Download as pdf or txt
Download as pdf or txt
You are on page 1of 10

Journal of Functional Foods 106 (2023) 105601

Contents lists available at ScienceDirect

Journal of Functional Foods


journal homepage: www.elsevier.com/locate/jff

Aporphine alkaloids identified from Xylopia aethiopica and their potential


hypoglycemic and hypolipidemic activities
Ye Liu a, Yawen Li a, b, Felix Wambua Muema a, b, Hui Zhang a, b, Armel Jackson Seukep a, c,
Mingquan Guo a, b, *, 1
a
CAS Key Laboratory of Plant Germplasm Enhancement and Specialty Agriculture, Wuhan Botanical Garden, Chinese Academy of Sciences, Wuhan 430074, China
b
University of Chinese Academy of Sciences, Beijing 100049, China
c
Department of Biomedical Sciences, Faculty of Health Sciences, University of Buea, P.O Box 63, Buea, Cameroon

A R T I C L E I N F O A B S T R A C T

Keywords: Xylopia aethiopica has been widely utilized as food condiments in West African countries, as well as traditional
Xylopia aethiopica medicine for diabetes, but the responsible components remain unknown. In this work, the probable active
Aporphine alkaloids components in the contributing fraction were screened using bio-affinity ultrafiltration strategy targeting
Bio-affinity ultrafiltration
α-glucosidase and pancreatic lipase. Then, 11 alkaloids were identified, 9 of which were aporphines, including a
Hypoglycemic
new one. Further enzyme inhibitory validation of these compounds discovered that compounds 1, 2, 6 and 11
Hypolipidemic
showed better α-glucosidase inhibition activity than positive control, among which compounds 1, 2 and 6
simultaneously possessed potent inhibition against pancreatic lipase. The hypoglycemic and hypolipidemic ca­
pacities of these screened compounds were verified by in vitro glucose consumption and lipid accumulation
HepG2 cell models. This work provided solid evidences for the anti-diabetic benefit of X. aethiopica, and several
aporphine alkaloids could be the responsible compounds for its promising hypoglycemic and hypolipidemic
activities.

1. Introduction digestion (Akolade et al., 2018), and ameliorate the oxidative stress in a
type 2 diabetes mellitus (T2DM) rat model (Mohammed and Islam,
Xylopia aethiopica, belonging to the family Annonaceae, mainly dis­ 2017). Meanwhile, it was found to be able to decrease the lipids level
tributes in the Savanna region of Africa (Yin et al., 2019). The fruits of and produce a favourable serum lipid profile to exert hypolipidemic
X. aethiopica, known as Ethiopian pepper, are widely used as a food effect (Adefegha et al., 2018); (Nwozo et al., 2011). However, most anti-
condiment in many local dishes, and is served as a pepper substitute diabetes research focuses on the X. aethiopica fruit extracts’ activity
(Adefegha et al., 2018). In addition to its application in garnishing the screening, while the exact functional components remain unknown.
taste of foods, X. aethiopica is also traditionally used as a medicinal agent It is well known that T2DM, a chronic metabolic disease character­
for the treatment of malaria, fungal infections, uterine fibroid, female ized by persistent hyperglycemia, remains a major public health prob­
infertility, and so on, which also have been proven by modern phar­ lem due to its wide damage to various organ systems leading to severe
macological study (Erhirhie and Moke, 2014); (Yin et al., 2019). Defi­ diabetes complications (Olokoba et al., 2012). The mechanism of T2DM
nitely, the hypoglycemic benefits of X. aethiopica fruits have been has been recognized as insulin secretion dysfunction and multiple organ
verified traditionally by its application alone or within poly-herbal insulin resistance, which lead to a series of pathological manifestations,
formulation in the treatment of diabetes (Diallo et al., 2012); (Ogbon­ such as hyperglycemia and dyslipidemia (Galicia-Garcia et al., 2020).
nia et al., 2010). Emerging pharmacological research also disclosed that Currently, many oral medications have been approved to manage the
the X. aethiopica fruits extracts could inhibit the glucose synthesis disease situation, but diet and exercise remain basic effective means for
related enzymes (Adeyeoluwa et al., 2020), delay carbohydrate diabetes control (Marin-Penalver et al., 2016). Hence, more and more

* Corresponding author at: Cixi Institute of Biomedical Engineering, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences,
Ningbo 315201, China.
E-mail address: guomingquan@nimte.ac.cn (M. Guo).
1
Current address: Cixi Institute of Biomedical Engineering, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo
315201, China.

https://doi.org/10.1016/j.jff.2023.105601
Received 20 March 2023; Received in revised form 17 May 2023; Accepted 28 May 2023
Available online 2 June 2023
1756-4646/© 2023 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/).
Y. Liu et al. Journal of Functional Foods 106 (2023) 105601

scientific researchers set their sights on the anti-diabetes natural herbs 2.3. Extraction and segmentation
or edible plants to search for natural effective agents, which could have
more possibility, and security using as drugs or dietary supplements (Xu The dried X. aethiopica fruits (5.2 kg) were shredded, and extracted in
et al., 2018). 75% methanol (25 L per time) by the multifunctional extraction appa­
Family Annonaceae is one of the main resources of aporphine alka­ ratus (YC-050, Shanghai Pilotech instrument and equipment Co., Ltd.,
loids, characterized by a tetracyclic biphenyl structure, which belongs to Shanghai, China) at 50 ◦ C (60 min per time) under reduced pressure.
isoquinoline alkaloids (Xin et al., 2018). The evidences for aporphine After being suspended in water, the concentrated extract was sequen­
alkaloids’ important role in the improvement of diabetes have been tially fractionated with petroleum ether, dichloromethane, and ethyl
accumulated a lot in the past. For example, the typical aporphine al­ acetate.
kaloids, nuciferine and pronuciferine derived from a functional food The dichloromethane fraction, with the proper polarity and amount,
Nelumbo nucifera, could ameliorate glucose and lipid metabolism (Ma was subjected to an atmospheric silica gel column eluting with a
et al., 2014), promote insulin secretion (Nguyen et al., 2012), and inhibit gradient of petroleum ether and dichloromethane from 1:0 to 0:1 to
the activity of α-glucosidase hence preventing diabetes (H. (Zhang et al., obtain eight fractions A − H. Fraction F was then sequentially parti­
2022). The phytochemical studies on X. aethiopica have revealed that tioned by the medium pressure liquid chromatography (MPLC, Shanghai
this plant is rich in terpenoids, especially the volatile class components Tauto Biotech Co., Ltd., TBP 5010, ODS C18, 50 µm), eluted with a
in the essential oil, which contributes to making food attractive and gradient of MeOH − H2O from 40% to 100% to afford three sub-
palatable (Obiloma et al., 2019). Most research interests in this plant lay fractions F1 − F3 (XA-F1 − XA-F3).
in the functions and components of fruits’ essential oil, while those
intended to the anti-diabetes activity were limited to aqueous fruit or 2.4. In vitro α-glucosidase inhibition assay
defatted extract (Mohammed and Islam, 2017); (Okwari et al., 2010).
Combined with our primary screening results, the primary segmentation The α-glucosidase inhibition ability was evaluated according to a
and activity screening disclosed the active fraction with the most po­ previous method reported by Hamid and coworkers with minor modi­
tential hypoglycemic and hypolipidemic activities might be attributed to fications (Hamid et al., 2015). Briefly, 40 µL phosphate buffer saline
the enrichment of aporphine alkaloids. Therefore, the biologically active (PBS) at pH 6.8, 25 µL α-glucosidase solution in PBS (0.2 U/mL) and 10
compounds in this functional food were subsequently subjected to ul­ µL test samples were mixed in a 96-well plate and incubated for 15 min
trafiltration with α-glucosidase and pancreatic lipase followed by the at 37 ◦ C. Then, the addition of 25 µL p-NPG (0.5 mM) initiated the re­
phytochemical isolation and identification. At last, and the biological action followed by further incubation (15 min) at 37 ◦ C. Finally, the
activities of these identified compounds were further verified by enzyme addition of 100 µL Na2CO3 solution (0.1 M) quenched the reaction, and
inhibition, HepG2 cell glucose consumption and triglyceride (TG) the absorbance was taken on a Tecan Infinite M200 PRO Microplate
lowering tests to explore the most potent compounds with hypoglycemic Reader (Männedorf, Switzerland) at 405 nm. As a positive check, acar­
and hypolipidemic activities, which revealed the chemical bases un­ bose was utilized. Four groups were set as a blank control (without
derlying its traditional use against diabetes. enzyme and sample), sample control (with sample only), 100% enzyme
activity control (with enzyme only) and test sample groups (with sample
2. Materials and methods and enzyme) in all the tests. The following computational formula was
used for α-glucosidase inhibition ability evaluation: inhibition rate % =
2.1. Plant material (ΔAc − ΔAs)/ΔAc × 100%, where the ΔAc means the absorbance differ­
ence value between 100% enzyme activity control and blank control
The air-dried fruits of X. aethiopica were collected from Cameroon by groups and the ΔAs means the absorbance difference value between test
Doctor Armel Jackson Seukep. The specimens (voucher specimen No. sample and sample control groups. All of the tests were conducted in
20200901) were deposited in Plant Chemical biology laboratory of triplicate.
Wuhan Botanical Garden, Chinese Academy of Sciences, and authenti­
cated by a senior taxonomist Professor Guangwan Hu from the same 2.5. In vitro pancreatic lipase inhibition assay
institution.
The pancreatic lipase inhibition ability test was carried out referring
2.2. Chemicals and reagents to a previously reported method (H. (Zhang et al., 2022). In brief, the
reaction mixture, which was composed of 90 µL PBS (pH 7.4), 60 µL
The α-glucosidase and its reaction substrate p-Nitrophenyl α-D-glu­ pancreatic lipase in PBS (1.0 mg/mL), and 10 µL test samples, was added
copyranoside (p-NPG) were purchased from Aladdin Biochemical in a 96-well plate, afterward incubated for 10 min (37 ◦ C). Subsequently,
Technology Company (Shanghai, China), p-Nitrophenyl palmitate (p- 40 µL substrate p-NPP (2.5 mM) for the enzyme was added to start the
NPP) and orlistat were bought in Macklin Biochemical Company reaction at 37 ◦ C, and the absorbance was read on a microplate reader at
(Shanghai, China), acarbose and porcine pancreatic lipase and were 405 nm after 20 min incubation. Orlistat served as the positive control.
bought from Yuanye Bio-Technology Company (Shanghai, China). The The groups setting and inhibition rate calculation methods were the
organic reagents in analytical grade used in the isolation work, Na2CO3 same as the α-glucosidase inhibition assay section and all the tests were
and other inorganic chemicals used in buffer solution preparation were performed in triplicate.
obtained from Sinopharm Chemical Reagent Company (Shanghai,
China), while the solvents (methanol and acetonitrile) in HPLC grade 2.6. Screening for potential ligands by ultrafiltration and HPLC-UV
were bought from TEDIA Company (Fairfield, Ohio, USA). The water analysis
used for HPLC was prepared by Millipore Ultrapure Water System
(Billerica, Massachusetts, USA). Various chromatographic fillers, such as The procedure for ultrafiltration (UF) screening referred to the
200–300 mesh of silica gel from Qingdao Marine Chemical Company method in the literature with appropriate adjustment (Chen et al.,
(Qingdao, Shandong, China), Sephadex LH-20 gel (GE Healthcare, 2020). In short, 100 µL XA-F2 (F2 fraction of dichloromethane extract
Sweden), 50 µm size of ODS gel from YMC Co., Ltd. (Kyoto, Japan), were from X. aethiopica fruits) solution (2.0 mg/mL in PBS) was pre-incubated
used for compounds separation and purification. for 30 min with α-glucosidase (4 U in 100 µL pH 6.8 PBS) or pancreatic
lipase (0.2 mg in 100 µL pH 7.4 PBS) enzyme solution at 37 ◦ C,
respectively. In the meantime, the sample with inactivated α-glucosi­
dase or pancreatic lipase, which had been boiled in water for 15 min,

2
Y. Liu et al. Journal of Functional Foods 106 (2023) 105601

parallelly performed as the negative control. Then, the combinations supernatant was changed to culture medium (DMEM added with 10%
were moved to the 0.5 mL ultrafiltration tubes (Millipore) with 30 kD fetal bovine serum and 1% penicillin–streptomycin mixture solution,
membrane size (α-glucosidase) or 10 kD (pancreatic lipase) then Hyclone, UT, USA) with compound solutions in the final concentration
centrifugation for 10 min (10000 rpm). Afterward, the filtrate was dis­ of 50 and 100 µM. 24 h later, the cell supernatant was discarded and
carded, while the residuum on the membrane was washed and centri­ DMEM with 10% CCK8 reagent (Beyotime, Shanghai, China) was added,
fuged by 200 µL PBS three times to eliminate the uncombined or and then the absorbance was measured at 450 nm after keeping out of
nonspecific binding compounds. Next, MeOH (200 µL) was added and the light for 1 h.
incubated for 10 min to release the binding ligands for these two en­
zymes. After three times repetitions and centrifugation at 10000 rpm 2.9. The glucose consumption capacity test
(10 min), these filtrates were dried and dissolved in 50 µL MeOH for the
HPLC-UV analysis. The glucose consumption capacity was evaluated according to the
The UF sample analysis was implemented on an Agilent 1220 HPLC reported method (H. (Zhang et al., 2022). Briefly, 100 µL cell suspension
system fitted with a Waters column (C18, 3.5 µm, 150 × 4.6 mm, Mil­ (8 × 103 cells) were seeded in 96-well plate for adherence for 12 h, then
ford, Massachusetts, USA) and 280 nm was chosen as the UV wave­ the cell supernatant was replaced by DMEM without serum to starve the
length. 10 µL of the prepared sample was injected and eluted by the H2O cells for another 12 h. Afterwards, the cell supernatant was discarded
(A) containing 0.1% (v/v) formic acid and ACN (B) as follows: 20%− and low glucose DMEM medium with sample solution was added. After
45% B in 0–15 min, 45%− 60% B in15 − 35 min, 60%− 85% B in 35–45 incubation for 24 h, the supernatant was taken for centrifuging for 10
min with the flow rate of 0.8 mL/min. min at 1000 rpm, then the upper layer was used for glucose content test
by glucose determination kit (Nanjing Jiancheng Bioengineering Insti­
2.7. Isolation and identification tute, China) at 505 nm. The metformin or medium without sample were
used as positive or negative control, respectively.
Based on the previous enzyme inhibition screening results, the XA-F2
fraction was subjected to column chromatography (Sephadex LH-20) 2.10. TG content determination in OA/PA induced HepG2 lipid
washing by MeOH to divide into five sub-fractions F21 − F25 according accumulation model
to the LC analysis under the UF-LC conditions as mentioned above. The
separation conditions of target compounds screened from UF-LC were HepG2 cell suspension was seeded in 12-well plate (1 × 105cells/1
purposefully determined by comparing them with UF-LC chromatogram mL), and the culture medium with sample solution and 0.125 mM free
under the same analytical conditions. Based on this tracking method, F21 fatty acid composed of oleic acid (OA) and palmitic acid (PA) in a ratio
was put through the semi-preparative column (PolyPak, 5 µm C18, 250 of 2:1 was added to replace the primary one 12 h later. After 24 h in­
× 10 mm, Zhejiang Fuli Analytical Instruments Co., Ltd., Zhejiang, cubation, the cells were digested by 0.25% trypsin (Hyclone, UT, USA)
China, flow rate: 3.0 mL/min) on a Qingbohua P2050 system (Beijing and centrifuged for 10 min at 1000 rpm. Then the cell sediment was
Qingbohua Science and Technology company, China) eluting with 55% lysed by radio-immunoprecipitation assay (RIPA) lysis buffer (Beyotime,
MeOH − H2O to obtain compounds 1 (15.4 mg), 2 (9.2 mg) and 11 (1.4 Shanghai, China) for 1 h and the supernatant was used for triglyceride
mg), and collected impure peak 2 was further purified on an Agilent (TG) content determination using TG content assay kit (Nanjing Jian­
1100 machine (Santa Clara, California, USA) with a YMC-Pack column cheng Bioengineering Institute, China). The remaining supernatant was
(C18, 250 × 10 mm, 5 µm, YMC, Tokyo, Japan), eluting with 30% ACN centrifuged by 12000 rpm for 15 min at 4 ◦ C to obtain the upper layer for
− H2O supplied with 0.1% FA (2.0 mL/min) to obtain compound 8 (2.4 protein content assay by bicinchoninic acid (BCA) method (Beyotime,
mg). The F22 sub-fraction was segmented by semi-preparative HPLC Shanghai, China). The TG content was expressed as the content of tri­
(58% MeOH − H2O, 3.0 mL/min) and subsequently purified on the glyceride per gram of protein and orlistat was used as positive control.
Agilent 1100 system (25% ACN − H2O with 0.1% FA, 2.0 mL/min) to get
compounds 3 (3.9 mg) and 5 (2.4 mg). Then F23 was chromatographed 2.11. Statistical analyses
over the semi-preparative HPLC system with an elution of 45% MeOH −
H2O (3.0 mL/min) to get compound 4 (2.0 mg). Fraction F24 was All activities tests were carried out at least three times and were
separated by the HPLC with elution of 60% MeOH − H2O (flow rate: 3.0 manifested as means ± standard deviation (SD). The IC50 values were
mL/min) to yield compounds 6 (4.5 mg) and 10 (3.5 mg). Subraction F25 calculated by the log (inhibitor) vs. nor-malized response–variable slope
was chromatographed successively on Qingbohua P2050 system (50% method in GraphPad Prism 5 software. The statistically significant dif­
MeOH − H2O, 3.0 mL/min) and Agilent 1100 system (30% ACN − H2O ference were analyzed by one-way ANOVA multiple comparisons of
with 0.1% FA, 2.0 mL/min) to yield compounds 7 (1.5 mg) and 9 (2.0 Duncan test at the significance level of p < 0.05 (n = 3) in SPSS Statistics
mg). 26.0 software or one-way ANOVA Newman-Keuls multiple comparison
The chemical structures of the above isolated compounds were test by GraphPad Prism 5 software. In addition, the NMR data were
identified by their 1D (1H NMR, 13C NMR, and DEPT 135 spectra) and analyzed by MestReNova, the chemical structures were drawn by
2D NMR (1H–1H COSY, HMBC, HSQC, and NOESY spectra), which were ChemDraw 14.0, and the ultrafiltration-HPLC chromatograms were
analyzed by a 600 MHz NMR spectrometer (Bruker Avance, Karlsruhe, drawn by OriginPro 9.0 software, respectively.
Germany). The HR-ESI-MS data were acquired on an Aglient 1290 in­
finity II 6530C Q-TOF mass spectrometer (Santa Clara, CA, USA) using a 3. Results and discussion
Waters ACQUITY C18 UPLC column (1.7 µm, Milford, MA, USA) under
the UF-LC chromatographic condition as mentioned before. The optical 3.1. The inhibition effects of XA-F2 fraction on α-glucosidase and
rotations were carried out on a polarimeter and the UV spectra were pancreatic lipase
tested by a Lambda 35 UV spectrometer (PerkinElmer, Waltham, Mas­
sachusetts, USA). α-Glucosidase is a carbohydrate hydrolytic enzyme presented in the
small intestine devoted to hydrolyzing poly- or oligosaccharides to
2.8. Cytotoxicity assessment of selected compounds on HepG2 cells monosaccharides, which results in the increase of blood glucose con­
centration (Papoutsis et al., 2021). The inhibitors against α-glucosidase
Before the further cellular experiments, the cytotoxicity of these could reduce the speed of digestion and absorption of carbohydrates to
compounds on HepG2 cells was assessed in advance. 100 µL cell sus­ maintain the postprandial blood glucose at a relatively lower level
pension (1 × 104 cells) were seeded in 96-well plate, 12 h later, the cell (Abbas et al., 2017). Pancreatic lipase is one of the most important

3
Y. Liu et al. Journal of Functional Foods 106 (2023) 105601

enzymes in the lipolysis procedure of dietary lipids to release free fatty compound complexes, which were subsequently intercepted by an ul­
acid, the excessive accumulation of which leads to dyslipidemia and trafiltration membrane with a certain pore size. The complexes were
obesity, the pathologic abnormality and major risk factor of T2DM unbound by incubating with methanol and the released ligands were
(Rajan et al., 2020). Reducing lipid absorption by inhibiting lipases is an then analyzed by HPLC.
efficient approach to the development of hypolipidemic drugs. Accord­ Accordingly, the XA-F2 original sample solution and compounds
ing to the above understanding, α-glucosidase and pancreatic lipase released from the binding complex produced by ligands and active or
enzymes have been regarded as therapeutic targets for diabetes man­ denatured enzymes were analyzed by HPLC, respectively. As shown in
agement, and the natural agents with inhibition effects on these two Fig. 2, eleven peaks, speculated as alkaloids based on their molecular
enzymes also have attracted attention of researchers. weight preliminarily (Table 1), were listed as the main ingredients of
In this study, the aqueous methanol extract was successively defatted XA-F2, and seven of them were screened out in the α-glucosidase based
through partitioned extraction by organic solvents with different po­ bio-affinity ultrafiltration, while six of them appeared in the pancreatic
larities and normal phase silica gel column chromatography to remove a lipase experiment. The relative affinity abilities between ligands
large amount of oil. Therein, the sub-fraction F was further chromato­ screened out and enzymes were described by the enrichment factor (EF)
graphed, and three fractions XA-F1 − XA-F3 were obtained. Considering value (Liu et al., 2021), which was defined as EF (%) = (As – Ac)/A0 ×
the crucial role of α-glucosidase and pancreatic lipase played in the 100%, wherein As, Ac and A0 represent the peak areas acquired from the
development of diabetes, the inhibiting activities of these fractions were incubation of XA-F2 solution with activated, denatured and without
evaluated and compared. tested enzyme, respectively. Accordingly, the EF values of these seven
As shown in Fig. 1, the XA-F2 fraction exhibited promising dose- peaks screened out for α-glucosidase were calculated and listed in
dependent inhibitory effects on α-glucosidase (IC50 141.5 ± 10.8 μg/ Table 1, among which peak 11 possessed the highest EF value of 6.07%,
mL) (Fig. 1a), which was lower than acarbose (196.4 ± 27.6 μg/mL), a indicating its best binding affinity with α-glucosidase, followed by peak
glucosidase inhibitor severed as the positive control, while the other two 2 (2.46%), peak 6 (2.39%) and peak 1 (2.20%). As for pancreatic lipase
fractions did not show any activities. Meanwhile, this fraction also (Table 1), six peaks were screened out and the best three ones possessed
showed best inhibition effect on pancreatic lipase with the IC50 value of close EF values displaying as 3.92% for peak 2, 3.63% for peak 1 and
204.7 ± 21.0 μg/mL (Fig. 1b) compared with XA-F1 fraction (IC50 value: 3.40% for peak 6, followed by peak 3 (2.43%) and peak 8 (2.39%). To
362.8 ± 73.4 μg/mL) and XA-F3 fraction (the inhibition rate was 40.4% some extent, the EF values reflected the affinity capacities of these
at maximum detected concentration 500 μg/mL), along with the posi­ compounds with target enzymes, the differences between which might
tive control possessing 0.048 ± 0.006 μg/mL. Similar screening has be attributed to the competitive interactions of these active ingredients
done to compare the α-glucosidase inhibition activity of X. aethiopica with enzymes (Chen et al., 2019), and the experimental verifications
extracts obtained by different extraction methods (Adeyeoluwa et al., were quite essential to reveal the exact inhibitory abilities of these li­
2020), while the best result was from the aqueous ethanol extract with gands against α-glucosidase and pancreatic lipase.
the IC50 value of 559.9 ± 44.9 μg/mL, which was much higher than that
of XA-F2. The above evidences indicated that the chromatographed
3.3. UF-HPLC guided isolation and structure elucidation of enzyme
enrichment method removed the inactive or less active substances suc­
ligands
cessfully, and the XA-F2 fraction might be the target functional fraction
to find effective enzyme ligands for the α-glucosidase and pancreatic
Guided by the UF-HPLC results, the XA-F2 fraction was segmented
lipase.
and purified by layers of progressive chromatography to obtain 11 al­
kaloids (1–11), corresponding to the UF-HPLC-UV results displayed in
3.2. Screening for the potential α-glucosidase and pancreatic lipase Table 1 and Fig. 2. The structures of these alkaloids were mainly
ligands from XA-F2 fraction by ultrafiltration determined by comprehensive 1D and 2D NMR analyses with the aid of
other spectral analytical methods. To the best of our knowledge, com­
In consideration of the simultaneous inhibitory effects on α-gluco­ pound 6 was an undescribed structure first found in this work, com­
sidase and pancreatic lipase, the XA-F2 fraction was subjected to rapid pounds 2, 4, 5 and 7–9 were first isolated from Xylopia genus, and
and preliminary screening for potential enzyme ligands by affinity compounds 10–11 were isolated from X. aethiopica for the first time,
ultrafiltration-HPLC method. Affinity ultrafiltration is a membrane which have been found in X. lemurica and X. ferruginea, respectively
technique with a certain molecular weight cut-off to separate target- (Nieto et al., 1975); Nik Abdullah (Zawawi and N. K., Ahmat, N.,
ligand complexes based on the interaction between small molecules Ahmad, R., Jaafar, F. M., & Ab Ghani, N, 2012), while compounds 1 and
and biomacromolecules. And it is an efficient tool for high throughput 3 have already been reported in X. aethiopica (Harrigan et al., 1994). The
screening bioactive components from complex mixtures (Wei et al., brief structure elucidation of compounds 6–8 was shown as follows.
2016). In this work, the XA-F2 fraction was incubated with the active Compound 6 was isolated as a yellowish-brown solid and its mo­
and denatured α-glucosidase or pancreatic lipase to generate enzyme- lecular formula was determined as C20H21NO5 according to the 13C NMR

Fig. 1. The inhibiting effects of XA-F2 fraction on α-glucosidase (a) or pancreatic lipase (b).

4
Y. Liu et al. Journal of Functional Foods 106 (2023) 105601

Fig. 2. The ultrafiltration-HPLC chromatograms of XA-F2 fraction without ultrafiltration (black line), ultrafiltration with activated (red line), and inactivated (blue
line) α-glucosidase or pancreatic lipase, respectively. (For interpretation of the references to colour in this figure legend, the reader is referred to the web version of
this article.)

Table 1
Identification of potential α-glucosidase or pancreatic lipase ligands in the XA-F2 fraction.
Peak No.a Rt (min) EF (%)b for α-glucosidase EF (%)b for pancreatic lipase [M + H]+ (m/z) Compound namec

1 13.85 2.20 3.63 276.0669 Liriodenine


2 16.12 2.46 3.92 336.0881 Oxonantenine
3 17.56 0.91 2.43 292.0978 Lysicamine
4 17.80 – – 256.1357 N-benzoyl-L-phenylalaninol
5 18.20 – – 308.0940 Peruvianine
6 18.40 2.39 3.40 356.1487 N-formyllaurotetanine
7 18.80 – – 370.1671 N-Acetyllaurotetanine
8 18.99 1.65 2.39 354.1327 N-Acetylnordomesticine
9 19.38 1.14 0.76 268.1351 N-trans-cinnamoyltyramine
10 19.92 – – 306.0753 Lanuginosine
11 20.30 6.07 – 306.0771 Atherospermidine
a
The peak No. and Retention time was corresponding to LC results.
b
Enrichment factor.
c
Identified by comparing with the corresponding isolated pure compounds.

data and HR-ESI-MS peak at m/z 733.2733 [2 M + Na]+ (calcd for Hz, H-5) and 3.28 (1H, m, H-5); 2.73 (1H, dd, J = 13.6, 4.3 Hz, H-7) and
733.2738). The signals in the 1H NMR spectrum (Table 2 and Fig. S2) 2.61 (1H, t, J = 13.6 Hz, H-7)], three methoxy groups at δH 3.81 (3H, s,
were attributed to an aldehyde hydrogen at δH 8.21, three aromatic 2-OCH3), 3.79 (3H, s, 10-OCH3), 3.60 (3H, s, 1-OCH3), with the assist of
singlet hydrogens at δH 7.93 (1H, s, H-11), 6.80 (1H, s, H-3) and 6.70 HSQC signals (Fig. S5). The 13C NMR spectrum (Table 2 and Fig. S3),
(1H, s, H-8), one methine at δH 4.60 (1H, dd, J = 13.6, 4.3 Hz, H-6), three with the aid of the DEPT 135 spectrum (Fig. S4), showed twenty carbons
methylene groups [δH 2.77 (2H, m, H-4); 3.91 (1H, 1H, dd, J = 12.8, 2.3 consisting of a N-formyl carbon at δC 162.3, twelve aromatic carbons

5
Y. Liu et al. Journal of Functional Foods 106 (2023) 105601

Table 2
1
H NMR and 13C NMR spectroscopic data for the major isomers of compounds 6-8.a
6a 7a 8a

position δH, (J in Hz) δC, type δH, (J in Hz) δC, type δH, (J in Hz) δC, type

1 144.2, C 146.1, C 150.7, C


1a 127.0, C 128.9, C 128.4, C
2 151.9, C 153.6, C 145.2, C
3 6.80, s 111.2, CH 6.79, s 111.8, CH 6.66, s 115.7, CH
3a 129.3, C 130.9, C 131.2, C
4 2.77, m (overlap) 30.3, CH2 2.79, m 31.4, CH2 2.82, m (overlap) 31.1, CH2
2.74, m 2.69, m (overlap)
5 3.91, dd (12.8, 2.3) 41.0, CH2 4.13, dd (12.8, 2.3) 43.2, CH2 4.10, dd (12.9, 1.8) 43.3, CH2
3.28, m (overlap) 2.90, td (14.3, 12.8, 4.3) 3.28, dd (12.9, 2.4)
6 4.60, dd (13.6, 4.3) 48.8, CH 4.95, m (overlap) 52.3, CH 4.89, dd (13.6, 4.2) 52.2, CH
6a 123.8, C 126.3, C 125.3, C
7 2.73, dd (13.6, 4.3) 33.0, CH2 2.84, dd (13.5, 4.2) 34.3, CH2 2.88, dd (13.6, 4.2) 35.0, CH2
2.61, t (13.6) 2.68, t (13.5) 2.69, m (overlap)
7a 129.5, C 131.6, C 132.5, C
8 6.70, s 115.4, CH 6.73, s 116.2, CH 6.78, s 109.6, CH
9 146.3, C 147.6, C 148.2, C
10 146.1, C 147.4, C 148.2, C
11 7.93, s 112.4, CH 8.08, s 113.4, CH 7.93, s 109.2, CH
11a 122.1, C 124.3, C 126.4, C
N-CHO 8.21, s 162.3, C
N-COCH3 172.0, C 172.1, C
2.24, s 22.3, CH3 2.23, s 22.3, CH3
1-OCH3 3.60, s 59.3, CH3 3.66, s 60.2, CH3
2-OCH3 3.81, s 55.8, CH3 3.91, s 56.6, CH3 3.57, s 60.2, CH3
10-OCH3 3.79, s 55.7, CH3 3.89, s 56.4, CH3
9,10-OCH2O 5.98, s 102.4, CH2
a
Data (δ) were measured in DMSO‑d6 for 6, in CD3OD for 7 and 8 by 600 MHz for 1H NMR and 150 MHz for 13C NMR. The data listed here were belonged to the
major isomers 6a, 7a and 8a, and data for 6b, 7b and 8b could be found in Table S1 in supplementary materials.

contributing to two benzene rings (δC 111.2–151.9), one methine carbon quite similar to that of 6, and 7 was first and only mentioned in literature
signal at δC 48.8, three methylene (δC 41.0, 33.0, 30.3) and three reported in 1965 (Pachler et al., 1965) without any referable data, so
methoxys (δC 59.3, 55.8, 55.7), corresponding to the proton signals. The here was a supplement for its NMR data and isomer manner. The major
HMBC cross-peaks from H-3 to C-2 and C-4, H-4 to C-3a and C-6a, H-6 to structure of 7 was also laurotetanine, along with the extra signals of N-
C-1a, C-6a and N-CHO, and H-7 to C-7a and C-11a (Fig. 3 and Fig. S7), COCH3 at δH 2.24 (3H, s) and δC 172.0, 22.3, so the planar structure of 7
combined with the 1H− 1H COSY correlations of H-4/H-5 and H-6/H-7 was determined as N-acetyl substituted laurotetanine. The 1D NMR of 7
(Fig. 3 and Fig. S6), indicated that compound 6 possessed an aporphine displayed the same isomer manner as 6, with a pair of amide bond
alkaloid skeleton. The three methoxy substituents attached to the ben­ isomers with the ratio of 3:1. The data of 7a and 7b was shown in Table 2
zene rings were determined by the HMBC correlations from OCH3 to C-1, and Table S1, and compound 7 was accordingly named as N-
C-2 and C-10, respectively, while the remaining oxygenated aromatic acetyllaurotetanine.
carbon at δC 146.3 substituted by a hydroxyl was deduced by the mo­ Compound 8 could be found in the Scifinder database, but without
lecular weight of 6. The above characteristic data were quite similar to any data and reference, so here gave the brief structure elucidation and
those of the known compound laurotetanine (Costa et al., 2013) except data supplement for it. The 1D NMR data of compound 8 was similar to
for the extra signals at δH 8.21 and δC 162.3, which were disclosed that of 7, except for the absence of two methoxy groups and the emer­
belonging to the N-formyl group by further analysis of the HMBC gence of a methylenedioxy group at δH 5.98 (2H, s) and δC 102.4 (Table 2
spectrum of 6, which showed the correlations from N-formyl to C-5 and and Fig. S15–S16). The HMBC correlations (Fig. 3 and S18) of OCH3/C-2
C-6. From the above findings, the planar structure of 6 should be N- and OCH2O/C-9, C-10 revealed that the remaining methoxy at δH 3.57
formyl substituted laurotetanine. (3H, s) and δC 60.2 was attached to C-2 and the methylenedioxy group
Interestingly, with a deep-going analysis of the NMR data, compound was attached to C-9 and C-10. The substituent of hydroxyl at the
6 exhibited two complete sets of signals with an approximate ratio of 2:1 oxygenated C-1 (δC 150.7) was deduced by the molecular formula of 8,
calculated by the 1H NMR signal integrals. The major isomer was labeled which was determined by ion peak at m/z 729.2413 [2 M + Na]+ (calcd
as 6a, while the minor one was marked as 6b (Fig. 3), and in consid­ for 729.2424) in HR-ESI-MS and 13C NMR. Based on the above analysis,
eration of the similar profile of 6b (Table S1), the planar structure the planar structure of 8 was elucidated as N-acetyl substituted nordo­
elucidation above was based on the data of 6a. This phenomenon is mesticine (Indra et al., 2002). Similarly, the NMR data of 8 could be
attributed to the partial double-bond property of the amide bond (Wang separated into two sets with a ratio of 3:1 (Table 2 and Table S1), the
et al., 2020). In the NOESY spectrum (Fig. S8), the aldehyde hydrogen of isomer manner of which was the same as that of 6. Consequently,
6a (δH 8.21) correlated to H-5 (δH 3.91), while that of 6b (δH 8.34) compound 8 was named as N-Acetylnordomesticine.
correlated to H-6 (δH 4.48), which indicated that the amide bond of 6a The other 8 known compounds yielded from XA-F2 were identified
was Z configuration and 6b was E configuration (Wang et al., 2020). as liriodenine (1) (Z. Z. (Zhang et al., 2002), Oxonantenine (2) (Teles
Meanwhile, the upfield moving of H-6 from δH 4.60 (6a) to δH 4.48 (6b) et al., 2015), lysicamine (3) (Teles et al., 2015), N-benzoyl-L-phenyl­
and the downfield moving of CHO from δH 8.21 (6a) to δH 8.34 (6b) alaninol (4) (Barrow and Sun, 1994), peruvianine (5) (Menachery and
were also consisted with the manners of reported isomers (Wang et al., Cava, 1981), N-trans-cinnamoyltyramine (9) (Yang et al., 2002), lanu­
2020). In addition, the absolute configuration of C-6 was determined as ginosine (10) (Nakano et al., 2013), atherospermidine (11) (Costa et al.,
S due to the positive optical rotation value of 6 (Wang et al., 2020). 2011), respectively, based on the careful analysis of their NMR data and
Thus, compound 6 was named as N-formyllaurotetanine. comparison with the reported spectroscopic data.
The 1D NMR data of compound 7 (Table 2 and Fig. S11–S12) was Data for N-formyllaurotetanine (6): tawny solid, [α]20 D + 325 (c 0.4,

6
Y. Liu et al. Journal of Functional Foods 106 (2023) 105601

Fig. 3. (a) Alkaloids 1–11 isolated from X. aethiopica (*new compound). (b) Key 1H− 1H COSY and HMBC correlations of compounds 6 and 8. (b) Isomers of
compounds 6–8.

MeOH); UV (MeOH), see Fig. S9, λmax (log ε) 215 (4.69), 274 (3.99), 302 ligands screened out by UF-HPLC, the in vitro inhibitory effects on
(3.67) nm; data for 1H NMR (600 MHz) and 13C NMR (150 MHz) in
DMSO‑d6 were listed in Table 2 and Table S1.
Data for N-Acetyllaurotetanine (7): white power; [α]20 D + 307 (c 0.1, Table 3
The α-glucosidase or pancreatic lipase inhibition activities of compounds 1–11.
MeOH); UV (MeOH), see Fig. S13, λmax (log ε) 214 (4.57), 282 (4.03),
305 (3.89) nm; data for 1H NMR (600 MHz) and 13C NMR (150 MHz) in Compound No. α-glucosidase pancreatic lipase
CD3OD were listed in Table 2 and Table S1. IC50 ± SD (µM) IC50 ± SD (µM)
Data for N-Acetylnordomesticine (8): white power; [α]20 D + 330 (c
1 148.8 ± 7.3b 306.2 ± 17.5b
0.2, MeOH); UV (MeOH), see Fig. S19, λmax (log ε) 217 (4.67), 284 2 193.3 ± 8.2c 358.2 ± 83.7bc
(4.01), 310 (4.23) nm; data for 1H NMR (600 MHz) and 13C NMR (150 3 – 421.2 ± 90.4c
MHz) in CD3OD were listed in Table 2 and Table S1. 6 115.6 ± 22.6ab 294.2 ± 49.0b
11 89.1 ± 3.7a –
Positive control Acarbose Orlistat
3.4. The inhibition effects validation of the isolated ligands on 304.2 ± 42.8d 0.096 ± 0.013a
α-glucosidase and pancreatic lipase a–d
Means the significant difference between these compounds and positive
control by one-way ANOVA Duncan’s multiple range test (DMRT) at the level of
To further validate the actual enzyme inhibition potential of the p < 0.05 (n = 3).

7
Y. Liu et al. Journal of Functional Foods 106 (2023) 105601

α-glucosidase and pancreatic lipase were tested for all the isolated inhibitors screening efficiency of the ultrafiltration method from com­
compounds (1–11). As the results shown in Table 3, compounds 1, 2, 6 plex naturally derived samples and provided several potential aporphine
and 11 possessed significant α-glucosidase inhibition activities, all the alkaloids ligands for T2DM.
IC50 values of which were lower than that of the oral hypoglycemic
α-glucosidase inhibitor acarbose, and showed statistical difference at the 3.5. The active ligands promoted glucose consumption in HepG2 cells
level of p < 0.05, while other compounds didn’t exhibit more than 50%
inhibition rate at the maximum tested concentration. Among the iso­ The most direct manifestation of diabetes is hyperglycemia, which is
lated compounds, 1, 3 and 9 have been reported for their α-glucosidase closely associated with the deficiency of glucose consumption. Anti-
inhibition activities, but only 1 showed potent inhibitory activity diabetes agents take effects through different mechanisms of action,
(Hamid et al., 2015), which was identical to the screening results in this but most of them could affect glucose consumption, including insulin
work. For pancreatic lipase, compounds 1, 2, 3 and 6 were verified to and metformin (Shi et al., 2019). In this study, HepG2 based glucose
exhibit promising inhibition effects (Table 3), and all the compounds consumption assay was used to verify the in vitro hypoglycemic effect of
were first reported for their lipase inhibitory abilities. Although the IC50 α-glucosidase enzyme inhibitors screened out (compounds 1, 2, 6 and
values were much higher than the positive control orlistat, that also 11). Firstly, the cytotoxicity of these compounds were evaluated at the
made sense for natural unmodified molecules and possible simultaneous concentration of 50 and 100 μM (Fig. 4a), as a result, the cell viability of
inhibition of multiple T2DM targets. HepG2 treated with compounds 2, 6 and 11 were all over 90% in the two
Comparing the results of the activity validation with those of UF-LC tested concentrations, except that compound 1 showed some toxicity to
screening, we found that the peaks screened out with higher EF values in HepG2 cells at 100 μM (cell viability was lower than 80%). As shown in
the ultrafiltration experiment did have better inhibitory activities. Peak Fig. 4b, the glucose content in the groups treated with 100 μM of com­
11, which possessed the distinguishing highest EF value of 6.07%, pounds 2, 6 and 11 significantly increased glucose consumption, and
showed the lowest IC50 value of 89.1 ± 3.7 against α-glucosidase, fol­ compounds 6 and 11 showed greater efficacy than that of hypoglycemic
lowed by peaks 1, 2 and 6 with comparatively lower EF values and agent metformin with a much higher concentration of 1 mM. In
higher IC50 values, while peaks missing in the UF experiment or with consideration of the toxicity of compound 1, same work was also per­
lower EF values did not show any potential in the enzyme inhibition formed at 50 μM, but it turned out that only compound 11 exhibited an
assays. A roughly similar trend was also observed in the pancreatic effect of increasing glucose consumption (Fig. S20). In conclusion,
lipase verification assays, as expected, the best three compounds 1, 2 compounds 2, 6 and 11, which possessed better α-glucosidase inhibition
and 6 with relatively close EF values were revealed to have excellent capacity than positive control, were confirmed for their great potenti­
lipase inhibition capacities. The above results confirmed the enzyme ality in glucose-lowering effect.

Fig. 4. (a) The cytotoxicity of compounds on HepG2 cells (***means p < 0.001). (b) The effects of compounds 2, 6, 11 (100 μM) and positive control metformin (1
mM) on glucose consumption of HepG2 cells (***means p < 0.001 compared with the blank group). (c) The effects of compounds 1, 2, 3, 6 (50 μM) and positive
control metformin (20 μM) on TG production in OA/PA induced HepG2 lipid accumulation cell model (###means p < 0.001 compared with the blank group,
***means p < 0.001, **means p < 0.01 compared with the model group).

8
Y. Liu et al. Journal of Functional Foods 106 (2023) 105601

3.6. The active ligands reduced TG production in OA/PA induced HepG2 Data availability
lipid accumulation model
Data will be made available on request.
The elevation of free fatty acid level emerged in many metabolic
diseases, like obesity and diabetes, which will lead to insulin resistance Acknowledgments
in insulin target organs, including liver. The mixture of OA and PA
induced lipid accumulation HepG2 cell model was used to evaluate the This work was partly supported by the National Natural Science
lipid lowering effects of selected compounds. As shown in Fig. 4a, Foundation of China (Grant No. 32200324 to Y. Liu).
compounds 1, 2, 3 and 6, lipase inhibitors screened out in XA-F2,
exhibited no significant cytotoxicity on HepG2 cells at 50 μM, which Appendix A. Supplementary data
was consequently chosen as the screening concentration. In Fig. 4c, OA/
PA induced a marked growing level of TG content in the model group, Supplementary data to this article can be found online at https://doi.
which was significantly decreased by the treatment of compounds 1, 2, org/10.1016/j.jff.2023.105601.
3, 6 and positive control orlistat (20 μM). The above results confirmed
the lipid-lowering effects of these pancreatic lipase inhibitors screened References
out.
Abbas, G., Al-Harrasi, A. S., & Hussain, H. (2017). Chapter 9 - α-Glucosidase enzyme
inhibitors from natural products. In G. Brahmachari (Ed.), Discovery and Development
4. Conclusion of Antidiabetic Agents from Natural Products (pp. 251–269). Elsevier.
Adefegha, S. A., Oboh, G., Olasehinde, T. A., & Boligon, A. A. (2018). Dietary
Many studies have confirmed the effective role X. aethiopica played supplementation with Ethiopian pepper (Xylopia aethiopica) modulates angiotensin-I
converting enzyme activity, antioxidant status and extenuates hypercholesterolemia
in diabetes, but the responsible components remained unknown. Hence, in high cholesterol fed Wistar rats. PharmaNutrition, 6(1), 9–16. https://doi.org/
in this work, polarity-guided segmentation along with α-glucosidase and 10.1016/j.phanu.2017.11.001
pancreatic lipase-based enzyme inhibition screening ascertained the Adeyeoluwa, T. E., Balogun, F. O., & Ashafa, A. O. T. (2020). In vitro comparative
assessment of the inhibitory effects of single and combined spices against glucose-
active fraction XA-F2, which was further subjected to UF-LC, disclosing synthesizing enzymes. Tropical Journal of Pharmaceutical Research, 19, 1209–1214.
the excellent bio-affinity of aporphine alkaloids with α-glucosidase and https://doi.org/10.4314/tjpr.v19i6.14
pancreatic lipase. UF-LC guided phytochemical isolation led to the un­ Akolade, J., Na’Allah, A., Sulyman, A., Abdulazeez, A., Atoti, A., & Isiaku, M. (2018).
Antidiabetic screening of phenolic-rich extracts of selected medicinal spices. Iranian
equivocal identification of 11 alkaloids (1–11), including a new one (6),
Journal of Science and Technology, 43, 1–11. https://doi.org/10.1007/s40995-017-
among which compounds 1, 2, 6 and 11 exhibited potent α-glucosidase 0410-y
inhibitory activities better than acarbose, while compounds 1, 2, 3 and 6 Barrow, C. J., & Sun, H. H. (1994). Spiroquinazoline, a novel substance-P inhibitor with a
showed potential suppressing effects on pancreatic lipase. Meanwhile, new carbon skeleton, isolated from Aspergillus flavipes. Journal of Natural Products, 57
(4), 471–476. https://doi.org/10.1021/Np50106a005
their glucose consumption and lipid-lowering capacity were validated, Chen, G. L., Fan, M. X., Wu, J. L., Li, N., & Guo, M. Q. (2019). Antioxidant and anti-
and compounds 2 and 6 showed concurrently glucose consumption inflammatory properties of flavonoids from lotus plumule. Food Chemistry, 277,
increasing and TG production reducing capacity, which might be 706–712. https://doi.org/10.1016/j.foodchem.2018.11.040
Chen, G. L., Xu, Y. B., Wu, J. L., Li, N., & Guo, M. Q. (2020). Hypoglycemic and
promising anti-diabetes agents. hypolipidemic effects of Moringa oleifera leaves and their functional chemical
In conclusion, dietary food or condiments contains numerous sec­ constituents. Food Chemistry, 333, Article 127478. https://doi.org/10.1016/j.
ondary metabolites, which attributes to their health benefits beyond foodchem.2020.127478
Costa, E. V., Dutra, L. M., Nepel, A., & Barison, A. (2013). Isoquinoline alkaloids from the
their nutritional function, particularly in favor of those diseases related leaves of Xylopia laevigata (Annonaceae). Biochemical Systematics and Ecology, 51,
to metabolism, such as obesity and diabetes. This work supplied new 331–334. https://doi.org/10.1016/j.bse.2013.10.005
evidences to support the anti-diabetes traditional use of X. aethiopica, Costa, E. V., Pinheiro, M. L. B., de Souza, A. D. L., Barison, A., Campos, F. R.,
Valdez, R. H., … Nakamura, C. V. (2011). Trypanocidal activity of oxoaporphine and
and disclosed its responsible components. In addition, two promising pyrimidine-beta-carboline alkaloids from the branches of Annona foetida Mart.
candidate molecules, including an undescribed one, exhibited good (Annonaceae). Molecules, 16(11), 9714–9720. https://doi.org/10.3390/
potential to be further developed as new natural therapeutics for the molecules16119714
Diallo, A., Traore, M. S., Keita, S. M., Balde, M. A., Keita, A., Camara, M., … Balde, A. M.
treatment of diabetes and obesity through simultaneously inhibiting
(2012). Management of diabetes in Guinean traditional medicine: An ethnobotanical
α-glucosidase and pancreatic lipase, increasing glucose consumption investigation in the coastal lowlands. Journal of ethnopharmacology, 144(2),
and reducing TG production, which might take cognizance of aporphine 353–361. https://doi.org/10.1016/j.jep.2012.09.020
alkaloids for their hypoglycemic and hypolipidemic activity except for Erhirhie, E. O., & Moke, G. E. (2014). Xylopia aethiopica: A review of its ethnomedicinal,
chemical and pharmacological properties. American Journal of Pharmtech Research, 4,
the frequently studied nuciferine and pronuciferine. 22–37.
Galicia-Garcia, U., Benito-Vicente, A., Jebari, S., Larrea-Sebal, A., Siddiqi, H.,
5. Ethics statement Uribe, K. B., … Martin, C. (2020). Pathophysiology of type 2 diabetes mellitus.
International journal of molecular sciences, 21(17), 6275. https://doi.org/10.3390/
ijms21176275
No animal or human experiments are involved in this work. Hamid, H. A., Yusoff, M. M., Liu, M., & Karim, M. R. (2015). α-Glucosidase and α-amylase
inhibitory constituents of Tinospora crispa: Isolation and chemical profile
confirmation by ultra-high performance liquid chromatography-quadrupole time-of-
CRediT authorship contribution statement flight/mass spectrometry. Journal of Functional Foods, 16, 74–80. https://doi.org/
10.1016/j.jff.2015.04.011
Ye Liu: Data curation, Funding acquisition, Methodology, Software, Harrigan, G. G., Gunatilaka, A. A. L., David, G. I., Chan, G. W., & Johnson, R. K. (1994).
Isolation of bioactive and other oxoaporphine alkaloids from two annonaceous
Writing – original draft. Yawen Li: Methodology, Validation. Felix plants, Xylopia aethiopica and Miliusa cf. banacea. Journal of Natural Products, 57(1),
Wambua Muema: Methodology, Software. Hui Zhang: Methodology, 68–73. https://doi.org/10.1021/np50103a009
Validation. Armel Jackson Seukep: Investigation, Resources. Min­ Indra, B., Matsunaga, K., Hoshino, O., Suzuki, M., Ogasawara, H., & Ohizumi, Y. (2002).
Structure-activity relationship studies with (+/-)-nantenine derivatives for alpha(1)-
gquan Guo: Conceptualization, Funding acquisition, Project adminis­
adrenoceptor antagonist activity. European Journal of Pharmacology, 437(3),
tration, Supervision, Writing – review & editing. 173–178. https://doi.org/10.1016/S0014-2999(02)01303-1
Liu, Y., Muema, F. W., Zhang, Y. L., & Guo, M. Q. (2021). Acyl quinic acid derivatives
screened out from Carissa spinarum by SOD-affinity ultrafiltration LC-MS and their
Declaration of Competing Interest
antioxidative and hepatoprotective activities. Antioxidants, 10(8), 1302. https://doi.
org/10.3390/Antiox10081302
The authors declare that they have no known competing financial Ma, C., Wang, J., Chu, H., Zhang, X., Wang, Z., Wang, H., & Li, G. (2014). Purification
interests or personal relationships that could have appeared to influence and characterization of aporphine alkaloids from leaves of Nelumbo nucifera Gaertn

the work reported in this paper.

9
Y. Liu et al. Journal of Functional Foods 106 (2023) 105601

and their effects on glucose consumption in 3T3-L1 adipocytes. International journal Papoutsis, K., Zhang, J. Y., Bowyer, M. C., Brunton, N., Gibney, E. R., & Lyng, J. (2021).
of molecular sciences, 15(3), 3481–3494. https://doi.org/10.3390/ijms15033481 Fruit, vegetables, and mushrooms for the preparation of extracts with alpha-amylase
Marin-Penalver, J. J., Martin-Timon, I., Sevillano-Collantes, C., & Del Canizo- and alpha-glucosidase inhibition properties: A review. Food Chemistry, 338, Article
Gomez, F. J. (2016). Update on the treatment of type 2 diabetes mellitus. World 128119. https://doi.org/10.1016/j.foodchem.2020.128119
Journal of Diabetes, 7(17), 354–395. https://doi.org/10.4239/wjd.v7.i17.354 Rajan, L., Palaniswamy, D., & Mohankumar, S. K. (2020). Targeting obesity with plant-
Menachery, M. D., & Cava, M. P. (1981). The alkaloids of Telitoxicum peruvianum. Journal derived pancreatic lipase inhibitors: A comprehensive review. Pharmacological
of Natural Products, 44(3), 320–323. https://doi.org/10.1021/Np50015a014 Research, 155, Article 104681. https://doi.org/10.1016/J.Phrs.2020.104681
Mohammed, A., & Islam, M. S. (2017). Antioxidant potential of Xylopia aethiopica fruit Shi, L. L., Jia, W. H., Zhang, L., Xu, C. Y., Chen, X., Yin, L., … Guan, H. D. (2019). Glucose
acetone fraction in a type 2 diabetes model of rats. Biomedicine & Pharmacotherapy, consumption assay discovers coptisine with beneficial effect on diabetic mice.
96, 30–36. https://doi.org/10.1016/j.biopha.2017.09.116 European Journal of Pharmacology, 859, Article 172523. https://doi.org/10.1016/J.
Nakano, D., Ishitsuka, K., Kamikawa, M., Matsuda, M., Tsuchihashi, R., Okawa, M., … Ejphar.2019.172523
Kinjo, J. (2013). Screening of promising chemotherapeutic candidates from plants Teles, M. N. D., Dutra, L. M., Barison, A., & Costa, E. V. (2015). Alkaloids from leaves of
against human adult T-cell leukemia/lymphoma (III). Journal of Natural Medicines, Annona salzmannii and Annona vepretorum (Annonaceae). Biochemical Systematics and
67(4), 894–903. https://doi.org/10.1007/s11418-013-0747-2 Ecology, 61, 465–469. https://doi.org/10.1016/j.bse.2015.07.016
Nguyen, K. H., Ta, T. N., Pham, T. H., Nguyen, Q. T., Pham, H. D., Mishra, S., & Wang, R. Z., Zhou, J., Shi, G. R., Liu, Y. F., & Yu, D. Q. (2020). Aporphine and
Nyomba, B. L. (2012). Nuciferine stimulates insulin secretion from beta cells-an in phenanthrene alkaloids with antioxidant activity from the roots of Stephania
vitro comparison with glibenclamide. Journal of Ethnopharmacology, 142(2), tetrandra. Fitoterapia, 143, Article 104551. https://doi.org/10.1016/J.
488–495. https://doi.org/10.1016/j.jep.2012.05.024 Fitote.2020.104551
Nieto, M., Leboeuf, M., & Cave, A. (1975). Isolation of the lanuginosine from a Wei, H., Zhang, X., Tian, X., & Wu, G. (2016). Pharmaceutical applications of affinity-
Madagascan Annonaceae. Xylopia lemurica. Phytochemistry, 14(11), 2508–2509. ultrafiltration mass spectrometry: Recent advances and future prospects. Journal of
https://doi.org/10.1016/0031-9422(75)80378-5 Pharmaceutical and Biomedical Analysis, 131, 444–453. https://doi.org/10.1016/j.
Zawawi, N. A., & N. K., Ahmat, N., Ahmad, R., Jaafar, F. M., & Ab Ghani, N. (2012). jpba.2016.09.021
Oxoaporphine alkaloids and flavonols from Xylopia ferruginea (Annonaceae). Xin, A., Liu, J., & Di, D. (2018). Research progress on aporphine alkaloids. Chinese
Biochemical Systematics and Ecology, 43, 7–9. https://doi.org/10.1016/j. Traditional and Herbal Drugs, 49(3), 712–724. https://doi.org/10.7501/j.issn.0253-
bse.2012.02.019 2670.2018.03.030
Nwozo, S. O., Orojobi, B. F., & Adaramoye, O. A. (2011). Hypolipidemic and antioxidant Xu, L., Li, Y., Dai, Y., & Peng, J. (2018). Natural products for the treatment of type 2
potentials of Xylopia aethiopica seed extract in hypercholesterolemic rats. Journal of diabetes mellitus: Pharmacology and mechanisms. Pharmacological Research, 130,
Medicinal Food, 14, 114–119. https://doi.org/10.1089/jmf.2008.0168 451–465. https://doi.org/10.1016/j.phrs.2018.01.015
Obiloma, A. A., Madu, W. C., Osuji, G. O., Ampim, P. A., Weerasooriya, A. D., & Yang, G., Zhao, S., & Li, Y. (2002). Studies on the chemical constituents of Lycianthes
Carson, L. E. (2019). Terpene-rich medicinal plant spices for flavoring of processed biflora. Acta Pharmaceutica Sinica, 37(6), 437–439. https://doi.org/10.16438/j.05
tropical food. American Journal of Plant Sciences, 10(4), 572–577. https://doi.org/ 13-4870.2002.06.010.
10.4236/ajps.2019.104041 Yin, X., Chávez León, M. A. S. C., Osae, R., Linus, L. O., Qi, L.-W., & Alolga, R. N. (2019).
Ogbonnia, S. O., Mbaka, G. O., Adekunle, A., Anyika, E. N., Gbolade, O. E., & Xylopia aethiopica seeds from two countries in west africa exhibit differences in their
Nwakakwa, N. (2010). Effect of a poly-herbal formulation, Okudiabet, on alloxan- proteomes, mineral content and bioactive phytochemical composition. Molecules, 24
induced diabetic rats. Agriculture Biology Journal North America, 1(2), 139–145. http (10), 1979. https://doi.org/10.3390/molecules24101979
s://doi.org/10.13140/RG.2.2.33629.72167. Zhang, H., Chen, G., Zhang, Y., Yang, M., Chen, J., & Guo, M. (2022). Potential
Okwari, O. O., Nneli, R. O., & Osim, E. E. (2010). Effect of aqueous fruit extract of Xylopia hypoglycemic, hypolipidemic, and anti-inflammatory bioactive components in
aethiopica on intestinal fluid and glucose transfer in rats. Nigerian journal of Nelumbo nucifera leaves explored by bioaffinity ultrafiltration with multiple targets.
physiological sciences, 25(2), 181–186. Food Chemistry, 375, Article 131856. https://doi.org/10.1016/j.
Olokoba, A. B., Obateru, O. A., & Olokoba, L. B. (2012). Type 2 diabetes mellitus: A foodchem.2021.131856
review of current trends. Oman Medical Journal, 27(4), 269–273. https://doi.org/ Zhang, Z. Z., ElSohly, H. N., Jacob, M. R., Pasco, D. S., Walker, L. A., & Clark, A. M.
10.5001/omj.2012.68 (2002). New sesquiterpenoids from the root of Guatteria multivenia. Journal of
Pachler, K. G. R., Arndt, R. R., & Baarschers, W. H. (1965). Nuclear magnetic resonance Natural Products, 65(6), 856–859. https://doi.org/10.1021/np0200717
of aporphine alkaloids. II. Structure of rogersine. Tetrahedron, 21(8), 2159. https://
doi.org/10.1016/s0040-4020(01)98351-6

10

You might also like