SlideShare a Scribd company logo
1 of 21
Download to read offline
Journal Pre-proof
Antidiabetic and Antioxidant Potentials of Pleurotus ostreatus
-derived Compounds: An in vitro and in silico approach
S.C. Nnemolisa , C.C. Chukwurah , S.C. Edeh , R.N. Aguchem ,
C.C. Chibuogwu , E.C. Aham , M.C. Chukwu , M.O. Obiora ,
D.E. Anyebe , I.U. Okagu
PII: S2772-753X(24)00035-2
DOI: https://doi.org/10.1016/j.focha.2024.100639
Reference: FOCHA 100639
To appear in: Food Chemistry Advances
Received date: 30 June 2023
Revised date: 10 December 2023
Accepted date: 6 February 2024
Please cite this article as: S.C. Nnemolisa , C.C. Chukwurah , S.C. Edeh , R.N. Aguchem ,
C.C. Chibuogwu , E.C. Aham , M.C. Chukwu , M.O. Obiora , D.E. Anyebe , I.U. Okagu , Antidia-
betic and Antioxidant Potentials of Pleurotus ostreatus -derived Compounds: An in vitro and in silico
approach, Food Chemistry Advances (2024), doi: https://doi.org/10.1016/j.focha.2024.100639
This is a PDF file of an article that has undergone enhancements after acceptance, such as the addition
of a cover page and metadata, and formatting for readability, but it is not yet the definitive version of
record. This version will undergo additional copyediting, typesetting and review before it is published
in its final form, but we are providing this version to give early visibility of the article. Please note that,
during the production process, errors may be discovered which could affect the content, and all legal
disclaimers that apply to the journal pertain.
© 2024 Published by Elsevier Ltd.
This is an open access article under the CC BY-NC-ND license
(http://creativecommons.org/licenses/by-nc-nd/4.0/)
1
Highlights
• Pleurotus ostreatus methanol extract (MEPO) contains many health-beneficial compounds
• MEPO showed stronger inhibition of α-glucosidase activity than α-amylase.
• MEPO showed potential for blood glucose regulation in in silico study.
2
Antidiabetic and Antioxidant Potentials of Pleurotus ostreatus -derived Compounds: An in vitro and
in silico approach
1
Nnemolisa, S.C., 1
Chukwurah, C.C., 1
Edeh, S.C., 1
Aguchem, R.N., 1,2
Chibuogwu*, C.C., 1
Aham, E.C.,
3
Chukwu, M.C., 1
Obiora, M.O., 1
Anyebe, D.E. and 1
Okagu, I.U.
1 Department of Biochemistry, Faculty of Biological Sciences, University of Nigeria, Nsukka, Enugu,
Nigeria.
2 Institute for Drug-Herbal Medicine-Excipient Research and Development, Faculty of Pharmaceutical
Sciences, University of Nigeria, Nsukka, Enugu, Nigeria.
3 Department of Genetics and Biotechnology, Faculty of Biological Sciences, University of Nigeria, Enugu,
Nsukka.
* Correspondence: christian.chibuogwu@unn.edu.ng (ORCID: 0000-0002-9091-4310) +2347069263943
Abstract
Many health benefits have reportedly been associated with mushroom consumption. This study determined
the chemical constituents of Pleurotus ostreatus methanol extract (MEPO) and investigated its antioxidant
and antidiabetic effects using in vitro and in silico approaches. The chemical composition of MEPO was
determined using the gas chromatography-flame ionization detector (GC-FID) technique, while 2,2-
Diphenyl-1-picrylhydrazyl (DPPH) and ferric reducing antioxidant power (FRAP) methods were used to
determine antioxidant activity. The antidiabetic activity was evaluated using α-amylase and α-glucosidase
inhibition assays, while molecular docking was done to give insight into the binding potentials of MEPO
constituents against α-amylase, α-glucosidase, and phosphoenolpyruvate (PEP) carboxykinase activities.
Thirteen compounds, including ephedrine, oxalate, rutin, naringin, and kaempferol, were identified in
MEPO. The extract showed moderate antioxidant activity, as observed from the DPPH (IC50 = 732.41
mg/ml) and FRAP studies. The extract also demonstrated stronger inhibition of α-glucosidase activity (IC50
= 246.58 mg/ml) than α-amylase activity (IC50 = 1074.05 mg/ml). Docking studies revealed that rutin and
naringin interacted effectively with amino acid residues crucial for α-amylase, α-glucosidase, and PEP
carboxykinase activities via hydrogen bonds. The result shows that MEPO is a rich store of beneficial
compounds which could be explored for the management of diabetes and associated complications.
Keywords: Mushroom, methanol extract, α-amylase, α-glucosidase, PEP carboxykinase.
1. Introduction
Diabetes mellitus (DM) is a group of chronic metabolic disorders characterized by an abnormal rise in
plasma glucose levels resulting from poor glucose metabolism due to abnormal insulin secretion, insulin
deficiency, and/or insulin insensitivity (Singh et al., 2022). In 2021, 536.6 million people were reportedly
living with diabetes, and a 46% increase (783.2 million) has been predicted by 2045 (Ogurtsova et al.,
2022). Most diabetics (80.6%, 432.7 million) reside in low- and middle-income nations and about half of
these individuals are more likely to experience complications because they are unaware that they have the
condition (Sun et al., 2022). These complications are the major causes of mortality in diabetic patients. One
of the factors contributing to the pathogenesis of diabetic complications from hyperglycemia is an
impairment of reactive oxygen species and antioxidant defense balance (Dehghan et al., 2016). This results
3
in a state known as oxidative stress. Indeed, studies have shown that hyperglycemia-induced oxidative
stress is implicated in vascular damage of diabetes subjects (Singh et al., 2022).
Numerous consequences of diabetes may be delayed or prevented with intensive postprandial
hyperglycemic management. However, the available treatment reflects an insufficient and less effective
glycemic control for diabetic patients (Abbas et al., 2019). This has led to the exploration of more effective
therapeutic targets and approaches to glycemic control. One of the therapeutic options presently explored
for effective glycemic control is the inhibition of α-amylase and α-glucosidase activities. The α-amylase is
a calcium metalloenzyme that plays an important role in starch digestion catalyzing the hydrolysis of α-(1,
4)-D-glycosidic linkages in starch and other polymers of glucose, producing smaller fragments (Sun et al.,
2020). These products are further degraded by α-glucosidase to absorbable monosaccharides which then
enters the bloodstream. Therefore, inhibiting these enzymes would hinder the absorption of starch, expand
intestinal sugar-holding time and delay the rate of glucose absorption and consequently decrease blood
sugar level. Acarbose, voglibose and migiltol are drugs used to inhibit the activities of these enzymes in
practice. However, they cause unwanted side effects such as abdominal discomfort (Alqahtani et al., 2020)
which hinders their use in the treatment of diabetes and its complications. This has therefore led to urgent
requirement for effective substitutes which would reduce diabetes complications with fewer side effects.
Phosphoenolpyruvate carboxykinase (PEPCK) is a key enzyme in gluconeogenesis that catalyzes the
carboxylation of pyruvate to form oxaloacetate in the gluconeogenic pathway (Gómez-Valadés et al., 2006).
Although this enzyme is widely implicated for its role in hepatic glucose production in diabetic conditions,
studies have shown that overexpression of PEPCK in adipose tissue is a major contributor to obesity
development due to increased glyceroneogenesis in the adipose tissue (Rines et al., 2016; Yu et al., 2021).
Therefore, it makes sense to target PEPCK activity for inhibition as a strategy in diabetes therapy.
For ages, nature has been a source of therapeutic agents and an astounding number of modern medications
have been identified from natural sources based on their usage in conventional medicine (Bhushan &
Kulshreshtha, 2018). The interest in natural substances have significantly increased in recent years, because
these substances are intended to be used as nutraceuticals to replace synthetic substances whose usage is
restricted due to their adverse effects (Omari et al., 2019). The nutraceutical potentials of mushroom have
been well documented since time immemorial; however, it is in the beginning of this century that the
nutritional and pharmacological properties of mushroom gathered much attention (Huang et al., 2022).
Mushrooms are well-known macro-fungus with a characteristic fruiting body that can either be hypogenous
or epigeous (Yadav & Negi, 2021). Mushroom cultivation has emerged as a promising agricultural business
that is land-independent with the capacity of transforming lignocellulosic waste into nutrient-rich foods, in
addition to significantly reducing environmental pollution. Nutritionally, mushroom contains a relatively
high amount of carbohydrate, protein, dietary fibers, minerals, vitamins and unsaturated fatty acids
(Nowakowski et al., 2021). Edible mushrooms are excellent sources of beneficial bioactive metabolites,
and these substances have broad-spectrum therapeutic applications, including but not limited to antiviral,
anti-inflammatory, anti-bacterial, anti-oxidant and antidiabetic effect (Mwangi et al., 2022; Suleiman et al.,
2022). Pleurotus genus is among the species with the highest production worldwide (Torres-Martínez et
al., 2022). This can be attributed to its short cultivation cycle, its ability to grow at moderate temperature
(25-30ºC) and a wide range of lignocellulosic materials (Cruz-Solorio et al., 2018). Of particular interest is
P. ostreatus which is the second most widely grown and consumed edible mushroom in the world. It is
considered a high quality food due to its high nutritional value and beneficial health effects (Lesa et al.,
4
2022). In the present study, the in vitro and in silico anti-diabetic potential of methanol extract of Pleurotus
ostreatus was investigated.
2. Materials and Methods
2.1 Collection and Authentication of Material
Pleurotus ostreatus together with wood sawdust and rice bran were the materials used for this study. Wood
sawdust and rice bran were obtained from timber and rice farmers in Enugu State, Nigeria and were
pulverized using a mechanical grinder. The mushroom was cultivated at Southeast Zonal Biotechnology
Center, University of Nigeria.
2.1.2 Chemicals and Reagents
The chemicals and reagents used in this study were of analytical grade.
2.2. Mushroom Cultivation
Mushroom was cultivated following the method described by Aguchem et al., (2022). 50 kg of the substrate
was prepared by mixing the substrate with 0.40 kg of CaCO3 before adding 38 liters of water to the mixed
substrate. The substrate mixture was then packed in a limpid refractory polythene bag, each tied with rubber
bans before sterilization at 121o
C for 15 minutes at 15 psi. After sterilization, the sterile packed substrates
were allowed to cool to room temperature. The substrates were then inoculated in a germ-free manner with
the spawn and then re-tied. The inoculated bags were then taken inside a darkroom and allowed some days
to ramify with the complete ramification of the inoculated bags clearly visible by the appearance of pin
heads and fungi hyphae. Incisions were gently made on the ramified mushroom bags in a germ-free manner
before they were transferred to the growth room for the production of fruiting body. During the course of
this process, the relative humidity and temperature were maintained at 74-86% and 25o
C respectively. By
watering the bags at least once daily, wetting the floor, keeping water in tray and ice blocks close to the
mushroom bags, humid air in the growth room was achieved. The fruiting bodies of the mushroom was
then harvested and used for analysis.
2.2.1 Preparation of Mushroom Extract
Fresh whole mushroom sample (100 g) was oven-dried at 50o
C and ground into fine particles and then
macerated in 400 ml of methanol at room temperature in an air-tight container. The container was left for
72 hours with intermittent shaking to increase the efficiency of extraction of the phytochemicals. This was
followed by filtration and concentration in vacuo to obtain the crude methanol extract of P. ostreatus
(MEPO) (Figure S1 (A-F) - supplementary materials).
2.2.2 Extraction of Chemical Constituents for GC-FID analysis
Extraction of chemical constituent for gas chromatography-flame ionization detector (GC-FID)
analysis was carried out following Boisa et al., (2020) and Ojukwu et al., (2021) methods, respectively. A
test tube containing 1 g of the sample was filled with 15 ml of methanol and 10 ml of 50% w/v KOH. The
test tube was then placed in a water bath set to 60°C for 60 minutes. After that, the content was moved to a
separatory funnel. 20 ml of ethanol, 10 ml of cold water, 10 ml of hot water and 3 ml of hexane was used
successively to wash the contents of the tube which was all transferred to the funnel. This extract was then
5
mixed with 10 ml of 10% v/v ethanol aqueous solution and rinsed three times. The solvent was evaporated
after the solution was dried with anhydrous sodium sulfate. The resulting sample was solubilized in 1000
µl of pyridine after which 200 µl (0.2 ml) of the solution was transferred to a vial for analysis.
2.2.3 Analysis of chemical constituent of MEPO
The chemical constituents of MEPO were analyzed on a BUCK M910 Gas chromatograph (GC) (BUCK
M910, PUB Scientist, USA) equipped with a flame ionization detector (FID). 0.1 ml of the extract was
injected into the GC machine. At an ionization voltage of 70 eV, injector temperature of 250°C and injector
mode was split with linear velocity 36.5 cm/sec and pressure was 57.5 kPa. The instrument was run in
electron impact mode. In principle, the detector uses a flame to ionize carbon-containing organic
compounds. During separation, the sample passes through a hydrogen-fueled flame in the GC column
which ionizes the carbon atoms in the sample.
2.2.4 Assay of the inhibitory effect of MEPO on α-amylase and α-glucosidase activities
The assay mixtures containing the solution of the enzyme (0.1 ml), acetate buffer (0.1 - 0.5 ml, 0.1 mol/l at
pH 5.5) and mushroom extract (32 - 160 mg/ml) were incubated for 15 minutes at room temperature. This
was followed by the addition of 0.5 ml of 2% starch and 2% sucrose to each test tube containing the
respective enzymes and incubation of the resulting mixture in a water bath at 50°C for 30 min. The reaction
was terminated with the addition of 1 ml of dinitrosalicylic acid (DNSA) reagent to each test tube and
placed in a boiling water bath for 10 minutes. To stabilize the color after heating, 1 ml of 1.4 mol/l Rochelle
salt (sodium potassium tartrate) was added to the test tubes immediately and the total volume of the solution
was adjusted to 4 ml with distilled water. The reaction mixtures were then cooled to room temperature and
absorbance was taken at 540 nm using a spectrophotometer (Spectron lab 2A, England). The test tube used
as control was prepared without mushroom extract. All tests were done in triplicate and the percentage of
inhibition of enzyme activity was calculated as Equation 1:
% inhibition =
(Abs.of control − Abs.of treatment)
(Abs.of control)
× 100 1
Where: Abs. of control = Absorbance of control at 540 nm; Abs. of treatment = Absorbance of sample
containing extract at 540 nm
2.2.5 In vitro antioxidant activity of MEPO
2.2.5.1 DPPH radical-scavenging assay
The DPPH radical-scavenging activity of MEPO was determined following the protocol of Sayah et al.,
(2017). Differently labelled test tubes received additions of MEPO at various mass concentrations (32 - 160
mg/ml) followed by the addition of MeOH to bring the volume to 250 𝜇l. Each test tube was then shaken
violently following the addition of 2 ml of a 0.18 mmol/l (0.005%) methanolic solution of DPPH after
which the mixture was allowed to stand in the dark for 30 minutes at room temperature. The same procedure
was used to prepare the control without any extract and MeOH served for baseline correction. Changes in
samples absorbances were taken at 517 nm using a spectrophotometer (Spectron lab 2A, England). DPPH
radical-scavenging potential was calculated using the method provided by Adebiyi et al., (2017).
6
2.2.5.2 Ferric-reducing antioxidant power assay
The method described by Alachaher et al., (2018) was used to determine the ferric reducing antioxidant
potential of MEPO. 1 ml solution containing 32, 64, 96, 128, and 160 mg/ml each of the extracts was
combined with potassium ferricyanide (5.0 ml, 1.0%) and sodium phosphate buffer (5.0 ml, 0.2 mol/l, pH
6.6). The mixture was incubated at 50°C for 20 minutes followed by the addition of 5 mL of 10%
trichloroacetic acid and centrifuged at 1000 xg (10 min at 5°C). The upper layer of the solution (5.0 ml)
was diluted with 5.0 ml of distilled water and 0.5 ml ferric chloride (0.1% w/v) and absorbance measured
at 700 nm using a spectrophotometer (Spectron lab 2A, England). The experiment was performed thrice
and scavenging activity calculated as in Equation 2.
Scavenging activity (%) =
(Abs.of control − Abs.of treatment)
(Abs.of control)
× 100 2
Where: Abs. of control = Absorbance of control at 700 nm; Abs. of treatment = Absorbance of sample
containing extract at 700 nm
2.3 In silico molecular docking studies
2.3.1 Ligands and protein preparation
Epicatechin, naringin, catechin, kaempferol, linamarin, rutin, resveratrol, spartein, ephedrine and standard
drug (Acarbose) were the ligands used in this study. The chemical structures of the ligands were retrieved
from NCBI PubChem database (https://www.ncbi.nlm.nih.gov/pccompound) in 2D (sdf) format and were
converted to 3D (PDB) and PDBQT structures using Discovery Studio software and AutoDock Tool,
respectively.
The selected crystal structures of the proteins, α-amylase (PDB entry: 1U33), PEP-carboxykinase (PDB
entry: 1KHB) and α-glucosidase (PDB entry: 5KZW) were obtained from the Protein Data Bank
(http://www.rcsb.org/pdb). The crystallographic water as well as the existing ligands were removed. The
crystal structures were changed to PDBQT format after the addition of hydrogen atom to the geometry. The
structures were refined, and their geometries optimized with the aid of AutoDock Tool.
2.3.2 Docking
Following the protocol described by Dundas et al. (2006), identification of the proteins active sites was
done using CASTp server (Computer Atlas of Surface Topology of protein). The X, Y, Z center coordinates
and X, Y, Z grid dimensions of proteins were selected (30 x 30 x 30 Å), optimized and thereafter docked.
Docking and image preparation was performed with AutoDock Vina (Scripps Research Institute, San
Diego, CA, USA), BIOVIA Discovery Studio and PyMOL (to generate 3D structures).
2.3.4 Drug-likeness and toxicity prediction
In silico prediction of molecular descriptors and drug-like properties of the MEPO phytochemicals was
estimated using an online tool on the SwissADME server (http://http://www.swissadme.ch/index.php). MW
(molecular weight), cLogP, HBD (number of hydrogen bond donors), HBA (number of hydrogen bond
acceptors), gastro-intestinal absorption, and nViolation (number of violations of Lipinski’s rule of five)
7
were calculated on the basis of Lipinski's rule of five as described by Daina et al., (2017). The ProTox
webserver was used to predict the toxicity profile of the MEPO phytochemicals.
2.4 Statistical analysis
The results obtained were analyzed using the Statistical Product and Service Solutions (SPSS) software
(version 20). Results were presented as mean ± standard deviation and tests for statistical significance done
using one way analysis of variance (ANOVA) at 95% confidence level and mean values with p≤ 0.05 was
taken to be significant. The IC50 values were estimated from the linear plot of inhibition (%) against
concentration using the equation of the line graph.
3. Results
3.1 Phytochemical constituents of methanol extract of Pleurotus ostreatus
The gas chromatography analysis of the methanol extract of Pleurotus ostreatus as presented in Table 1
showed the presence of thirteen (13) compounds. Ephedrine was found to be the highest followed by
oxalate. Rutin, naringin, kaempferol, epicatechin, phytate, resveratrol, catechin were also detected (Figure
S2 - Supplementary materials).
Table 1: phytochemical constituents of methanol extract of Pleurotus ostreatus
Compounds Concentration (𝝁g/ml)
Linamarin 8.03 ± 0.05
Catechin 4.27 ± 0.16
Sapogenin 7.97 ± 0.04
Rutin 11.19 ± 0.45
Spartein 8.18 ± 0.14
Flavan-3-ol 4.34 ± 0.15
Naringin 10.40 ± 0.15
Resveratrol 7.38 ± 0.35
Kaempferol 9.02 ± 0.13
Phytate 7.65 ± 0.29
Epicatechin 7.72 ± 0.10
Oxalate 16.68 ± 0.34
Ephedrine 33.77 ± 0.92
8
3.2 In vitro antidiabetic activity of the mushroom extract
The result presented in Table 2 shows the percentage inhibition of α-amylase and α-glucosidase activity by
MEPO. The result showed a concentration-related inhibitory effect of the extract on α-amylase and α-
glucosidase activity with IC50 value of 1074.05 mg/ml and 246.58 mg/ml respectively. The highest
inhibitory effect was observed with the highest concentration of the extract (160 mg/ml) and vice versa.
Table 2: Inhibitory effect of MEPO on α-amylase and α-glucosidase activity in vitro
Sample concentration
(mg/ml)
α-Amylase activity
(%)
α-Glucosidase activity
(%)
32 4.8±0.18 33.87±0.63
64 5.1± 0.16 36.53± 0.09
96 5.14±0.11 36.69±0.6
128 8.79±0.23 41.9±0.41
160 10.03±0.46 43.43±0.16
IC50 = 1074.05 mg/ml IC50 = 246.58
mg/ml
3.3. In vitro antioxidant effects of methanolic extract of P. ostreatus
The DPPH radical-scavenging activity and ferric reducing antioxidant power of MEPO is shown in Table
3. MEPO demonstrated DPPH radical-scavenging activities in a concentration-dependent manner with an
IC50 value of 732.41 mg/ml; the highest DPPH radical-scavenging effect was observed with the highest
concentration. The same trend is also obtainable with the ferric reducing antioxidant power of the extract.
Table 3: In vitro DPPH radical-scavenging activity and ferric reducing antioxidant power of MEPO
Sample concentration (mg/ml) % Quenching of DPPH radical FRAP
32 9.73 ± 0.10a
77.00 ± 0.58a
64 9.58 ± 0.07a
84.93 ± 0.31b
96 12.21 ± 0.25b
87.18 ± 0.68c
128 14.88 ± 0.20c
89.46 ± 0.11d
160 16.48 ± 0.11d
98.13 ± 0.54e
IC50 = 732.41 mg/ml
3.4. In silico molecular docking studies
A molecular docking study was carried out to elucidate the binding interactions of MEPO phytochemicals
at the active sites of α-amylase, α-glucosidase, and PEP carboxykinase to garner an understanding into the
in vitro results. From Table 4, favorable overall binding energies were observed in ranges of -5.9 to -9.8
kcal/mol for α-amylase (1U33), -5.7 to -9.3 kcal/mol for α-glucosidase and -5.4 to -9.8 kcal/ mol for PEP
carboxykinase (1KHB). Epicatechin, naringin and rutin had the best binding energies across all three targets
when compared to Acarbose. On the other hand, kaempferol and linamarin showed better binding energies
for α-amylase and PEP carboxykinase only when compared to the Acarbose.
9
Table 4: Binding energies of selected compounds and standard inhibitors against protein targets
Ligands
α-amylase
(1U33)
Proteins (Kcal/mol)
α-glucosidase
(5KZW)
PEP carboxykinase
(1KHB)
Epicatechin -9.8 -8.2 -9.3
Naringin -9.4 -9.3 -9.8
Catechin -9.3 -7.2 -7.8
Kaempferol -9.3 -6.9 -8.0
Linamarin -9.0 -7.3 -8.6
Rutin -8.7 -8.8 -9.6
Resveratrol -8.1 -6.9 -7.5
Spartein -7.7 -6.5 -6.6
Ephedrine -5.9 -5.7 -5.4
Standard (Acarbose) -7.5 -8.3 -7.5
3.5 Drug-likeness prediction of the MEPO phytochemicals
Table 5 shows the drug-likeness of MEPO phytochemicals. Apart from naringin and rutin, all the MEPO
phytochemicals tested had MW less than 500, HBA < 10 and HBD not more than 5. All the compounds
showed reasonable water solubility while only naringin and rutin violated Lipinski’s rule for orally
administered drugs similar to the standard drug.
10
Table 5: Drug-likeness Prediction of the MEPO phytochemicals
Phytochemicals MW
(g/mol)
No.
HBA
No.
HBD
cLogP
value
GIA No. violation Water solubility
Epicatechin 290.27 6 5 0.85 High No Soluble
Naringin 580.53 14 8 -0.79 Low 3 Soluble
Catechin 290.27 6 5 0.85 High No Soluble
Kaempferol 286.24 6 4 1.58 High No Soluble
Linamarin 309.32 4 0 2.87 High No Moderately
soluble
Rutin 610.52 16 10 -1.29 High 3 Soluble
Resveratrol 228.24 3 3 2.48 High No Soluble
Spartein 234.38 2 0 2.36 Low No Soluble
Ephedrine 165.23 2 2 1.45 Low No Very Soluble
Acarbose 645.60 19 14 -6.09 Low 3 Highly Soluble
MW: Molecular weight (<500), HBA: Number of Hydrogen Bond Acceptors (≤10), HBD: Number of
Hydrogen Bond Donor (≤5), cLogP ≤ 5, No. Violation: Number of Violation of Lipinski’s rule.
3.6 Toxicity study of MEPO phytochemicals
Apart from ephedrine, linamarin and resveratrol, all the MEPO phytochemicals had LD50 greater than 2000
mg/kg. The toxicity prediction showed relative safety of MEPO phytochemicals except for rutin and
naringin which showed potentials for immuno-toxicity, like Acarbose. Linamarin also showed potential for
hepatotoxicity like Acarbose.
11
Table 6: Toxicity Properties of the MEPO phytochemicals
Compounds Predicted
LD50 value
(mg/kg)
Prediction
accuracy
(%)
Hepato-
toxicity
Immuno
-toxicity
Cytotoxi
city
Carcino
genicity
Mutagen
icity
Epicatechin 10000 100% Inactive Inactive Inactive Inactive Inactive
Naringin 2300 70.97% Inactive Active Inactive Inactive Inactive
Catechin 10000 70.97% Inactive Inactive Inactive Inactive Inactive
Kaempferol 3919 100% Inactive Inactive Inactive Inactive Inactive
Linamarin 400 100% Active Inactive Inactive Inactive Inactive
Rutin 5000 100% Inactive Active Inactive Inactive Inactive
Resveratrol 1560 68.07% Inactive Inactive Inactive Inactive Inactive
Spartein 2850 100% Inactive Inactive Inactive Inactive Inactive
Ephedrine 404 100% Inactive Inactive Inactive Inactive Inactive
Acarbose 24000 100% Active Active Inactive Inactive Inactive
4. Discussion
Despite available pharmaceutical interventions, the current projections of the incidence of diabetes and its
associated complications have necessitated the search for alternative therapeutic approaches. In this study,
in vitro and in silico antidiabetic potential of methanol extract of P. ostreatus (MEPO) was evaluated. Our
results demonstrated that MEPO significantly inhibited α-amylase and α-glucosidase activities in a
concentration-dependent manner. α -amylase and α-glucosidase are two enzymes that play key role in
carbohydrate metabolism. While α-amylase facilitates the catabolism of starch, releasing absorbable
molecules, α-glucosidase catalyzes the end step of starch and disaccharides digestion from human diet
(Kazeem et al., 2013). The uncontrolled activity of these enzymes is one of the contributors to postprandial
hyperglycemia and has been implicated in diabetes development and complications. Hence, inhibiting their
activities has become a strategy in blood glucose control. The inhibition of these enzymes’ activities is
indicative of the potential of MEPO as a promising candidate for glycemic control in diabetes management
and this is supported by previous studies. Karim et al., (2020) and Johnny & Okon, (2013) reported anti-
hyperglycemic effects of solvent extracts of P. ostreatus, using in vivo models and from our result,
inhibition of these enzymes may be one mechanism in which P. ostreatus exerts its anti-hyperglycemic
effect.
Oxidative stress, occasioned by excessive free radical production, is implicated in both the development
and progression of many disease conditions (Yaribeygi et al., 2020); thus, antioxidant therapy remains a
viable option to mitigate the oxidative damage that is associated with many diseases. DPPH radical-
scavenging and FRAP assays are common test methods used to determine the antioxidant capacity of test
substances. Antioxidant compounds are able to stabilize/quench DPPH radicals resulting in a change of
color of reaction mixture from purple to yellow, while in the FRAP assay, antioxidant compounds prevent
12
the oxidation of ferrous ion (Fe2+
) to ferric ion (Fe3+
) (Omari et al., 2019). Our result showed that MEPO
moderately scavenged DPPH radicals and reduced Fe3+
to Fe2+
in a concentration-related manner, indicating
that MEPO has phytochemicals with antioxidant properties. In agreement with our result, Mishra et al.,
(2022) and Mkhize et al., (2022) also reported good DPPH radical-scavenging activity and FRAP,
respectively, of solvent extract of P. ostreatus. The same trend was also reported by Menaga et al., (2014)
and Leong et al., (2021) for extracts of other mushroom species.
To identify the phytochemicals potentially involved in the anti-diabetic and antioxidant potentials of
MEPO, we used gas chromatography instrument coupled with a flame ionization detector (GC-FID)
ephedrine, oxalate, cyanogenic glycoside, rutin, naringin, kaempferol, epicatechin, phytate and catechin,
among others. The presence of these compounds indicates that MEPO is a rich store of health-beneficial
bioactive compounds. For example, catechin, epicatechin, rutin, and kaempferol are polyphenolic
compounds known for their antioxidant and antidiabetic properties (Ademiluyi et al., 2018; Chukwuma et
al., 2022). Thus, the presence of these phytochemicals in MEPO could have individually or in combination
contributed to the antioxidant and antidiabetic potentials observed in this study.
To clarify the contribution of each of the identified compounds to the antidiabetic properties of MEPO, we
conducted molecular docking studies. Nine MEPO phytochemicals (naringin, kaempferol, resveratrol,
linamarin, rutin, epicatechin, catechin, spartein, and ephedrine) were docked against α-amylase, α-
glucosidase and PEPCK to provide insights into the ligand-complexes orientations and interactions between
these phytochemicals and the enzymes implicated in diabetes based on the binding energies. MEPO
phytochemicals demonstrated favorable binding energies (negative binding scores) against α-amylase, α-
glucosidase and PEPCK. Negative binding scores usually indicate correctly docked compounds into target
proteins’ crystal structure and higher negative scores usually indicate stronger binding affinities toward
targets (Kumar et al., 2019).
The active site of every enzyme consists of amino acid residues with three different functions, viz, catalytic
residues (responsible for the ligand conversion to products), ligand-binding residues (responsible for the
binding of the ligand to active site), and structural residues (responsible for the maintenance of the active
site structure in its functional state). Three amino acids, Asp300, Glu233, and Asp197 are the catalytic
residues of α-amylase active site. Our study showed that naringin and epicatechin had better interactions
with α-amylase active site’s residues compared to the other MEPO phytochemicals docked. Naringin had
nine hydrogen (H)-bond interactions within the active site with His305, Asp300, Glu233, Asp197, Glu240,
Lys200, Thr163, Gln63, and Trp59. Likewise, epicatechin had five hydrogen bond interactions with
His299, Asp300, Glu233, Thr163 and Gln63. A visual representation of the 3D and 2D of the docked
ligand conformations at the active site of α-amylase (Figure 1, see appendix) indicated that interactions
occurred with the key residues at the active site, like Acarbose which had four H-bond interactions with
Asp197, Asp300, Glu233 and Asn53. Naringin and rutin showed higher binding energies for α-glucosidase
when compared to Acarbose. Naringin formed H-bonds with Arg411, Ser676, Leu678, Asp518, Met519
and Arg600. We also observed that among the six H-bond interactions observed with naringin, 2-H bonds
(Arg600 and Asp518) were also observed with Acarbose. Similarly, rutin showed H-bond interactions with
Asp616, His674, Asp404, Trp481, Asp518, and Ser679. Naringin and epicatechin showed higher binding
energies (-9.8 kcal/mol and -9.3 kcal/mol, respectively) for PEPCK compared to Acarbose (-7.5 kcal/mol).
Epicatechin formed H-bond interactions with Asn403, Ala287, Cys288, Gly289, Thr291, Val335, Lys290,
Arg436 and Asn292. Naringin also had H-bond interactions with Ala287, Gly289, Cys288, Asn292,
Lys290, Pro402 and Arg87, compared to Acarbose that formed H-bond interactions with Asn292, Phe333,
13
Asp311, Lys244, Lys290, Thr291, Cys288, Gly289. Hence, because of the more negative binding energies
exhibited by the phytochemicals coupled with their interactions with higher number of active site amino
acid residue may explain why MEPO constituents are better in regulating blood glucose level than
Acarbose. This may suggest that MEPO constituent might serve as an effective alternative in diabetes
management.
According to the Lipinski's rule, a drug must have an HBD (hydrogen bond donors) count of ≤ 5 and an
HBA (hydrogen bond acceptor) of ≤ 10 in order for it to be orally active (Lipinski, 2016). All the MEPO
phytochemicals and standard drug passed the oral bioactivity test, except for rutin and naringin which had
high HBDs (10 and 8, respectively) and high HBAs (16 and 14, respectively). Similarly, rutin and naringin
displayed immunotoxicity potentials, while linamarin showed hepatotoxic effects, unlike other MEPO
phytochemicals that are predicted to be safe. Future studies should establish the safety profile of MEPO
and its phytochemicals using cellular and in vivo models.
Conclusion
Methanol extract of Pleurotus ostreatus (MEPO) is rich in health-enhancing phytochemicals and
demonstrated moderate antioxidant and antidiabetic activities as evidenced by in vitro and in silico
molecular docking studies. The presence of the identified constituents and their respective interactions with
enzymes implicated in blood glucose elevation suggest that the P. ostreatus-derived compounds may be a
promising therapeutic strategy for managing blood sugar levels, especially in diabetic conditions. Further
studies are warranted to confirm the above beneficial potentials using in vivo models.
Data Availability
The data supporting the outcome of this present study are included within the article.
Declaration of Competing Interest
None
Acknowledgement
None.
REFERENCES
Abbas, G., Al Harrasi, A., Hussain, H., Hamaed, A., & Supuran, C. T. (2019). The management of diabetes
mellitus-imperative role of natural products against dipeptidyl peptidase-4, α-glucosidase and sodium-
dependent glucose co-transporter 2 (SGLT2). Bioorganic Chemistry, 86(February), 305–315.
https://doi.org/10.1016/j.bioorg.2019.02.009
Adebiyi, O. E., Olayemi, F. O., Ning-Hua, T., & Guang-Zhi, Z. (2017). In vitro antioxidant activity, total
phenolic and flavonoid contents of ethanol extract of stem and leaf of Grewia carpinifolia. Beni-Suef
University Journal of Basic and Applied Sciences, 6(1), 10–14.
https://doi.org/10.1016/j.bjbas.2016.12.003
Ademiluyi, A. O., Aladeselu, O. H., Oboh, G., & Boligon, A. A. (2018). Drying alters the phenolic
constituents, antioxidant properties, α-amylase, and α-glucosidase inhibitory properties of Moringa
(Moringa oleifera) leaf. Food Science and Nutrition, 6(8), 2123–2133.
https://doi.org/10.1002/fsn3.770
14
Aguchem, R. N., Chibuogwu, C. C., Okolo, B. O., Oyeagu, U., Etim, V. E., Anaele, E. N., & Njoku, O. U.
(2022). Nutrient and Antinutrient Composition of Pleurotus ostreatus Grown on Different Substrates.
69. https://doi.org/10.3390/iecps2021-11955
Alachaher, F. Z., Dali, S., Dida, N., & Krouf, D. (2018). Comparison of phytochemical and antioxidant
properties of extracts from flaxseed (Linum usitatissimum) using different solvents. International
Food Research Journal, 25(1), 75–82.
Alqahtani, A. S., Hidayathulla, S., Rehman, T., Elgamal, A. A., Dib, R. A. El, & Alajmi, M. F. (2020).
Alpha-Amylase and Alpha-Glucosidase Enzyme Inhibition and Antioxidant Potential of 3-
Oxolupenal and Katononic Acid Isolated from Nuxia oppositifolia. Biomolecules, 10(61), 2–19.
https://pubmed.ncbi.nlm.nih.gov/31905962/
Bhushan, A., & Kulshreshtha, M. (2018). The Medicinal Mushroom Agaricus bisporus: Review of
Phytopharmacology and Potential Role in the Treatment of Various Diseases. Journal of Nature and
Science of Medicine, 1, 4–9. https://doi.org/10.4103/JNSM.JNSM
Boisa N, Nwachoko N, Bull OS, & James FA. (2020). Physical Composition, Proximate, Phytochemical
and Impact of Coconut Oil on Lipid Profile of Albino Rats. IOSR Journal of Applied Chemistry, 13(7),
51–56. https://doi.org/10.9790/5736-1307015156
Chukwuma, I. F., Nworah, F. N., Apeh, V. O., Omeje, K. O., Nweze, E. J., Asogwa, C. D., & Ezeorba, T.
P. C. (2022). Phytochemical Characterization, Functional Nutrition, and Anti-Diabetic Potentials of
Leptadenia hastata (pers) Decne Leaves: In Silico and In Vitro Studies. Bioinformatics and Biology
Insights, 16. https://doi.org/10.1177/11779322221115436
Cruz-Solorio, A., Villanueva-Arce, R., Garín-Aguilar, M. E., Leal-Lara, H., & Valencia-del Toro, G.
(2018). Functional properties of flours and protein concentrates of 3 strains of the edible mushroom
Pleurotus ostreatus. Journal of Food Science and Technology, 55(10), 3892–3901.
https://doi.org/10.1007/s13197-018-3312-x
Daina, A., Michielin, O., & Zoete, V. (2017). SwissADME: A free web tool to evaluate pharmacokinetics,
drug-likeness and medicinal chemistry friendliness of small molecules. Scientific Reports, 7(January),
1–13. https://doi.org/10.1038/srep42717
Dehghan, H., Sarrafi, Y., & Salehi, P. (2016). Antioxidant and antidiabetic activities of 11 herbal plants
from Hyrcania region, Iran. Journal of Food and Drug Analysis, 24(1), 179–188.
https://doi.org/10.1016/j.jfda.2015.06.010
Dundas, J., Ouyang, Z., Tseng, J., Binkowski, A., Turpaz, Y., & Liang, J. (2006). CASTp: Computed atlas
of surface topography of proteins with structural and topographical mapping of functionally annotated
residues. Nucleic Acids Research, 34(WEB. SERV. ISS.), 116–118.
https://doi.org/10.1093/nar/gkl282
Gómez-Valadés, A. G., Vidal-Alabró, A., Molas, M., Boada, J., Bermúdez, J., Bartrons, R., & Perales, J.
C. (2006). Overcoming diabetes-induced hyperglycemia through inhibition of hepatic
phosphoenolpyruvate carboxykinase (GTP) with RNAi. Molecular Therapy, 13(2), 401–410.
https://doi.org/10.1016/j.ymthe.2005.08.026
Huang, K., El-Seedi, H. R., & Xu, B. (2022). Critical review on chemical compositions and health-
promoting effects of mushroom Agaricus blazei Murill. Current Research in Food Science, 5(July),
2190–2203. https://doi.org/10.1016/j.crfs.2022.10.029
Johnny, I., & Okon, J. (2013). Antidiabetic Effect of Pleurotus ostreatus (Jacq.ex Fr) kumm. Mushroom on
Alloxan-induced Diabetic Rats. Indian Journal of Pharmaceutical and Biological Research, 1(02),
15
31–36. https://doi.org/10.30750/ijpbr.1.2.6
Karim, R., Rahman, F., Rahman, R., Tamannaa, Z., Ghose, D. K., Islam, N., Hossain, T., Islam, R., &
Rahman, A. (2020). Hypoglycemic and antidyslipidemic potential of pleurotusostreatus in
streptozotocin-induced diabetic rats. Journal of Advanced Biotechnology and Experimental
Therapeutics, 3(1), 49–55. https://doi.org/10.5455/jabet.2020.d106
Kazeem, M. I., Adamson, J. O., & Ogunwande, I. A. (2013). Modes of inhibition of α-amylase and α-
glucosidase by aqueous extract of morinda lucida benth leaf. BioMed Research International, 2013.
https://doi.org/10.1155/2013/527570
Kumar, A. R., Merlin, N. J., Arundhathi, R. P., & Dharan, S. S. (2019). In-silico Molecular Docking
Evaluation of Plumbagine Derivatives for Anticancer Activity. 11(7), 2676–2678.
Leong, C. C., Ho, W. Y., Yeap, S. K., Krishnen, G., Chong, Z. X., Ho, J. S., Lim, P. T., & Ten, S. T. (2021).
Assessment of phylogenetic, growth, and antioxidant capacity of Pleurotus spp. in Malaysia. Journal
of Food Processing and Preservation, 45(6), 1–13. https://doi.org/10.1111/jfpp.15554
Lesa, K. N., Khandaker, M. U., Mohammad, F., Iqbal, R., Sharma, R., Islam, F., Mitra, S., & Emran, T.
Bin. (2022). Nutritional Value , Medicinal Importance , and Health-Promoting Effects of Dietary
Mushroom ( Pleurotus ostreatus ). 2022.
Lipinski, C. A. (2016). Rule of five in 2015 and beyond: Target and ligand structural limitations, ligand
chemistry structure and drug discovery project decisions. Advanced Drug Delivery Reviews, 101, 34–
41. https://doi.org/10.1016/j.addr.2016.04.029
Menaga, D., Rajakumar, S., & Ayyasamy, P. M. (2014). Free radical scavenging activity of methanolic
extract of brown alga Spatoglossum asperum. International Journal of Pharmacy and Pharmaceutical
Sciences, 5(4), 601–606.
Mishra, V., Tomar, S., Yadav, P., Vishwakarma, S., & Singh, M. P. (2022). Elemental Analysis,
Phytochemical Screening and Evaluation of Antioxidant, Antibacterial and Anticancer Activity of
Pleurotus ostreatus through In Vitro and In Silico Approaches. Metabolites, 12(9).
https://doi.org/10.3390/metabo12090821
Mkhize, S. S., Cedric Simelane, M. B., Mongalo, I. N., & Pooe, O. J. (2022). The Effect of Supplementing
Mushroom Growing Substrates on the Bioactive Compounds, Antimicrobial Activity, and
Antioxidant Activity of Pleurotus ostreatus. Biochemistry Research International, 2022.
https://doi.org/10.1155/2022/9436614
Mwangi, R. W., Macharia, J. M., Wagara, I. N., & Bence, R. L. (2022). The antioxidant potential of
different edible and medicinal mushrooms. Biomedicine and Pharmacotherapy, 147(October 2021),
112621. https://doi.org/10.1016/j.biopha.2022.112621
Nowakowski, P., Markiewicz-Żukowska, R., Bielecka, J., Mielcarek, K., Grabia, M., & Socha, K. (2021).
Treasures from the forest: Evaluation of mushroom extracts as anti-cancer agents. Biomedicine and
Pharmacotherapy, 143(June). https://doi.org/10.1016/j.biopha.2021.112106
Ogurtsova, K., Guariguata, L., Barengo, N. C., Ruiz, P. L. D., Sacre, J. W., Karuranga, S., Sun, H., Boyko,
E. J., & Magliano, D. J. (2022). IDF diabetes Atlas: Global estimates of undiagnosed diabetes in adults
for 2021. Diabetes Research and Clinical Practice, 183(2022), 1–9.
https://doi.org/10.1016/j.diabres.2021.109118
Ojukwu, S. U., FA, O., & CE, U. (2021). GC-FID phytochemical analysis of selected antidiabetic herbal
preparations sold in Onitsha metropolis, Anambra state, Nigeria. Journal of Medicinal Plants Studies,
9(4), 60–67. https://doi.org/10.22271/plants.2021.v9.i4a.1313
16
Omari, N. El, Sayah, K., Fettach, S., Blidi, O. El, Bouyahya, A., Faouzi, M. E. A., Kamal, R., & Barkiyou,
M. (2019). Evaluation of in vitro antioxidant and antidiabetic activities of Aristolochia longa extracts.
Evidence-Based Complementary and Alternative Medicine, 2019.
https://doi.org/10.1155/2019/7384735
Rines, A. K., Sharabi, K., Tavares, C. D. J., & Puigserver, P. (2016). Targeting hepatic glucose metabolism
in the treatment of type 2 diabetes. Nature Reviews Drug Discovery, 15(11), 786–804.
https://doi.org/10.1038/nrd.2016.151
Sayah, K., Marmouzi, I., Naceiri Mrabti, H., Cherrah, Y., & Faouzi, M. E. A. (2017). Antioxidant Activity
and Inhibitory Potential of Cistus salviifolius (L.) and Cistus monspeliensis (L.) Aerial Parts Extracts
against Key Enzymes Linked to Hyperglycemia. BioMed Research International, 2017.
https://doi.org/10.1155/2017/2789482
Singh, A., Kukreti, R., Saso, L., & Kukreti, S. (2022). Pathways and Type 2 Diabetes. Molecules, 27, 950–
969.
Suleiman, W. B., Shehata, R. M., & Younis, A. M. (2022). In vitro assessment of multipotential therapeutic
importance of Hericium erinaceus mushroom extracts using different solvents. Bioresources and
Bioprocessing, 9(1). https://doi.org/10.1186/s40643-022-00592-6
Sun, H., Saeedi, P., Karuranga, S., Pinkepank, M., Ogurtsova, K., Duncan, B. B., Stein, C., Basit, A., Chan,
J. C. N., Mbanya, J. C., Pavkov, M. E., Ramachandaran, A., Wild, S. H., James, S., Herman, W. H.,
Zhang, P., Bommer, C., Kuo, S., Boyko, E. J., & Magliano, D. J. (2022). IDF Diabetes Atlas: Global,
regional and country-level diabetes prevalence estimates for 2021 and projections for 2045. Diabetes
Research and Clinical Practice, 183, 109119. https://doi.org/10.1016/j.diabres.2021.109119
Sun, L., Wang, Y., & Miao, M. (2020). Inhibition of α -amylase by polyphenolic compounds : Substrate
digestion , binding interactions and nutritional intervention. Trends in Food Science & Technology,
104(February), 190–207. https://doi.org/10.1016/j.tifs.2020.08.003
Torres-Martínez, B. D. M., Vargas-Sánchez, R. D., Torrescano-Urrutia, G. R., Esqueda, M., Rodríguez-
Carpena, J. G., Fernández-López, J., Perez-Alvarez, J. A., & Sánchez-Escalante, A. (2022). Pleurotus
Genus as a Potential Ingredient for Meat Products. Foods, 11(6), 1–13.
https://doi.org/10.3390/foods11060779
Yadav, D., & Negi, P. S. (2021). Bioactive components of mushrooms: Processing effects and health
benefits. Food Research International, 148(July), 110599.
https://doi.org/10.1016/j.foodres.2021.110599
Yaribeygi, H., Sathyapalan, T., Atkin, S. L., & Sahebkar, A. (2020). Review Article Molecular Mechanisms
Linking Oxidative Stress and Diabetes Mellitus. 2020.
Yu, S., Meng, S., Xiang, M., & Ma, H. (2021). Phosphoenolpyruvate carboxykinase in cell metabolism:
Roles and mechanisms beyond gluconeogenesis. Molecular Metabolism, 53(May), 101257.
https://doi.org/10.1016/j.molmet.2021.101257
17
Ai
Bi
Di
Ci
Aii
Bii
Cii
Dii
18
Figure 1. An observation of the molecular interaction (left: 2D and right: 3D) between compounds with the
highest binding energies and standard against α-amylase. Epicatechin (Ai, Aii), Naringin (Bi, Bii), Catechin
(Ci, Cii), Standard (Acarbose) (Di, Dii).
Ai
Bi
Ci Cii
Bii
Aii
19
Figure 2. An observation of the molecular interaction (left: 2D and right: 3D) between compounds with the
highest binding energies and standard against α-glucosidase. Epicatechin (Ai, Aii), Naringin (Bi, Bii),
Rutin (Ci, Cii), Standard (Acarbose) (Di, Dii).
Di
Ai
Bi
Ci
Dii
Aii
Bii
Cii
20
Figure 3. An observation of the molecular interaction (left: 2D and right: 3D) between compounds with the
highest binding energies and standard against PEP carboxykinase. Epicatechin (Ai, Aii), Naringin (Bi,
Bii), Rutin (Ci, Cii), Standard (Acarbose) (Di, Dii).
Author contribution
Chibuogwu, C.C., Okagu, I.U. and Aham, E.C. conceived and designed the study. Nnemolisa,
S.C., Chukwurah, C.C, Aguchem, R.N., Edeh, S.C. and Obiora, M.O carried out the experiment.
Aguchem, R.O. and Anyebe, D.E. analyzed the data. Chukwu, M.C. carried out the Molecular
docking studies. Nnemolisa, S.C. and Chibuogwu, C.C. drafted the manuscript. Okagu, I.U. and
Aham, E.C. edited and revised the manuscript. All authors read and approved the final manuscript.
Declaration of Competing Interest
None declared.
Di Dii

More Related Content

Similar to Food chemistry avances en la producción de antibioticos

SF424R-R_biosketch_VerC
SF424R-R_biosketch_VerCSF424R-R_biosketch_VerC
SF424R-R_biosketch_VerC
Brooks Hylemon
 
2010 effect of pistachio diet on lipid parameters, endothelial function
2010 effect of pistachio diet on lipid parameters, endothelial function2010 effect of pistachio diet on lipid parameters, endothelial function
2010 effect of pistachio diet on lipid parameters, endothelial function
Agrin Life
 
Bioinformatic Analysis of Selected Natural Compounds for Antidiabetic Potential
Bioinformatic Analysis of Selected Natural Compounds for Antidiabetic PotentialBioinformatic Analysis of Selected Natural Compounds for Antidiabetic Potential
Bioinformatic Analysis of Selected Natural Compounds for Antidiabetic Potential
ijtsrd
 
NUTRIGENOMICS : PERIODONTAL IMPLICATIONS,
NUTRIGENOMICS : PERIODONTAL IMPLICATIONS, NUTRIGENOMICS : PERIODONTAL IMPLICATIONS,
NUTRIGENOMICS : PERIODONTAL IMPLICATIONS,
Shilpa Shiv
 
2010 exenatide and weight loss
2010 exenatide and weight loss2010 exenatide and weight loss
2010 exenatide and weight loss
Agrin Life
 
Hypoglycaemia and improved testicular parameters in Sesamum radiatum treated ...
Hypoglycaemia and improved testicular parameters in Sesamum radiatum treated ...Hypoglycaemia and improved testicular parameters in Sesamum radiatum treated ...
Hypoglycaemia and improved testicular parameters in Sesamum radiatum treated ...
lukeman Joseph Ade shittu
 
The effects of curcumin supplementation on liver enzymes, lipid
The effects of curcumin supplementation on liver enzymes, lipidThe effects of curcumin supplementation on liver enzymes, lipid
The effects of curcumin supplementation on liver enzymes, lipid
wahyu purnama
 
Urine protein relates with likeness of lentils
Urine protein relates with likeness of lentilsUrine protein relates with likeness of lentils
Urine protein relates with likeness of lentils
BRNSSPublicationHubI
 

Similar to Food chemistry avances en la producción de antibioticos (20)

Effect of zinc supplementation on glycemic control, lipid
Effect of zinc supplementation on glycemic control, lipidEffect of zinc supplementation on glycemic control, lipid
Effect of zinc supplementation on glycemic control, lipid
 
Estudio científico de la UV demuestra los beneficios de consumir arándanos an...
Estudio científico de la UV demuestra los beneficios de consumir arándanos an...Estudio científico de la UV demuestra los beneficios de consumir arándanos an...
Estudio científico de la UV demuestra los beneficios de consumir arándanos an...
 
SF424R-R_biosketch_VerC
SF424R-R_biosketch_VerCSF424R-R_biosketch_VerC
SF424R-R_biosketch_VerC
 
LACTOWHEY powder: Provides body’s defense against cancer
LACTOWHEY powder: Provides body’s defense against cancerLACTOWHEY powder: Provides body’s defense against cancer
LACTOWHEY powder: Provides body’s defense against cancer
 
2010 effect of pistachio diet on lipid parameters, endothelial function
2010 effect of pistachio diet on lipid parameters, endothelial function2010 effect of pistachio diet on lipid parameters, endothelial function
2010 effect of pistachio diet on lipid parameters, endothelial function
 
Crimson Publishers-Biomarker-Guided Nutritional Intervention to Treat Autism
Crimson Publishers-Biomarker-Guided Nutritional Intervention to Treat AutismCrimson Publishers-Biomarker-Guided Nutritional Intervention to Treat Autism
Crimson Publishers-Biomarker-Guided Nutritional Intervention to Treat Autism
 
Probiotics
ProbioticsProbiotics
Probiotics
 
The role of NSAIDs in periodontal disease progression
The role of NSAIDs in periodontal disease progressionThe role of NSAIDs in periodontal disease progression
The role of NSAIDs in periodontal disease progression
 
Vitafoods Asia 2017 - Presentations
Vitafoods Asia 2017 - PresentationsVitafoods Asia 2017 - Presentations
Vitafoods Asia 2017 - Presentations
 
Bioinformatic Analysis of Selected Natural Compounds for Antidiabetic Potential
Bioinformatic Analysis of Selected Natural Compounds for Antidiabetic PotentialBioinformatic Analysis of Selected Natural Compounds for Antidiabetic Potential
Bioinformatic Analysis of Selected Natural Compounds for Antidiabetic Potential
 
kayachikitsa CME Gadag abstarcts
kayachikitsa CME Gadag abstarctskayachikitsa CME Gadag abstarcts
kayachikitsa CME Gadag abstarcts
 
Ellagic Acid and Its Metabolites as Potent and Selective Allosteric Inhibitor...
Ellagic Acid and Its Metabolites as Potent and Selective Allosteric Inhibitor...Ellagic Acid and Its Metabolites as Potent and Selective Allosteric Inhibitor...
Ellagic Acid and Its Metabolites as Potent and Selective Allosteric Inhibitor...
 
Probiotics and Hypertension.pdf
Probiotics and Hypertension.pdfProbiotics and Hypertension.pdf
Probiotics and Hypertension.pdf
 
NUTRIGENOMICS : PERIODONTAL IMPLICATIONS,
NUTRIGENOMICS : PERIODONTAL IMPLICATIONS, NUTRIGENOMICS : PERIODONTAL IMPLICATIONS,
NUTRIGENOMICS : PERIODONTAL IMPLICATIONS,
 
2010 exenatide and weight loss
2010 exenatide and weight loss2010 exenatide and weight loss
2010 exenatide and weight loss
 
Hypoglycaemia and improved testicular parameters in Sesamum radiatum treated ...
Hypoglycaemia and improved testicular parameters in Sesamum radiatum treated ...Hypoglycaemia and improved testicular parameters in Sesamum radiatum treated ...
Hypoglycaemia and improved testicular parameters in Sesamum radiatum treated ...
 
Effects of Whole Grains on Coronary Heart Disease Risk
Effects of Whole Grains on Coronary Heart Disease RiskEffects of Whole Grains on Coronary Heart Disease Risk
Effects of Whole Grains on Coronary Heart Disease Risk
 
The effects of curcumin supplementation on liver enzymes, lipid
The effects of curcumin supplementation on liver enzymes, lipidThe effects of curcumin supplementation on liver enzymes, lipid
The effects of curcumin supplementation on liver enzymes, lipid
 
Anti diabetic property_of_moringa_oleifera
Anti diabetic property_of_moringa_oleiferaAnti diabetic property_of_moringa_oleifera
Anti diabetic property_of_moringa_oleifera
 
Urine protein relates with likeness of lentils
Urine protein relates with likeness of lentilsUrine protein relates with likeness of lentils
Urine protein relates with likeness of lentils
 

Recently uploaded

Call Girls in Ramesh Nagar Delhi 💯 Call Us 🔝9953056974 🔝 Escort Service
Call Girls in Ramesh Nagar Delhi 💯 Call Us 🔝9953056974 🔝 Escort ServiceCall Girls in Ramesh Nagar Delhi 💯 Call Us 🔝9953056974 🔝 Escort Service
Call Girls in Ramesh Nagar Delhi 💯 Call Us 🔝9953056974 🔝 Escort Service
9953056974 Low Rate Call Girls In Saket, Delhi NCR
 
result management system report for college project
result management system report for college projectresult management system report for college project
result management system report for college project
Tonystark477637
 
FULL ENJOY Call Girls In Mahipalpur Delhi Contact Us 8377877756
FULL ENJOY Call Girls In Mahipalpur Delhi Contact Us 8377877756FULL ENJOY Call Girls In Mahipalpur Delhi Contact Us 8377877756
FULL ENJOY Call Girls In Mahipalpur Delhi Contact Us 8377877756
dollysharma2066
 
VIP Call Girls Ankleshwar 7001035870 Whatsapp Number, 24/07 Booking
VIP Call Girls Ankleshwar 7001035870 Whatsapp Number, 24/07 BookingVIP Call Girls Ankleshwar 7001035870 Whatsapp Number, 24/07 Booking
VIP Call Girls Ankleshwar 7001035870 Whatsapp Number, 24/07 Booking
dharasingh5698
 

Recently uploaded (20)

ONLINE FOOD ORDER SYSTEM PROJECT REPORT.pdf
ONLINE FOOD ORDER SYSTEM PROJECT REPORT.pdfONLINE FOOD ORDER SYSTEM PROJECT REPORT.pdf
ONLINE FOOD ORDER SYSTEM PROJECT REPORT.pdf
 
University management System project report..pdf
University management System project report..pdfUniversity management System project report..pdf
University management System project report..pdf
 
Water Industry Process Automation & Control Monthly - April 2024
Water Industry Process Automation & Control Monthly - April 2024Water Industry Process Automation & Control Monthly - April 2024
Water Industry Process Automation & Control Monthly - April 2024
 
Call Girls Pimpri Chinchwad Call Me 7737669865 Budget Friendly No Advance Boo...
Call Girls Pimpri Chinchwad Call Me 7737669865 Budget Friendly No Advance Boo...Call Girls Pimpri Chinchwad Call Me 7737669865 Budget Friendly No Advance Boo...
Call Girls Pimpri Chinchwad Call Me 7737669865 Budget Friendly No Advance Boo...
 
Call Girls in Ramesh Nagar Delhi 💯 Call Us 🔝9953056974 🔝 Escort Service
Call Girls in Ramesh Nagar Delhi 💯 Call Us 🔝9953056974 🔝 Escort ServiceCall Girls in Ramesh Nagar Delhi 💯 Call Us 🔝9953056974 🔝 Escort Service
Call Girls in Ramesh Nagar Delhi 💯 Call Us 🔝9953056974 🔝 Escort Service
 
VIP Model Call Girls Kothrud ( Pune ) Call ON 8005736733 Starting From 5K to ...
VIP Model Call Girls Kothrud ( Pune ) Call ON 8005736733 Starting From 5K to ...VIP Model Call Girls Kothrud ( Pune ) Call ON 8005736733 Starting From 5K to ...
VIP Model Call Girls Kothrud ( Pune ) Call ON 8005736733 Starting From 5K to ...
 
Glass Ceramics: Processing and Properties
Glass Ceramics: Processing and PropertiesGlass Ceramics: Processing and Properties
Glass Ceramics: Processing and Properties
 
(INDIRA) Call Girl Meerut Call Now 8617697112 Meerut Escorts 24x7
(INDIRA) Call Girl Meerut Call Now 8617697112 Meerut Escorts 24x7(INDIRA) Call Girl Meerut Call Now 8617697112 Meerut Escorts 24x7
(INDIRA) Call Girl Meerut Call Now 8617697112 Meerut Escorts 24x7
 
result management system report for college project
result management system report for college projectresult management system report for college project
result management system report for college project
 
FULL ENJOY Call Girls In Mahipalpur Delhi Contact Us 8377877756
FULL ENJOY Call Girls In Mahipalpur Delhi Contact Us 8377877756FULL ENJOY Call Girls In Mahipalpur Delhi Contact Us 8377877756
FULL ENJOY Call Girls In Mahipalpur Delhi Contact Us 8377877756
 
Call for Papers - International Journal of Intelligent Systems and Applicatio...
Call for Papers - International Journal of Intelligent Systems and Applicatio...Call for Papers - International Journal of Intelligent Systems and Applicatio...
Call for Papers - International Journal of Intelligent Systems and Applicatio...
 
Unit 1 - Soil Classification and Compaction.pdf
Unit 1 - Soil Classification and Compaction.pdfUnit 1 - Soil Classification and Compaction.pdf
Unit 1 - Soil Classification and Compaction.pdf
 
VIP Call Girls Ankleshwar 7001035870 Whatsapp Number, 24/07 Booking
VIP Call Girls Ankleshwar 7001035870 Whatsapp Number, 24/07 BookingVIP Call Girls Ankleshwar 7001035870 Whatsapp Number, 24/07 Booking
VIP Call Girls Ankleshwar 7001035870 Whatsapp Number, 24/07 Booking
 
The Most Attractive Pune Call Girls Budhwar Peth 8250192130 Will You Miss Thi...
The Most Attractive Pune Call Girls Budhwar Peth 8250192130 Will You Miss Thi...The Most Attractive Pune Call Girls Budhwar Peth 8250192130 Will You Miss Thi...
The Most Attractive Pune Call Girls Budhwar Peth 8250192130 Will You Miss Thi...
 
UNIT-II FMM-Flow Through Circular Conduits
UNIT-II FMM-Flow Through Circular ConduitsUNIT-II FMM-Flow Through Circular Conduits
UNIT-II FMM-Flow Through Circular Conduits
 
Java Programming :Event Handling(Types of Events)
Java Programming :Event Handling(Types of Events)Java Programming :Event Handling(Types of Events)
Java Programming :Event Handling(Types of Events)
 
UNIT - IV - Air Compressors and its Performance
UNIT - IV - Air Compressors and its PerformanceUNIT - IV - Air Compressors and its Performance
UNIT - IV - Air Compressors and its Performance
 
KubeKraft presentation @CloudNativeHooghly
KubeKraft presentation @CloudNativeHooghlyKubeKraft presentation @CloudNativeHooghly
KubeKraft presentation @CloudNativeHooghly
 
UNIT-IFLUID PROPERTIES & FLOW CHARACTERISTICS
UNIT-IFLUID PROPERTIES & FLOW CHARACTERISTICSUNIT-IFLUID PROPERTIES & FLOW CHARACTERISTICS
UNIT-IFLUID PROPERTIES & FLOW CHARACTERISTICS
 
Thermal Engineering-R & A / C - unit - V
Thermal Engineering-R & A / C - unit - VThermal Engineering-R & A / C - unit - V
Thermal Engineering-R & A / C - unit - V
 

Food chemistry avances en la producción de antibioticos

  • 1. Journal Pre-proof Antidiabetic and Antioxidant Potentials of Pleurotus ostreatus -derived Compounds: An in vitro and in silico approach S.C. Nnemolisa , C.C. Chukwurah , S.C. Edeh , R.N. Aguchem , C.C. Chibuogwu , E.C. Aham , M.C. Chukwu , M.O. Obiora , D.E. Anyebe , I.U. Okagu PII: S2772-753X(24)00035-2 DOI: https://doi.org/10.1016/j.focha.2024.100639 Reference: FOCHA 100639 To appear in: Food Chemistry Advances Received date: 30 June 2023 Revised date: 10 December 2023 Accepted date: 6 February 2024 Please cite this article as: S.C. Nnemolisa , C.C. Chukwurah , S.C. Edeh , R.N. Aguchem , C.C. Chibuogwu , E.C. Aham , M.C. Chukwu , M.O. Obiora , D.E. Anyebe , I.U. Okagu , Antidia- betic and Antioxidant Potentials of Pleurotus ostreatus -derived Compounds: An in vitro and in silico approach, Food Chemistry Advances (2024), doi: https://doi.org/10.1016/j.focha.2024.100639 This is a PDF file of an article that has undergone enhancements after acceptance, such as the addition of a cover page and metadata, and formatting for readability, but it is not yet the definitive version of record. This version will undergo additional copyediting, typesetting and review before it is published in its final form, but we are providing this version to give early visibility of the article. Please note that, during the production process, errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain. © 2024 Published by Elsevier Ltd. This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/)
  • 2. 1 Highlights • Pleurotus ostreatus methanol extract (MEPO) contains many health-beneficial compounds • MEPO showed stronger inhibition of α-glucosidase activity than α-amylase. • MEPO showed potential for blood glucose regulation in in silico study.
  • 3. 2 Antidiabetic and Antioxidant Potentials of Pleurotus ostreatus -derived Compounds: An in vitro and in silico approach 1 Nnemolisa, S.C., 1 Chukwurah, C.C., 1 Edeh, S.C., 1 Aguchem, R.N., 1,2 Chibuogwu*, C.C., 1 Aham, E.C., 3 Chukwu, M.C., 1 Obiora, M.O., 1 Anyebe, D.E. and 1 Okagu, I.U. 1 Department of Biochemistry, Faculty of Biological Sciences, University of Nigeria, Nsukka, Enugu, Nigeria. 2 Institute for Drug-Herbal Medicine-Excipient Research and Development, Faculty of Pharmaceutical Sciences, University of Nigeria, Nsukka, Enugu, Nigeria. 3 Department of Genetics and Biotechnology, Faculty of Biological Sciences, University of Nigeria, Enugu, Nsukka. * Correspondence: christian.chibuogwu@unn.edu.ng (ORCID: 0000-0002-9091-4310) +2347069263943 Abstract Many health benefits have reportedly been associated with mushroom consumption. This study determined the chemical constituents of Pleurotus ostreatus methanol extract (MEPO) and investigated its antioxidant and antidiabetic effects using in vitro and in silico approaches. The chemical composition of MEPO was determined using the gas chromatography-flame ionization detector (GC-FID) technique, while 2,2- Diphenyl-1-picrylhydrazyl (DPPH) and ferric reducing antioxidant power (FRAP) methods were used to determine antioxidant activity. The antidiabetic activity was evaluated using α-amylase and α-glucosidase inhibition assays, while molecular docking was done to give insight into the binding potentials of MEPO constituents against α-amylase, α-glucosidase, and phosphoenolpyruvate (PEP) carboxykinase activities. Thirteen compounds, including ephedrine, oxalate, rutin, naringin, and kaempferol, were identified in MEPO. The extract showed moderate antioxidant activity, as observed from the DPPH (IC50 = 732.41 mg/ml) and FRAP studies. The extract also demonstrated stronger inhibition of α-glucosidase activity (IC50 = 246.58 mg/ml) than α-amylase activity (IC50 = 1074.05 mg/ml). Docking studies revealed that rutin and naringin interacted effectively with amino acid residues crucial for α-amylase, α-glucosidase, and PEP carboxykinase activities via hydrogen bonds. The result shows that MEPO is a rich store of beneficial compounds which could be explored for the management of diabetes and associated complications. Keywords: Mushroom, methanol extract, α-amylase, α-glucosidase, PEP carboxykinase. 1. Introduction Diabetes mellitus (DM) is a group of chronic metabolic disorders characterized by an abnormal rise in plasma glucose levels resulting from poor glucose metabolism due to abnormal insulin secretion, insulin deficiency, and/or insulin insensitivity (Singh et al., 2022). In 2021, 536.6 million people were reportedly living with diabetes, and a 46% increase (783.2 million) has been predicted by 2045 (Ogurtsova et al., 2022). Most diabetics (80.6%, 432.7 million) reside in low- and middle-income nations and about half of these individuals are more likely to experience complications because they are unaware that they have the condition (Sun et al., 2022). These complications are the major causes of mortality in diabetic patients. One of the factors contributing to the pathogenesis of diabetic complications from hyperglycemia is an impairment of reactive oxygen species and antioxidant defense balance (Dehghan et al., 2016). This results
  • 4. 3 in a state known as oxidative stress. Indeed, studies have shown that hyperglycemia-induced oxidative stress is implicated in vascular damage of diabetes subjects (Singh et al., 2022). Numerous consequences of diabetes may be delayed or prevented with intensive postprandial hyperglycemic management. However, the available treatment reflects an insufficient and less effective glycemic control for diabetic patients (Abbas et al., 2019). This has led to the exploration of more effective therapeutic targets and approaches to glycemic control. One of the therapeutic options presently explored for effective glycemic control is the inhibition of α-amylase and α-glucosidase activities. The α-amylase is a calcium metalloenzyme that plays an important role in starch digestion catalyzing the hydrolysis of α-(1, 4)-D-glycosidic linkages in starch and other polymers of glucose, producing smaller fragments (Sun et al., 2020). These products are further degraded by α-glucosidase to absorbable monosaccharides which then enters the bloodstream. Therefore, inhibiting these enzymes would hinder the absorption of starch, expand intestinal sugar-holding time and delay the rate of glucose absorption and consequently decrease blood sugar level. Acarbose, voglibose and migiltol are drugs used to inhibit the activities of these enzymes in practice. However, they cause unwanted side effects such as abdominal discomfort (Alqahtani et al., 2020) which hinders their use in the treatment of diabetes and its complications. This has therefore led to urgent requirement for effective substitutes which would reduce diabetes complications with fewer side effects. Phosphoenolpyruvate carboxykinase (PEPCK) is a key enzyme in gluconeogenesis that catalyzes the carboxylation of pyruvate to form oxaloacetate in the gluconeogenic pathway (Gómez-Valadés et al., 2006). Although this enzyme is widely implicated for its role in hepatic glucose production in diabetic conditions, studies have shown that overexpression of PEPCK in adipose tissue is a major contributor to obesity development due to increased glyceroneogenesis in the adipose tissue (Rines et al., 2016; Yu et al., 2021). Therefore, it makes sense to target PEPCK activity for inhibition as a strategy in diabetes therapy. For ages, nature has been a source of therapeutic agents and an astounding number of modern medications have been identified from natural sources based on their usage in conventional medicine (Bhushan & Kulshreshtha, 2018). The interest in natural substances have significantly increased in recent years, because these substances are intended to be used as nutraceuticals to replace synthetic substances whose usage is restricted due to their adverse effects (Omari et al., 2019). The nutraceutical potentials of mushroom have been well documented since time immemorial; however, it is in the beginning of this century that the nutritional and pharmacological properties of mushroom gathered much attention (Huang et al., 2022). Mushrooms are well-known macro-fungus with a characteristic fruiting body that can either be hypogenous or epigeous (Yadav & Negi, 2021). Mushroom cultivation has emerged as a promising agricultural business that is land-independent with the capacity of transforming lignocellulosic waste into nutrient-rich foods, in addition to significantly reducing environmental pollution. Nutritionally, mushroom contains a relatively high amount of carbohydrate, protein, dietary fibers, minerals, vitamins and unsaturated fatty acids (Nowakowski et al., 2021). Edible mushrooms are excellent sources of beneficial bioactive metabolites, and these substances have broad-spectrum therapeutic applications, including but not limited to antiviral, anti-inflammatory, anti-bacterial, anti-oxidant and antidiabetic effect (Mwangi et al., 2022; Suleiman et al., 2022). Pleurotus genus is among the species with the highest production worldwide (Torres-Martínez et al., 2022). This can be attributed to its short cultivation cycle, its ability to grow at moderate temperature (25-30ºC) and a wide range of lignocellulosic materials (Cruz-Solorio et al., 2018). Of particular interest is P. ostreatus which is the second most widely grown and consumed edible mushroom in the world. It is considered a high quality food due to its high nutritional value and beneficial health effects (Lesa et al.,
  • 5. 4 2022). In the present study, the in vitro and in silico anti-diabetic potential of methanol extract of Pleurotus ostreatus was investigated. 2. Materials and Methods 2.1 Collection and Authentication of Material Pleurotus ostreatus together with wood sawdust and rice bran were the materials used for this study. Wood sawdust and rice bran were obtained from timber and rice farmers in Enugu State, Nigeria and were pulverized using a mechanical grinder. The mushroom was cultivated at Southeast Zonal Biotechnology Center, University of Nigeria. 2.1.2 Chemicals and Reagents The chemicals and reagents used in this study were of analytical grade. 2.2. Mushroom Cultivation Mushroom was cultivated following the method described by Aguchem et al., (2022). 50 kg of the substrate was prepared by mixing the substrate with 0.40 kg of CaCO3 before adding 38 liters of water to the mixed substrate. The substrate mixture was then packed in a limpid refractory polythene bag, each tied with rubber bans before sterilization at 121o C for 15 minutes at 15 psi. After sterilization, the sterile packed substrates were allowed to cool to room temperature. The substrates were then inoculated in a germ-free manner with the spawn and then re-tied. The inoculated bags were then taken inside a darkroom and allowed some days to ramify with the complete ramification of the inoculated bags clearly visible by the appearance of pin heads and fungi hyphae. Incisions were gently made on the ramified mushroom bags in a germ-free manner before they were transferred to the growth room for the production of fruiting body. During the course of this process, the relative humidity and temperature were maintained at 74-86% and 25o C respectively. By watering the bags at least once daily, wetting the floor, keeping water in tray and ice blocks close to the mushroom bags, humid air in the growth room was achieved. The fruiting bodies of the mushroom was then harvested and used for analysis. 2.2.1 Preparation of Mushroom Extract Fresh whole mushroom sample (100 g) was oven-dried at 50o C and ground into fine particles and then macerated in 400 ml of methanol at room temperature in an air-tight container. The container was left for 72 hours with intermittent shaking to increase the efficiency of extraction of the phytochemicals. This was followed by filtration and concentration in vacuo to obtain the crude methanol extract of P. ostreatus (MEPO) (Figure S1 (A-F) - supplementary materials). 2.2.2 Extraction of Chemical Constituents for GC-FID analysis Extraction of chemical constituent for gas chromatography-flame ionization detector (GC-FID) analysis was carried out following Boisa et al., (2020) and Ojukwu et al., (2021) methods, respectively. A test tube containing 1 g of the sample was filled with 15 ml of methanol and 10 ml of 50% w/v KOH. The test tube was then placed in a water bath set to 60°C for 60 minutes. After that, the content was moved to a separatory funnel. 20 ml of ethanol, 10 ml of cold water, 10 ml of hot water and 3 ml of hexane was used successively to wash the contents of the tube which was all transferred to the funnel. This extract was then
  • 6. 5 mixed with 10 ml of 10% v/v ethanol aqueous solution and rinsed three times. The solvent was evaporated after the solution was dried with anhydrous sodium sulfate. The resulting sample was solubilized in 1000 µl of pyridine after which 200 µl (0.2 ml) of the solution was transferred to a vial for analysis. 2.2.3 Analysis of chemical constituent of MEPO The chemical constituents of MEPO were analyzed on a BUCK M910 Gas chromatograph (GC) (BUCK M910, PUB Scientist, USA) equipped with a flame ionization detector (FID). 0.1 ml of the extract was injected into the GC machine. At an ionization voltage of 70 eV, injector temperature of 250°C and injector mode was split with linear velocity 36.5 cm/sec and pressure was 57.5 kPa. The instrument was run in electron impact mode. In principle, the detector uses a flame to ionize carbon-containing organic compounds. During separation, the sample passes through a hydrogen-fueled flame in the GC column which ionizes the carbon atoms in the sample. 2.2.4 Assay of the inhibitory effect of MEPO on α-amylase and α-glucosidase activities The assay mixtures containing the solution of the enzyme (0.1 ml), acetate buffer (0.1 - 0.5 ml, 0.1 mol/l at pH 5.5) and mushroom extract (32 - 160 mg/ml) were incubated for 15 minutes at room temperature. This was followed by the addition of 0.5 ml of 2% starch and 2% sucrose to each test tube containing the respective enzymes and incubation of the resulting mixture in a water bath at 50°C for 30 min. The reaction was terminated with the addition of 1 ml of dinitrosalicylic acid (DNSA) reagent to each test tube and placed in a boiling water bath for 10 minutes. To stabilize the color after heating, 1 ml of 1.4 mol/l Rochelle salt (sodium potassium tartrate) was added to the test tubes immediately and the total volume of the solution was adjusted to 4 ml with distilled water. The reaction mixtures were then cooled to room temperature and absorbance was taken at 540 nm using a spectrophotometer (Spectron lab 2A, England). The test tube used as control was prepared without mushroom extract. All tests were done in triplicate and the percentage of inhibition of enzyme activity was calculated as Equation 1: % inhibition = (Abs.of control − Abs.of treatment) (Abs.of control) × 100 1 Where: Abs. of control = Absorbance of control at 540 nm; Abs. of treatment = Absorbance of sample containing extract at 540 nm 2.2.5 In vitro antioxidant activity of MEPO 2.2.5.1 DPPH radical-scavenging assay The DPPH radical-scavenging activity of MEPO was determined following the protocol of Sayah et al., (2017). Differently labelled test tubes received additions of MEPO at various mass concentrations (32 - 160 mg/ml) followed by the addition of MeOH to bring the volume to 250 𝜇l. Each test tube was then shaken violently following the addition of 2 ml of a 0.18 mmol/l (0.005%) methanolic solution of DPPH after which the mixture was allowed to stand in the dark for 30 minutes at room temperature. The same procedure was used to prepare the control without any extract and MeOH served for baseline correction. Changes in samples absorbances were taken at 517 nm using a spectrophotometer (Spectron lab 2A, England). DPPH radical-scavenging potential was calculated using the method provided by Adebiyi et al., (2017).
  • 7. 6 2.2.5.2 Ferric-reducing antioxidant power assay The method described by Alachaher et al., (2018) was used to determine the ferric reducing antioxidant potential of MEPO. 1 ml solution containing 32, 64, 96, 128, and 160 mg/ml each of the extracts was combined with potassium ferricyanide (5.0 ml, 1.0%) and sodium phosphate buffer (5.0 ml, 0.2 mol/l, pH 6.6). The mixture was incubated at 50°C for 20 minutes followed by the addition of 5 mL of 10% trichloroacetic acid and centrifuged at 1000 xg (10 min at 5°C). The upper layer of the solution (5.0 ml) was diluted with 5.0 ml of distilled water and 0.5 ml ferric chloride (0.1% w/v) and absorbance measured at 700 nm using a spectrophotometer (Spectron lab 2A, England). The experiment was performed thrice and scavenging activity calculated as in Equation 2. Scavenging activity (%) = (Abs.of control − Abs.of treatment) (Abs.of control) × 100 2 Where: Abs. of control = Absorbance of control at 700 nm; Abs. of treatment = Absorbance of sample containing extract at 700 nm 2.3 In silico molecular docking studies 2.3.1 Ligands and protein preparation Epicatechin, naringin, catechin, kaempferol, linamarin, rutin, resveratrol, spartein, ephedrine and standard drug (Acarbose) were the ligands used in this study. The chemical structures of the ligands were retrieved from NCBI PubChem database (https://www.ncbi.nlm.nih.gov/pccompound) in 2D (sdf) format and were converted to 3D (PDB) and PDBQT structures using Discovery Studio software and AutoDock Tool, respectively. The selected crystal structures of the proteins, α-amylase (PDB entry: 1U33), PEP-carboxykinase (PDB entry: 1KHB) and α-glucosidase (PDB entry: 5KZW) were obtained from the Protein Data Bank (http://www.rcsb.org/pdb). The crystallographic water as well as the existing ligands were removed. The crystal structures were changed to PDBQT format after the addition of hydrogen atom to the geometry. The structures were refined, and their geometries optimized with the aid of AutoDock Tool. 2.3.2 Docking Following the protocol described by Dundas et al. (2006), identification of the proteins active sites was done using CASTp server (Computer Atlas of Surface Topology of protein). The X, Y, Z center coordinates and X, Y, Z grid dimensions of proteins were selected (30 x 30 x 30 Å), optimized and thereafter docked. Docking and image preparation was performed with AutoDock Vina (Scripps Research Institute, San Diego, CA, USA), BIOVIA Discovery Studio and PyMOL (to generate 3D structures). 2.3.4 Drug-likeness and toxicity prediction In silico prediction of molecular descriptors and drug-like properties of the MEPO phytochemicals was estimated using an online tool on the SwissADME server (http://http://www.swissadme.ch/index.php). MW (molecular weight), cLogP, HBD (number of hydrogen bond donors), HBA (number of hydrogen bond acceptors), gastro-intestinal absorption, and nViolation (number of violations of Lipinski’s rule of five)
  • 8. 7 were calculated on the basis of Lipinski's rule of five as described by Daina et al., (2017). The ProTox webserver was used to predict the toxicity profile of the MEPO phytochemicals. 2.4 Statistical analysis The results obtained were analyzed using the Statistical Product and Service Solutions (SPSS) software (version 20). Results were presented as mean ± standard deviation and tests for statistical significance done using one way analysis of variance (ANOVA) at 95% confidence level and mean values with p≤ 0.05 was taken to be significant. The IC50 values were estimated from the linear plot of inhibition (%) against concentration using the equation of the line graph. 3. Results 3.1 Phytochemical constituents of methanol extract of Pleurotus ostreatus The gas chromatography analysis of the methanol extract of Pleurotus ostreatus as presented in Table 1 showed the presence of thirteen (13) compounds. Ephedrine was found to be the highest followed by oxalate. Rutin, naringin, kaempferol, epicatechin, phytate, resveratrol, catechin were also detected (Figure S2 - Supplementary materials). Table 1: phytochemical constituents of methanol extract of Pleurotus ostreatus Compounds Concentration (𝝁g/ml) Linamarin 8.03 ± 0.05 Catechin 4.27 ± 0.16 Sapogenin 7.97 ± 0.04 Rutin 11.19 ± 0.45 Spartein 8.18 ± 0.14 Flavan-3-ol 4.34 ± 0.15 Naringin 10.40 ± 0.15 Resveratrol 7.38 ± 0.35 Kaempferol 9.02 ± 0.13 Phytate 7.65 ± 0.29 Epicatechin 7.72 ± 0.10 Oxalate 16.68 ± 0.34 Ephedrine 33.77 ± 0.92
  • 9. 8 3.2 In vitro antidiabetic activity of the mushroom extract The result presented in Table 2 shows the percentage inhibition of α-amylase and α-glucosidase activity by MEPO. The result showed a concentration-related inhibitory effect of the extract on α-amylase and α- glucosidase activity with IC50 value of 1074.05 mg/ml and 246.58 mg/ml respectively. The highest inhibitory effect was observed with the highest concentration of the extract (160 mg/ml) and vice versa. Table 2: Inhibitory effect of MEPO on α-amylase and α-glucosidase activity in vitro Sample concentration (mg/ml) α-Amylase activity (%) α-Glucosidase activity (%) 32 4.8±0.18 33.87±0.63 64 5.1± 0.16 36.53± 0.09 96 5.14±0.11 36.69±0.6 128 8.79±0.23 41.9±0.41 160 10.03±0.46 43.43±0.16 IC50 = 1074.05 mg/ml IC50 = 246.58 mg/ml 3.3. In vitro antioxidant effects of methanolic extract of P. ostreatus The DPPH radical-scavenging activity and ferric reducing antioxidant power of MEPO is shown in Table 3. MEPO demonstrated DPPH radical-scavenging activities in a concentration-dependent manner with an IC50 value of 732.41 mg/ml; the highest DPPH radical-scavenging effect was observed with the highest concentration. The same trend is also obtainable with the ferric reducing antioxidant power of the extract. Table 3: In vitro DPPH radical-scavenging activity and ferric reducing antioxidant power of MEPO Sample concentration (mg/ml) % Quenching of DPPH radical FRAP 32 9.73 ± 0.10a 77.00 ± 0.58a 64 9.58 ± 0.07a 84.93 ± 0.31b 96 12.21 ± 0.25b 87.18 ± 0.68c 128 14.88 ± 0.20c 89.46 ± 0.11d 160 16.48 ± 0.11d 98.13 ± 0.54e IC50 = 732.41 mg/ml 3.4. In silico molecular docking studies A molecular docking study was carried out to elucidate the binding interactions of MEPO phytochemicals at the active sites of α-amylase, α-glucosidase, and PEP carboxykinase to garner an understanding into the in vitro results. From Table 4, favorable overall binding energies were observed in ranges of -5.9 to -9.8 kcal/mol for α-amylase (1U33), -5.7 to -9.3 kcal/mol for α-glucosidase and -5.4 to -9.8 kcal/ mol for PEP carboxykinase (1KHB). Epicatechin, naringin and rutin had the best binding energies across all three targets when compared to Acarbose. On the other hand, kaempferol and linamarin showed better binding energies for α-amylase and PEP carboxykinase only when compared to the Acarbose.
  • 10. 9 Table 4: Binding energies of selected compounds and standard inhibitors against protein targets Ligands α-amylase (1U33) Proteins (Kcal/mol) α-glucosidase (5KZW) PEP carboxykinase (1KHB) Epicatechin -9.8 -8.2 -9.3 Naringin -9.4 -9.3 -9.8 Catechin -9.3 -7.2 -7.8 Kaempferol -9.3 -6.9 -8.0 Linamarin -9.0 -7.3 -8.6 Rutin -8.7 -8.8 -9.6 Resveratrol -8.1 -6.9 -7.5 Spartein -7.7 -6.5 -6.6 Ephedrine -5.9 -5.7 -5.4 Standard (Acarbose) -7.5 -8.3 -7.5 3.5 Drug-likeness prediction of the MEPO phytochemicals Table 5 shows the drug-likeness of MEPO phytochemicals. Apart from naringin and rutin, all the MEPO phytochemicals tested had MW less than 500, HBA < 10 and HBD not more than 5. All the compounds showed reasonable water solubility while only naringin and rutin violated Lipinski’s rule for orally administered drugs similar to the standard drug.
  • 11. 10 Table 5: Drug-likeness Prediction of the MEPO phytochemicals Phytochemicals MW (g/mol) No. HBA No. HBD cLogP value GIA No. violation Water solubility Epicatechin 290.27 6 5 0.85 High No Soluble Naringin 580.53 14 8 -0.79 Low 3 Soluble Catechin 290.27 6 5 0.85 High No Soluble Kaempferol 286.24 6 4 1.58 High No Soluble Linamarin 309.32 4 0 2.87 High No Moderately soluble Rutin 610.52 16 10 -1.29 High 3 Soluble Resveratrol 228.24 3 3 2.48 High No Soluble Spartein 234.38 2 0 2.36 Low No Soluble Ephedrine 165.23 2 2 1.45 Low No Very Soluble Acarbose 645.60 19 14 -6.09 Low 3 Highly Soluble MW: Molecular weight (<500), HBA: Number of Hydrogen Bond Acceptors (≤10), HBD: Number of Hydrogen Bond Donor (≤5), cLogP ≤ 5, No. Violation: Number of Violation of Lipinski’s rule. 3.6 Toxicity study of MEPO phytochemicals Apart from ephedrine, linamarin and resveratrol, all the MEPO phytochemicals had LD50 greater than 2000 mg/kg. The toxicity prediction showed relative safety of MEPO phytochemicals except for rutin and naringin which showed potentials for immuno-toxicity, like Acarbose. Linamarin also showed potential for hepatotoxicity like Acarbose.
  • 12. 11 Table 6: Toxicity Properties of the MEPO phytochemicals Compounds Predicted LD50 value (mg/kg) Prediction accuracy (%) Hepato- toxicity Immuno -toxicity Cytotoxi city Carcino genicity Mutagen icity Epicatechin 10000 100% Inactive Inactive Inactive Inactive Inactive Naringin 2300 70.97% Inactive Active Inactive Inactive Inactive Catechin 10000 70.97% Inactive Inactive Inactive Inactive Inactive Kaempferol 3919 100% Inactive Inactive Inactive Inactive Inactive Linamarin 400 100% Active Inactive Inactive Inactive Inactive Rutin 5000 100% Inactive Active Inactive Inactive Inactive Resveratrol 1560 68.07% Inactive Inactive Inactive Inactive Inactive Spartein 2850 100% Inactive Inactive Inactive Inactive Inactive Ephedrine 404 100% Inactive Inactive Inactive Inactive Inactive Acarbose 24000 100% Active Active Inactive Inactive Inactive 4. Discussion Despite available pharmaceutical interventions, the current projections of the incidence of diabetes and its associated complications have necessitated the search for alternative therapeutic approaches. In this study, in vitro and in silico antidiabetic potential of methanol extract of P. ostreatus (MEPO) was evaluated. Our results demonstrated that MEPO significantly inhibited α-amylase and α-glucosidase activities in a concentration-dependent manner. α -amylase and α-glucosidase are two enzymes that play key role in carbohydrate metabolism. While α-amylase facilitates the catabolism of starch, releasing absorbable molecules, α-glucosidase catalyzes the end step of starch and disaccharides digestion from human diet (Kazeem et al., 2013). The uncontrolled activity of these enzymes is one of the contributors to postprandial hyperglycemia and has been implicated in diabetes development and complications. Hence, inhibiting their activities has become a strategy in blood glucose control. The inhibition of these enzymes’ activities is indicative of the potential of MEPO as a promising candidate for glycemic control in diabetes management and this is supported by previous studies. Karim et al., (2020) and Johnny & Okon, (2013) reported anti- hyperglycemic effects of solvent extracts of P. ostreatus, using in vivo models and from our result, inhibition of these enzymes may be one mechanism in which P. ostreatus exerts its anti-hyperglycemic effect. Oxidative stress, occasioned by excessive free radical production, is implicated in both the development and progression of many disease conditions (Yaribeygi et al., 2020); thus, antioxidant therapy remains a viable option to mitigate the oxidative damage that is associated with many diseases. DPPH radical- scavenging and FRAP assays are common test methods used to determine the antioxidant capacity of test substances. Antioxidant compounds are able to stabilize/quench DPPH radicals resulting in a change of color of reaction mixture from purple to yellow, while in the FRAP assay, antioxidant compounds prevent
  • 13. 12 the oxidation of ferrous ion (Fe2+ ) to ferric ion (Fe3+ ) (Omari et al., 2019). Our result showed that MEPO moderately scavenged DPPH radicals and reduced Fe3+ to Fe2+ in a concentration-related manner, indicating that MEPO has phytochemicals with antioxidant properties. In agreement with our result, Mishra et al., (2022) and Mkhize et al., (2022) also reported good DPPH radical-scavenging activity and FRAP, respectively, of solvent extract of P. ostreatus. The same trend was also reported by Menaga et al., (2014) and Leong et al., (2021) for extracts of other mushroom species. To identify the phytochemicals potentially involved in the anti-diabetic and antioxidant potentials of MEPO, we used gas chromatography instrument coupled with a flame ionization detector (GC-FID) ephedrine, oxalate, cyanogenic glycoside, rutin, naringin, kaempferol, epicatechin, phytate and catechin, among others. The presence of these compounds indicates that MEPO is a rich store of health-beneficial bioactive compounds. For example, catechin, epicatechin, rutin, and kaempferol are polyphenolic compounds known for their antioxidant and antidiabetic properties (Ademiluyi et al., 2018; Chukwuma et al., 2022). Thus, the presence of these phytochemicals in MEPO could have individually or in combination contributed to the antioxidant and antidiabetic potentials observed in this study. To clarify the contribution of each of the identified compounds to the antidiabetic properties of MEPO, we conducted molecular docking studies. Nine MEPO phytochemicals (naringin, kaempferol, resveratrol, linamarin, rutin, epicatechin, catechin, spartein, and ephedrine) were docked against α-amylase, α- glucosidase and PEPCK to provide insights into the ligand-complexes orientations and interactions between these phytochemicals and the enzymes implicated in diabetes based on the binding energies. MEPO phytochemicals demonstrated favorable binding energies (negative binding scores) against α-amylase, α- glucosidase and PEPCK. Negative binding scores usually indicate correctly docked compounds into target proteins’ crystal structure and higher negative scores usually indicate stronger binding affinities toward targets (Kumar et al., 2019). The active site of every enzyme consists of amino acid residues with three different functions, viz, catalytic residues (responsible for the ligand conversion to products), ligand-binding residues (responsible for the binding of the ligand to active site), and structural residues (responsible for the maintenance of the active site structure in its functional state). Three amino acids, Asp300, Glu233, and Asp197 are the catalytic residues of α-amylase active site. Our study showed that naringin and epicatechin had better interactions with α-amylase active site’s residues compared to the other MEPO phytochemicals docked. Naringin had nine hydrogen (H)-bond interactions within the active site with His305, Asp300, Glu233, Asp197, Glu240, Lys200, Thr163, Gln63, and Trp59. Likewise, epicatechin had five hydrogen bond interactions with His299, Asp300, Glu233, Thr163 and Gln63. A visual representation of the 3D and 2D of the docked ligand conformations at the active site of α-amylase (Figure 1, see appendix) indicated that interactions occurred with the key residues at the active site, like Acarbose which had four H-bond interactions with Asp197, Asp300, Glu233 and Asn53. Naringin and rutin showed higher binding energies for α-glucosidase when compared to Acarbose. Naringin formed H-bonds with Arg411, Ser676, Leu678, Asp518, Met519 and Arg600. We also observed that among the six H-bond interactions observed with naringin, 2-H bonds (Arg600 and Asp518) were also observed with Acarbose. Similarly, rutin showed H-bond interactions with Asp616, His674, Asp404, Trp481, Asp518, and Ser679. Naringin and epicatechin showed higher binding energies (-9.8 kcal/mol and -9.3 kcal/mol, respectively) for PEPCK compared to Acarbose (-7.5 kcal/mol). Epicatechin formed H-bond interactions with Asn403, Ala287, Cys288, Gly289, Thr291, Val335, Lys290, Arg436 and Asn292. Naringin also had H-bond interactions with Ala287, Gly289, Cys288, Asn292, Lys290, Pro402 and Arg87, compared to Acarbose that formed H-bond interactions with Asn292, Phe333,
  • 14. 13 Asp311, Lys244, Lys290, Thr291, Cys288, Gly289. Hence, because of the more negative binding energies exhibited by the phytochemicals coupled with their interactions with higher number of active site amino acid residue may explain why MEPO constituents are better in regulating blood glucose level than Acarbose. This may suggest that MEPO constituent might serve as an effective alternative in diabetes management. According to the Lipinski's rule, a drug must have an HBD (hydrogen bond donors) count of ≤ 5 and an HBA (hydrogen bond acceptor) of ≤ 10 in order for it to be orally active (Lipinski, 2016). All the MEPO phytochemicals and standard drug passed the oral bioactivity test, except for rutin and naringin which had high HBDs (10 and 8, respectively) and high HBAs (16 and 14, respectively). Similarly, rutin and naringin displayed immunotoxicity potentials, while linamarin showed hepatotoxic effects, unlike other MEPO phytochemicals that are predicted to be safe. Future studies should establish the safety profile of MEPO and its phytochemicals using cellular and in vivo models. Conclusion Methanol extract of Pleurotus ostreatus (MEPO) is rich in health-enhancing phytochemicals and demonstrated moderate antioxidant and antidiabetic activities as evidenced by in vitro and in silico molecular docking studies. The presence of the identified constituents and their respective interactions with enzymes implicated in blood glucose elevation suggest that the P. ostreatus-derived compounds may be a promising therapeutic strategy for managing blood sugar levels, especially in diabetic conditions. Further studies are warranted to confirm the above beneficial potentials using in vivo models. Data Availability The data supporting the outcome of this present study are included within the article. Declaration of Competing Interest None Acknowledgement None. REFERENCES Abbas, G., Al Harrasi, A., Hussain, H., Hamaed, A., & Supuran, C. T. (2019). The management of diabetes mellitus-imperative role of natural products against dipeptidyl peptidase-4, α-glucosidase and sodium- dependent glucose co-transporter 2 (SGLT2). Bioorganic Chemistry, 86(February), 305–315. https://doi.org/10.1016/j.bioorg.2019.02.009 Adebiyi, O. E., Olayemi, F. O., Ning-Hua, T., & Guang-Zhi, Z. (2017). In vitro antioxidant activity, total phenolic and flavonoid contents of ethanol extract of stem and leaf of Grewia carpinifolia. Beni-Suef University Journal of Basic and Applied Sciences, 6(1), 10–14. https://doi.org/10.1016/j.bjbas.2016.12.003 Ademiluyi, A. O., Aladeselu, O. H., Oboh, G., & Boligon, A. A. (2018). Drying alters the phenolic constituents, antioxidant properties, α-amylase, and α-glucosidase inhibitory properties of Moringa (Moringa oleifera) leaf. Food Science and Nutrition, 6(8), 2123–2133. https://doi.org/10.1002/fsn3.770
  • 15. 14 Aguchem, R. N., Chibuogwu, C. C., Okolo, B. O., Oyeagu, U., Etim, V. E., Anaele, E. N., & Njoku, O. U. (2022). Nutrient and Antinutrient Composition of Pleurotus ostreatus Grown on Different Substrates. 69. https://doi.org/10.3390/iecps2021-11955 Alachaher, F. Z., Dali, S., Dida, N., & Krouf, D. (2018). Comparison of phytochemical and antioxidant properties of extracts from flaxseed (Linum usitatissimum) using different solvents. International Food Research Journal, 25(1), 75–82. Alqahtani, A. S., Hidayathulla, S., Rehman, T., Elgamal, A. A., Dib, R. A. El, & Alajmi, M. F. (2020). Alpha-Amylase and Alpha-Glucosidase Enzyme Inhibition and Antioxidant Potential of 3- Oxolupenal and Katononic Acid Isolated from Nuxia oppositifolia. Biomolecules, 10(61), 2–19. https://pubmed.ncbi.nlm.nih.gov/31905962/ Bhushan, A., & Kulshreshtha, M. (2018). The Medicinal Mushroom Agaricus bisporus: Review of Phytopharmacology and Potential Role in the Treatment of Various Diseases. Journal of Nature and Science of Medicine, 1, 4–9. https://doi.org/10.4103/JNSM.JNSM Boisa N, Nwachoko N, Bull OS, & James FA. (2020). Physical Composition, Proximate, Phytochemical and Impact of Coconut Oil on Lipid Profile of Albino Rats. IOSR Journal of Applied Chemistry, 13(7), 51–56. https://doi.org/10.9790/5736-1307015156 Chukwuma, I. F., Nworah, F. N., Apeh, V. O., Omeje, K. O., Nweze, E. J., Asogwa, C. D., & Ezeorba, T. P. C. (2022). Phytochemical Characterization, Functional Nutrition, and Anti-Diabetic Potentials of Leptadenia hastata (pers) Decne Leaves: In Silico and In Vitro Studies. Bioinformatics and Biology Insights, 16. https://doi.org/10.1177/11779322221115436 Cruz-Solorio, A., Villanueva-Arce, R., Garín-Aguilar, M. E., Leal-Lara, H., & Valencia-del Toro, G. (2018). Functional properties of flours and protein concentrates of 3 strains of the edible mushroom Pleurotus ostreatus. Journal of Food Science and Technology, 55(10), 3892–3901. https://doi.org/10.1007/s13197-018-3312-x Daina, A., Michielin, O., & Zoete, V. (2017). SwissADME: A free web tool to evaluate pharmacokinetics, drug-likeness and medicinal chemistry friendliness of small molecules. Scientific Reports, 7(January), 1–13. https://doi.org/10.1038/srep42717 Dehghan, H., Sarrafi, Y., & Salehi, P. (2016). Antioxidant and antidiabetic activities of 11 herbal plants from Hyrcania region, Iran. Journal of Food and Drug Analysis, 24(1), 179–188. https://doi.org/10.1016/j.jfda.2015.06.010 Dundas, J., Ouyang, Z., Tseng, J., Binkowski, A., Turpaz, Y., & Liang, J. (2006). CASTp: Computed atlas of surface topography of proteins with structural and topographical mapping of functionally annotated residues. Nucleic Acids Research, 34(WEB. SERV. ISS.), 116–118. https://doi.org/10.1093/nar/gkl282 Gómez-Valadés, A. G., Vidal-Alabró, A., Molas, M., Boada, J., Bermúdez, J., Bartrons, R., & Perales, J. C. (2006). Overcoming diabetes-induced hyperglycemia through inhibition of hepatic phosphoenolpyruvate carboxykinase (GTP) with RNAi. Molecular Therapy, 13(2), 401–410. https://doi.org/10.1016/j.ymthe.2005.08.026 Huang, K., El-Seedi, H. R., & Xu, B. (2022). Critical review on chemical compositions and health- promoting effects of mushroom Agaricus blazei Murill. Current Research in Food Science, 5(July), 2190–2203. https://doi.org/10.1016/j.crfs.2022.10.029 Johnny, I., & Okon, J. (2013). Antidiabetic Effect of Pleurotus ostreatus (Jacq.ex Fr) kumm. Mushroom on Alloxan-induced Diabetic Rats. Indian Journal of Pharmaceutical and Biological Research, 1(02),
  • 16. 15 31–36. https://doi.org/10.30750/ijpbr.1.2.6 Karim, R., Rahman, F., Rahman, R., Tamannaa, Z., Ghose, D. K., Islam, N., Hossain, T., Islam, R., & Rahman, A. (2020). Hypoglycemic and antidyslipidemic potential of pleurotusostreatus in streptozotocin-induced diabetic rats. Journal of Advanced Biotechnology and Experimental Therapeutics, 3(1), 49–55. https://doi.org/10.5455/jabet.2020.d106 Kazeem, M. I., Adamson, J. O., & Ogunwande, I. A. (2013). Modes of inhibition of α-amylase and α- glucosidase by aqueous extract of morinda lucida benth leaf. BioMed Research International, 2013. https://doi.org/10.1155/2013/527570 Kumar, A. R., Merlin, N. J., Arundhathi, R. P., & Dharan, S. S. (2019). In-silico Molecular Docking Evaluation of Plumbagine Derivatives for Anticancer Activity. 11(7), 2676–2678. Leong, C. C., Ho, W. Y., Yeap, S. K., Krishnen, G., Chong, Z. X., Ho, J. S., Lim, P. T., & Ten, S. T. (2021). Assessment of phylogenetic, growth, and antioxidant capacity of Pleurotus spp. in Malaysia. Journal of Food Processing and Preservation, 45(6), 1–13. https://doi.org/10.1111/jfpp.15554 Lesa, K. N., Khandaker, M. U., Mohammad, F., Iqbal, R., Sharma, R., Islam, F., Mitra, S., & Emran, T. Bin. (2022). Nutritional Value , Medicinal Importance , and Health-Promoting Effects of Dietary Mushroom ( Pleurotus ostreatus ). 2022. Lipinski, C. A. (2016). Rule of five in 2015 and beyond: Target and ligand structural limitations, ligand chemistry structure and drug discovery project decisions. Advanced Drug Delivery Reviews, 101, 34– 41. https://doi.org/10.1016/j.addr.2016.04.029 Menaga, D., Rajakumar, S., & Ayyasamy, P. M. (2014). Free radical scavenging activity of methanolic extract of brown alga Spatoglossum asperum. International Journal of Pharmacy and Pharmaceutical Sciences, 5(4), 601–606. Mishra, V., Tomar, S., Yadav, P., Vishwakarma, S., & Singh, M. P. (2022). Elemental Analysis, Phytochemical Screening and Evaluation of Antioxidant, Antibacterial and Anticancer Activity of Pleurotus ostreatus through In Vitro and In Silico Approaches. Metabolites, 12(9). https://doi.org/10.3390/metabo12090821 Mkhize, S. S., Cedric Simelane, M. B., Mongalo, I. N., & Pooe, O. J. (2022). The Effect of Supplementing Mushroom Growing Substrates on the Bioactive Compounds, Antimicrobial Activity, and Antioxidant Activity of Pleurotus ostreatus. Biochemistry Research International, 2022. https://doi.org/10.1155/2022/9436614 Mwangi, R. W., Macharia, J. M., Wagara, I. N., & Bence, R. L. (2022). The antioxidant potential of different edible and medicinal mushrooms. Biomedicine and Pharmacotherapy, 147(October 2021), 112621. https://doi.org/10.1016/j.biopha.2022.112621 Nowakowski, P., Markiewicz-Żukowska, R., Bielecka, J., Mielcarek, K., Grabia, M., & Socha, K. (2021). Treasures from the forest: Evaluation of mushroom extracts as anti-cancer agents. Biomedicine and Pharmacotherapy, 143(June). https://doi.org/10.1016/j.biopha.2021.112106 Ogurtsova, K., Guariguata, L., Barengo, N. C., Ruiz, P. L. D., Sacre, J. W., Karuranga, S., Sun, H., Boyko, E. J., & Magliano, D. J. (2022). IDF diabetes Atlas: Global estimates of undiagnosed diabetes in adults for 2021. Diabetes Research and Clinical Practice, 183(2022), 1–9. https://doi.org/10.1016/j.diabres.2021.109118 Ojukwu, S. U., FA, O., & CE, U. (2021). GC-FID phytochemical analysis of selected antidiabetic herbal preparations sold in Onitsha metropolis, Anambra state, Nigeria. Journal of Medicinal Plants Studies, 9(4), 60–67. https://doi.org/10.22271/plants.2021.v9.i4a.1313
  • 17. 16 Omari, N. El, Sayah, K., Fettach, S., Blidi, O. El, Bouyahya, A., Faouzi, M. E. A., Kamal, R., & Barkiyou, M. (2019). Evaluation of in vitro antioxidant and antidiabetic activities of Aristolochia longa extracts. Evidence-Based Complementary and Alternative Medicine, 2019. https://doi.org/10.1155/2019/7384735 Rines, A. K., Sharabi, K., Tavares, C. D. J., & Puigserver, P. (2016). Targeting hepatic glucose metabolism in the treatment of type 2 diabetes. Nature Reviews Drug Discovery, 15(11), 786–804. https://doi.org/10.1038/nrd.2016.151 Sayah, K., Marmouzi, I., Naceiri Mrabti, H., Cherrah, Y., & Faouzi, M. E. A. (2017). Antioxidant Activity and Inhibitory Potential of Cistus salviifolius (L.) and Cistus monspeliensis (L.) Aerial Parts Extracts against Key Enzymes Linked to Hyperglycemia. BioMed Research International, 2017. https://doi.org/10.1155/2017/2789482 Singh, A., Kukreti, R., Saso, L., & Kukreti, S. (2022). Pathways and Type 2 Diabetes. Molecules, 27, 950– 969. Suleiman, W. B., Shehata, R. M., & Younis, A. M. (2022). In vitro assessment of multipotential therapeutic importance of Hericium erinaceus mushroom extracts using different solvents. Bioresources and Bioprocessing, 9(1). https://doi.org/10.1186/s40643-022-00592-6 Sun, H., Saeedi, P., Karuranga, S., Pinkepank, M., Ogurtsova, K., Duncan, B. B., Stein, C., Basit, A., Chan, J. C. N., Mbanya, J. C., Pavkov, M. E., Ramachandaran, A., Wild, S. H., James, S., Herman, W. H., Zhang, P., Bommer, C., Kuo, S., Boyko, E. J., & Magliano, D. J. (2022). IDF Diabetes Atlas: Global, regional and country-level diabetes prevalence estimates for 2021 and projections for 2045. Diabetes Research and Clinical Practice, 183, 109119. https://doi.org/10.1016/j.diabres.2021.109119 Sun, L., Wang, Y., & Miao, M. (2020). Inhibition of α -amylase by polyphenolic compounds : Substrate digestion , binding interactions and nutritional intervention. Trends in Food Science & Technology, 104(February), 190–207. https://doi.org/10.1016/j.tifs.2020.08.003 Torres-Martínez, B. D. M., Vargas-Sánchez, R. D., Torrescano-Urrutia, G. R., Esqueda, M., Rodríguez- Carpena, J. G., Fernández-López, J., Perez-Alvarez, J. A., & Sánchez-Escalante, A. (2022). Pleurotus Genus as a Potential Ingredient for Meat Products. Foods, 11(6), 1–13. https://doi.org/10.3390/foods11060779 Yadav, D., & Negi, P. S. (2021). Bioactive components of mushrooms: Processing effects and health benefits. Food Research International, 148(July), 110599. https://doi.org/10.1016/j.foodres.2021.110599 Yaribeygi, H., Sathyapalan, T., Atkin, S. L., & Sahebkar, A. (2020). Review Article Molecular Mechanisms Linking Oxidative Stress and Diabetes Mellitus. 2020. Yu, S., Meng, S., Xiang, M., & Ma, H. (2021). Phosphoenolpyruvate carboxykinase in cell metabolism: Roles and mechanisms beyond gluconeogenesis. Molecular Metabolism, 53(May), 101257. https://doi.org/10.1016/j.molmet.2021.101257
  • 19. 18 Figure 1. An observation of the molecular interaction (left: 2D and right: 3D) between compounds with the highest binding energies and standard against α-amylase. Epicatechin (Ai, Aii), Naringin (Bi, Bii), Catechin (Ci, Cii), Standard (Acarbose) (Di, Dii). Ai Bi Ci Cii Bii Aii
  • 20. 19 Figure 2. An observation of the molecular interaction (left: 2D and right: 3D) between compounds with the highest binding energies and standard against α-glucosidase. Epicatechin (Ai, Aii), Naringin (Bi, Bii), Rutin (Ci, Cii), Standard (Acarbose) (Di, Dii). Di Ai Bi Ci Dii Aii Bii Cii
  • 21. 20 Figure 3. An observation of the molecular interaction (left: 2D and right: 3D) between compounds with the highest binding energies and standard against PEP carboxykinase. Epicatechin (Ai, Aii), Naringin (Bi, Bii), Rutin (Ci, Cii), Standard (Acarbose) (Di, Dii). Author contribution Chibuogwu, C.C., Okagu, I.U. and Aham, E.C. conceived and designed the study. Nnemolisa, S.C., Chukwurah, C.C, Aguchem, R.N., Edeh, S.C. and Obiora, M.O carried out the experiment. Aguchem, R.O. and Anyebe, D.E. analyzed the data. Chukwu, M.C. carried out the Molecular docking studies. Nnemolisa, S.C. and Chibuogwu, C.C. drafted the manuscript. Okagu, I.U. and Aham, E.C. edited and revised the manuscript. All authors read and approved the final manuscript. Declaration of Competing Interest None declared. Di Dii