You are on page 1of 8

Journal of Biomolecular Structure and Dynamics

ISSN: (Print) (Online) Journal homepage: https://www.tandfonline.com/loi/tbsd20

Understanding hypocholesterolemic activity of


soy isoflavones: Completing the puzzle through
computational simulations

Feri Eko Hermanto, Warsito Warsito, Muhaimin Rifa’i & Nashi Widodo

To cite this article: Feri Eko Hermanto, Warsito Warsito, Muhaimin Rifa’i & Nashi Widodo
(2022): Understanding hypocholesterolemic activity of soy isoflavones: Completing the puzzle
through computational simulations, Journal of Biomolecular Structure and Dynamics, DOI:
10.1080/07391102.2022.2148752

To link to this article: https://doi.org/10.1080/07391102.2022.2148752

© 2022 The Author(s). Published by Informa View supplementary material


UK Limited, trading as Taylor & Francis
Group

Published online: 28 Nov 2022. Submit your article to this journal

Article views: 907 View related articles

View Crossmark data Citing articles: 1 View citing articles

Full Terms & Conditions of access and use can be found at


https://www.tandfonline.com/action/journalInformation?journalCode=tbsd20
JOURNAL OF BIOMOLECULAR STRUCTURE AND DYNAMICS
https://doi.org/10.1080/07391102.2022.2148752

Understanding hypocholesterolemic activity of soy isoflavones: Completing the


puzzle through computational simulations
Feri Eko Hermantoa , Warsito Warsitob,c, Muhaimin Rifa’ia,d and Nashi Widodoa,d
a
Department of Biology, Faculty of Mathematics and Natural Sciences, Universitas Brawijaya, Malang, Indonesia; bDepartment of Chemistry,
Faculty of Mathematics and Natural Sciences, Universitas Brawijaya, Malang, Indonesia; cEssential Oil Institute, Universitas Brawijaya,
Malang, Indonesia; dBiosystem Study Center, Universitas Brawijaya, Malang, Indonesia
Communicated by Ramaswamy H. Sarma

ABSTRACT ARTICLE HISTORY


The hypocholesterolemic activity of soy isoflavones has been studied, but the exact mechanism under- Received 14 March 2022
lying the activity remains unclear. This study reveals the proposed mechanism of the cholesterol-low- Accepted 12 November 2022
ering effect of soy isoflavones by computational simulations. Daidzin, Glycitin, Genistin, Daidzein,
KEYWORDS
Glycitein, Genistein, Glyceollin I, Glyceollin II, and Glyceollin III were selected to be analyzed their inter-
Genistein; Genistin; HMGCR
action with 3-Hydroxy-3-Methyl-Glutaryl-Coenzyme A Reductase (HMGCR) and Sterol Regulatory inhibitor; hypercholesterol-
Element-Binding Protein 2 (SREBP2) as key factors in cholesterol biosynthesis as well as Proprotein emia; isoflavones
Convertase Subtilisin/Kexin type 9 (PCSK9) as a common target for hypercholesterolemia. Protein-iso-
flavones interaction was analyzed using AutoDock. According to binding energy calculations, a total of
five out of those nine isoflavones, including Glycitin, Genistin, Genistein, Glyceollin II, and Glyceollin III,
were favored to be a HMGCR inhibitor but not with SREBP2 and PCSK9. Those isoflavones were then
compared with Simvastatin as known inhibitor of HMGCR. Isoflavone with binding energy lower than
Simvastatin then directed to molecular dynamics using YASARA and headed into toxicity estimations.
Almost all of those isoflavones could bind with HMGCR with better stability than Simvastatin accord-
ing to molecular dynamics simulations. Toxicity prediction filtered two out of the five isoflavones men-
tioned earlier as the proper candidate to be an HMGCR inhibitor. Those isoflavones were Genistin and
Genistein. In summary, the hypocholesterolemic activity of soy isoflavones may occur by blocking the
cholesterol biosynthesis pathway.

CONTACT Nashi Widodo widodo@ub.ac.id Biosystem Study Center, Universitas Brawijaya, Jl. Veteran, Malang, 65145, Indonesia
Supplemental data for this article can be accessed online at https://doi.org/10.1080/07391102.2022.2148752.
ß 2022 The Author(s). Published by Informa UK Limited, trading as Taylor & Francis Group
This is an Open Access article distributed under the terms of the Creative Commons Attribution-NonCommercial-NoDerivatives License (http://creativecommons.org/licenses/by-nc-nd/4.
0/), which permits non-commercial re-use, distribution, and reproduction in any medium, provided the original work is properly cited, and is not altered, transformed, or built upon in
any way.
2 F. E. HERMANTO ET AL.

1. Introduction Table 1. The binding energy of each isoflavone with HMGCR, PCSK9,
and SREBP2.
Hypercholesterolemia accounts for major public health prob- Binding Energy (kcal/mol)
lems in modern society (Taddei et al., 2020). People with Compound PubChem CID HMGCR PCSK9 SREBP2
hypercholesterolemia have a higher risk of developing car- Daidzin 107971 7.15 4.35 5.35
diovascular diseases (Soran et al., 2018; Zarate et al., 2016). Glycitin 187808 8.23 4.85 5.65
Besides conventional medicinal therapy, food intervention Genistin 5281377 8.93 4.68 6.08
Daidzein 5281708 7.06 3.94 5.24
also mitigates or accompanies medical treatment (Mannu Glycitein 5317750 6.64 5.60 4.91
et al., 2013). In particular, soybean consumption was discov- Genistein 5280961 7.69 3.87 4.56
ered to have hypocholesterolemic activity (Ramdath et al., Glyceollin I 162807 7.35 5.35 4.81
Glyceollin II 181883 7.85 5.74 4.86
2017; Sirtori et al., 1995; Wong et al., 1998), suggesting a Glyceollin III 11954193 9.04 4.49 5.68
promising aspect in hypercholesterolemia treatment Simvastatin 54454 7.54 – –
using food.
Beyond its high amino acid contents, soybeans also have 2004; Sung et al., 2004). In addition, Simvastatin was used as
high bioactive compounds, majorly from isoflavones (Wang a control molecule for HMGCR inhibitor, according to the
et al., 2013). Dietary isoflavones have been reported to have previous study (Marahatha et al., 2021). On the other hand,
a protective role in regulating plasma cholesterol levels dur- three dimensional (3 D) structures of isoflavones and
ing the clinical phase (Herwana et al., 2020; Taku et al., Simvastatin were retrieved from PubChem, while the protein
2007). This result is supported by the experimental evidence structure was downloaded from Protein Data Bank (PDB)
that isoflavones reduced LDL cholesterol in blood plasma with the following identity: HMGCR (PDB ID: 1DQ8), PCSK9
(Huang et al., 2013; Lee et al., 2007; Teixeira Damasceno (PDB ID: 5OCA), and SREBP2 (PDB ID: 1UKL).
et al., 2007). On the other hand, an animal model experiment
discovered that isoflavones treatment altered SREBP expres-
sion and several of its downstream genes (Mullen et al., 2.2. Binding energy calculation
2004). Further investigations also tried to uncover the lipid-
Binding energy was calculated using AutoDock 4.0 in PyRx
lowering properties of the isoflavones by altering PPAR, ATP-
8.0 software using Lamarckian Genetic Algorithm (Dallakyan
Binding Cassette super-family G member 5 (ABCG5), and
& Olson, 2015; Morris et al., 1998, 2009). The grid setting was
ABCG8 expressions (Huang et al., 2013; Mezei et al., 2003). determined according to key residues for each protein
Unfortunately, the authors still hypothesized the exact mech- (Gustafsen et al., 2017; Istvan et al., 2000; Lee et al., 2003).
anism of the hypolipidemic activity of the isoflavones. The grid center position was set as follows: x: 11.8747, y:
Besides, the actual compound from the isoflavones group 17.447, z: 2.9508 for HMGCR; x: 46.3085, y: 10.8373, z:
involved in cholesterol-lowering activity remains unknown. It 23.6289 for PCSK9; and x: 23.9692, y: 6.1774, z: 101.417 for
is still hypothesized that all isoflavones have a beneficial role SREBP2 with 0.375 Å spacing for all docking simulations.
in maintaining normal plasma cholesterol. Nevertheless, since
chemical structure influences its interaction and activity
towards biological macromolecules (Lewandowski et al., 2.3. Docking analysis and structure visualization
2020), the mechanism leaves a gap to be filled.
Binding energy, types of the chemistry of residue interaction,
Several pathways have been suggested to regulate
and hydrophobicity surface were considered as the parame-
plasma cholesterol levels, including blocking de novo choles-
ters of docking results. A complex with binding energy lower
terol biosynthesis through inhibition of HMGCR (Boucher &
than Simvastatin was considered for molecular dynamics
Vogel, 2016; Ma et al., 2019) or SREBP2 (Tang et al., 2011)
simulation. Finally, the complex structure of each protein-lig-
and preventing Low-Density Lipoprotein Receptor (LDLR)
and was visualized using Biovia Discovery Studio 2019.
degradation through inhibition of PCSK9 activity (Gustafsen
et al., 2017; Ma et al., 2019). Thus, targeting those proteins
may unveil the hypolipidemic activity of the isoflavones. 2.4. Molecular dynamic simulation and binding energy
Therefore, this study will explores the mechanism underlying calculations
hypocholesterolemic activity of soy-isoflavone as well as
YASARA version 19.12.14 software (Krieger & Vriend, 2015)
complements and comprehends aforementioned pro-
with AMBER14 force field (Maier et al., 2015) was employed
posed mechanism.
to perform molecular dynamics simulation using the follow-
ing condition: pH 7.4, 0.9% NaCl concentration, 310 K tem-
2. Materials and method perature, 0.997 water density, 1 atm pressure, and 100 ns
running time. The Root-Mean-Square Deviation (RMSD) and
2.1. Data mining of isoflavone and protein structures Root-Mean-Square Fluctuation (RMSF) outputs were used for
The list of nine isoflavones was selected according to the determining structural stability and rigidity. Also, number of
previously identified from elicited soybean (Atho’illah et al. hydrogen bond was analyzed. In addition, free binding
2019). HMGCR, SREBP2, and PCSK9 were selected as the tar- energy was calculated using YASARA macros with Boundary
get based on the previous studies related to cholesterol Fast method. The DEnergy was estimated according to the
metabolism regulation (Mannino et al., 2021; Mullen et al., following sum:
JOURNAL OF BIOMOLECULAR STRUCTURE AND DYNAMICS 3

Figure 1. Amino acid interactions of daidzin (A), Glycitin (B), Genistin (C), Daidzein (D), Glycitein (E), Genistein (F), Glyceollin I (G), Glyceollin II (H), Glyceollin III (I),
and Simvastatin (J) with HMGCR.


DEbinding ¼ DEreceptor þ DEligand  DEcomplex cytotoxic properties were estimated according to the prob-
ability score.

3. Results and discussion


2.5. Toxicity prediction
3.1. Isoflavones interaction with target proteins
ProTox II (Banerjee et al., 2018) was used to determine the
toxicity profile of selected isoflavones. Lethal dose 50 (LD50) Molecular docking simulation revealed that all of the isofla-
and toxicity class were measured, while probability to have vones interacted with HMGCR at lower binding energy than
hepatotoxic, carcinogenic, immunotoxic, mutagen, and other target proteins, i.e., PCSK9 and SREBP2. In addition,
4 F. E. HERMANTO ET AL.

Figure 2. The RMSD of the selected compounds describes the stability of interaction of HMGCR and Glycitin, Genistin, Genistein, and Simvastatin, respectively. The
RMSD of Protein Backbone (A) describes the stability of the protein, the RMSD of Ligand Conformation (B) describes the stability of ligand structure during its inter-
action with the protein, and the RMSD of Ligand Movement (C) describes the stability of the ligand during the interaction with the protein. Additionally, RMSF val-
ues of selected compounds specified on the active site residues (D) and all residues in chain A (E) and chain B (F) of HMGCR. Also, average number of hydrogen
bond (G) and its fluctuations during the simulation time (H) were described along with binding energy of selected isoflavones (I).

Glycitin, Genistin, Genistein, Glyceollin II, and Glyceollin III dynamics of isoflavones were more stable compared to the
have binding energy lower than Simvastatin as control mole- Simvastatin during the binding to HMGCR (Figure 2B).
cules for HMGCR inhibitor (Table 1). The isoflavones with However, the RMSD of ligand movement described that
lower binding energy than Simvastatin, except Genistein, Glyceollin III might be detached during simulation, indicated
also bounded in the hydrophobic area of the HMGCR. by the dramatic escalation of the RMSD at around 50 ns. On
Several catalytic residues of HMGCR (Istvan et al., 2000) also the other hand, other isoflavones, particularly Genistein and
interacted with these isoflavones, including ARG590, GLU559, Glyceollin II, have better interaction stability with the HMGCR
LEU853, ASP690, LYS691, ASN755, and ASP767 (Figure 1). On than the Simvastatin (Figure 2C).
the other hand, Genistein showed more hydrogen bonds Residual fluctuations represented by RMSF showed no dif-
than the other four isoflavones (Supplementary Table 1). ference among isoflavones and Simvastatin (Figure 2D-F).
These results agree with the previous studies describing the Catalytic residues also stayed stable in the complex of
potential of isoflavones as competitive inhibitors of the HMGCR with isoflavones and Simvastatin, suggesting no
HMGCR (Sung et al., 2004). Accordingly, Glycitin, Genistin, alteration of residual fluctuation at the catalytic sites (Figure
Genistein, Glyceollin II, and Glyceollin III interactions with 2D). RMSF value represented that isoflavones’ binding to the
HMGCR were further analyzed using molecular dynam- HMGCR did not influence the stability of each residue, par-
ics simulation. ticularly catalytic residues of HMGCR. Therefore, all of the iso-
To understand the structural dynamics of the selected iso- flavones may bound with great affinity to the HMGCR to act
flavones with HMGCR, molecular dynamics were employed. as its inhibitor.
The RMSD of the protein backbone value showed that the Since hydrogen bond has a vital role in protein-ligand sta-
interactions of isoflavones and Simvastatin did not alter the bility, the number of hydrogen bond will influence the bind-
structural backbone of HMGCR as all of those ligands have a ing of isoflavones to the HMGCR (Fu et al., 2018; Patil et al.,
value of less than 3 Å (Figure 2A). Meanwhile, the structural 2010). Genistin, Genistein and Glyceollin II have greater
JOURNAL OF BIOMOLECULAR STRUCTURE AND DYNAMICS 5

Table 2. Calculations on free binding energy of HMGCR with every selected ligands.
D Energy (kJ/mol)
Ligand Binding Potential Receptor Solvation Receptor Potential Ligand Solvation Ligand Potential Complex Solvation Complex
Simvastatin 20.158 43412.268 53826.624 53.461 77.618 43554.937 53687.954
±53.645 ±1023.807 ±1328.619 ±21.366 ±10.157 ±1025.471 ±1307.768
Glycitin 128.494 43161.864 54532.992 5.128 118.893 43373.147 54317.237
±56.788 ±908.797 ±1150.590 ±19.458 ±14.247 ±934.553 ±1176.149
Genistin 11.839 42178.139 53519.153 24.437 108.176 42372.696 53469.048
±35.886 ±874.601 ±1232.478 ±19.916 ±17.380 ±877.777 ±1229.739
Genistein 281.768 41574.401 54952.332 94.421 427.697 41804.823 54773.417
±59.381 ±888.422 ±1130.330 ±32.179 ±37.839 ±885.184 ±1129.452
Glyceollin II 70.18 41602.929 54606.344 65.247 150.833 41848.156 54507.017
±40.326 ±888.576 ±1192.082 ±17.687 ±12.381 ±890.733 ±1192.142
Glyceollin III 54.989 42107.701 53848.603 110.211 122.275 42113.457 53799.921
±65.678 ±895.644 ±1169.889 ±19.653 ±29.479 ±894.903 ±1171.589
Notes: Each value was presented in mean ± standard deviation.

Figure 3. Toxicity classification (A) and probability to induce several toxicity properties (B) of selected isoflavones.

number of formed hydrogen bond than other isoflavones cellular metabolic utilization (Goldstein & Brown, 2009).
and Simvastatin (Figure 2G-H). This data also supported by Therefore, soy isoflavones may have hypolipidemic activity
the RMSD of ligand movement where Genistein and through alterations of HMGCR expression and activity at the
Glyceollin II remains stable and better than Simvastatin as transcriptomic and proteomic levels.
control inhibitor. However, binding energy calculations
described that Genistin was the most stable one even com-
pared to the Simvastatin. Among all of the complexes,
3.2. Toxicity profile of selected isoflavones
Genistein had lowest energy than other complex (Table 2,
Figure 2I). Toxicity prediction revealed that Glycitin, Genistin, and
Despite many experiments and clinical studies conducted Genistein were the less toxic isoflavones among selected iso-
to evaluate the hypolipidemic activity of soy-isoflavones, the flavones. Glyceollin III became the most toxic isoflavone with
exact mechanism is still poorly understood. This study LD50 25 mg/kg according to the ProTox II estimations
revealed that Genistin, Genistein, and Glyceollin II might per- (Banerjee et al., 2018). Glyceollin II was classified as class 4
form as HMGCR inhibitors through their interaction with with slight toxicity profile based on LD50 calculation (Figure
HMGCR catalytic residues. The interaction remains stable 3A). Further, Glycitin, Glyceollin II, and Glyceollin III showed
compared to Simvastatin, suggesting that those compounds high probability to have immunotoxic effect (Figure 3B). In
have excellent potential to block uncontrolled cholesterol view of that, Genistin and Genistein were suggested as the
biosynthesis in hyperlipidemia. Isoflavones, mainly Genistein, best HMGCR inhibitors with the lowest toxicity profile.
have been evaluated as an HMGCR inhibitor in vitro as Statin
(Sung et al., 2004). As a rate-limiting factor of cholesterol bio-
synthesis, HMGCR inhibition alleviated plasma cholesterol
4. Conclusion
levels (Lee et al., 2007). The HMGCR blocking activity was
also proven to have an atheroprotective role in vivo Genistin and Genistein may have excellent properties as
(Mansouri et al., 2021). Advance shrinking of cholesterol lev- HMGCR competitive inhibitors with the highest affinities and
els leads to upregulation of LDLR (Notarnicola et al., 2008) low toxicity profiles. The cholesterol-regulating mechanism of
through the SREBP2 pathway at the transcription scale (Lu soy isoflavones may be achieved by inhibiting cholesterol
et al., 2019), thus allowing plasma cholesterol clearance for biosynthesis by blocking HMGCR activity.
6 F. E. HERMANTO ET AL.

Acknowledgement Krieger, E., & Vriend, G. (2015). New ways to boost molecular dynamics
simulations. Journal of Computational Chemistry, 36(13), 996–1007.
The authors thank to Dr. Dinia R. Dwijayanti for her constructive com- https://doi.org/10.1002/jcc.23899
ments and suggestions during the manuscript arrangement. Lee, S.-O., Renouf, M., Ye, Z., Murphy, P. A., & Hendrich, S. (2007).
Isoflavone glycitein diminished plasma cholesterol in female golden
Syrian hamsters. Journal of Agricultural and Food Chemistry, 55(26),
Disclosure statement 11063–11067. https://doi.org/10.1021/jf070972r
Lee, S. J., Sekimoto, T., Yamashita, E., Nagoshi, E., Nakagawa, A.,
The authors declare no competing interest in this work.
Imamoto, N., Yoshimura, M., Sakai, H., Chong, K. T., Tsukihara, T., &
Yoneda, Y. (2003). The structure of importin-beta bound to SREBP-2:
Nuclear import of a transcription factor. Science (New York, N.Y.),
Funding 302(5650), 1571–1575. https://doi.org/10.1126/science.1088372
This research was funded by Ministry of Education, Culture, Research, Lewandowski, W., Lewandowska, H., Golonko, A.,  Swiderski, G., Swisłocka,
and Technology, Republic of Indonesia under PMDSU research scheme R., & Kalinowska, M. (2020). Correlations between molecular structure
funding (grant No. 438.31/UN10.C10/TU/2021 and 612.11/UN10.C10/ and biological activity in “logical series” of dietary chromone deriva-
TU/2022). tives. PLoS ONE, 15(8), e0229477. https://doi.org/10.1371/journal.pone.
0229477
Lu, R., Zheng, Z., Yin, Y., & Jiang, Z. (2019). Effect of Genistein on choles-
ORCID terol metabolism-related genes in HepG2 cell. Journal of Food Science,
84(8), 2330–2336. https://doi.org/10.1111/1750-3841.14725
Feri Eko Hermanto http://orcid.org/0000-0002-6955-3688 Ma, S., Sun, W., Gao, L., & Liu, S. (2019). Therapeutic targets of hyperchol-
Muhaimin Rifa’i http://orcid.org/0000-0001-5731-2951 esterolemia: HMGCR and LDLR. Diabetes, Metabolic Syndrome and
Nashi Widodo http://orcid.org/0000-0002-1126-498X Obesity : targets and Therapy, 12, 1543–1553. https://doi.org/10.2147/
DMSO.S219013
Maier, J. A., Martinez, C., Kasavajhala, K., Wickstrom, L., Hauser, K. E., &
References Simmerling, C. (2015). ff14SB: Improving the accuracy of protein side
chain and backbone parameters from ff99SB. Journal of Chemical
Atho’illah, M. F., Safitri, Y. D., Nur’aini, F. D., Savitri, R. U., Rahayu, S., Theory and Computation, 11(8), 3696–3713. https://doi.org/10.1021/
Widyarti, S., & Rifa’i, M. (2019). Evaluation of glyceollin accumulation acs.jctc.5b00255
and antioxidant properties on soybean (Glycine max L.) through com- Mannino, G., Iovino, P., Lauria, A., Genova, T., Asteggiano, A.,
bination of different biotic elicitor and light. Scientific Study & Notarbartolo, M., Porcu, A., Serio, G., Chinigo , G., Occhipinti, A.,
Research Chemistry & Chemical Engineering, Biotechnology, Food Capuzzo, A., Medana, C., Munaron, L., & Gentile, C. (2021). Bioactive
Industry 20, 199–208. https://doi.org/10.13140/RG.2.2.19614.28483 triterpenes of protium heptaphyllum gum resin extract display choles-
Banerjee, P., Eckert, A. O., Schrey, A. K., & Preissner, R. (2018). ProTox-II: A terol-lowering potential. International Journal of Molecular Sciences,
webserver for the prediction of toxicity of chemicals. Nucleic Acids 22(5), 2664. https://doi.org/10.3390/ijms22052664
Research, 46(W1), W257–W263. https://doi.org/10.1093/nar/gky318 Mannu, G. S., Zaman, M. J. S., Gupta, A., Rehman, H. U., & Myint, P. K.
Boucher, P., & Vogel, H. G. (2016). Inhibition of cholesterol biosynthesis. (2013). Evidence of lifestyle modification in the management of
In F. J. Hock (Ed.), Drug discovery and evaluation: Pharmacological hypercholesterolemia. Current Cardiology Reviews, 9(1), 2–14. https://
assays (pp. 2247–2271). Springer International Publishing. doi.org/10.2174/157340313805076313
Dallakyan, S., & Olson, A. J. (2015). Small-molecule library screening by Mansouri, A., Vahdati, A., Nematbakhsh, M., & Moshtaghian, J. (2021).
docking with PyRx. Methods in molecular biology (Clifton, N.J.), 1263, Hypercholesterolemia role of vitamin D3 and genistein effect in
243–250. https://doi.org/10.1007/978-1-4939-2269-7_19 reducing atherosclerosis in rat. Hormozgan Med J, 19, 306–314.
Fu, Y., Zhao, J., & Chen, Z. (2018). Insights into the molecular mecha- Marahatha, R., Basnet, S., Bhattarai, B. R., Budhathoki, P., Aryal, B.,
nisms of protein-ligand interactions by molecular docking and Adhikari, B., Lamichhane, G., Poudel, D. K., & Parajuli, N. (2021).
molecular dynamics simulation: A case of oligopeptide binding pro- Potential natural inhibitors of xanthine oxidase and HMG-CoA reduc-
tein. Computational and Mathematical Methods in Medicine, 2018, tase in cholesterol regulation: In silico analysis. BMC Complementary
3502514. https://doi.org/10.1155/2018/3502514 Medicine and Therapies, 21(1), 1. https://doi.org/10.1186/s12906-020-
Goldstein, J. L., & Brown, M. S. (2009). History of discovery: The LDL 03162-5
receptor. Arteriosclerosis, Thrombosis, and Vascular Biology, 29(4), Mezei, O., Banz, W. J., Steger, R. W., Peluso, M. R., Winters, T. A., & Shay,
N. (2003). Soy Isoflavones exert antidiabetic and hypolipidemic effects
431–438. https://doi.org/10.1161/ATVBAHA.108.179564
through the PPAR Pathways in obese zucker rats and murine RAW
Gustafsen, C., Olsen, D., Vilstrup, J., Lund, S., Reinhardt, A., Wellner, N.,
264.7 cells. The Journal of Nutrition, 133(5), 1238–1243. https://doi.org/
Larsen, T., Andersen, C. B. F., Weyer, K., Li, J., Seeberger, P. H., Thirup,
10.1093/jn/133.5.1238
S., Madsen, P., & Glerup, S. (2017). Heparan sulfate proteoglycans pre-
Morris, G. M., Huey, R., Lindstrom, W., Sanner, M. F., Belew, R. K.,
sent PCSK9 to the LDL receptor. Nature Communications, 8(1), 503.
Goodsell, D. S., & Olson, A. J. (2009). AutoDock4 and AutoDockTools4:
https://doi.org/10.1038/s41467-017-00568-7
Automated docking with selective receptor flexibility. Journal of
Herwana, E., Pusparini, P., & Graciela, A. (2020). High dietary daidzein
Computational Chemistry, 30(16), 2785–2791. https://doi.org/10.1002/
intake lowers cholesterol levels among post-menopausal women.
jcc.21256
Universa Med, 39(1), 47–54. https://doi.org/10.18051/UnivMed.2020. Morris, G. M., Goodsell, D. S., Halliday, R. S., Huey, R., Hart, W. E., Belew,
v39.47-54 R. K., & Olson, A. J. (1998). Automated docking using a Lamarckian
Huang, H., Xie, Z., Boue, S. M., Bhatnagar, D., Yokoyama, W., Yu, L. L., & genetic algorithm and an empirical binding free energy function. J
Wang, T. T. Y. (2013). Cholesterol-lowering activity of soy-derived gly- Comput Chem 19:1639–1662. https://doi.org/10.1002/(SICI)1096-
ceollins in the golden Syrian hamster model. Journal of Agricultural 987X(19981115)19:14 < 1639::AID-JCC10 > 3.0.CO;2-B
and Food Chemistry, 61(24), 5772–5782. https://doi.org/10.1021/ Mullen, E., Brown, R. M., Osborne, T. F., & Shay, N. F. (2004). Soy
jf400557p Isoflavones Affect sterol regulatory element binding proteins (SREBPs)
Istvan, E. S., Palnitkar, M., Buchanan, S. K., & Deisenhofer, J. (2000). and SREBP-regulated genes in HepG2 cells. The Journal of Nutrition,
Crystal structure of the catalytic portion of human HMG-CoA reduc- 134(11), 2942–2947. https://doi.org/10.1093/jn/134.11.2942
tase: Insights into regulation of activity and catalysis. The EMBO Notarnicola, M., Messa, C., Orlando, A., D’Attoma, B., Tutino, V.,
Journal, 19(5), 819–830. https://doi.org/10.1093/emboj/19.5.819 Rivizzigno, R., & Caruso, M. G. (2008). Effect of genistein on
JOURNAL OF BIOMOLECULAR STRUCTURE AND DYNAMICS 7

cholesterol metabolism-related genes in a colon cancer cell line. cholesterol. Nature, 582(7810), 73–77. https://doi.org/10.1038/s41586-
Genes & Nutrition, 3(1), 35–40. https://doi.org/10.1007/s12263-008- 020-2338-1
0082-5 Taku, K., Umegaki, K., Sato, Y., Taki, Y., Endoh, K., & Watanabe, S. (2007).
Patil, R., Das, S., Stanley, A., Yadav, L., Sudhakar, A., & Varma, A. K. (2010). Soy isoflavones lower serum total and LDL cholesterol in humans: a
Optimized hydrophobic interactions and hydrogen bonding at the meta-analysis of 11 randomized controlled trials. The American
target-ligand interface leads the pathways of drug-designing. PloS Journal of Clinical Nutrition, 85(4), 1148–1156. https://doi.org/10.1093/
One, 5(8), e12029. https://doi.org/10.1371/journal.pone.0012029 ajcn/85.4.1148
Ramdath, D. D., Padhi, E. M. T., Sarfaraz, S., Renwick, S., & Duncan, A. M. Tang, J.-J., Li, J.-G., Qi, W., Qiu, W.-W., Li, P.-S., Li, B.-L., & Song, B.-L.
(2017). Beyond the cholesterol-lowering effect of soy protein: A (2011). Inhibition of SREBP by a small molecule, betulin, improves
review of the effects of dietary soy and its constituents on risk factors hyperlipidemia and insulin resistance and reduces atherosclerotic pla-
for cardiovascular disease. Nutrients, 9(4), 324. https://doi.org/10.3390/ ques. Cell Metabolism, 13(1), 44–56. https://doi.org/10.1016/j.cmet.
nu9040324 2010.12.004
Sirtori, C. R., Lovati, M. R., Manzoni, C., Monetti, M., Pazzucconi, F., & Teixeira Damasceno, N. R., Apolinario, E., Dias Flauzino, F., Fernandes, I.,
Gatti, E. (1995). Soy and cholesterol reduction: Clinical experience. The
& Abdalla, D. S. P. (2007). Soy isoflavones reduce electronegative low-
Journal of Nutrition, 125(3 Suppl), 598S–605S. https://doi.org/10.1093/
density lipoprotein (LDL(-)) and anti-LDL (-) autoantibodies in experi-
jn/125.suppl_3.598S
mental atherosclerosis. European Journal of Nutrition, 46(3), 125–132.
Soran, H., Adam, S., Mohammad, J. B., Ho, J. H., Schofield, J. D., Kwok, S.,
https://doi.org/10.1007/s00394-006-0640-9
Siahmansur, T., Liu, Y., Syed, A. A., Dhage, S. S., Stefanutti, C., Donn,
Wang, Q., Ge, X., Tian, X., Zhang, Y., Zhang, J., & Zhang, P. (2013). Soy
R., Malik, R. A., Banach, M., & Durrington, P. N. (2018).
Hypercholesterolaemia – practical information for non-specialists. isoflavone: The multipurpose phytochemical (Review). Biomedical
Archives of Medical Science : AMS, 14(1), 1–21. https://doi.org/10.5114/ Reports, 1(5), 697–701. https://doi.org/10.3892/br.2013.129
aoms.2018.72238 Wong, W. W., Smith, E. O., Stuff, J. E., Hachey, D. L., Heird, W. C., &
Sung, J. H., Lee, S.-J., Park, K. H., & Moon, T. W. (2004). Isoflavones inhibit Pownell, H. J. (1998). Cholesterol-lowering effect of soy protein in nor-
3-hydroxy-3-methylglutaryl coenzyme a reductase in vitro. Bioscience, mocholesterolemic and hypercholesterolemic men. The American
Biotechnology, and Biochemistry, 68(2), 428–432. https://doi.org/10. Journal of Clinical Nutrition, 68(6 Suppl), 1385S–1389S. https://doi.org/
1271/bbb.68.428 10.1093/ajcn/68.6.1385S
Taddei, C., Zhou, B., Bixby, H., Carrillo-Larco, R. M., Danaei, G., Jackson, Zarate, A., Manuel-Apolinar, L., Saucedo, R., Hernandez-Valencia, M., &
R. T., Farzadfar, F., Sophiea, M. K., Di Cesare, M., Iurilli, M. L. C., Basurto, L. (2016). Hypercholesterolemia as a risk factor for cardiovas-
Martinez, A. R., Asghari, G., Dhana, K., Gulayin, P., Kakarmath, S., cular disease: Current controversial therapeutic management. Archives
Santero, M., Voortman, T., Riley, L. M., Cowan, M. J., … Ezzati, M. of Medical Research, 47(7), 491–495. https://doi.org/10.1016/j.arcmed.
(2020). Repositioning of the global epicentre of non-optimal 2016.11.009

You might also like