fito P (2011) Functional foods development: trends and technologies [J]. Trends Food Sci Technol 22:498–508

    Article  CAS  Google Scholar 

  6. Rajanbabu V, Chen JY (2011) Antiviral function of tilapia hepcidin 1–5 and its modulation of immune-related gene expressions against infectious pancreatic necrosis virus (IPNV) in Chinook salmon embryo (CHSE)-214 cells. Fish Shellfish Immunol 30:39–447

    Article  CAS  Google Scholar 

  7. Mann KG, Lorand L (1992) Introduction: blood coagulation [J]. Methods Enzymol 222:1–10

    Google Scholar 

  8. Furie B, Furie BC (1992) Molecular and cellular biology of blood coagulation [J]. N Engl J Med 326:800–806

    Article  CAS  Google Scholar 

  9. Davie EW (1995) Biochemical and molecular aspects of the coagulation cascade [J]. Thromb Haemost 74:1–610

    CAS  Google Scholar 

  10. Ricardo LH, Walter SH (1960) Proteolytic and polymerase activity of thrombin [J]. Am J Physisiol 198:173–179

    Google Scholar 

  11. Sidelmann JJ, Gram J, Jespersen J, Kluft C (2000) Fibrin clot formation and lysis: basic mechanisms [J]. Semin Thromb Hemost 26:605–618

    Article  CAS  Google Scholar 

  12. Chen H, Lyne PD, Giordanetto F, Lovell T, Jin L (2006) On evaluating molecular-docking methods for pose prediction and enrichment factors [J]. J Chem Inf Model 46:401–415

    Article  CAS  Google Scholar 

  13. Je JY, Park PJ, Byun HG, Jung WK, Kim SK (2005) Angiotensin I converting enzyme (ACE) inhibitory peptide derived from the sauce of fermented blue mussel, Mytilus edulis [J]. Biores Technol 96:1624–1629

    Article  CAS  Google Scholar 

  14. Dai ZY, Zhang YP, Zhang H, Lu YB (2012) Preparation and characterization of mussel (Mytilus edulis) protein hydrolysates with angiotensin-I-converting enzyme (ACE) inhibitory activity by enzymatic hydrolysis [J]. J Food Biochem 36:66–74

    Article  CAS  Google Scholar 

  15. Kumar JV, Chen WY, Tsai JJP, Hu WP, et al (2013) Molecular simulation methods for selecting thrombin-binding aptamers [J]. Inf Technol Converg 253:743–749 (Springer, Netherlands)

  16. Wei JT, Chiang BH (2009) Bioactive peptide production by hydrolysis of porcine blood proteins in a continuous enzymatic membrane reactor [J]. J Sci Food Agric 89:372–37817

    Article  CAS  Google Scholar 

  17. Adler-Nissen J (1982) Limited enzymic degradation of proteins: a new approach in the industrial application of hydrolases [J]. J Chem Technol Biotechnol 32:138–156

    Article  CAS  Google Scholar 

  18. Guérard F, Dufossé L, Broise DDL, Binet A (2001) Enzymatic hydrolysis of proteins from yellowfin tuna (Thunnus albacares) wastes using Alcalase [J]. J Mol Catal B Enzym 11:1051–1059

    Article  Google Scholar 

  19. Nasri R, Amor IB, Bougatef A, Nedjar-Arroume N, Dhulster P, Gargouri J et al (2012) Anticoagulant activities of goby muscle protein hydrolysates [J]. Food Chem 133:835–841

    Article  CAS  Google Scholar 

  20. Rojas-Ronquillo R, Cruz-Guerrero A, Flores-Nájera A, Rodríguez-Serrano G, Gómez-Ruiz L, Reyes-Grajeda JP et al (2012) Antithrombotic and angiotensin-converting enzyme inhibitory properties of peptides released from bovine casein by Lactobacillus casei Shirota [J]. Int Dairy J 26:147–154

    Article  CAS  Google Scholar 

  21. Yan Z, Li J, Li S et al (2015) Impact of lignin removal on the enzymatic hydrolysis of fermented sweet sorghum bagasse [J]. Appl Energy 160:641–647

    Article  CAS  Google Scholar 

  22. Lassoued I, Mora L, Barkia A et al (2015) Bioactive peptides identified in thornback ray skin’s gelatin hydrolysates by proteases from Bacillus subtilis and Bacillus amyloliquefacien [J]. J Proteom 128:8–17

    Article  CAS  Google Scholar 

  23. Zhou C, Zhong Q, Rhodes LV et al (2012) Proteomic analysis of acquired tamoxifen resistance in MCF-7 cells reveals expression signatures associated with enhanced migration [J]. Breast Cancer Res 14:R45

    Article  CAS  Google Scholar 

  24. Burton LJ, Rivera M, Hawsawi O et al (2016) Muscadine grape skin extract induces an unfolded protein response-mediated autophagy in prostate cancer cells: a TMT-based quantitative proteomic analysis [J]. PLoS One 11:e0164115

    Article  Google Scholar 

  25. Liu R, Zheng W, Li J, Wang L, Wu H, Wang X et al (2015) Rapid identification of bioactive peptides with antioxidant activity from the enzymatic hydrolysate of Mactra veneriformis by UHPLC–Q-TOF mass spectrometry [J]. Food Chem 167:484–489

    Article  CAS  Google Scholar 

  26. Savitski MM, Wilhelm M, Hahne H et al (2015) A scalable approach for protein false discovery rate estimation in large proteomic data sets [J]. Mol Cell Proteom 14:2394–2404

    Article  CAS  Google Scholar 

  27. Gupta S, Kapoor P, Chaudhary K, Gautam A, Kumar R, Raghava GP (2013) In silico approach for predicting toxicity of peptides and proteins [J]. PLoS One 8:e73957

    Article  CAS  Google Scholar 

  28. Rawendra RDS, Chang CI, Chen HH, Huang TC, Hsu JL (2013) A novel angiotensin converting enzyme inhibitory peptide derived from proteolytic digest of Chinese soft-shelled turtle egg white proteins [J]. J Proteom 94:359–36929

    Article  CAS  Google Scholar 

  29. Neumann T, Junker HD, Keil O, Burkert K, Ottleben H, Gamer J, Metz G (2005) Discovery of thrombin inhibitor fragments from chemical microarray screening [J]. Lett Drug Des Discov 2:590–594

    Article  CAS  Google Scholar 

  30. Corralrodríguez MÁ, Bock PE, Hernándezcarvajal E, Gutiérrezgallego R, Fuentesprior P (2011) Structural basis of thrombin-mediated factor V activation: the Glu666-Glu672 sequence is critical for processing at the heavy chain-B domain junction [J]. Blood 117:7164–7173

    Article  Google Scholar 

  31. Koh CY, Kumar S, Kazimirova M, Nuttall PA, Radhakrishnan UP, Kim S et al (2011) Crystal structure of thrombin in complex with S-variegin: insights of a novel mechanism of inhibition and design of tunable thrombin inhibitors [J]. PLoS One 6:e26367

    Article  CAS  Google Scholar 

  32. He D, Gu D, Huang Y et al (2009) Separation and purification of phenolic acids and myricetin from black currant by high-speed countercurrent chromatography [J]. J Liq Chromatogr Relat Technol 32:3077–3088

    Article  CAS  Google Scholar 

  33. Ling JY, Zhang GY, Cui ZJ et al (2007) Supercritical fluid extraction of quinolizidine alkaloids from Sophora flavescens Ait. and purification by high-speed counter-current chromatography [J]. J Chromatogr A 1145:123–127

    Article  CAS  Google Scholar 

  34. Wu G, Robertson DH, Vieth M (2003) Detailed analysis of grid-based molecular docking: a case study of CDOCKER-A CHARMm-based MD docking algorithm [J]. J Comput Chem 24:1549–1562

    Article  CAS  Google Scholar 

  35. Nasria R, Amorb IB, Bougatefa A, Nedjar-Arroume N, Dhulster P, Gargouri J (2012) Anticoagulant activities of goby muscle protein hydrolysates [J]. Food Chem 133(3):835–841

    Article  Google Scholar 

  36. Dodt J, Seemuller U, Maschler R, Fritz H (1985) The complete covalent structure of hirudin. Localization of the disulfide bonds [J]. Bio Chem Hoppe-Seyler 366:379–385

    Article  CAS  Google Scholar 

  37. Mao SJ, Yates MT, Owen TJ, Krstenansky JL (1988) Interaction hirudin with thrombin: identification of a minimal bindingdomain of hirudin that inhibits clotting activity. Biochem US 27(21):8170–8173

    Article  CAS  Google Scholar 

  38. Biela A, Khayat M, Tan H, Kong J, Heine A, Hangauer D, Klebe G (2012) Impact of ligand and protein desolvation on ligand binding to the S1 pocket of throm-bin. J Mol Biol 418:350–366

    Article  CAS  Google Scholar 

Download references

Acknowledgements

This study was financially supported by the National Natural Science Foundation of China (31371805), the State key research and development plan “Modern Food Processing and Food Storage and Transportation Technology and Equipment (No. 2017YFD0400200). The authors appreciated the NeoTrident Technology Limited. (Beijing, China) for the guide of molecular docking.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Ming Du.

Ethics declarations

Conflict of interest

All the authors have no conflict of interest for this article.

Compliance with ethics requirements

This article does not contain any studies with human or animal subjects.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Feng, L., Tu, M., Qiao, M. et al. Thrombin inhibitory peptides derived from Mytilus edulis proteins: identification, molecular docking and in silico prediction of toxicity. Eur Food Res Technol 244, 207–217 (2018). https://doi.org/10.1007/s00217-017-2946-7

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s00217-017-2946-7

Keywords

Navigation