Download as pdf or txt
Download as pdf or txt
You are on page 1of 14

Received: 16 April 2021 Revised: 28 July 2021 Accepted: 1 August 2021

DOI: 10.1002/pca.3086

RESEARCH ARTICLE

Screening and characterisation of potential antioxidant,


hypoglycemic and hypolipidemic components revealed in
Portulaca oleracea via multi-target affinity ultrafiltration LC–MS
and molecular docking

Hui Zhang1,2,3,4 | Guilin Chen1,3,4 | Jinpeng Yang5 | Chunlei Yang5 |


1,3,4
Mingquan Guo

1
CAS Key Laboratory of Plant Germplasm
Enhancement and Specialty Agriculture, Abstract
Wuhan Botanical Garden, Chinese Academy of Introduction: Portulaca oleracea is a commonly used nutritional vegetable and tradi-
Sciences, Wuhan, China
2 tional herbal medicine with plenty of nutrients and manifold pharmacological activi-
College of Life Sciences, University of
Chinese Academy of Sciences, Beijing, China ties. However, the potential active ingredients for its remarkable antioxidant,
3
Sino-Africa Joint Research Centre, Chinese hypoglycemic and hypolipidemic activities remain unexplored.
Academy of Sciences, Wuhan, China
4
Objectives: The present study aims to systematically evaluate the antioxidant activi-
Innovation Academy for Drug Discovery and
Development, Chinese Academy of Sciences, ties of different extracts of P. oleracea and screen bioactive ligands that can interact
Shanghai, China
with α-glucosidase, pancreatic lipase, and superoxide dismutase (SOD).
5
Tobacco Research Institute of Hubei
Province, Wuhan, China
Methods: In this research, the antioxidant activities of different parts of P. oleracea
and their corresponding total phenolic content (TPC) and total flavonoid content
Correspondence
Mingquan Guo, CAS Key Laboratory of Plant
(TFC) were systematically determined. Subsequently, a multi-target affinity ultrafiltra-
Germplasm Enhancement and Specialty tion method was developed using affinity ultrafiltration with SOD, α-glucosidase, and
Agriculture, Wuhan Botanical Garden, Chinese
Academy of Sciences, Wuhan, China.
pancreatic lipase coupled to liquid chromatography–mass spectrometry (UF-LC–MS).
Email: guomq@wbgcas.cn Later, molecular docking was used to further investigate the possible interaction
Chunlei Yang, Tobacco Research Institute of mechanism between these ligands and target enzymes.
Hubei Province, Wuhan, China.
Email: ycl193737@163.com Results: Among them, the ethyl acetate (EA) fraction showed the highest antioxidant
activity along with the highest TPC and TFC, and four compounds in the EA fraction
Funding information
Natural Science Foundation of Hubei Province, were quickly retrieved as potential SOD, α-glucosidase, and pancreatic lipase ligands,
Grant/Award Numbers: 2019CFB254, respectively. Molecular docking revealed that these potential ligands exhibited strong
2019CFB254 to G. Chen; Natural Science
Foundation of China, Grant/Award Number: binding ability and inhibitory activities on SOD, α-glucosidase, and pancreatic lipase.
81903791 to G. Chen Conclusion: The present study revealed that P. oleracea can be used as a functional
food with excellent antioxidant, hypoglycemic and hypolipidemic effects. Meanwhile,
the integrated strategy based on multi-target UF-LC–MS and molecular docking also
provided a powerful tool and a multidimensional perspective for further exploration
of active ingredients in P. oleracea responsible for the antioxidant, hypoglycemic and
hypolipidemic activities.

Hui Zhang, Guilin Chen and Jinpeng Yang contributed equally to this work.

Phytochemical Analysis. 2021;1–14. wileyonlinelibrary.com/journal/pca © 2021 John Wiley & Sons, Ltd. 1
2 ZHANG ET AL.

KEYWORDS
antioxidant, diabetes, obesity, pancreatic lipase, Portulaca oleracea, SOD, UF-LC–MS,
α-glucosidase

1 | I N T RO DU CT I O N been reported that P. oleracea is rich in variable active constituents


including polysaccharides, alkaloids, polyphenols, fatty acids, and
Diabetes mellitus (DM) is a metabolic disorder syndrome flavonoids,19 and possesses many biological activities, such as
characterised by hyperglycemia with high morbidity and high mortal- antioxidant, antibacterial, anti-inflammatory, hypoglycemic, and
20,21
ity, especially type 2 diabetes (T2DM), which accounts for more than hypolipidemic, Traditionally, P. oleracea was widely used in the
90% of the total amount.1 It is estimated that about five million treatment of cardiovascular and cerebrovascular diseases, diabetes,
people died from diabetes with the financial outlay of $850 billion in and their correlated complications.22 A previous study revealed that
2017.2 Obesity is a metabolic disease associated with genes, environ- P. oleracea extracts played an hypoglycemic role by improving insulin
3
ment, lifestyle, inflammation and other factors. The global epidemic resistance in mice with type 2 diabetes.23 Besides, the aqueous
of obesity and diabetes has led to a significant increase in the number extracts of P. oleracea inhibited hyperglycemia and diabetic vascular
of people worldwide with metabolic syndrome.4 Key enzymes inflammation in db/db mice, prevented the development of diabetic
involved in the pathogenesis and the treatment of T2DM and obesity endothelial dysfunction, and thus delayed the development of
have been studied extensively, among which α-glucosidase and lipase diabetes and vascular complications.24,25 Meanwhile, its seed extracts
have attracted worldwide attention. Effective α-glucosidase inhibi-
5
have been reported to exhibit significant hypolipidemic activity in
tors, such as acarbose,6 voglibose,7 and miglitol,8 served as the main diabetic rats,26 and extracts exerted excellent inhibitory activity
tactic in the field of diabetes and its complications.9,10 Pancreatic against α-glucosidase and pancreatic lipase.27,28 However, the respon-
lipase is also an important target enzyme for obesity. Orlistat is the sible bioactive compounds of P. oleracea against those enzymes and
only lipase inhibitor drug registered for the treatment of obesity.11 their underlying mechanisms are far from known.
Moreover, epidemiological and clinical research results disclosed that In this context, the development of efficient screening methods
increased oxidative stress in adipose tissue is both the instigator and and strategies to reveal the bioactive ingredients from natural prod-
the consequence of obesity and its related metabolic syndrome.12 ucts and their underlying mechanism has become an urgent need.
Excessive reactive oxygen species trigger oxidative stress, which Ultrafiltration liquid chromatography–mass spectrometry (UF-LC–MS)
causes significant damage to the body by activating cellular stress is a high-throughput screening method that combines ultrafiltration
pathways, ultimately leading to diabetic, obesity, and cardiovascular and LC–MS. It has attracted numerous attention due to its potential
13
disease. Therefore, further exploration of the potential of to quickly, efficiently, and sensitively screen bioactive ingredients
antioxidants in the earlier processes is necessary for the prevention from complex mixtures.29,30 In the present study, different fractions
and treatment of diabetes, obesity and other metabolic diseases. with diverse antioxidant activity, total phenolic content (TPC), and
Although α-glycosidase and lipase have been revealed as total flavonoid content (TFC) were firstly evaluated, and the correla-
effective enzyme targets for diabetes and obesity, there is still few tion between the antioxidant activity of P. oleracea and its TPC
enzyme inhibitors in clinical use, thus searching for new drug and TFC was explored through correlation analysis. Subsequently,
candidates is always to be appreciated, especially from natural superoxide dismutase (SOD), α-glucosidase, and pancreatic lipase
sources for their tremendous structural and functional diversity.14 were selected as the targets to screen potential ligands from the
For example, Moringa oleifera leaf extracts significantly inhibited fractions with the strongest antioxidant activity through UF-LC–MS.
the activities of α-glucosidase and pancreatic lipase, as well as Ultimately molecular docking was employed to revealing the interac-
enhanced the glucose consumption of 3T3-L1 cells.15 Additionally, tion mechanism between these potential ligands and enzymes. Hence,
α-lipoic acid, a typical natural antioxidant,16 is widely applied to this study provides practical guidance and a theoretical basis for the
treat diabetes and its complications by reducing blood lipids, in-depth exploration of the potential applications of P. oleracea as an
protein oxidation products, and protecting pancreatic cells from effective functional food for the treatment of diabetes, obesity and
damage by ameliorating oxidative stress.17 To some extent, the rich other metabolic diseases.
and diverse active ingredients of edible/medicinal plants bring us
more choices and hopes.
Portulaca oleracea, a kind of annual herb widely distributed all 2 | EX PE RI MENT AL
over the world, is a well-received vegetable with abundant ω-3 fatty
acids, dietary minerals, vitamins, and other nutrients. Meanwhile, 2.1 | Chemicals and reagents
P. oleracea is also a traditional herbal medicine and has not only been
listed as one of the most commonly used medicinal plants but also α-Glucosidase from yeast was purchased from Kamai Shu Biotechnol-
named “Global Panacea” by the World Health Organisation.18 It has ogy (Shanghai, China). Lipase from porcine pancreas and SOD from
ZHANG ET AL. 3

bovine erythrocytes were purchased from Yuanye Bio-Technology ± standard deviation). DPPH free radical scavenging ability was
Co., Ltd (Shanghai, China). Ultrafiltration membranes (0.5 mL, 10 and expressed by inhibition rate (%) and half maximal inhibitory concentra-
30 kDa) were purchased from Millipore Co. Ltd (Bedford, MA, USA). tion (IC50) value, and calculated as follows:
Chromatographic grade acetonitrile and formic acid for high-
performance LC–MS liquid phase, and 2,2-diphenyl-1-picrylhydrazyl DPPH  free radical scavenging effect ð%Þ ¼ ½ðAC  AS Þ=AC   100 ð1Þ
(DPPH), 2,20 -azino-bis-(3-ethylbenzothiazoline-6-sulphonic acid)
(ABTS), and 1,3,5-tri(2-pyridyl)-2.4.6-triazine (TPTZ) for antioxidant where AC and AS represent the absorbance value of the blank control
analysis, were purchased from Sigma-Aldrich Group (Shanghai, China). and the tested sample control.
The water for the LC–MS analysis was prepared with a pure water
meter from Nanjing EPED Technology Development Co., Ltd (Nanjing,
China). Rutin was purchased from J&K Scientific Ltd (Beijing, China), 2.4.2 | Determination of ABTS assay
all other analytical solvents and chemicals were purchased from
Sinopharm Chemical Reagent Co., Ltd (Shanghai, China). A slightly improved method reported by Zhu et al. was used to con-
duct ABTS free radical scavenging experiments of the CE and differ-
ent extraction parts of P. oleracea.32 The ABTS solution (7mM in
2.2 | Plant materials water) was mixed with potassium persulfate (4.9 mM in water) in an
equal amount (v/v) and reacted in darkness for 12 to 16 hours as the
Portulaca oleracea was collected from a farm in Zhengzhou, Henan working solution (ABTS+). The ABTS+ solution was properly diluted
province, in August 2018. The specimen was identified by Prof. with methanol to ensure an absorbance of approximately 0.700
Guangwan Hu, a botanist at Wuhan Botanical Garden, Chinese ± 0.100 at 734 nm. Then, 190 μL ABTS+ solution was mixed with
Academy of Sciences. The fresh plant materials were dried in an oven 10 μL samples, and the absorbance at 734 nm was recorded after
under 40 C, crushed with a granulator, and stored in refrigerators at incubation in the dark for 30 minutes. Methanol and vitamin C

4 C until use (voucher specimen number: ZH-2018-08-002). (31.25–1000 μM) were used as blank and positive controls, respec-
tively. The results of ABTS scavenging activity were calculated using
the earlier calculation (equation 1).
2.3 | Preparation of different extracts from
P. oleracea
2.4.3 | Determination of ferric reducing antioxidant
Initially, 4 L 90% (v/v) ethanol was added to 400 g dry powders of power (FRAP) assay
P. oleracea, soaked at room temperature for 12 hours. After that, they
were ultrasonically extracted (200 W, 40 kHz) for 30 minutes, the The ferric reducing antioxidant power (FRAP) assay was conducted by
extracts were filtered. The earlier steps were repeated three times. the method described by Xu et al. with slight modifications.31 The
Then, the crude extract (CE) was concentrated, and the residue was FRAP reagent was produced by mixing acetate buffer (300 mM,
dissolved in water and successively partitioned by petroleum ether pH 3.6), TPTZ solution (10 mM) in hydrochloric acid (HCl, 40 mM),
(PE), dichloromethane (DCM), ethyl acetate (EA), and n-butanol (n-Bu) and iron(III) chloride (FeCl3, 20 mM) at a ratio of 10:1:1 (v/v/v). Next,
until the organic phase was colorless. Finally, the extracts of each part 10 μL suitably diluted samples, 30 μL ultrapure water, and 260 μL
were collected and concentrated with a rotary evaporator at reduced FRAP reagent were added in turn and then incubated at 37 C for
pressure, freeze-dried into dry powders under vacuum, and kept at 10 minutes. The absorbance of the mixture was recorded at 593 nm.

4 C till use. Iron sulphate (FeSO4, 31.25–1000 μM) was used to establish a
calibration curve. The activity of FRAP was expressed as milligrams of
Fe2+ per gram of the sample (mg Fe2+/g sample).
2.4 | Evaluation of the antioxidant capacity of
P. oleracea
2.5 | Determinations of total phenolic content
2.4.1 | Determination of DPPH assay (TPC)

For the antioxidant activity of P. oleracea samples against DPPH free TPC was calculated using the Folin–Ciocalteu method reported by
radicals were determined according to a previous study reported by Deng et al.33 Namely, 20 μL of the diluted sample, 20 μL Folin–
Xu et al. 31
with slight modifications. Initially, 10 μL samples or the pos- Ciocalteu (in pure water, 25%, v/v), 100 μL sodium carbonate
itive control solutions of vitamin C (31.25–1000 μM) were mixed with (Na2CO3, 1 M), and 20 μL ultrapure water were added into the reac-
190 μL of DPPH (100 μM) and incubated for 30 minutes in darkness tion system successively and incubated in the dark at room tempera-
at room temperature. Then, the absorbance was measured at 517 nm. ture for 1 hour. The absorbance was recorded at 760 nm. Gallic acid
The final results were expressed as the mean of three replicates (mean (5–40 μg/mL) was used as the standard to establish a calibration
4 ZHANG ET AL.

curve. The results of TPC were expressed in milligrams equivalent of for HPLC elution consisted of 0.1% (v/v) formic acid aqueous solution
gallic acid per milligram of dry sample (mg GAE/g sample). (mobile phase A) and acetonitrile (mobile phase B). The gradient con-
ditions for the EA fraction were as follows: 5–20% (B) in 0–
15 minutes; 20–22% (B) in 15–24 minutes, 22–24% (B) in 24–
2.6 | Determinations of total flavonoid content 26 minutes, and 24–26% (B) in 26–46 minutes. Furthermore, 15 μL
(TFC) sample was injected at the flow rate of 0.8 mL/min. The chromato-
gram of EA fraction was obtained at the wavelength of 320 nm. The
The TFC of ethanol extracts and different fractions of P. oleracea was ESI-MS/MS analysis was performed in the negative ion mode by a
determined by colorimetry, following the method described by Zou TSQ Quantum Access MAX MS equipped with ESI interface under
et al. with minor modifications.34 Briefly, 60 μL of sample solution the following conditions: the temperatures of vaporiser and capillary
were mixed with 360 μL of methanol and 20 μL of sodium nitrite were 350 C and 250 C, respectively; the gas pressure of sheath gas
(NaNO2) solution (5%, w/v). After incubation for 6 minutes, 40 μL of (nitrogen gas, N2) and axu gas (helium, He) was 40 psi and 10 psi,
aluminium chloride (AlCl3, 10%, w/v) was added to the tubes and respectively; the source voltage was 3000 V and the cone voltage and
shaken gently for 6 minutes. Next, 40 μL of 10% AlCl3 was added to collision energy were 40 V and 10 V, respectively; mass range (m/z)
stand for another 6 minutes. Finally, 120 μL of sodium hydroxide was from 150 to 1500, spray voltage was 3 kV. MS data (mass range
(NaOH) solution (4%, w/v) was added and reacted for 15 minutes, and from m/z 100–1000) was obtained in the full-scan mode. The
the resultant absorbance was measured at 510 nm. The results of TFC preliminary identification of compounds in the EA fraction of
were expressed in milligrams equivalent of rutin per milligram of dry P. oleracea was carried out by comparing the parent ions, MS
sample (mg RE/g sample). fragments, and retention times with the references.

2.7 | Procedures for screening and identifying 2.8 | Molecular docking analysis
potential ligands based on UF-LC–MS
According to the paper published by Chen and Guo,35 SOD (PDB:
2.7.1 | Affinity ultrafiltration screening 2C9V), α-glucosidase (PDB: 3A4A), and pancreatic lipase (PDB: 1LPB)
were simulated with corresponding potential ligands in P. oleracea by
The screening process of potential ligands was conducted based on a molecular docking. Initially, the three-dimensional (3D) structures of
method reported by Chen et al. with slight modification.15 Briefly, the three enzymes were downloaded from the RCSB protein databank
180 μL EA fraction of P. oleracea was incubated with 20 μL SOD (2 U, (www.rcsb.org), and ChemBio3D Ultra (version 12.0) was used to
pH 7.8), α-glucosidase (6 U, pH 6.8), and pancreatic lipase (2 mg/mL, establish the 3D structure of the ligands with the lowest energy. The

pH 7.4) in phosphate-buffered saline PBS buffer at 37 C for 1 hour, molecular docking process was simulated and optimised as follows.
respectively. The mixed solution was then transferred to an ultrafiltra- The structure of the enzymes and their ligands were prepared by
tion membrane of 30 kD (for SOD and α-glucosidase) or 10 kD (for removing the water molecules and adding the hydrogen atoms, and
pancreatic lipase), centrifuged at 10000 rpm for 10 minutes and washed the energy is then minimised in the MMFF94 force field. The grid box
three times with appropriate PBS buffer to remove the non-specific was centred on the active site of the enzyme with dimensions size of
binding components. Then, 200 μL acetonitrile (90%, v/v) was added 60  60  60 points, and the coordinate axes were center_X: 22.346,
and incubated for 20 minutes to release compounds bound to the center_Y: 15.739, and center_Z: 15.492 (SOD); center_X: 21.519,
enzyme, followed by the centrifugation at 10000 rpm for 10 minutes. center_Y: 7.702, and center_Z: 23.554 (α-glucosidase); center_X:
Finally, the filtrates were collected, lyophilised, and redissolved with 5.998, center_Y: 18.427, and center_Z: 37.754 (pancreatic lipase).
40 μL methanol for high-performance liquid chromatography ultraviolet Then, AutoDock Tools 1.5.6 was used to dock the ligands to the
electrospray ionisation tandem mass spectrometry (HPLC-UV/ESI-MS/ active sites of enzymes to create usable conformations. Subsequent
MS) analysis. The denatured enzyme which was incubated in boiling docking calculations were carried out through 2.5  106 energy
water for 10 minutes was served as negative control. assessments coupled with 30 independent runs of lamarckian genetic
algorithms. AutoDock Tools 1.5.6 and the Discovery Studio 4.1
software were employed for the analysis and visualisation of the
2.7.2 | HPLC-UV/ESI-MS/MS analysis docking results.

The chemical composition of EA fraction from P. oleracea was not


clear. In this part, Thermo Accela 600 HPLC system coupled with a 2.9 | Statistical analysis
TSQ Quantum Access MAX MS was acquired for the analysis and
identification. The separation of EA fraction was conducted with a All assays were repeated at least in triplicate, and the results were
HPLC system equipped with Waters Sunfire RP-C18 column (4.6 mm expressed as mean ± standard deviation. SPSS data analysis software
 250 mm, 5 μm; Waters, Wexford, Ireland). The mobile phase used version 24 was used for regression analysis of IC50 values, and Origin
ZHANG ET AL. 5

2021 software was used for significant difference analysis with the interact with free radicals before they attack the cells to prevent fur-
one-way analysis of variance (ANOVA). Difference was considered ther damages.36 Portulaca oleracea is rich in polyphenolic compounds,
significant at P ≤ 0.05. and its content fluctuates greatly in different growing stages37 and
different extraction processes.38 Flavonoids are also a major compo-
nent of P. oleracea.39 Just as shown in Figure 2A, The EA fraction con-
3 | RESULTS AND DISCUSSION tained the most abundant phenolics (46.67 ± 0.99 mg GAE/g dw),
which was more than four times higher than that of CE (P < 0.01), and
3.1 | In vitro antioxidant activity of P. oleracea water fraction showed the lowest total polyphenols of 7.35 ± 0.21 mg
GAE/g dry weight (DW). Similarly, the content of total flavonoids in
Considering the diverse scavenging methods of reactive oxygen spe- different fractions also showed a certain diversity. As shown in
cies and the complexity of natural phytochemicals, a series of Figure 2B, the EA fraction remained the highest of 86.28 ± 9.21 mg
methods have been developed to comprehensively evaluate the anti- RE/g DW, significantly lower than that of the CE fraction (36.30
oxidant activity of phytochemicals.31 In this work, three methods ± 2.57 mg RE/g DW), while water fraction was still the lowest (13.22
were applied to evaluate and compare the antioxidant activities of the ± 0.95 mg RE/g DW). These results further validate our findings
CE from P. oleracea together with its PE, DCM, EA, n-Bu, and water earlier., and provide a clear clue for us to explore the potential active
fractions. The results are shown in Figure 1. Compared with CE, EA ingredients for its remarkable antioxidant and hypoglycemic activities
fraction showed significantly stronger radical scavenging capacity from EA fraction.
against DPPH with the IC50 value of 142.70 ± 6.48 μg/mL, and also
stronger than the other four fractions, respectively. Ascorbic acid
(Vc) was used as positive controls in these assays with the IC50 values 3.3 | Correlation analysis between antioxidant
of 3.68 ± 0.12 μg/mL. Meanwhile, the EA fraction also showed the activities and phytochemical components
strongest potential antioxidant activities in FRAP assay with the mg
Fe2+/g of 11.37 ± 1.40 mg Fe2+/g, which was not only significantly To evaluate the relationships between antioxidant activities (DPPH,
higher than that of CE (3.50 ± 0.25 mg Fe2+/g), but also better than ABTS, and FRAP) of P. oleracea and its TPC and TFC, the correlation
the other four fractions. Besides, ABTS assays showed that DCM frac- analysis was conducted and the results are shown in Table 1. On the
tion had the strongest scavenging activity with IC50 at 24.49 one hand, significant correlations were found among the three experi-
± 1.52 μg/mL, followed by the EA fraction with IC50 at 45.16 mental results for the determination of antioxidant activities, the
± 1.67 μg/mL. Other fractions along with Vc (4.55 ± 0.25 μg/mL) also Pearson correlation coefficients (R2) between DPPH with ABTS and
showed considerable scavenging activities. Based on the results from FRAP were 0.887 (P < 0.05), 0.831 (P < 0.05), the R2 between ABTS
these three methods, the EA fraction showed the strongest antioxi- and FRAP was 0.842 (P < 0.05), indicating that these three methods
dant capacity as compared with the other four fractions. Thus, the EA were reliable and interchangeable. On the other hand, there were also
fraction was selected for further studies in this work. strong correlations between antioxidant activities and chemical com-
positions. The DPPH and ABTS values were highly negative correlated
with TPC with the R2 at 0.843 (P < 0.05) and 0.941 (P < 0.01),
3.2 | Total phenolic and flavonoid contents of respectively. While the FRAP values showed an extremely significant
P. oleracea (P < 0.01, R2 0.956) positive correlation with the TPC. The R2 between
the total flavonoids with three antioxidant methods and the total
Antioxidant compounds, mainly polyphenols and flavonoids, serve as polyphenol content were 0.786, 0.576, 0.809, and 0.707, respec-
the first line of defense against free radicals. These compounds tively. This indicated that the overall trend was consistent, but not

F I G U R E 1 Antioxidant activities of the crude extract (CE), petroleum ether (PE), dichloromethane (DCM), ethyl acetate (EA), and n-butanol (n-
Bu) and water (H2O) fractions of Portulaca oleracea. (A) The half maximal inhibitory concentration (IC50) values of 2,2-diphenyl-1- picrylhydrazyl
(DPPH) radical scavenging assay; (B) the IC50 values of 2,20 -azino-bis-(3-ethylbenzothiazoline-6-sulphonic acid) (ABTS) radical scavenging assay;
(C) ferric reducing antioxidant power (FRAP) assay. ** P < 0.01, && P < 0.01 as compared with the CE and ascorbic acid (Vc), respectively
6 ZHANG ET AL.

F I G U R E 2 (A) Total phenolic content (TPC) and (B) total flavonoid content (TFC) of Portulaca oleracea. GAE/g DW: gallic acid equivalent per
gram of dry weight; RE/g DW: rutin equivalent per gram of dry weight. ** P < 0.01, as compared with the crude extract (CE)

T A B L E 1 Pearson correlation coefficients (R2) among the various oxidative stress damage induced by free radicals.43 Extracts of
antioxidant activities and phytochemical contents of Portulaca P. oleracea reduced postprandial blood glucose by effectively reducing
oleracea
α-amylase and α-glucosidase activities,28 and also showed dose-
ABTS FRAP TPC TFC dependent antioxidant and anti-inflammatory activities against lipo-
DPPH 0.887 * 0.831 * 0.843 * 0.786 polysaccharide (LPS) induced lung injury in rats.18 In addition, it can
ABTS 0.842 * 0.941 ** 0.576 also greatly inhibit the activity of lipase.29 The earlier analysis indi-

FRAP 0.956 ** 0.809 cated that it was of theoretical and practical significance to explore
the antioxidant, hypoglycemic, and hypolipidemic activities of
TPC 0.707
P. oleracea by targeting SOD, α-glucosidase, and pancreatic lipase.
Note: * and ** indicate the correlation is significant at the level of 0.05 and
Ultrafiltration can be applied to effectively and sensitively screen
0.01, respectively.
DPPH, 2,2-diphenyl-1-picrylhydrazyl; ABTS, 2,20 -azino-bis- the specific bioactive ingredients from the complicated extracts. The
(3-ethylbenzothiazoline-6-sulphonic acid); FRAP, ferric reducing binding ability of the ligand to the enzyme was measured by compar-
antioxidant power; TPC, total phenolic content; TFC, total flavonoid ing the relative binding affinity (RBA), which was obtained by calculat-
content. ing the ratio of the peak area after incubation with active and
inactivated enzymes. Compound with an RBA greater than 1.5 was
considered as a potential ligand.44
significant. Taken together, the earlier results implied that polyphenols The RBA was calculated as follows:
might be the main contributors to the antioxidant capacity of
P. oleracea, which was consistent with the report of Voynikov et al.40 RBA ¼ AS =AC ð2Þ

where AS and AC represent the peak areas of samples incubated with


3.4 | UF-LC–MS analysis activated and inactivated enzymes, respectively. Just as shown in
Table 2 and Figure 3, for SOD, genistein-40 -O-glucoside (peak 18) and
3.4.1 | Screening for the potential superoxide N-acetyl-L-phenylalanine (peak 15) exhibited higher RBAs with SOD,
dismutase, α-glucosidase, and pancreatic lipase ligands with the RBA values of 1.62 and 1.57, other peaks also show good
in P. oleracea higher RBA values, but their configuration needs to be further deter-
mined. For α-glucosidase, three of the 17 compounds showed higher
α-Glucosidase and pancreatic lipase are key enzymes that catalyse the RBA values (more than 1.5), including, feruloylmalic acid (peak 17),
digestion of carbohydrates and fat,41 its inhibitors provide deeper N-(carboxyacetyl)phenylalanine (peak 14), genistein-40 -O-glucoside
insights into the treatment of diabetes, obesity, and other metabolic (peak 18), and the corresponding RBA values were 1.70, 1.53, and
diseases. Free radicals (a group of harmful reactive oxygen species) 1.52, respectively. For pancreatic lipase, N-(carboxyacetyl)phenylala-
are highly reactive small molecules that can damage DNA, proteins, nine (peak 14) exhibited the highest RBA value of 2.08, followed by
carbohydrates, and lipids. These damages could structurally lead to the genistein-40 -O-glucoside (peak 18) of 1.86, dhurrin 60 -glucoside
cell damage or apoptosis, which ultimately leads to chronic diseases (peak 19) of 1.77. On the whole, genistein-40 -O-glucoside (peak 18)
such as diabetes and atherosclerosis. 42
Exogenous antioxidant showed higher RBA values to SOD, α-glucosidase, and pancreatic
enzymes, such as SOD, a major antioxidant metalloenzyme responsi- lipase, indicating that it may exert antioxidant, hypoglycemic, and
ble for scavenging reactive oxygen species, can protect cells from hypolipidemic activities through multiple targets. The SOD, pancreatic
ZHANG ET AL. 7

lipase, and α-glucosidase inhibitory activities of these five compounds and their representative MS/MS spectra fragments are listed in
have not been measured in any of the references so far. Table 2. In addition, the chemical structures of the compounds
identified in the EA fraction of P. oleracea were tentatively
identified as shown later, and part of them was also presented in
3.4.2 | Characterisation of potential superoxide Figure 4.
dismutase, α-glucosidase, and pancreatic lipase ligands Peak 1 displayed a deprotonated molecular ion [M  H] of m/z
based on LC–MS 195. Two major fragment ions at m/z 177 [M  H  H2O] and
m/z 159 [M  H  2H2O] indicated the continuous release of the
The main peaks of EA fraction were identified and characterised H2O group from [M  H]. The product ions observed at m/z
by HPLC-ESI-MS/MS. The MS and MS/MS data of these peaks, 129 [M  H  H2O  CH2O] and m/z 99 [M  H  H2O  CO2]

such as retention time (RT), deprotonated molecular ion [M  H] , were suggestive for the successive losses of CH2O and CO2 moieties

T A B L E 2 The mass spectrometry (MS) data and relative binding affinities (RBAs) of compounds identified from the ethyl acetate (EA) fraction
of Portulaca oleracea

RBA
a
Peak RT MS/MS Superoxide Pancreatic
numbera (min) [M  H] Formula fragments Name dismutase α-Glucosidase lipase
1 3.25 195 C6H12O7 195, 177, 159, Gluconic acid 1.44 — 1.31
129, 99, 87
2 4.67 133 C4H6O5 133, 115, 89, 71 Malic acid 1.38 — —
3 5.59 188 C10H7NO3 188, 170, 144, 2-Hydroxyquinoline- — —
128, 102, 59 3-carboxylic acid
4 6.48 117 C4H6O4 117, 99, 73, 55 Succinic acid — —
5 11.68 315 — 315, 153, 152, Unknown 1.58 1.36 0.95
109, 108
6 12.22 341 C15H18O9 341, 179, 135 Caffeic acid-hexose 2.29 — —
7 13.28 355 C16H20O9 355, 193, 178, Ferulic acid hexose-I 1.34 1.25 0.92
149, 134
8 15.14 353 C16H18O9 191, 173, 171 Chlorogenic acid 1.17 1.26
9 16.65 355 C16H20O9 193, 178, 149, Ferulic acid hexose-II 1.60 0.65 0.94
134
10 17.25 278 — 278, 216, 162, Unknown — —
132, 119
11 18.71 236 C11H11NO5 236, 192, 174, N-Benzoylaspartic acid — —
148, 120, 77
12 22.44 502 C24H26O11N 340, 296, 268, Oleracein A — —
194, 145
13 22.98 401 — 401, 341, 221, Unknown 1.24 —
177, 113
14 23.54 250 C12H13NO5 206, 164, 147, N-(Carboxyacetyl) 1.53 2.08
103, 91 phenylalanine
15 25.26 206 C11H13NO3 206, 164, 147, N-Acetyl-L- 1.57 1.38 1.12
103, 91 phenylalanine
16 25.64 163 C9H8O3 163, 119 p-Coumaric acid 1.29 — —
17 27.57 309 C14H14O8 193, 178, 149, Feruloylmalic acid 1.34 1.70 1.42
134, 117
18 30.37 431 C21H20O10 431, 269, 89 Genistein-40 -O- 1.62 1.52 1.86
glucoside
19 31.11 472 C14H17NO7 310, 161, 148 Dhurrin 60 -glucoside 1.31 1.24 1.77
20 39.18 456 C20H27NO11 294, 148, 145 4-Hydroxybenzyl 1.54 1.25 1.41
cyanide-Rha-Glc
21 41.09 486 — 324, 175, 148 Unknown 1.47 1.19 1.32
22 42.95 175 — Unknown 1.45 1.14 1.26

(Continues)
8 ZHANG ET AL.

F I G U R E 3 The ultrafiltration
high-performance liquid
chromatography (UF-HPLC)
chromatograms of the potential
superoxide dismutase (SOD),
α-glucosidase, and pancreatic
lipase ligands in Portulaca
oleracea. The black, red, and blue
lines represent the HPLC
chromatograms of P. oleracea
without (A), with activated and
inactivated SOD (B), with
activated and inactivated
α-glucosidase (C), with activated
and inactivated pancreatic lipase
(D), respectively [Colour figure
can be viewed at
wileyonlinelibrary.com]

at m/z 159. Peak 2 revealed the molecular ion [M  H] at m/z 1 and peak 2 can be tentatively identified as gluconic acid and malic
133, similar fragmentations of [M  H  H2O] and [M  H  CO2] acid, which were previously isolated from P. oleracea.45 Peak 4 showed
were observed at m/z 115 and 89. Another major fragment ion m/z product ions at m/z 117 [M  H], 99 [M  H  H2O], 73 [M  H 
71 was obtained by the loss of H2O from m/z 89. By comparing the CO2], 55 [M  H  CO2  H2O], all of which were 16 Da less than
fragmentation information of MS with previous references, peak the corresponding product ions of peak 2, indicating the loss of –OH
ZHANG ET AL. 9

group from malic acid. These fragments are consistent with those Peak 6 produced a deprotonated molecular ion at m/z 341
reported in the literature,45 hence peak 4 was tentatively assigned as [M  H], and the appearance of the major product ions at m/z
succinic acid. 179 [M  H  C6H10O5] were due to loss of a hexose
Peak 3 displayed a deprotonated molecular ion at m/z 188. Two residue. Another obvious fragment at m/z 135 [M  H  C6H10O5
distinguished fragments at m/z 170 [M  H  H2O] and m/z  CO2] was due to the loss of the CO2 (44 Da) group from m/z
144 [M  H  CO2] were generated by the loss of H2O and CO2. 179. Peak 6 was supposed to be caffeic acid-hexose by comparing
Hence peak 3 was tentatively assigned as 2-hydroxyquinoline- the information on fragment ions in the reported reference.45
3-carboxylic acid, which has been detected in the aerial parts of Peaks 7 and 9 showed the same deprotonated molecular ions at
P. oleracea.46 m/z 355 [M  H], suggesting that they were structural isomers. The

FIGURE 4 Chemical structures of the compounds identified in the ethyl acetate (EA) fraction of Poertulaca oleracea
10 ZHANG ET AL.

appearance of the major product ions at m/z 193 [M  H  103 showed a loss of CO2 (44 Da) group at m/z 147. The fragment at
C6H10O5] was due to the loss of hexose residue, the fragment ions m/z 71 was due to the loss of CH2 (14 Da) group at m/z 103. Thus, peak
at m/z 178 and 149 were due to the subsequent neutral losses of – 15 can be tentatively identified as N-acetyl-L-phenylalanine, which was
CH3 (15 Da) and CO2 (44 Da) group, respectively. Product ion at m/z previously detected in P. oleracea.47 Likewise, peak 14 exhibited a

134 [M  H  C6H10O5  CO2] was due to the continued loss of molecular ion at m/z 250 [M  H], which was 44 Da higher than that
the CO2 (44 Da) group from m/z 178. Thus, peaks 7 and 9 were of peak 15, meanwhile, showed the same fragment ions at m/z
presumed to be isomers of ferulic acid hexose and were tentatively 206 [M – H  CO2], m/z 164 [M – H  CO2  COCH3], m/z
identified as ferulic acid hexose-I and ferulic acid hexose- II. 45,47
147 [M  H  CO2  COCH3  NH3], m/z 103 [M – H  CO2 
Peak 8 displayed a deprotonated molecular ion at m/z COCH3  NH3  CO2], m/z 91 [M – H  CO2  COCH3  NH3
353 [M  H]. Its main fragment ions at m/z 191, and m/z 173 were  CO2  CH2], respectively. Consequently, peak 14 was annotated as
produced by quinic acid. By comparing the MS fragments with the liter- N-(carboxyacetyl)phenylalanine, which was previously reported in
ature, peak 8 was tentatively assigned as chlorogenic acid, previously P. oleracea.47 Peak 12 exhibited its molecular ion at m/z 502 [M  H]
40
isolated from P. oleracea. Peak 16, displayed its deprotonated molecu- and characteristic MS/MS fragment at m/z 340 for the respective losses
lar ion at m/z 163 [M  H]. Further fragments appearing at m/z of glucoside moieties. The fragment ion at m/z 296 showed a loss of
119 showed a loss of CO2 (44 Da) group. Hence, peak 16 was assigned CO2 (44 Da) group at m/z 340. Another characteristic MS/MS fragment
as p-coumaric acid, which was previously isolated from P. oleracea.20 was observed at m/z 194 [M – H  C9H6O2], indicative of the loss of
Peak 11 produced a deprotonated molecular ion at m/z 236 p-coumaroyl. Consequently, peak 12 was annotated as oleracein A, pre-
[M  H]. Two distinguished fragments at m/z 192 [M – H  44] and viously isolated from P. oleracea.48

m/z 148 [M  H  2CO2] were generated, which indicated the contin- Peak 17 gave a deprotonated molecular ion signal [M  H] at
uous loss of CO2 (44 Da) group. The fragment at m/z 174 was caused m/z 309, its characteristic product ion was obtained at m/z 193.
by the loss of the H2O group on m/z 192 [M  H  CO2]. Another MS/MS product ions at m/z 178 and 149 inferred the loss of CH3
fragment ion at m/z 120 was obtained by losing a C2H4 from m/z (15 Da) and CO2 (44 Da) moiety of m/z 193. Another fragment ion at
148 [M – H – 44  44]. Hence, peak 11 was tentatively identified as m/z 134 suggested the loss of CO2 (44 Da) moiety of m/z 178. The
N-benzoylaspartic acid. Peak 15 displayed its molecular ion at m/z presence of these ions suggested that Peak 17 was feruloylmalic acid,
206 [M  H] together with fragment ion at m/z 164 [M – H  42], previously isolated in P. oleracea.20
owing to the loss of –COCH3 moiety. Fragment appearing at m/z Peak 18 showed a deprotonated molecular ion [M  H] at m/z
147 indicated the loss of NH3 moieties. The fragment at m/z 431, which produced daughter ions at m/z 269 [M – H  C6H10O5]

TABLE 3 Molecular docking analysis of the potential superoxide dismutase (SOD), α-glucosidase, and pancreatic lipase ligands in Portulaca
oleracea

SOD α-Glucosidase Pancreatic lipase

Peak BE IC50 Hydrogen- BE IC50 Hydrogen- BE IC50 Hydrogen-


no. Name (kcal/mol) (μM) bond atoms (kcal/mol) (μM) bond atoms (kcal/mol) (μM) bond atoms
14 N- — — — 5.09 185.88 5.82 54.57 GLN244,
(Carboxyacetyl) LYS239,
phenylalanine LYS238
15 N-Acetyl-L- 5.46 99.57 LYS3 — — — — — —
phenylalanine
17 Feruloylmalic — — — 4.55 462.71 GLN279, — — —
acid HIS280,
SER157,
ASN415,
LYS156,
LEU313
18 Genistein-40 -O- 6.07 35.70 LYS136, 8.11 1.14 ASP69, 6.27 25.24 GLN244,
glucoside HIS63, TYR158, ASN88,
ASN65 ASP215, ASP249,
ARG442, SER333,
ASP352 ARG265
19 Dhurrin 60 - — — — — — — 4.32 678.29 ASP249,
glucoside ASP257,
ASP331,
ARG265,
LYS268

Note: BE, binding energy; IC50, half maximal inhibitory concentration.


ZHANG ET AL. 11

due to the loss of glucoside residue. Thus, peak 19 was tentatively rupture of O-linkage on the glucoside residue. By comparing with
identified as genistein-40 -O-glucoside, which was previously reported the fragmentation data in previous literature, peak 19 was tenta-
in P. oleracea.39
tively identified as dhurrin 60 -glucoside.49 Similar fragmentation
Peak 19 gave a deprotonated molecular ion at m/z 472, and patterns and fragments were observed at peaks 20 and 21. Peak
its MS/MS fragments ions at m/z 310 [M – H  Glc] and 20 showed the molecular ion [M  H] at m/z 456, its product
148 [M – H – Glc  Glc] implied the presence of two glucoside ions at m/z 294 [M – H  Glc] and m/z 148 [M – H – Glc 
residues. Another obvious product ion m/z 161 may be due to the Rha] indicated the presence of rhamnose and glucose residue.

F I G U R E 5 Three-dimensional (A) and two-dimensional (B) docking models of genistein-40 -O-glucoside with superoxide dismutase (SOD) and
three-dimensional (C) and two-dimensional (D) docking models of genistein-40 -O-glucoside with α-glucosidase [Colour figure can be viewed at
wileyonlinelibrary.com]
12 ZHANG ET AL.

Product ion m/z 145 may be due to the rupture of O-linkage on It was worth mentioning that different hydrogen-bonds were
the rhamnose. Therefore, peak 20 was tentatively deduced as formed between three potential ligands and diverse amino acid resi-
4-hydroxybenzyl cyanide-Rha-Glc, which has not yet been dues of α-glucosidase. Among them, there were common amino acid
identified from P. oleracea and in current databases. residues between three ligands and lipase, suggesting that the interac-
tions among those three bioactive components with lipase may be
competitive. Meanwhile, in the docking simulation process, other
3.5 | Molecular docking studies forces such as van der Waals interaction, hydrophobic interaction,
and Coulomb interaction also provided vital role in the interactions
Molecular docking can be employed to quickly simulate the ligand– between the earlier potential bioactive components and enzymes,
target interaction mechanisms at the molecular level through especially pancreatic lipase. In this sense, the present study provided
computer simulation, including the active site, binding mode, and a convenient method to explore the interaction mechanisms of SOD,
energy, and reveal the potential relationship between chemical α-glucosidase, and pancreatic lipase with potential ligands in
structures and activity, which has become a broad and powerful P. oleracea, and is of considerable significance for the development
approach in the field of new drug discovery and development. The of active components from P. oleracea with hypoglycemic and
inhibitory activities of P. oleracea extracts on α-glucosidase50
and pan- hypolipidemic activities.
creatic lipase28 have been widely reported. In addition, five potential
SOD, α-glucosidase, and pancreatic lipase ligands were screened AC KNOW LEDG EME NT S
separately by the earlier UF-LC–MS method, but the interaction This work was jointly supported by the Natural Science Foundation of
mechanisms between these ligands and enzymes remain unclear. In China (Grant no. 81903791 to G. Chen) and the Natural Science
this study, molecular docking was hired to investigate the binding Foundation of Hubei Province (Grant No. 2019CFB254 to G. Chen).
mode of SOD, α-glucosidase, and pancreatic lipase with its ligands. The funders played no roles in the study design, data collection and
The binding energy (BE), in silico estimated IC50 values, and hydrogen analysis, and decision to publish. The authors would like to thank Prof.
bonds are shown in Table 3. Guangwan Hu for his assistance with the authentication of the plant
As shown in Table 3, those three potential α-glucosidase sample.
ligands screened out exhibited low BE and estimated IC50 values,
basically confirming the higher RBA revealed by UF-LC–MS in CONFLIC T OF INT ER E ST
Table 2. In addition, according to the BE and IC50 values, the ranks The authors declare no conflict of interest.
of the binding strength and enzyme inhibitory activity were in the
following order: genistein-40 -O-glucoside (peak 18), feruloylmalic DATA AVAILABILITY STAT EMEN T
acid (peak 17), and N-(carboxyacetyl)phenylalanine (peak 14), which No data are available
were consistent with the corresponding rankings of the number of
OR CID
hydrogen bonds, suggesting that more hydrogen bonds might con-
Hui Zhang https://orcid.org/0000-0001-9166-570X
tribute to the interaction strength and enzyme inhibitory activity to
Mingquan Guo https://orcid.org/0000-0002-6627-5627
some extent. For example, genistein-40 -O-glucoside exhibited the
lowest BE of 4.90 kcal/mol and the lowest estimated IC50 value
RE FE RE NCE S
at 1.14 μM. At the same time, a larger number of hydrogen bonds
1. Liu S, Li D, Huang B, Chen Y, Lu X, Wang Y. Inhibition of pancreatic
were also observed between genistein-40 -O-glucoside and α-gluco-
lipase, α-glucosidase, α-amylase, and hypolipidemic effects of the
sidase. It formed six hydrogen bonds with residues on ASP69, total flavonoids from Nelumbo nucifera leaves. J Ethnopharmacol.
TYR158, ASP215, ASP352, and ARG442. For pancreatic lipase, the 2013;149(1):263-269.
orders of the binding strength and enzyme inhibitory activity were: 2. Cho NH, Shaw JE, Karuranga S, et al. IDF Diabetes atlas: global esti-
mates of diabetes prevalence for 2017 and projections for 2045. Dia-
genistein-40 -O-glucoside (peak 18), N-(carboxyacetyl)phenylalanine
betes Res Clin Pract. 2018;138:271-281.
(peak 14), and dhurrin 60 -glucoside (peak 19), and which were basi- 3. Hamre K. Obesity: Multiple factors contribute. Nature. 2013;
cally consistent with the corresponding rankings of the RBA 493(7433):480.
revealed by UF-LC–MS in Table 2. Among them, genistein-40 -O- 4. Eckel RH, Grundy SM, Zimmet PZ. The metabolic syndrome. Lancet.
2005;365(9468):1415-1428.
glucoside (peak 18) showed the most potential bonding ability to
5. Peng X, Zhang G, Liao Y, Gong D. Inhibitory kinetics and mecha-
pancreatic lipase by binding to the adjacent residue, and the nism of kaempferol on α-glucosidase. Food Chem. 2016;190:
corresponding BE and estimated IC50 value were 6.27 kcal/mol 207-215.
and 25.24 μM, respectively. The other two compounds bind to 6. Tsunosue M, Mashiko N, Ohta Y, et al. An alpha-glucosidase inhibitor,
pancreatic lipase mainly by van der Waals interaction, hydrophobic acarbose treatment decreases serum levels of glyceraldehyde-derived
advanced glycation end products (ages) in patients with type 2 diabe-
interaction, and Coulomb interaction rather than hydrogen bonds.
tes. Clin Exp Med. 2010;10(2):139-141.
The three-dimensional docking models of genistein-40 -O-glucoside 7. Chen XL, Zheng YG, Shen YC. Voglibose (Basen AO-128), one of the
with the α-glucosidase and pancreatic lipase active site are shown most important alpha-glucosidase inhibitors. Curr Med Chem. 2006;
in Figure 5. 13(1):109-116.
ZHANG ET AL. 13

8. Campbell LK, Baker DE, Campbell RK. Miglitol: assessment of its role 27. Park JE, Han JS. Portulaca oleracea L. extract lowers postprandial
in the treatment of patients with diabetes mellitus. Ann Pharmacother. hyperglycemia by inhibiting carbohydrate-digesting enzymes. J Life
2000;34(11):1291-1301. Sci. 2018;28:421-428.
9. Liu X, Feng J, Li Y. Preparation of carbon-functionalized 28. Conforti F, Perri V, Menichini F, et al. Wild mediterranean dietary
magnetic graphene/mesoporous silica composites for selective plants as inhibitors of pancreatic lipase. Phytother Res. 2012;26(4):
extraction of miglitol and voglibose in rat plasma. Talanta. 2018; 600-604.
182:405-413. 29. Chen GL, Huang BX, Guo MQ. Current advances in screening for bio-
10. van de Laar FA, Lucassen PL, Akkermans RP, van de Lisdonk EH, active components from medicinal plants by affinity ultrafiltration
Rutten GE, van Weel C. α-Glucosidase Inhibitors for Patients With mass spectrometry. Phytochem Anal. 2018;29(4):375-386.
Type 2 Diabetes: Results from a Cochrane systematic review and 30. Fan MX, Chen GL, Sun BQ, et al. Screening for natural inhibitors of
meta-analysis. Diabetes Care. 2005;28(1):154-163. human topoisomerases from medicinal plants with bio-affinity ultrafil-
11. Shirai K, Tanaka M, Fujita T, et al. Reduction of excessive visceral tration and LC-MS. Phytochem Rev. 2020;19(5):1231-1261.
fat and safety with 52-week administration of lipase inhibitor 31. Xu YB, Chen GL, Guo MQ. Antioxidant and anti-inflammatory activi-
orlistat in Japanese: Long-term clinical study. Adv Ther. 2019;36(1): ties of the crude extracts of Moringa oleifera from kenya and their cor-
217-231. relations with flavonoids. Antioxidants (Basel). 2019;8(8):296.
12. Furukawa S, Fujita T, Shimabukuro M, et al. Increased oxidative stress 32. Zhu MZ, Liu T, Zhang CY, Guo MQ. Flavonoids of lotus (Nelumbo
in obesity and its impact on metabolic syndrome. J Clin Invest. 2004; nucifera) seed embryos and their antioxidant potential. J Food Sci.
114(12):1752-1761. 2017;82(8):1834-1841.
13. Panigrahy SK, Bhatt R, Kumar A. Reactive oxygen species: Sources, 33. Deng J, Liu R, Lu Q, et al. Biochemical properties, antibacterial and
consequences and targeted therapy in type 2 diabetes. J Drug Target. cellular antioxidant activities of buckwheat honey in comparison to
2017;25(2):93-101. manuka honey. Food Chem. 2018;252:243-249.
14. Harvey AL. Natural products in drug discovery. Drug Discov Today. 34. Zou Y, Chang SKC, Gu Y, Qian SY. Antioxidant activity and phenolic
2008;13(19-20):894-901. compositions of lentil (Lens culinaris var. Morton) extract and its frac-
15. Chen GL, Xu YB, Wu JL, Li N, Guo MQ. Hypoglycemic and hypo- tions. J Agric Food Chem. 2011;59(6):2268-2276.
lipidemic effects of Moringa oleifera leaves and their functional chemi- 35. Chen GL, Guo MQ. Rapid screening for α-glucosidase inhibitors from
cal constituents. Food Chem. 2020;333:127478. Gymnema sylvestre by affinity ultrafiltration-HPLC-MS. Front
16. Choi SW, Ho CK. Antioxidant properties of drugs used in type 2 dia- Pharmacol. 2017;8:228.
betes management: Could they contribute to, confound or conceal 36. Lobo V, Patil A, Phatak A, Chandra N. Free radicals, antioxidants and
effects of antioxidant therapy? Redox Rep. 2018;23(1):1-24. functional foods: impact on human health. Pharmacogn Rev. 2010;
17. Ghibu S, Craciun CE, Rusu R, et al. Impact of alpha-lipoic acid chronic 4(8):118-126.
discontinuous treatment in cardiometabolic disorders and oxidative 37. Saffaryazdi A, Ganjeali A, Farhoosh R, Cheniany M. Variation in
stress induced by fructose intake in rats. Antioxidants. 2019; phenolic compounds, α-linolenic acid and linoleic acid contents and
8(12):636. antioxidant activity of purslane (Portulaca oleracea L.) during
18. Miao L, Tao H, Peng Y, et al. The anti-inflammatory potential of Portu- phenological growth stages. Physiol Mol Biol Plants. 2020;26(7):
laca oleracea L. (purslane) extract by partial suppression on NF-κB and 1519-1529.
MAPK activation. Food Chem. 2019;290:239-245. 38. Gallo M, Conte E, Naviglio D. Analysis and comparison of the antioxi-
19. Zhou Y, Xin H, Rahman K, Wang S, Peng C, Zhang H. Portulaca dant component of Portulaca oleracea leaves obtained by different
oleracea L.: A review of phytochemistry and pharmacological effects. solid–liquid extraction techniques. Antioxidants (Basel). 2017;6(3):64.
Biomed Res Int. 2015;2015:925631. 39. Liang X, Li L, Tian J, et al. A rapid extraction and analysis method for
20. Nemzer B, Al-Taher F, Abshiru N. Phytochemical composition and the simultaneous determination of 26 bioflavonoids in Portulaca
nutritional value of different plant parts in two cultivated and wild oleracea L. Phytochem Anal. 2014;25(6):537-543.
purslane (Portulaca oleracea L.) genotypes. Food Chem. 2020;320: 40. Voynikov Y, Gevrenova R, Balabanova V, Doytchinova I, Nedialkov P,
126621. Zheleva-Dimitrova D. LC-MS analysis of phenolic compounds and
21. Hozayen W. Effects of aqueous purslane (Portulaca oleracea) extract oleraceins in aerial parts of Portulaca oleracea L. J Appl Bot Food Qual.
and fish oil on gentamicin nephrotoxicity in albino rats. Nature Sci. 2019;92:298-312.
2011;9:47-62. 41. Sun H, Zhang Y, Ding W, et al. Inhibitory activity evaluation and
22. Rahdar P. The studying effect of drought stress on germination, pro- mechanistic studies of tetracyclic oxindole derivatives as alpha-
line, sugar, lipid, protein and chlorophyll content in purslane (Portulaca glucosidase inhibitors. Eur J Med Chem. 2016;123:365-378.
oleracea L.) Leaves. J Med Plant Res. 2012;6(9):1539-1547. 42. Abeyrathne N, Huang X, Ahn DU. Antioxidant, angiotensin-
23. Lee JH, Park JE, Han JS. Portulaca oleracea L. extract reduces hyper- converting enzyme inhibitory activity and other functional properties
glycemia via PI3k/Akt and AMPK pathways in the skeletal muscles of of egg white proteins and their derived peptides – a review. Poult Sci.
C57BL/Ksj-db/db mice. J Ethnopharmacol. 2020;260:112973. 2018;97(4):1462-1468.
24. Lee AS, Lee YJ, Lee SM, et al. Portulaca oleracea ameliorates diabetic 43. Sheng Y, Abreu IA, Cabelli DE, et al. Superoxide dismutases and
vascular inflammation and endothelial dysfunction in db/db mice. Evid superoxide reductases. Chem Rev. 2014;114(7):3854-3918.
Based Complement Alternat Med. 2012;2012:741824. 44. Munigunti R, Mulabagal V, Caldero  n AI. Screening of natural com-
25. Zidan Y, Bouderbala S, Djellouli F, Lacaille-Dubois MA, pounds for ligands to PfTrxR by ultrafiltration and LC-MS based bind-
Bouchenak M. Portulaca oleracea reduces triglyceridemia, ing assay. J Pharm Biomed Anal. 2011;55(2):265-271.
cholesterolemia, and improves lecithin: Cholesterol acyltransferase 45. Fernández-Poyatos MDP, Llorent-Martínez EJ, Ruiz-Medina A. Phy-
activity in rats fed enriched-cholesterol diet. Phytomedicine. 2014; tochemical composition and antioxidant activity of Portulaca oleracea:
21(12):1504-1508. Influence of the steaming cooking process. Foods. 2021;10(1):94.
26. Nazeam JA, El-Hefnawy HM, Omran G, Singab AN. Chemical profile 46. Yan J, Sun LR, Zhou ZY, et al. Homoisoflavonoids from the medicinal
and antihyperlipidemic effect of Portulaca oleracea L. seeds in plant Portulaca oleracea. Phytochemistry. 2012;80:37-41.
streptozotocin-induced diabetic rats. Nat Prod Res. 2018;32(12): 47. Yang Y, Zhao XJ, Pan Y, Zhou Z. Identification of the chemical compo-
1484-1488. sitions of ponkan peel by ultra performance liquid chromatography
14 ZHANG ET AL.

coupled with quadrupole time-of-flight mass spectrometry. Anal


Methods. 2016;8(4):893-903. How to cite this article: Zhang H, Chen G, Yang J, Yang C,
48. Farag MA, Shakour ZTA. Metabolomics driven analysis of 11 Portulaca
Guo M. Screening and characterisation of potential
leaf taxa as analysed via UPLC-ESI-MS/MS and chemometrics. Phyto-
chemistry. 2019;161:117-129.
antioxidant, hypoglycemic and hypolipidemic components
49. Selmar D, Irandoost Z, Wray V. Dhurrin-60 -glucoside, a cyanogenic revealed in Portulaca oleracea via multi-target affinity
diglucoside from Sorghum bicolor. Phytochemistry. 1996;43(3): ultrafiltration LC–MS and molecular docking. Phytochemical
569-572. Analysis. 2021;1-14. doi:10.1002/pca.3086
50. Sicari V, Loizzo MR, Tundis R, Mincione A, Pellicano TM. Portulaca
oleracea L. (purslane) extracts display antioxidant and hypoglycaemic
effects. J Appl Bot Food Qual. 2018;91:39-46.

You might also like