Skip to content
Publicly Available Published by De Gruyter May 27, 2019

Investigation and isolation of peptide based antiglycating agents from various sources

Çeşitli Kaynaklardan Peptid Temelli Antiglikasyon Ajanlarının İzolasyonu ve Araştırılması
  • Hatice K. Kani , Ebru K. Kocazorbaz ORCID logo EMAIL logo and Figen Zihnioglu

Abstract

Background

In this work, peptide based antiglycation agents from various sources against the advanced glycation endproducts (AGE) formation was investigated.

Materials and methods

As a source of peptides with deglycating activity, Glycine max, Hordeum vulgare, Triticum aestivum, Avena sativa, Prunus dulcis ve Juglans regia were used. The metal chelating activity and antioxidant activity were determined by Cu(II) chelating activity and CUPRAC (Cupric Reducing Antioxidant Capacity) methods. Antidiabetic activity was evaluated through BSA-glucose model.

Results

Most of the extracts obtained have inhibitory activity against AGE formation. Among all plant peptide isolates soybean was found to be most efficient by means of antiglycating (IC50 1.33 μg/mL), antioxidant (28.2 ± 1.4 μmol AAE/mg) and metal chelation activity (55%).

Conclusion

As a result, this study can provide preliminary data to literature to support researches those focused on peptide based glycation inhibitors and discovery of potent AGE inhibitory peptides.

Öz

Amaç

Bu çalışmada, ileri glikasyon son ürünleri (AGE) ile oluşabilecek protein hasarlarının minimize edilmesi amacı ile çeşitli kaynaklardan antiglikasyon etkinliği olan peptid yapıda bileşiklerin araştırılması hedeflendi.

Gereç ve Yöntem

Deglikasyon etkinliği olabilecek endojen peptidlerin eldesinde kaynak olarak Glycine max, Hordeum vulgare, Triticum aestivum, Avena sativa, Prunus dulcis ve Juglans regia kullanıldı. Peptid izolatlarının deglikasyon aktiviteleri BSA-Glukoz modeli ile, antioksidan kapasitesi ve metal bağlama etkinliği ise CUPRAC (bakır(II) iyonu indirgeme antioksidan kapasite) ve bakır şelatasyon metodları kullanılarak incelenmiştir.

Bulgular

Çalışılan ekstrakların çoğunda AGE oluşumuna karşı inhibitör aktivitesi gözlendi. İncelenen tüm parametreler açısından değerlendirildiğinde ise soya peptid izolatının en iyi deglikasyon (IC50 1.33 μg/mL), antioksidan (28.2 ± 1.4 μmolAAE/mg) ve metal bağlama etkinliği (% 55) olduğu saptanmış olup daha etkin ve seçimli inhibitörlerin geliştirilmesi açısından önemli olduğu düşünüldü.

Sonuç

Sonuç olarak, peptid bazlı glikasyon inhibitörlerinin ve güçlü AGE peptit inhibitörlerin keşfedilmesine yönelik araştırmaları desteklemek için literatürlere katkı sağlayacağı düşünüldü.

Introduction

Non-enzymatic glycation occurs through the reaction of reducing sugars with free amino groups of proteins. Initially the product called Schiff’s base is constituted. Later more stable products called Amadori compounds are formed which constitution is followed by advanced glycation [1]. Recent studies have shown that formation of advanced glycation end products (AGE) is autooxidative process dependent, affecting the reactive oxygen species (ROS) formation [2], [3], [4], [5], [6]. Accumulation of AGE products alters the structure and function of tissue proteins and as a result this takes part in aging, aging dependent diseases, and pathogenesis of important diseases [7], [8], [9], [10]. For this reason, prevention of non-enzymatic glycation and formation of AGE products have become a prior interest in recent years. Various endogenous and exogenous resource based compounds have been mechanistically evaluated in this regard. Especially dipeptides such as carnosine and serine analogs, compounds such as pyridoxamine, aminoguanidine, pyridoxal-5-phosphate, and n-acetyl cysteine have been evaluated for their antioxidant, metal chelating, and deglycation activities [10], [11], [12], [13], [14], [15], [16]. For this regard, obtainment of most possible potent compounds with metal chelating and deglycating activities. For this aim, isolation of especially peptide based compounds from histidine and cysteine rich protein sources was aimed.

Materials and methods

Dried plants seeds were commercially ordered and then identified in Ege University Department of Biology – Botany. All the chemicals used in this study are of analytical grade.

Extraction of peptides from various sources

All plant species used in this study [Soybean (Glycine max), Barley (Hordeum vulgare), Wheat (Triticum aestivum), Oat (Avena sativa), Almond (Prunus dulcis) and Walnut (Juglans regia)] were minced until the material became powder. For each sample, 1 g of sample were mixed with various solvents (20 mM sodium acetate pH 9, 20 mM sodium acetate pH 5, 5% TCA, 1 M acetic acid in 10% acetonitrile.) and stirred at room temperature for 1 h at 175 rpm. Mixtures were homogenated and filtered through glass wool under vacuum. The extracts were centrifuged at +4°C for 15 min at 9000 g. Cold acetone was added to the mixture at a 1:1 (v/v) ratio and centrifugation was repeated.

Inhibition of in vitro protein glycation

The methodology was based on that of Wu and Yen [17]. BSA (10 mg/mL) was incubated with glucose (500 mM) in phosphate buffered-saline (PBS) (3mL total volume, pH 7.4) and extract containing 0.02% sodium azide at 60°C. All the reagent and samples were sterilized by filtration through 0.2μm membrane filters. The protein, the sugar and the prospective inhibitor were included in the mixture simultaneously. Aminoguanidine was used as an inhibitor positive control. Reactions without any inhibitor were also setup. Each solution was kept in the dark in a capped tube. After 2 h of incubation, fluorescence intensity (excitation wavelength of 370nm and emission wave-length of 440nm) was measured for the test solutions.

% inhibition values were calculated according to the following formula.

% inhibition=[(AcontrolAblank)/Acontrol]×100

Determination of IC50 values of peptide extracts

The solvent system that yielded the highest deglycation was chosen for extraction of all plant samples. For the determination IC50 values of all peptide isolates, inhibition assays were done at five different protein concentrations with three replicates. Results were compared in percentages of glycation and inhibition. Using the inhibition results and linear regression formulations, IC50 value was calculated as the protein concentration that inhibits the 50% of the glycation activity. IC50 values for carnosine and aminoguanidine were determined too for comparison.

2,4,6-Trinitrobenzenesulfonic acid (TNBS) method

For determination of free amino groups in proteins and other molecules, various methods such as TNBS and o-Phthalaldehyde (OPA) are used. TNBS method was chosen for its sensitivity, for detection of peptides [18]. The peptide isolates were dissolved in hot 1% sodium dodecyl sulfate. A sample solution (0.250 mL) was mixed with 2 mL of 0.2125 M sodium phosphate buffer (pH 8.2) and 2 mL of 0.1% TNBS and the mixture was incubated in dark for 60 min at 50°C. The reaction was quenched by adding 4 mL of 0.1 N HCl and the absorbance was measured at 340 nm using Multiskan™ FC Microplate Photometer.

Total thiol groups assay

Thiol groups of the peptide preparates were analyzed spectrophotometrically via Ellman method [19]. Reagent mixture contains 10 μL sample, 180 μL 0.1 M pH 8.2 Tris-HCl, and 100 μL Ellman reagent prepared in the same buffer (60 mg 5,5′-dithiobis-(2-nitrobenzoic acid) (DTNB)/100 mL). The absorbance of yellow colored product of the reaction, 2-nitro-5-mercapto-benzoic acid, was measured at 405 nm via microplate reader (Thermo Scientific™ Multiskan™, FC, USA). Glutathione (GSH) standard with a concentration range of 2–20 μg/mL was used for calculation total thiol of the samples.

Determination of metal chelating activity

To determine the metal chelating capacity of the peptide isolates with deglycation activity, copper-chelation assay [20]. Copper (II) sulfate was dissolved in 10 mM KCl containing 30 mM hydroxylamine hydrochloride buffer pH 5. One milliliter copper solution (3 mM) and 400 μL 1 mM murexide is added on 1 mL of different dilutions of samples. The mixture is incubated 3 min at room temperature. Proportion of absorbance at 485 nm to absorbance at 530 nm gives the metal chelation activity. Using a copper (II) sulfate standard with a range of 0.025–0.125 mM, free copper amounts were calculated. Assays were done in three replicates.

%Chelation (bound copper)=[(FcontrolFsample)/Fcontrol]×100

Determination of total antioxidant capacity

This assay is based on reduction of copper (II) to copper (I) by antioxidants [21]. Ten millimeter CuCl2·2H2O, 7.5 mM neocuproin, and 1 M ammonium acetate pH 7 were respectively added to the wells of microplate, each with an equal volume of 50 μL. Mixtures were preincubated for 15 min at 37°C and absorbance was measured at 450 nm. Ascorbic acid was used as a standard, with a concentration range of 20–200 μmol/mL. Assays were done in three replicates.

Investigation of peptide isolates by RP-HPLC

The peptides were eluted by a linear gradient from 98% to 2% solvent A (0.1% trifluoroacetic acid (TFA) in deionised water) in solvent B (0.1% TFA in 90%, v/v, acetonitrile in deionised water) over 20 min. Separations were conducted at 40°C at a flow rate of 1 mL/min. The eluted peptides were detected at 215 nm.

Results and discussion

Diabetes is a multifactorial disease which affects world population at an increasing rate. For this reason, aside from preventive life styles, treatment based approaches are also arising. It is known that extended periods of hyperglycemia increases non-enzymatic glycooxidation based damage to protein structure and function, resulting in protein aggregation based diabetic complications and take role in diseases such as Alzheimer’s disease, cataract formation, rheumatoid arthritis, neurodegenerative diseases, and pathogenesis of important diseases [22], [23], [24]. For this reason, approaches for minimization of non-enzymatic glycation and formation of advanced glycation end products are of prior importance. Studies regarding compounds with deglycation activities especially for carnosine and its analogs are present in the literature [25], [26], [27], [28], [29]. But currently compound characterization studies for deglycation are mainly limited for natural sources. These studies have mainly involved characterization of total plant extract for deglycation activity [30], [31], [32], [33]. In this study, plant species with proteins of high cysteine and histidine contents (Glycine max, Hordeum vulgare, Triticum aestivum, Avena sativa, Prunus dulcis and Juglans regia) were used for extraction of endogenous peptides. Determination of the sample with the highest biological activity and its further characterization was aimed. This study is expected to be preliminary for the future studies regarding the discovery and characterization of peptide based deglycating compounds and to reveal possibly effective AGE peptide inhibitors as the main outcome.

Peptide extraction from various sources

First the extraction method was optimized to obtain bioactive compounds with highest yield. Four different extraction conditions were tried. The prepared extracts were vacuum dried yield was calculated to be the dry weight obtained per gram of plant sample. Using four different extraction conditions, increasing protein concentration linearly decrease AGE formation for extracts prepared with sodium acetate pH 5 buffer and TCA. Extracts prepared with sodium acetate pH 9 buffer and acid/acetonitrile solvent did not increasingly inhibit the glycation at higher protein concentrations than 40 μg/mL.

According to the results, the TCA extraction method that yielded the lowest IC50 value for deglycation was selected for further studies.

Determination of deglycation activities and other properties of peptide isolates

All the isolates were dissolved in appropriate solvent according to the downstream assays. First, effect of protein concentration on AGE formation were studied (Figure 1).

Figure 1: Curves and linear regressions of AGE inhibition activities of (A) walnut peptide extract, (B) almond peptide extract, (C) wheat peptide extract, (D) oat peptide extract and (E) barley peptide extract.
Figure 1:

Curves and linear regressions of AGE inhibition activities of (A) walnut peptide extract, (B) almond peptide extract, (C) wheat peptide extract, (D) oat peptide extract and (E) barley peptide extract.

Inhibitory activity results have shown that aside from the walnut derived one, every peptide isolates was potent for deglycation. Moreover, oat, barley, wheat, and soya derived peptide isolates were more potent inhibitors of AGE formation than the standards aminoguanidine and carnosine. IC50 values of the peptide isolates were given in Table 1.

Table 1:

IC50 values of the peptide isolates.

IC50 (μg/mL)aLinear regressi on (%inhibition)aR2
Hordeum vulgare (Barley)6.870±0.2y=10.480x−22.000.9900
Avena sativa (Oat)9.280±1.8y=5.095x+2.68540.9832
Glycine max. (Soybean)1.330±0.8y=10.615x+35.8460.6228
Triticum aestivum (Wheat)7.680±1.1y=1.7938x+36.2030.9819
Prunus dulcis (Almond)7.310±0.3y=7921x+36.8810.9478
Juglans regia (Walnut)107.71±2.1y=0.3672x+10.4480.9352
Aminoguanidine30.04±0.60.9852x+20.3930.9483
Carnosine29.13±0.51.3492x+10.6890.9016
  1. aValues are expressed as means±SD.

It was observed that soy peptide isolate has the highest antioxidant activity, metal chelating capacity, and free amino and thiol group content when compared to the other plants those were used in this study (Table 2). Oat and barley hydrolyzates are also have similarly strong activities. When compared to the known deglycating and antioxidant compounds carnosine and amino guanidine, almond and wheat peptide isolates were stronger, with higher free amino and thiol group content and metal chelating capacity. On the other hand, almond and barley peptide isolates were surpassed by soy and oat peptide isolates.

Table 2:

Analysis of other properties of peptide extracts.

Free thiol groupa (μg/mg protein)Free amino groupa (μmol/mg protein)Antioxidant activitya (μmol AAE/mgprotein)Metal chelationa (%)
H. vulgare (Barley)22.4±0.3556.0±0.9223.5±0.0253
A. sativa (Oat)27.8±2.652.1±1.0226.3±0.7348
G. max (Soya)31.2±1.4131.0±2.828.2±1.455
T.aestivum (Wheat)21.3±0.2353.2±1.055.2±0.4545
P. dulcis (Almond)26.4±1.7693±2.347.1±0.0351
J. regia (Walnut)22.6±2.953.2±0.065.4±1.248
  1. aValues are expressed as means±SD.

The alleged mechanism of prevention of glycation by these compounds are thought to be caused by interference of the compounds to Schiff’s base formation, thus blocking AGE formation. As a result, five peptide isolates (soy, barley, almond, wheat, and oat) are thought to have remarkable deglycation activities. Among them, especially soy peptide isolate is considered to be valuable in terms of antioxidant activity and metal chelating capacity too. RP-HPLC separations of the soybean peptide isolate was performed in order to analyse the complexity of the peptide preparate, purification and enrichment of more potent peptides among the isolated pool and initiate structural studies. For this purpose, a sample chromatogram post optimization of separation studies is shown in Figure 2.

Figure 2: HPLC profile of the soybean isolate (Supelco ODS C18 (4.5×250), gradient: 98% A (0.1% TFA), % 2 B (Acetonitrile/0.1% TFA), 20 min 60% B, 1 mL/min).
Figure 2:

HPLC profile of the soybean isolate (Supelco ODS C18 (4.5×250), gradient: 98% A (0.1% TFA), % 2 B (Acetonitrile/0.1% TFA), 20 min 60% B, 1 mL/min).

Examination of the chromatograms taken at three different wavelengths by DAD detector were examined and it has been observed that the sensitivity was higher in the chromatogram in which the peptide bonds were measured. The eluate collected between the 4th–5th min of the run showed a dense peak. The collected fraction has exhibited 74% AGE inhibitory activity.

Conclusion

Bioactive proteins and peptides are recently among important research topics. Studies have revealed that bioactive peptides take part in many important physiological processes, which has lead to discovery of new peptides and chemical synthesis of them. Development of new bioanalytical techniques have further facilitated discovery of a hormones and neuropeptides. Solid phase and classical solution phase techniques allowed chemical synthesis of oligopeptides. This allowed rapid and high fidelity production of bioactive peptides when compared to the molecular biological and genetic engineering techniques, which involve in vivo synthesis and demand thorough purification. Through these developments, important steps were taken for explanation of biochemical events and drug development studies. In this study, isolation and obtainment of bioactive peptides were of priority. Briefly, results have shown that respectively antiglycation activity of soya, barley, wheat, and almond significant when compared to their standards carnosine and aminoguanidine. Soya peptide isolate has shown the highest deglycation, antioxidant, and metal chelating activities, providing important results for development of more potent and specific inhibitors. Purification and structural analysis of products those inhibit AGE formation along with use of bioinformatics tools, similar peptides can be discovered or designed, ultimately resulting in a wider repertoire of products in this field. In vivo pharmacokinetic studies will also be required as further characterization steps.

It is clear that this study provides a valuable preliminary data to literature and can become a template for choosing a starting material for the design of compounds with desired bioactivities such as metal chelation, antioxidant, deglycation, etc. for advanced researches.

Acknowledgements

This work was supported by the Scientific and Technological Research Council of Turkey (TÜBİTAK, Project No. 214Z300). The authors thank Ege University, Faculty of Science project coordination (Project no: 15/FEN/O53) for their financial support.

  1. Conflict of interest: The authors have no conflict of interest.

References

1. Altan N. Diabetes mellitus and oxidative stress. Turk J Biochem 2006;31:51–6.Search in Google Scholar

2. Brewer GJ. Risks of copper and iron toxicity during aging in humans. Chem Res Toxicol 2009; 23:319–26.10.1021/tx900338dSearch in Google Scholar PubMed

3. Van Campenhout A, Van Campenhout C, Lagrou AR, Manuel-Y-Keenoy B. Effects of in vitro glycation on Fe3+ binding and Fe3+ isoforms of transferrin. Clin Chem 2004;50:1640–9.10.1373/clinchem.2004.033811Search in Google Scholar PubMed

4. Chen CL, Guangyuan HO. Generation of proline-specific Maillard compounds by the reaction of 2-deoxyglucose with proline. J Agric Food Chem 1997;45:2996–9.10.1021/jf970197nSearch in Google Scholar

5. Price DL, Rhett PM, Thorpe SR, Baynes JW. Chelating activity of advanced glycation end-product (AGE) inhibitors. J Biol Chem 2001;276:48967–72.10.1074/jbc.M108196200Search in Google Scholar PubMed

6. Ramonaitytė DT, Keršienė M, Adams A, Tehrani KA, De Kimpe N. The interaction of metal ions with Maillard reaction products in a lactose–glycine model system. Food Res Int 2009;42:331–6.10.1016/j.foodres.2008.12.008Search in Google Scholar

7. Sattarahmady N, Moosayi-Moyahedi AA, Ahmad F, Hakimelahi GH, Habibi-Rezaei M, Saboury AA, et al. Formation of the molten globule-like state during prolonged glycation of human serum albumin. Biochim Biophys Acta 2007;1770:933–42.10.1016/j.bbagen.2007.02.001Search in Google Scholar PubMed

8. Christen Y. Oxidative stress and Alzheimer disease. Am J Clin Nutr 2000;71:621–9.10.1093/ajcn/71.2.621sSearch in Google Scholar PubMed

9. Dukic-Stefanovic S, Schinzel R, Riederer P, Münch G. AGES in brain ageing: age-inhibitors as neuroprotective and anti-dementia drugs? Biogerontology 2001;2:19–34.10.1023/A:1010052800347Search in Google Scholar

10. Nawale RB, Mourya VK, Bhise SB. Non-enzymatic glycation of proteins: a cause for complications in diabetes. Indian J Biochem Biophys 2006;43:337–43.Search in Google Scholar

11. Ardestani A, Yazdanparast R. Cyperus rotundus suppresses AGE formation and protein oxidation in a model of fructose-mediated protein glycoxidation. Int J Biol Macromol 2007;41:572–8.10.1016/j.ijbiomac.2007.07.014Search in Google Scholar PubMed

12. Belia F, Amorini AM, La Mendola D, Vecchio G, Tavazzi B, Giardina B, et al. New glycosidic derivatives of histidine-containing dipeptides with antioxidant properties and resistant to carnosinase activity. Eur J Med Chem 2008;43:373–80.10.1016/j.ejmech.2007.03.038Search in Google Scholar PubMed

13. Brownson C, Hipkiss AR. Carnosine reacts with a glycated protein. Free Radic Biol Med 2000;28:1564–70.10.1016/S0891-5849(00)00270-7Search in Google Scholar

14. Chetyrkin SV, Mathis ME, Ham AJ, Hachey DL, Hudson BG, Voziyan PA. Propagation of protein glycation damage involves modification of tryptophan residues via reactive oxygen species: inhibition by pyridoxamine. Free Radic Biol Med 2008;44:1276–85.10.1016/j.freeradbiomed.2007.09.016Search in Google Scholar PubMed

15. Culbertson SM, Vassilenko EI, Morrison LD, Ingold KU. Paradoxical impact of antioxidants on post-amadori glycoxidation: counterintuitive increase in the yields of pentosidine and entosidine and Nϵ-carboxymethyllysine using a novel mulltifunctional pyridoxamine derivative. J Biol Chem 2003;278:38384–94.10.1074/jbc.M305099200Search in Google Scholar PubMed

16. Hobart LJ, Seibel I, Yeargans GS, Seidler NW. Anti-crosslinking properties of carnosine: significance of histidine. Life Sci 2004;75:1379–89.10.1016/j.lfs.2004.05.002Search in Google Scholar PubMed

17. Wu CH, Yen GC. Inhibitory effect of naturally occurring flavonoids on the formation of advanced glycation endproducts. J Agric Food Chem 2005;53:3167–73.10.1021/jf048550uSearch in Google Scholar PubMed

18. Adler-Nissen J. Determination of the degree of hydrolysis of food protein hydrolysates by trinitrobenzenesulfonic acid. J Agric Food Chem 1979;27:1256–62.10.1021/jf60226a042Search in Google Scholar PubMed

19. Kocazorbaz KE, Un NR, Erdag A, Zihnioglu F. Inhibitory effects of some bryophytes on glutathione-s-transferase. Curr Enzym Inhib 2017;13:34–40.10.2174/1573408012666160628081431Search in Google Scholar

20. Ruiz-Roca B, Navarro MP, Seiquer I. Antioxidant properties and metal chelating activity of glucose-lysine heated mixtures: relationships with mineral absorption across Caco-2 cell monolayers. J Agric Food Chem 2008;56:9056–63.10.1021/jf801718hSearch in Google Scholar PubMed

21. Apak R, Güçlü K, Demirata B, Özyürek M, Çelik SE, Bektaşoğlu B, et al. Comparative evaluation of various total antioxidant capacity assays applied to phenolic compounds with the CUPRAC assay. Molecules 2007;12:1496–547.10.3390/12071496Search in Google Scholar PubMed PubMed Central

22. Saraswat M, Reddy PY, Muthenna P, Reddy GB. Prevention of non-enzymic glycation of proteins by dietary agents: prospects for alleviating diabetic complications. Br J Nutr 2008;101:1714–21.10.1017/S0007114508116270Search in Google Scholar PubMed

23. Srinivas SM, Harohally NV. Improved synthesis of lysine-and arginine-derived Amadori and Heyns products and in vitro measurement of their angiotensin I-converting enzyme inhibitory activity. J Agric Food Chem 2012;60:1522–7.10.1021/jf204185ySearch in Google Scholar PubMed

24. Zhang EY, Swaan PW. Determination of membrane protein glycation in diabetic tissue. AAPS PharmSci 1999;1:27–33.10.1208/ps010420Search in Google Scholar PubMed PubMed Central

25. Metz TO, Alderson NL, Thorpe SR, Baynes JW. Pyridoxamine, an inhibitor of advanced glycation and lipoxidation reactions: a novel therapy for treatment of diabetic complications. Arch Biochem Biophys 2003;419:41–9.10.1016/j.abb.2003.08.021Search in Google Scholar PubMed

26. Pepper ED, Farrell MJ, Nord G, Finkel SE. Antiglycation effects of carnosine and other compounds on the long-term survival of Escherichia coli. Appl Environ Microbiol 2010;76:7925–30.10.1128/AEM.01369-10Search in Google Scholar PubMed PubMed Central

27. Reddy VP, Garrett MR, Perry G, Smith MA. Carnosine: a versatile antioxidant and antiglycating agent. Science’s Sage Ke 2005;18:12–8.10.1126/sageke.2005.18.pe12Search in Google Scholar PubMed

28. Tabakman R, Lazaovici P, Kohen R. Neuroprotective effects of carnosine and homocarnosine on pheochromocytoma PC12 cells exposed to ischemia. J Neurosci Res 2002;68:463–9.10.1002/jnr.10228Search in Google Scholar PubMed

29. Voziyan PA, Khalifah RG, Thibaudeau C, Yıldız A, Jacob J, Serianni AS, et al. Modification of proteins in vitro by physiological levels of glucose pyrodoxamine inhibits conversion of amadori intermediate to advanced glycation end-products through binding of redox metal ions. J Biol Chem 2003;278:46616–24.10.1074/jbc.M307155200Search in Google Scholar PubMed

30. Choudhary MI, Maher S, Begum A, Abbaskhan A, Ali S, Khan A, et al. Characterization and antiglycation activity of phenolic constituents from Viscum album (European Mistletoe). Chem Pharm Bull 2010;58:980–2.10.1248/cpb.58.980Search in Google Scholar PubMed

31. Gutierrez RM. Inhibition of advanced glycation end-product formation by Origanum majorana L. in vitro and in streptozotocin-induced diabetic rats. Evid Based Complement Alternat Med 2012;2012:1–8.10.1155/2012/598638Search in Google Scholar PubMed PubMed Central

32. Harris CS, Beaulleu LP, Fraser MH, McIntyre KL, Owen PL, Martineau LC, et al. Inhibition of advanced glycation end product formation by medicinal plant extracts correlates with phenolic metabolites and antioxidant activity. Planta Med 2011;77:196–204.10.1055/s-0030-1250161Search in Google Scholar PubMed

33. Kaewnarin K, Shank L, Niamsup H, Rakariyatham N. Inhibitory effects of Lamiaceae plants on the formation of advanced glycation endproducts (AGEs) in model proteins. J Med Bioeng 2013;2:224–7.10.12720/jomb.2.4.224-227Search in Google Scholar

Received: 2018-07-18
Accepted: 2018-11-27
Published Online: 2019-05-27
Published in Print: 2019-10-25

©2019 Walter de Gruyter GmbH, Berlin/Boston

Downloaded on 7.6.2024 from https://www.degruyter.com/document/doi/10.1515/tjb-2018-0294/html
Scroll to top button