ArticlePDF Available

GC–MS and LC-TOF–MS profiles, toxicity, and macrophage-dependent in vitro anti-osteoporosis activity of Prunus africana (Hook f.) Kalkman Bark

Springer Nature
Scientific Reports
Authors:
  • University of Science and Technology South Korea

Abstract and Figures

Abstract Osteoporosis affects millions of people worldwide. As such, this study assessed the macrophage-dependent in vitro anti-osteoporosis, phytochemical profile and hepatotoxicity effects in zebrafish larvae of the stem bark extracts of P. africana. Mouse bone marrow macrophages (BMM) cells were plated in 96-well plates and treated with P. africana methanolic bark extracts at concentrations of 0, 6.25, 12.5, 25, and 50 µg/ml for 24 h. The osteoclast tartrate-resistant acid phosphatase (TRAP) activity and cell viability were measured. Lipopolysaccharides (LPS) induced Nitrite (NO) and interleukin-6 (IL-6) production inhibitory effects of P. africana bark extracts (Methanolic, 150 µg/ml) and β-sitosterol (100 µM) were conducted using RAW 264.7 cells. Additionally, inhibition of IL-1β secretion and TRAP activity were determined for chlorogenic acid, catechin, naringenin and β-sitosterol. For toxicity study, zebrafish larvae were exposed to different concentrations of 25, 50, 100, and 200 µg/ml P. africana methanolic, ethanolic and water bark extracts. Dimethyl sulfoxide (0.05%) was used as a negative control and tamoxifen (5 µM) and dexamethasone (40 µM or 80 µM) were positive controls. The methanolic P. africana extracts significantly inhibited (p
This content is subject to copyright. Terms and conditions apply.
1
Vol.:(0123456789)
Scientic Reports | (2022) 12:7044 | https://doi.org/10.1038/s41598-022-10629-7
www.nature.com/scientificreports
GC–MS
and LC‑TOF–MS proles, toxicity,
and macrophage‑dependent
in vitro anti‑osteoporosis activity
of Prunus africana (Hook f.)
Kalkman Bark
Richard Komakech1,2,5, Ki‑Shuk Shim3, Nam‑Hui Yim4, Jun Ho Song1, Sungyu Yang1,
Goya Choi1, Jun Lee1,2, Yong‑goo Kim1, Francis Omujal5, Denis Okello1,2,
Moses Solomon Agwaya5, Grace Nambatya Kyeyune5, Hyemin Kan6, Kyu‑Seok Hwang6,
Motlalepula Gilbert Matsabisa7 & Youngmin Kang1,2*
Osteoporosis aects millions of people worldwide. As such, this study assessed the macrophage‑
dependent in vitro anti‑osteoporosis, phytochemical prole and hepatotoxicity eects in zebrash
larvae of the stem bark extracts of P. africana. Mouse bone marrow macrophages (BMM) cells were
plated in 96‑well plates and treated with P. africana methanolic bark extracts at concentrations of
0, 6.25, 12.5, 25, and 50 µg/ml for 24 h. The osteoclast tartrate‑resistant acid phosphatase (TRAP)
activity and cell viability were measured. Lipopolysaccharides (LPS) induced Nitrite (NO) and
interleukin‑6 (IL‑6) production inhibitory eects of P. africana bark extracts (Methanolic, 150 µg/
ml) and β‑sitosterol (100 µM) were conducted using RAW 264.7 cells. Additionally, inhibition of
IL‑1β secretion and TRAP activity were determined for chlorogenic acid, catechin, naringenin and
β‑sitosterol. For toxicity study, zebrash larvae were exposed to dierent concentrations of 25, 50,
100, and 200 µg/ml P. africana methanolic, ethanolic and water bark extracts. Dimethyl sulfoxide
(0.05%) was used as a negative control and tamoxifen (5 µM) and dexamethasone (40 µM or 80 µM)
were positive controls. The methanolic P. africana extracts signicantly inhibited (p < 0.001) TRAP
activity at all concentrations and at 12.5 and 25 µg/ml, the extract exhibited signicant (p < 0.05) BMM
cell viability. NO production was signicantly inhibited (all p < 0.0001) by the sample. IL‑6 secretion
was signicantly inhibited by P. africana methanolic extract (p < 0.0001) and β‑sitosterol (p < 0.0001)
and further, chlorogenic acid and naringenin remarkably inhibited IL‑1β production. The P. africana
methanolic extract signicantly inhibited RANKL‑induced TRAP activity. The phytochemical study of
P. africana stem bark revealed a number of chemical compounds with anti‑osteoporosis activity. There
was no observed hepatocyte apoptosis in the liver of zebrash larvae. In conclusion, the stem bark of
P. africana is non‑toxic to the liver and its inhibition of TRAP activity makes it an important source for
future anti‑osteoporosis drug development.
OPEN
1Herbal Medicine Resources Research Center, Korea Institute of Oriental Medicine (KIOM), 111 Geonjae-ro,
Naju-si, Jeollanam-do 58245, Republic of Korea. 2University of Science and Technology (UST), Korean
Convergence Medicine Major, KIOM campus, 1672 Yuseongdae-ro, Yuseong-gu, Daejeon 34054, Republic of
Korea. 3Korea Institute of Oriental Medicine (KIOM), 1672 Yuseongdae-ro, Yuseong-gu, Daejeon 34054, Republic
of Korea. 4Korean Medicine Application Center, Korea Institute of Oriental Medicine, 70 Cheomdan-ro, Dong-gu,
Daegu 41062, Republic of Korea. 5Natural Chemotherapeutics Research Institute (NCRI), Ministry of Health,
P.O. Box 4864, Kampala, Uganda. 6Bio and Drug Discovery Division, Korea Research Institute of Chemical
Technology, Daejeon, Republic of Korea. 7IKS Research Group, Department of Pharmacology, Faculty of Health
Sciences, University of the Free State, Bloemfontein 9301, Free State, South Africa. *email: ymkang@kiom.re.kr
Content courtesy of Springer Nature, terms of use apply. Rights reserved
2
Vol:.(1234567890)
Scientic Reports | (2022) 12:7044 | https://doi.org/10.1038/s41598-022-10629-7
www.nature.com/scientificreports/
Osteoporosis is a silent but one of the major global health problems characterized by deterioration of bone micro-
architecture and low bone mass1,2. It is one of the leading causes of morbidity in older people above 40 years3. is
condition occurs when the rate of bone resorption is higher than the rate of bone formation and consequently
presents a greater risk of fractures for the persons suering from it. Factors such as aging, sex steroid deciency,
and as well as menopause in women have been associated with a higher risk of osteoporosis4. Activated mac-
rophages have also been implicated in the pathogenesis of osteoporosis by stimulating the development of osteo-
clastogenesis-associated bone loss5. Cytokines such as tumor necrosis factor alpha (TNFα), interleukin-1 (IL-1β)
and interleukin-6 (IL-6) are pro-inammatory and play central role in inammation of which IL-6 is the most
important in chronic inammatory and autoimmune diseases, cytokine storm and cancer6,7. IL-6 is fundamental
in a number of processes including bone metabolism, inammation, hematopoiesis7. IL-6 is also implicated in
mediation of IL-1 eects, a potent bone resorption stimulator8. Although a number of conventional methods
have been employed in the treatment of osteoporosis including bisphophates and estrogen hormonal therapy,
adverse eects associated with these therapies that are reported to limit their use include; gastrointestinal tract
disturbances and burning sensation9. Exploration of other avenues including the use of natural products in the
treatment of osteoporosis have been suggested to oer a better alternative with lesser adverse side eects9. Herbal
medicines have been used over the years to prevent and treat osteoporosis condition2. e anti-osteoporosis of
herbal products has been attributed to the secondary metabolites including alkaloids, terpenes, steroids, and
phenolic compounds10,11 and these have led to the development of a number of drugs over the years with great
therapeutic activities12. Prunus africana (Hook f.) Kalkman (Family Rosaceae), commonly called African cherry
is an evergreen plant endemic in sub-Saharan Africa13, and contains a number of secondary metabolites like
terpenes, alkaloids, phenolic compounds, and sterols in its stem bark14. For centuries, P. africana has been used
in Africa to treat myriad of diseases including prostate cancer, hyperplasia, diabetes, malaria, and inammatory
conditions15. Despite its immense medicinal uses and with a wide array of secondary metabolites, P. africana
has not been investigated for anti-osteoporosis activity yet it is used by the persons with prostate cancer who are
vulnerable to suer from osteoporosis16,17. Hence, this study evaluated the phytochemistry P. africana bark extract
and it’s in vitro anti-osteoporosis activity based on osteoclast tartrate-resistant acid phosphatase (TRAP) as a
cytochemical marker of osteoclasts. However, due to the toxicity associated with some of the herbal medicines
such as liver damage18 and owing to association of liver health and osteoporosis19, this study also evaluated the
hepatotoxicity of P. africana bark extracts in zebrash (Danio rerio) larvae.
Materials and methods
Plant material and preparation of extract. e study was conducted in accordance to the relevant
institutional, national, and international guidelines and legislation. e stem bark of P. africana (1 kg) was
obtained from P. africana tree (Fig.1a,b) in the herbal garden of Natural Chemotherapeutics Research Institute,
Ministry of Health, Uganda. e study material was identied by Dr. Sungyu Yang at Korea Institute of Oriental
medicine (KIOM) and a voucher specimen (number KIOM201901022377) of the sample was deposited in the
Korean Herbarium of Standard Herbal Resources (Index Herbarium code: KIOM) at KIOM, South Korea. e
stem bark (Fig.1c) was dried in an oven at 40°C and then ground using a steel pulverizing machine (250G
New Type Pulverizing Machine, Model RT-N04-2V, Taiwan) to obtain a ne powder (Fig.1d). 500g of the
ne powder sample was extracted by maceration using 1,500ml of methanol. e extract was ltered using
Whatman lter No. 1 aer 24h. and concentrated under a vacuum reduced pressure at 40°C, 70rpm, using an
EYELA N-1200B (Tokyo Rikakikai Co. Ltd, Japan) ecient rotary evaporator. e concentrated extract was then
vacuum dried and yielded 60g of extract. e resultant dried extract was then used in the subsequent TRAP
assay, cell viability assay, and experiments on the production of inammatory factors.
Gas chromatography‑mass spectrometry (GC–MS) sample preparation and analysis. e P.
africana bark sample was extracted in 100% methanol by sonication for 30min. e extract was then prepared
at 50µg/L; ltered through a 0.2μm syringe membrane lter from Whatman Ltd (Maidstone, UK) and subjected
to GC–MS analysis. e analysis was performed in a 7890B GC–MS system (Agilent Technologies, Atlanta, GA,
USA), coupled with a 7977B model mass detector (Agilent Technologies, Atlanta, GA, USA) using DB-5 MS
capillary column (30m × 0.25mm × 0.25μm). Chromatographic conditions were as follows: the extract (1μL)
was injected in split mode with a ratio of 1/20 at 250°C; oven initial temperature was 50°C and increased 110°C
during 5min, followed by heating at a rate of 7°C/min at 300°C. e mass analyzer was set to scan from 30 to
600 amu. Peak identication was carried out by comparison of the experimental mass spectrum in the National
Institute of Standards and Technology (NIST) and Wiley GC–MS libraries.
Liquid chromatography time‑of ight mass spectrometry (LC‑TOF–MS) analysis. e P. af ri-
cana bark sample was extracted and prepared as in “Gas chromatography-mass spectrometry (GC–MS) sample
preparation and analysis” above. e LC-TOF–MS analysis was performed on an Agilent 1290 innity II system
coupled with an AB SCIEX Triple TOF 5600 mass spectrometer equipped with electrospray ionization. Gemini®
C18 (250mm × 4.6mm i.d., 5μm, Phenomenex, USA) was used for column separation. e column temperature
was maintained at 40°C, the ow rate was 1.0ml/min, and the injection volume was 10μl. e optimal mobile
phase consisted of a linear gradient system of (A) 0.1% formic acid in water and (B) 0.1% formic acid in ace-
tonitrile, 0–2min, 3% B; 2–30min, 3–35% B; 30–31min, 35–50% B; 31–35min, maintained 50% B; 35–40min,
100% B; 40–45min, maintained 100% B. Positive mode was applied in the ESI source with the following param-
eters: gas 1 = 50 psi, gas 2 = 50 psi, temperature = 500°C, and 5500V ion spray voltage with 30 psi curtain gas.
Intact protonated molecular ions [M-Na]+ were detected via TOF–MS scan (100 psi declustering potential, 10V
collision energy, 100–2000Da TOF MS scan range, and 250ms accumulation time). Negative mode was applied
Content courtesy of Springer Nature, terms of use apply. Rights reserved
3
Vol.:(0123456789)
Scientic Reports | (2022) 12:7044 | https://doi.org/10.1038/s41598-022-10629-7
www.nature.com/scientificreports/
in the ESI source with the following parameters: gas 1 = 50 psi, gas 2 = 50 psi, temperature = 500°C, and − 4500V
ion spray voltage with 30 psi curtain gas. Intact protonated molecular ions [M-H] were detected via TOF–MS
scan (− 100 psi declustering potential, 10V collision energy, 100–2000Da TOF MS scan range, and 250ms
accumulation time).
Inhibitory eect of P. africana on osteoclastogenesis. Cell culture and authentication. Mouse bone
marrow macrophages (BMMs) were isolated from the tibia and femur of mice (male ICR mouse, 7weeks old) by
ushing with PBS as describe in previous study20. Aer over-night incubation in non-coated culture dish, non-
attached cells, which were regarded as BMMs, were collected and cultured in proliferation medium [α-MEM
medium with 10% FBS and macrophage-colony stimulating factor (M-CSF) (60ng/mL)] for 7days. To dier-
entiate osteoclasts, BMMs (1 × 104 cells/well, 96-well plates) were cultured in α-MEM medium containing 10%
FBS, M-CSF (60ng/mL), and RANKL (100ng/mL) for 4days.
TRAP assay and BMM cell viability. is was performed following the previously described method21. e
measurement of osteoclast TRAP activity was based on the generation of absorbance by incubating BMM cells
with TRAP buer (50mM sodium tartrate, 0.12M sodium acetate, pH 5.2) and p-nitrophenyl phosphate (1mg/
ml) for 15min. For TRAP staining, the BMM cells were incubated with TRAP buer containing naphthol
AS-MX phosphate (0.1mg/ml) and Fast Red Violet (0.5mg/ml). e BMM cells were then cultured with P.
africana methanolic extract at 0, 6.25, 12.5, 25, and 50µg/ml concentrations and 50μM of β-sitosterol, chloro-
genic acid, catechin and naringenin each in the presence of RANKL for 6days. e osteoclast TRAP activity was
determined using a colorimetric assay with p-nitrophenyl phosphate as a substrate. e cell viability was deter-
Figure1. Prunus africana medicinal plant. (a) P. africana tree. (b) Stem of P. africana with part of its bark
harvested for medicine purpose. (c) Harvested and dried P. africana stem bark. (d) Pulverized P. africana stem
bark.
Content courtesy of Springer Nature, terms of use apply. Rights reserved
4
Vol:.(1234567890)
Scientic Reports | (2022) 12:7044 | https://doi.org/10.1038/s41598-022-10629-7
www.nature.com/scientificreports/
mined using Cell Counting Kit-8 (CCK) (WST-8/CCK8; Dojindo), according to the manufacturer’s instructions.
For the measurement of cell viability, cells were plated in 96-well plates and treated with P. africana extracts
concentrations as above for 24h. Aer incubating with the CCK solutions and the cells for 1h, the absorbance
was measured at 450nm using a microplate reader (Versa Max) and results were presented as a percentage of
the vehicle control.
Nitrite (NO) assay and RAW 264.7 cell viability. Murine macrophage RAW 264.7 cells were cultured
in DMEM medium supplemented with 100 U/mL of penicillin, 100 ug/mL of streptomycin, and 10% heat-inac-
tivated FBS. e nitrite concentration in the supernatant from cultured cells was analyzed using the Griess reac-
tion test. RAW 264.7 cells were plated at a density of 5 × 104 cells/mL in 96-well culture plates, pre-incubated with
samples (P. africana methanolic extracts (150μg/mL), β-sitosterol, chlorogenic acid, catechin and naringenin
(100μM each) for 3h, and stimulated with LPS (200ng/ml) for 24h. Griess reagent (1% sulfanilamide, 0.1%
N-1-napthylethylenediamine dihydrochloride, and 2.5% phosphoric acid) was mixed with an equal volume of
cell supernatant, and absorbance was measured at 570nm using the ELISA reader. Sodium nitrite was used as a
standard. Dexamethasone was used as a positive control (40 or 80μM).
Enzyme‑linked immunosorbent assay (ELISA). e concentrations of the inammatory cytokines
IL-1β (R&D, USA) and IL-6 (MyBiosource, USA) in culture supernatant was determined using ELISA antibody
kits following the manufacturer1s protocol (MyBiosource, USA). RAW264.7 cells were grown in 96-well culture
plates at a density of 5 × 104 cells/mL, pre-incubated with samples for 3h, and stimulated with LPS for 6h (IL-
1β) or 24h (IL-6). e cytokines produced in each sample were calculated from standard curves using known
concentrations of recombinant cytokines for each ELISA antibody kit.
Hepatotoxicity assay in zebrash (Danio rerio) larvae. is was performed following the previously
described method21. All methods were carried out in accordance with relevant guidelines and regulations. All
experimental protocols were approved by Korea Research Institute of Chemical Technology research ethics com-
mittee and conducted in compliance with the ARRIVE guidelines. Zebrash larvae were used for this study as
previously described22. At 96h post-fertilization (hpf), the larvae were transferred to a transparent 24-well plate
(N = 10/well) with 1ml of embryonic medium. e larvae were then exposed to increasing concentrations of 25,
50, 100, and 200µg/ml of P. africana methanolic and ethanolic extracts and water extract from 90 to 120 hpf.
Dimethyl sulfoxide (DMSO) was used as a negative control while 5μM of tamoxifen (Sigma-Aldrich, St. Louis,
MO, USA) was used as a positive control. To obtain images, the larvae were anesthetized in tricaine (Sigma-
Aldrich), mounted in 3% methyl cellulose (Sigma-Aldrich), and observed under a Leica MZ10 F stereomicro-
scope equipped with a Leica DFC425 camera and Leica application Suite soware (version 4.5).
Statistical analysis. Data were represented as the mean ± standard deviation. Statistical signicance
between groups was analyzed using the Student’s t-test and p-values < 0.05 were considered statistically signi-
cant.
Results
GC/MS analysis. e GC/MS analysis of the P. africana extract was based on mass spectra, retention times,
and quality ratio analysis revealed the presence of 32 components (Table1) including 3-Furanmethanol (1),
Dihydroxyacetone (2), Benzoic acid, methyl ester (3), 4H-Pyran-4-one,2,3-dihydro-3,5-dihydroxy-6-methyl-
(4), benzoic acid (5), Catechol (6), 4-Vinylphenol (7), 5-Hydroxymethyl-2-furaldhyde (8), Isosorbide (9),
Phenol, 2,6-dimethoxy- (10), 4-Hydroxy-3-methoxybenzaldehyde (11), 3,4-Altrosan (12), Mandelamide (13),
Vanillic acid (14), Benzenepropanol, 4-hydroxy-3-methoxy- (15), Benzaldehyde, 4-hydroxy-3,5-dimethoxy-
(16), 4-(hydroxymethyl)-2,6-dimethoxyphenol (17), (E)-4-(3-Hydroxyprop-1-en-1-yl)-2-methoxyphenol (18),
6-Hydroxy-5-triuoromethylcyclohexa-1,3-diene (19), Benzoic acid, 4-hydroxy-3,5-dimethoxy- (20), Isopro-
pyl myristate (21), Sorbitol (22), n-Hexadecanoic acid (23), 9,12-Octadecadienoic acid (Z,Z)- (24), Oleic acid
(25), Octadecanoic acid (26), Benzyl, beta-d-glucoside (27), 9-Octadecenamide, (Z)- (28), (R)-alpha-(beta-D-
glucopyranosyloxy)benzene-acetonitrile (29), 13-Docosenamide, (Z)- (30), Squalene (31), Beta-Sitosterol (32).
LC‑TOF–MS analysis. Based on the chemical proling by LC-TOF–MS analysis, the various phytochemi-
cals were detected from the P. africana extract. In the positive ion mode, as a result of analysis using retention
index libraries, 65 peaks with a library score over 90% were identied from the P. africana extract (Supple-
mentary TableY). Among them, 24 components showed reliable mass (over 98% of library score) in the P.
africana extract, especially, 7 components (Astragalin, Chlorogenic acid, Coproporphyrin I, Hyperin, Luteolo-
side, Mesoporphyrin IX and Naringenin), showed the accurate mass according to result of 100% library score
(Table2). In the negative ion mode, 72 peaks with a library score over 90% were identied from the P. africana
extract (Supplementary TableZ). Among them, 29 components showed the reliable mass (over 98% of library
score) in the P. africana extract, especially, 5 components (Pedunculoside, Luteoloside, Hexadecanedioic acid,
Guanosin, Betulonic acid and Naringenin), showed the accurate mass according to result of 100% library score
(Table2). In the present study, among the identied primary and secondary metabolic components, Catechin
showed the largest peak area among the identied primary and secondary metabolic components in the P. af ri-
cana extract (Supplementary TableY and Z).
Content courtesy of Springer Nature, terms of use apply. Rights reserved
5
Vol.:(0123456789)
Scientic Reports | (2022) 12:7044 | https://doi.org/10.1038/s41598-022-10629-7
www.nature.com/scientificreports/
Table 1. Phytochemical components identied in the stem bark of Prunus africana methanolic extract by GC/
MS analysis.
Peak no Identied compound tR (min) % of total Quality (%)
1 3-Furanmethanol 5.96 0.39 93
2 Dihydroxyacetone 7.01 2.22 74
3 Benzoic acid, methyl ester 12.83 0.46 91
4 4H-Pyran-4-one,2,3-dihydro-3,5-dihydroxy-6-methyl- 13.95 1.13 95
5 benzoic acid 14.68 14.02 91
6Catechol 15.17 4.74 91
7 4-Vinylphenol 15.61 0.77 72
8 5-Hydroxymethyl-2-furaldhyde 15.81 0.31 89
9 Isosorbide 17.10 0.98 93
10 Phenol, 2,6-dimethoxy- 18.39 0.41 94
11 4-Hydroxy-3-methoxybenzaldehyde 19.30 0.41 93
12 3,4-Altrosan 20.84 3.30 76
13 Mandelamide 21.54 0.40 93
14 Vanillic acid 22.13 0.93 95
15 Benzenepropanol, 4-hydroxy-3-methoxy- 23.61 2.14 92
16 Benzaldehyde, 4-hydroxy-3,5-dimethoxy- 23.83 0.75 91
17 4-(hydroxymethyl)-2,6-dimethoxyphenol 24.57 0.17 87
18 (E)-4-(3-Hydroxyprop-1-en-1-yl)-2-methoxyphenol 25.03 0.18 91
19 6-Hydroxy-5-triuoromethylcyclohexa-1,3-diene 25.98 3.65 59
20 Benzoic acid, 4-hydroxy-3,5-dimethoxy- 26.13 0.29 96
21 Isopropyl myristate 26.31 0.40 99
22 Sorbitol 27.95 0.05 91
23 n-Hexadecanoic acid 28.21 4.95 99
24 9,12-Octadecadienoic acid (Z,Z)- 30.51 0.20 99
25 Oleic acid 30.58 0.76 98
26 Octadecanoic acid 30.87 0.47 97
27 Benzyl, beta-d-glucoside 31.90 0.32 87
28 9-Octadecenamide, (Z)- 33.35 1.95 99
29 (R)-alpha-(beta-D-glucopyranosyloxy) benzene-acetonitrile 35.10 6.60 83
30 13-Docosenamide, (Z)- 37.88 6.49 99
31 Squalene 38.38 1.09 99
32 Beta-Sitosterol 43.55 8.37 99
Table 2. Phytochemical components identied in the stem bark of P. africana methanolic extract by LC-TOF–
MS analysis.
No Name Mass ([M-Na]+) Founded mass RT (min) Founded RT (min) Peak area Library score (%)
1 Astragalin 449.1083 449.1083 23.25 23.22 1129.49 100.00
2 Chlorogenic acid 355.1024 355.1024 13.16 13.14 2289.52 100.00
3Coproporphyrin I 655.4932 655.4961 1.19 1.32 1974.83 100.00
4 Hyperin 465.1033 465.1033 23.12 23.12 3329.70 100.00
5 Luteoloside 449.1787 449.1787 22.56 22.56 4448.29 100.00
6 Mesoporphyrin IX 567.2814 567.2809 42.57 42.56 1217.89 100.00
7 Naringenin 273.0762 273.0762 33.74 33.73 6559.30 100.00
No Name Mass ([M-H]–) Founded mass RT (min) Founded RT (min) Peak area Library score (%)
1Pedunculo-
side + HCOOH 695.3693 695.3696 34.39 34.38 20,498.21 100.00
2 Luteoloside 447.0696 447.0695 23.18 23.21 1513.48 100.00
3 Hexadecanedioic acid 285.1910 285.1909 40.49 40.48 4854.99 100.00
4 Guanosine 282.0683 282.0684 6.66 6.65 7969.05 100.00
5 Betulonic acid 453.2775 453.2776 42.97 42.97 1209.40 100.00
Content courtesy of Springer Nature, terms of use apply. Rights reserved
6
Vol:.(1234567890)
Scientic Reports | (2022) 12:7044 | https://doi.org/10.1038/s41598-022-10629-7
www.nature.com/scientificreports/
TRAP assay and BMM viability. TRAP activity was signicantly (p < 0.001) inhibited compared to the
control at concentrations of 6.25, 12.5, 25, and 50µg/ml methanolic P. africana stem bark extracts (Fig.2A).
Prunus africana methanolic stem bark extract had a signicant (p < 0.05) simulative eect at concentrations
of 12.5 and 25µg/ml on the cell viability of BMM cells (Fig.2B) compared to the control. However, at a higher
concentration of 50µg/ml the methanolic stem bark extract, the cell viability reduced to 70% compared to the
control. e high viability of the BMM cells may indicate the non-cytotoxicity of P. africana bark.
e eects of β-sitosterol, chlorogenic acid, catechin and naringenin (50μM each) on RANKL-induced TRAP
activity representing osteoclastogenesis were evaluated. We found that these compounds signicantly inhibited
RANKL-induced TRAP activity without showing cell toxicity (Fig.3).
Inhibitory eect on NO production. Since NO production is correlated with various inammatory dis-
eases, we determined the suppressive eects of samples (P. africana (PA) methanolic extract, β-sitosterol, chlo-
rogenic acid, catechin and naringenin) on NO levels in RAW264.7 cells via LPS stimulation. To determine NO
levels in the supernatant, cells were pre-treated with samples for 3h, followed by stimulation with LPS for 24h,
and then measured using Griess reagent. As the positive control, dexamethasone showed strong suppressive
eect on NO secretion upon LPS stimulation. All the investigated samples dramatically inhibited NO produc-
tion aer LPS stimulation (Fig.3b). All samples did not signicantly aect cell viability and β-sitosterol that
increased it (Fig.3a).
Inhibitory eect on IL‑6 and IL‑1β levels. e eects of samples on inammatory cytokine, IL-6, secre-
tion in macrophages were evaluated using enzyme-linked immunosorbent assay (ELISA). IL-6 secretion was
signicantly inhibited by PA-methanol (p < 0.001) and β-sitosterol (p < 0.0001) (Fig.3c). Chlorogenic acid and
naringenin, but not catechin, signicantly inhibited LPS-induced IL-1β level as shown in Fig.3d.
Hepatotoxicity in zebrash larvae. In this study, the zebrash larvae exposure was done from 96 to 120
hpf and those exposed to DMSO showed no liver cell death (shown by white dash line) (Fig.4A) but tamoxifen
treatment resulted in liver cell death (shown by red dash line) (Fig.4B). At a concentration of 50 and 100µg/
ml water extract of P. africana, 30% and 10% of the zebrash larvae survived at 120 hpf respectively and death
of hepatocytes was not observed in them (Fig.4C). However, at a higher concentration of 200µg/ml P. a fr i-
cana water extract, 100% larvae mortality was observed before 120 hpf. At a concentration of 25µg/ml etha-
nolic extract, 50% of the zebrash larvae survived and hepatocytes death was not observed in them at 120 hpf
(Fig.4D). However, at higher concentrations of 50 and 100µg/ml, 100% larvae mortality rate was observed at
120 hpf. 100% larvae mortality was observed for larvae exposed to P. africana bark methanolic extracts at various
concentrations of 25, 50, and 100µg/ml before the 120 hpf.
Discussion
TRAP is a specic and reliable cytochemical marker used as a measure of activated macrophages23. e bone
marrow macrophages play a major role in the activation and formation of osteoclastsand hence important for
the pathogenesis of osteoporosis5. Osteoporosis reects increased osteoclast function relative to that of osteo-
blasts and hence the pharmacological arrest of osteoclasts is a mainstay in the treatment of systemic bone loss24.
TRAP activity is an important cytochemical marker of osteoclasts and its concentration in the serum is utilized
as a biochemical and histochemical marker of osteoclast function and degree of bone resorption25. erefore,
as a measure of osteoclast number and bone resorption, TRAP plays a vital role in osteoporosis diagnosis and
prognosis23. And as evidenced in the previous study, the suppressing of TRAP pathway prevented ovariectomy-
induced osteoporosis in vivo26. erefore, the signicant (p < 0.001) inhibition compared to the control at con-
centrations of 6.25, 12.5, 25, and 50µg/ml methanolic P. africana stem bark extracts is an indication of P. africana
anti-osteoporosis eects. Additionally, chlorogenic acid, catechin, naringenin and β-sitosterol that are present
Figure2. Eect of P. africana on TRAP activity in BMM. e BMM were cultured with P. africana bark extracts
in the presence of RANKL for 6days and TRAP activity of osteoclasts measured by colorimetric assay using
p-nitrophenyl phosphate as a substrate. Cell viability was determined using Cell Counting Kit-8 following
manufacturer’s instruction. *p < 0.05 and ***p < 0.001.
Content courtesy of Springer Nature, terms of use apply. Rights reserved
7
Vol.:(0123456789)
Scientic Reports | (2022) 12:7044 | https://doi.org/10.1038/s41598-022-10629-7
www.nature.com/scientificreports/
in P. africana stem bark have been demonstrated in our study to remarkably inhibit RANKL-induced TRAP
activity explaining indeed the antiosteoporotic potential of the plant stem bark.
e anti-osteoporosis activity of P. africana bark extracts may be attributed to some of the compounds present
in it including astragalin, hyperin, luteoloside, mesoporphyrin, naringenin, chlorogenic acid, β-sitosterol and
catechin (Tables1 and 2, and supplementary TablesYand Z). Additionally, previous studies have also showed
that P. africana stem bark is indeed rich in these compounds with anti-osteoporosis activity (Table3). Astragalin
demonstrated estrogenic anti-osteoporosis activity and signicantly increased proliferation in osteoblastic cells
(UMR-106)27. In another study, Astragalin promoted dierentiation in MC3T3-E1 osteoblastic cells through
activation of MAPK and BMP pathways and promoted invivo bone formation28. Further, a number of other
studies have showed anti-osteoporosis activity of Astragalin10,29,30, and its presence in P. africana stem bark as
conrmed in our study shows that it indeed contributes to the anti-osteoporosis activity of the plant stem bark.
Hyperin was one of the major 3 chemical compounds in Cuscuta chinensis that showed anti-osteoporosis eects29.
In fact in their study, Tao, etal.29 ascribed the anti-osteoporosis eect of C. chinensis to hyperin as a result of its
high positive correlation to anti-osteoporosis activity. Hyperin was also reported to markedly increase alkaline
phosphatase (ALP) activity in osteoblast cells31.
Luteoloside, a natural compound is known to suppress activity of osteoclasts thus could potentially be used
for treating bone metabolism disorders including osteoporosis32. Luteoloside in a previous study was showed
to possess strong inhibition against LPS induced osteolysis in an invivo study32. Further, it was also dem-
onstrated to suppress dierentiation of RANKL-induced osteoclast and decrease bone resorption tendency
dose dependently32. e anti-osteoclastic and anti-resorptive actions of luteoloside were not only through
blockage of NFATc1 activity and debilitation of RANKL-mediated Ca2+signaling but also through MAPK and
NF-κB pathways32. Mesoporphyrin, a porphyrin derivative has been reported to possess anti-inammatory
activity through inhibiting IL-6 production33. Since IL-6 potently activates osteoclasts and is responsible for
bone resorption34, mesoporphyrin IX may thus possess anti-osteoporosis activity through inhibition of IL-6
production.
Figure3. Eects of samples (PA-methanol, β-sitosterol, chlorogenic acid, catechin, and naringenin) on (a)
cell viability, (b-d) the production of inammatory factors (nitric oxide, IL-6, or IL-1β), or (e) TRAP activity.
Aer 3h pre-incubation of samples, RAW 264.7 cells were treated with LPS for 6 to 48h depending on the
assay condition. (a) Cell viability was measured using a CCK assay. (b) Nitric oxide content in the medium was
determined using Griess reagent assay; (c) IL-6 and (d) IL-1β cytokine levels in the medium were measured
using ELISA kit. (e) TRAP activity was examined by using TRAP buer containing naphthol AS-MX phosphate.
Positive control: 40 or 80μM dexamethasone. As a control, cells were incubated with the vehicle alone. *
p < 0.05, ** p < 0.01 and *** p < 0.001.
Content courtesy of Springer Nature, terms of use apply. Rights reserved
8
Vol:.(1234567890)
Scientic Reports | (2022) 12:7044 | https://doi.org/10.1038/s41598-022-10629-7
www.nature.com/scientificreports/
Naringenin has been reported to signicantly inhibit osteoclastogenesis with inhibitions of up to 96 ± 1%
at 50μg/ml35. Naringenin has also been demonstrated to not only signicantly inhibit secretion of monocyte
chemoattractant protein-1, interleukin (IL)-1α and IL-23 but also markedly decrease release of a bone resorp-
tion activity indicator, helical peptide 620–633, thus greatly inhibiting osteoclastic bone resorption and human
osteoclastogenesis35. La, etal.35 indicates that naringenin could be used to treat bone-related diseases such as
Table 3. Summary of the previous studies on anti-osteoporosis eects of some of the compounds identied in
P. africana.
S/No Phytochemical compounds Compound structure Anti-osteoporosis eects Reference
(a) Vanillic acid
Improves bone mineral density and bone mineral content; Protects the
trabecular structure from being degraded by osteoclasts 43,44
(b) Sorbitol
Retards bone resorption 45
(c) Octadecanoic acid (Stearic acid)
Inhibits osteoclastogenesis invitro 46
(d) β-sitosterol
Inhibits osteoporosis through retardation of acute inammation 47
Figure4. Hepatotoxicity assay in zebrash larvae at 120 hpf. (A) DMSO as a negative control did not induce
hepatotoxicity. (B) Tamoxifen induced liver cell death (shown by red arrow). (C) 100µg/ml P. africana water did
not induce hepatotoxicity. (D) 25µg/ml P. africana ethanol did not induce hepatotoxicity.
Content courtesy of Springer Nature, terms of use apply. Rights reserved
9
Vol.:(0123456789)
Scientic Reports | (2022) 12:7044 | https://doi.org/10.1038/s41598-022-10629-7
www.nature.com/scientificreports/
osteoporosis. In the current study, naringenin was observed to signicantly inhibit LPS-induced NO production
to a level even greater than the eect of dexamethasone (positive control) and also remarkably suppress IL-1β
generation. Since increased NO level inhibits growth and dierentiation of osteoblasts36, and IL-1β stimulates
bone resorption37, naringenin suppression of NO and IL-1β production signies that it is important in prevent-
ing osteoporosis. In another study, naringenin was showed to signicantly promote osteogenic dierentiation38.
Naringenin being present in the stem bark of P. africana contributes to its anti-osteoporosis activity. Catechin
was identied as possessing the strongest osteogenic eects from a batch of herbal ingredients used in traditional
Chinese medicine using human mesenchymal stem cells (hMSCs)39. Catechin was reported to increase the activ-
ity of ALP, deposition of calcium and Runx2 mRNA expression among others39. It was thus proposed to enhance
osteogenesis through increasing protein phosphatases 2A (PP2A) level inhibiting extracellular signal-regulated
kinase (ERK) signaling in hMSCs39. In another study, catechin rich extract was demonstrated to promote for-
mation and enhance survival of osteoblasts and inhibiting the activity and growth of osteoclasts40. Similar to
our ndings, catechin in previous studies has been documented to suppress NO production in LPS stimulated
macrophages41,42. us, our study suggests that catechin exerts antiosteroporotic eect by eliminating the inhibi-
tory eect of NO on growth and dierentitation of osteoblasts. e presence of catechin in the stem bark of P.
africana enhances its anti-osteoporosis potential.
Vanillic acid has been reported to exhibit anti-osteoporotic activity by inhibitory eects on bone resorption48;
improving bone mineral density and bone mineral contentand as well as biomechanical stability43 and protect-
ing trabecular structure from degradation by osteoclasts in ovariectomized postmenopausal mice44. Sorbitol; a
sugar alcohol with a sweet taste has been observed to retard bone resorption in Sprague Dawley male rats45. e
positive eects on osteoporosis prevention by fruits including that of Prunus domestica and Prunus salicina may
partly be due to the presence of sorbitol compound in them1. Recent studies suggest that inammation is one of
the key factors that inuence bone turnover, leading to osteoporosis3. erefore, the potent anti-inammatory
activity of β-sitosterol47; a compound present in P. africana stem bark may further explain its anti-osteoporosis
activity. Stearic acid and oleic acid have been reported to inhibit osteoclastogenesis in bone marrow cultures and
RAW264.7 cells46. Chlorogenic acid was observed to improve the bone quality by modifying the bone mineral
density and trabecular microarchitecture in an ovariectomy rat model49. Furthermore, chlorogenic acid was also
observed to promote proliferation of osteoblast precursors and osteoblastic dierentiation in ovariectomized
rats50. In the present study, chlorogenic acid exhibited signicant inhibition of LPS-induced NO production and
secretion of IL-1β. Interleukin-1 is a very powerful stimulator of bone resorption and is well known to inhibit
bone formation51. e signicant inhibition of IL-1β and NO production by chlorogenic acid indicates that it
plays an important role in preventing and treating osteoporosis. In addition, our study showed that chlorogenic
acid had a signicant inhibition of RANKL-induced TRAP activity giving further evidence of its antiosteoporotic
potential.
In light of these results, the inhibition of the TRAP activity by P. africana bark extracts may therefore be due
to these dierent important compounds in it including chlorogenic acid, catechin, naringenin and β-sitosterol.
ese ndings therefore provide valuable insight in to the anti-osteoporosis potential of P. africana.
Excess production of NO in the body system plays a vital part in pathogenesis of inammatory diseases
including osteoporosis21. It has been reported previously that increased production of NO is a contributing fac-
tor to osteoporosis pathogenesis21,52. us, a potential therapeutic pathway for managing the disease is through
suppression of NO production as indicated by Komakech, etal.21. In this study, the P. africana methanolic
extract (each 150µg/ml) signicantly (p < 0.0001) inhibited NO production actually more than the positive
standard, dexamethasone (40µM). e chemical compounds, chlorogenic acid, catechin, naringenin and
β-sitosterol (100µM each) in RAW264.7 cells also signicantly suppressed LPS-induced NO production. Stud-
ies have reported that high NO concentrations have great inhibitory eects on growth and dierentiation of
osteoblasts36,5355. is has been suggested to be partly as a result of NO pro-apoptotic eects on osteoblasts56.
e signicant inhibition of LPS-induced NO production by PA extracts and its chemical constituents clearly
demonstrates its antiosteoporotic potential by eliminating the inhibitory eects of NO to growth and dierentia-
tion of osteoblasts. Chlorogenic acid, catehin, naringenin and β-sitosterol did not show toxic eects.
In our current study, PA-methanolic extract (150µg/ml) and β-sitosterol (100µM) just like the positive con-
trol (dexamethasone [40µM and 80µM]) in RAW264.7 cells were observed to signicantly inhibit LPS-induced
IL-6 secretion. is therefore suggests that methanol extracted phytochemical compounds from P. africana stem
bark including β-sitosterol exhibited antiosteoporotic potential by inhibiting the production of IL-6 within the
bone microenvironment. IL-6 is known to potently activate osteoclasts and is responsible for bone resorption34.
Pro-inammatory cytokines notably IL-6 are vital in normal processes of bone remodeling and pathogenesis of
osteoporosis in elderly persons and during perimenopause34. Production of IL-6 induces eminent lytic lesions
along with diuse osteoporosis typical of the disease34. Before menopause, estrogen in bone marrow regulates the
expression of most notably IL-634. IL-6 levels are known to increase with age in not only humans but monkeys
and mice and in the trend with osteoporosis34.
Liver toxicity from herbal and dietary supplements is a common phenomenon and is a leading cause of a num-
ber of underlying liver diseases57. Osteoporosis is a frequent complication in patients with liver complications58.
Indeed, decreased trophic factors such as insulin growth factor in the liver due to liver toxicity or chronic dis-
eases including diabetes my result in osteoblast dysfunction19. erefore, ensuring a healthy liver is fundamental
in maintaining a balanced body biological processes including prevention of bone loss. Zebrash larvae is an
important model system for the evaluation of the liver toxicity when an organism is exposed to a toxicant59.
Liver organogenesis in zebrash initiates at 30 hpf on the le-hand side of the embryo and an enlarged liver bud
connects with the intestine and functionally matures until 72 hpf60. Treatment of the zebrash larvae with liver
toxicants such as tamoxifen reduces liver transparency due to the liver cell death61. In this study, 200µg/ml water
extract and 25µg/ml methanolic extract exhibited maximum concentrations for the acute toxicity. us, tests at
Content courtesy of Springer Nature, terms of use apply. Rights reserved
10
Vol:.(1234567890)
Scientic Reports | (2022) 12:7044 | https://doi.org/10.1038/s41598-022-10629-7
www.nature.com/scientificreports/
100µg/ml water extract and 25µg/ml ethanolic extract was used to determine hepatotoxicity in zebrash larvae.
And although each experimental group showed larval mortality, the P. africana extracts did not induce hepato-
toxicity because living larvae did not show liver-specic cell death. Considering that the extraction method can
inuence the composition of extracts62, this may explain dierence in the mortality rate of the zebrash larvae
in the methanolic and water extracts of the P. africana stem bark. Previous studies also showed that P. africana
bark extract administered at 1000mg/kg body weight had no visible deleterious eects on BALB/c mice63 and
showed mild hepatotoxicity and nephrotoxicity in Sprague–Dawley rats64. ese observations therefore showed
that the stem bark of P. africana its non-toxic within a low dose range.
Conclusions
e macrophage-dependent anti-osteoporosis activity of P. africana bark may be attributed to the synergistic
action of the various phytochemicals in its stem bark including chlorogenic acid, catehin, naringenin, vanillic
acid, sorbitol, octadecanoic acid (stearic acid), and β-sitosterol. NO production was signicantly inhibited (all
p < 0.0001) by P. africana methanolic, chlorogenic acid, catehin, naringenin, and β-sitosterol. IL-6 secretion was
signicantly inhibited by P. africana methanolic extract (p < 0.0001) and β-sitosterol (p < 0.0001) and in addi-
tion, chlorogenic acid and naringenin remarkably inhibited IL-1β production. All samples displayed signicant
inhibition of RANKL-induced TRAP activity. Although the methanolic extract of P. africana bark exhibited
potent anti-osteoporosis activity, we recommend that future studies should carry out isolation of the individual
chemicals or group of chemicals that are/is responsible for its anti-osteoporosis activity. Nonetheless, this study
has demonstrated that P. africana bark extracts have no overt hepatotoxic eects in zebrash larvae at a given
dose range and oers a basis for future studies and medicine development with anti-osteoporosis therapeutic
application.
Data availability
e data for this current study are available from the corresponding author upon reasonable request.
Received: 1 June 2021; Accepted: 24 March 2022
References
1. Higgs, J., Derbyshire, E. & Styles, K. Nutrition and osteoporosis prevention for the orthopaedic surgeon: A wholefoods approach.
EFORT Open Rev. 2, 300–308 (2017).
2. An, J. et al. Natural products for treatment of osteoporosis: e eects and mechanisms on promoting osteoblast-mediated bone
formation. Life Sci. 147, 46–58 (2016).
3. Ginaldi, L., Mengoli, L. P. & De Martinis, M. Handbook on Immunosenescence 1329–1352 (Springer, 2009).
4. Sözen, T., Özışık, L. & Başaran, N. Ç. An overview and management of osteoporosis. Eur. J. Rheumatol. 4, 46 (2017).
5. Yang, D.-H. & Yang, M.-Y. e role of macrophage in the pathogenesis of osteoporosis. Int. J. Mol. Sci. 20, 2093 (2019).
6. Hirano, T. Interleukin 6 and its receptor: Ten years later. Int. Rev. Immunol. 16, 249–284 (1998).
7. Hirano, T. IL-6 in inammation, autoimmunity and cancer. Int. Immunol. 33, 127–148 (2021).
8. Ota, N. et al. A nucleotide variant in the promoter region of the interleukin-6 gene associated with decreased bone mineral density.
J. Hum. Genet. 46, 267–272 (2001).
9. Suvarna, V. et al. Bone health and natural products-an insight. Front. Pharmacol. 9, 981 (2018).
10. Jia, M. et al. Potential antiosteoporotic agents from plants: a comprehensive review. Evid.-Based Complement. Altern. Med. 2012,
2 (2012).
11. Evelyn, S. S. & Chitra, V. Medicinal plants for the treatment of postmenopausal osteoporosis. Biomed. Pharmacol. J. 12, 1561 (2019).
12. Rates, S. M. K. Plants as source of drugs. Tox icon 39, 603–613 (2001).
13. Jimu, L. reats and conservation strategies for the African cherry (Prunus africana) in its natural range-A review. J. Ecol. Nat.
Environ. 3, 118–130 (2011).
14. Nyamai, D. et al. Phytochemical prole of Prunus africana stem bark from Kenya. J. Pharmacogn. Nat. Products 1, 8 (2015).
15. Komakech, R. & Kang, Y. Ethnopharmacological potential of African cherry [Prunus africana]. J. Herb. Med. 17, 100283 (2019).
16. Smith, M. R. Osteoporosis during androgen deprivation therapy for prostate cancer. Urology 60, 79–85 (2002).
17. Daniell, H. W. et al. Progressive osteoporosis during androgen deprivation therapy for prostate cancer. J. Urol. 163, 181–186 (2000).
18. Amadi, C. N. & Orisakwe, O. E. Herb-induced liver injuries in developing nations: An update. Toxics 6, 24 (2018).
19. Kalaitzoglou, E., Popescu, I., Bunn, R. C., Fowlkes, J. L. & railkill, K. M. Eects of type 1 diabetes on osteoblasts, osteocytes, and
osteoclasts. Curr. Osteoporos. Rep. 14, 310–319 (2016).
20. Lee, J.-H. et al. Trolox prevents osteoclastogenesis by suppressing RANKL expression and signaling. J. Biol. Chem. 284, 13725–13734
(2009).
21. Komakech, R. et al. Invitro antiosteoporosis activity and hepatotoxicity evaluation in zebrash larvae of bark extracts of Prunus
jamasakura medicinal plant. Evid.-Based Complement. Altern. Med. 2020, 2 (2020).
22. Westereld, M. (University of Oregon Press Eugene, OR, 2000).
23. Janckila, A. J. & Yam, L. T. Biology and clinical signicance of tartrate-resistant acid phosphatases: New perspectives on an old
enzyme. Calcif. Tissue Int. 85, 465–483 (2009).
24. Teitelbaum, S. L. Osteoclasts: What do they do and how do they do it?. Am. J. Pathol. 170, 427–435 (2007).
25. Blumer, M. J. et al. Role of tartrate-resistant acid phosphatase (TRAP) in long bone development. Mech. Dev. 129, 162–176 (2012).
26. Yıldırım, M. et al. White tea reduced bone loss by suppressing the TRAP/CTX pathway in ovariectomy-induced osteoporosis
model rats. Cells Tissues Organs 209, 64–74 (2020).
27. Yang, L., Chen, Q., Wang, F. & Zhang, G. Antiosteoporotic compounds from seeds of Cuscuta chinensis. J. Ethnopharmacol. 135,
553–560 (2011).
28. Liu, L. et al. Astragalin promotes osteoblastic dierentiation in MC3T3-E1 cells and bone formation invivo. Front. Endocrinol.
10, 228 (2019).
29. Tao, Y., Chen, L., Pan, M., Zhu, F. & Yan, J. Tracing anti-osteoporosis components from raw and salt-processed semen of Cuscuta
chinensis by employing a biochemometrics strategy that integrates ultrasonic-assisted extraction, quantitation, ecacy assessment
in zebrash, and grey relationship analysis. J. Sep. Sci. 44, 3229–3236 (2021).
30. Riaz, A. et al. Astragalin: A bioactive phytochemical with potential therapeutic activities. Adv. Pharmacol. Sci. 2018, 2 (2018).
Content courtesy of Springer Nature, terms of use apply. Rights reserved
11
Vol.:(0123456789)
Scientic Reports | (2022) 12:7044 | https://doi.org/10.1038/s41598-022-10629-7
www.nature.com/scientificreports/
31. Yang, H. M., Shin, H.-K., Kang, Y.-H. & Kim, J.-K. Cuscuta chinensis extract promotes osteoblast dierentiation and mineraliza-
tion in human osteoblast-like MG-63 cells. J. Med. Food 12, 85–92 (2009).
32. Song, F. et al. Luteoloside prevents lipopolysaccharide-induced osteolysis and suppresses RANKL-induced osteoclastogenesis
through attenuating RANKL signaling cascades. J. Cell. Physiol. 233, 1723–1735 (2018).
33. Takaoka, Y., Matsuura, S., Boda, K. & Nagai, H. e eect of mesoporphyrin on the production of cytokines by inammatory cells
invitro. Jpn. J. Pharmacol. 80, 33–40 (1999).
34. Ershler, W. B., Harman, S. M. & Keller, E. T. Immunologic aspects of osteoporosis. Dev. Comp. Immunol. 21, 487–499 (1997).
35. L a, V., Tanabe, S. & Grenier, D. Naringenin inhibits human osteoclastogenesis and osteoclastic bone resorption. J. Periodontal Res.
44, 193–198 (2009).
36. Ralston, S. H., Todd, D., Helfrich, M., Benjamin, N. & Grabowski, P. S. Human osteoblast-like cells produce nitric oxide and express
inducible nitric oxide synthase. Endocrinology 135, 330–336 (1994).
37. Amano, S., Kawakami, K., Iwahashi, H., Kitano, S. & Hanazawa, S. Functional role of endogenous CD14 in lipopolysaccharide-
stimulated bone resorption. J. Cell. Physiol. 173, 301–309 (1997).
38. Wang, X. et al. Osteogenic eects of avonoid aglycones from an osteoprotective fraction of Drynaria fortunei—an invitro ecacy
study. Phytomedicine 18, 868–872 (2011).
39. Wei, Y. et al. Catechin stimulates osteogenesis by enhancing PP2A activity in human mesenchymal stem cells. Osteoporos. Int. 22,
1469–1479 (2011).
40. He, Y. et al. Glucagon like peptide 2 has a positive impact on osteoporosis in ovariectomized rats. Life Sci. 226, 47–56 (2019).
41. Guruvayoorappan, C. & Kuttan, G. (+)-Catechin inhibits tumour angiogenesis and regulates the production of nitric oxide and
TNF-α in LPS-stimulated macrophages. Innate Immun. 14, 160–174 (2008).
42. Li, T., Li, F., Liu, X., Liu, J. & Li, D. Synergistic anti-inammatory eects of quercetin and catechin via inhibiting activation of
TLR4–MyD88-mediated NF-κB and MAPK signaling pathways. Phytother. Res. 33, 756–767 (2019).
43. Wang, Y. G., Jiang, L. B. & Gou, B. Protective eect of vanillic acid on ovariectomy-induced osteoporosis in rats. Afr. J. Tradit.
Complement. Altern. Med. 14, 31–38 (2017).
44. Tanaka, T. et al. Anti-osteoporotic eects of syringic acid and vanilic acid in the extracts of waste beds aer mushroom cultivation.
J. Biosci. Bioeng. 128, 622–629 (2019).
45. Mattila, P. T., Svanberg, M. J., Mäkinen, K. K. & Knuuttila, M. L. Dietary xylitol, sorbitol and D-mannitol but not erythritol retard
bone resorption in rats. J. Nu tr. 126, 1865–1870 (1996).
46. Cornish, J. et al. Modulation of osteoclastogenesis by fatty acids. Endocrinology 149, 5688–5695 (2008).
47. Chauhan, S. et al. In-vitro osteoblast proliferation and in-vivo anti-osteoporotic activity of Bombax ceiba with quantication of
Lupeol, gallic acid and β-sitosterol by HPTLC and HPLC. BMC Complement. Altern. Med. 18, 1–12 (2018).
48. Pereira, J. V., Modesto-Filho, J. & deFAgra, M. & Barbosa-Filho, J. M.,. Plant and plant-derived compounds employed in prevention
of the osteoporosis. Acta Farmaceut. Bonaerense 21, 223–234 (2002).
49. Min, J. et al. Analysis of anti-osteoporosis function of chlorogenic acid by gene microarray proling in ovariectomy rat model.
Biosci. Rep. 38, 20180775 (2018).
50. Zhou, R. P. et al. Chlorogenic acid prevents osteoporosis by Shp2/PI3K/Akt pathway in ovariectomized rats. PLoS ONE 11,
e0166751 (2016).
51. Gür, A. et al. Possible pathogenetic role of new cytokines in postmenopausal osteoporosis and changes during calcitonin plus
calcium therapy. Rheumatol. Int. 22, 194–198 (2002).
52. Armour, K. E., Van’T Hof, R. J., Grabowski, P. S., Reid, D. M. & Ralston, S. H. Evidence for a pathogenic role of nitric oxide in
inammation-induced osteoporosis. J. Bone Miner. Res. 14, 2137–2142 (1999).
53. Mancini, L., Moradi-Bidhendi, N., Becherini, L., Martineti, V. & MacIntyre, I. e biphasic eects of nitric oxide in primary rat
osteoblasts are cGMP dependent. Biochem. Biophys. Res. Commun. 274, 477–481 (2000).
54. Damoulis, P. D. & Hauschka, P. V. Cytokines induce nitric oxide production in mouse osteoblasts. Biochem. Biophys. Res. Commun.
201, 924–931 (1994).
55. Van’T Hof, R. J. & Ralston, S. H. Nitric oxide and bone. Immunology 103, 255–261 (2001).
56. Mogi, M., Kinpara, K., Kondo, A. & Togari, A. Involvement of nitric oxide and biopterin in proinammatory cytokine-induced
apoptotic cell death in mouse osteoblastic cell line MC3T3-E1. Biochem. Pharmacol. 58, 649–654 (1999).
57. Navarro, V. J. et al. Liver injury from herbal and dietary supplements. Hepatology 65, 363–373 (2017).
58. Guañabens, N. & Parés, A. Osteoporosis in chronic liver disease. Liver Int. 38, 776–785 (2018).
59. Scholz, S. Zebrash embryos as an alternative model for screening of drug-induced organ toxicity. Arch. Toxicol. 87, 767–769
(2013).
60. Korzh, S. et al. Requirement of vasculogenesis and blood circulation in late stages of liver growth in zebrash. BMC Dev. Biol. 8,
1–15 (2008).
61. Nam, H.-S. et al. Expression of miRNA-122 induced by liver toxicants in zebrash. BioMed Res. Int. 2016, 2 (2016).
62. iagarajan, S. K. et al. Evaluation of the eect of aqueous Momordica charantia Linn. extract on zebrash embryo model through
acute toxicity assay assessment. Evid.-Based Complement. Altern. Med. 2019, 2 (2019).
63. Karani, L., Tolo, F., Karanja, S. & Khayeka, C. Safety of Prunus africana and Warburgia ugandensis in asthma treatment. S. Afr. J.
Bot. 88, 183–190 (2013).
64. Gathumbi, P., Mwangi, J., Mugera, G. & Njiro, S. Biochemical and haematological changes mediated by a chloroform extract of
Prunus africana stem bark in rats. Pharm. Biol. 38, 374–378 (2000).
Acknowledgements
is research was funded by the framework of International Cooperation Program (Korea-South Africa Coop-
erative Research Project for Excavation of Candidate Resources of Complementary and Alternative Medicine)
managed by National Research Foundation of Korea (Grant no. 2017093655 and KIOM: D17470). Additionally,
this work was also supported by Development of Foundational Techniques for the Domestic Production of Herbal
Medicines (K18405), Development of Sustainable Application for Standard Herbal Resources (KSN2012320),
Korea Institute of Oriental Medicine through the Ministry of Science and ICT, Republic of Korea. Partially, this
work was also supported by a grant from the Ministry of Trade, Industry and Energy, Republic of Korea (Grant
no. 2019-10063396).
Author contributions
R.K. conceived the original research plans, collected test materials, and wrote this manuscript. K.S. conducted
the experiments on TRAP assays. N.H.Y. and J.L. conducted the extractions of the samples and carried out the
chemical proling. S.K.Y., J.H.S., and G.C. carried out the botanical authentication of the P.africana used in this
study and wrote the manuscript. Y.G.K. performed the statistical analysis and wrote this manuscript. F.O., A.M.,
and G.N.K. collected the sample, wrote, and revised the manuscript. D.O. wrote and revised the manuscript. H.K.
Content courtesy of Springer Nature, terms of use apply. Rights reserved
12
Vol:.(1234567890)
Scientic Reports | (2022) 12:7044 | https://doi.org/10.1038/s41598-022-10629-7
www.nature.com/scientificreports/
and K.S.H. conducted the hepatotoxicity assay on zebrash. M.G.M. conducted experiments on phytochemical
proling and revised the manuscript. Y.K. technically supervised all the experiments and is the corresponding
author. All authors reviewed the manuscript.
Competing interests
e authors declare no competing interests.
Additional information
Supplementary Information e online version contains supplementary material available at https:// doi. org/
10. 1038/ s41598- 022- 10629-7.
Correspondence and requests for materials should be addressed to Y.K.
Reprints and permissions information is available at www.nature.com/reprints.
Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and
institutional aliations.
Open Access is article is licensed under a Creative Commons Attribution 4.0 International
License, which permits use, sharing, adaptation, distribution and reproduction in any medium or
format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the
Creative Commons licence, and indicate if changes were made. e images or other third party material in this
article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the
material. If material is not included in the article’s Creative Commons licence and your intended use is not
permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from
the copyright holder. To view a copy of this licence, visit http:// creat iveco mmons. org/ licen ses/ by/4. 0/.
© e Author(s) 2022
Content courtesy of Springer Nature, terms of use apply. Rights reserved
1.
2.
3.
4.
5.
6.
Terms and Conditions
Springer Nature journal content, brought to you courtesy of Springer Nature Customer Service Center GmbH (“Springer Nature”).
Springer Nature supports a reasonable amount of sharing of research papers by authors, subscribers and authorised users (“Users”), for small-
scale personal, non-commercial use provided that all copyright, trade and service marks and other proprietary notices are maintained. By
accessing, sharing, receiving or otherwise using the Springer Nature journal content you agree to these terms of use (“Terms”). For these
purposes, Springer Nature considers academic use (by researchers and students) to be non-commercial.
These Terms are supplementary and will apply in addition to any applicable website terms and conditions, a relevant site licence or a personal
subscription. These Terms will prevail over any conflict or ambiguity with regards to the relevant terms, a site licence or a personal subscription
(to the extent of the conflict or ambiguity only). For Creative Commons-licensed articles, the terms of the Creative Commons license used will
apply.
We collect and use personal data to provide access to the Springer Nature journal content. We may also use these personal data internally within
ResearchGate and Springer Nature and as agreed share it, in an anonymised way, for purposes of tracking, analysis and reporting. We will not
otherwise disclose your personal data outside the ResearchGate or the Springer Nature group of companies unless we have your permission as
detailed in the Privacy Policy.
While Users may use the Springer Nature journal content for small scale, personal non-commercial use, it is important to note that Users may
not:
use such content for the purpose of providing other users with access on a regular or large scale basis or as a means to circumvent access
control;
use such content where to do so would be considered a criminal or statutory offence in any jurisdiction, or gives rise to civil liability, or is
otherwise unlawful;
falsely or misleadingly imply or suggest endorsement, approval , sponsorship, or association unless explicitly agreed to by Springer Nature in
writing;
use bots or other automated methods to access the content or redirect messages
override any security feature or exclusionary protocol; or
share the content in order to create substitute for Springer Nature products or services or a systematic database of Springer Nature journal
content.
In line with the restriction against commercial use, Springer Nature does not permit the creation of a product or service that creates revenue,
royalties, rent or income from our content or its inclusion as part of a paid for service or for other commercial gain. Springer Nature journal
content cannot be used for inter-library loans and librarians may not upload Springer Nature journal content on a large scale into their, or any
other, institutional repository.
These terms of use are reviewed regularly and may be amended at any time. Springer Nature is not obligated to publish any information or
content on this website and may remove it or features or functionality at our sole discretion, at any time with or without notice. Springer Nature
may revoke this licence to you at any time and remove access to any copies of the Springer Nature journal content which have been saved.
To the fullest extent permitted by law, Springer Nature makes no warranties, representations or guarantees to Users, either express or implied
with respect to the Springer nature journal content and all parties disclaim and waive any implied warranties or warranties imposed by law,
including merchantability or fitness for any particular purpose.
Please note that these rights do not automatically extend to content, data or other material published by Springer Nature that may be licensed
from third parties.
If you would like to use or distribute our Springer Nature journal content to a wider audience or on a regular basis or in any other manner not
expressly permitted by these Terms, please contact Springer Nature at
onlineservice@springernature.com
... [31,32] Beta-sitosterol can also inhibit macrophage IL-6 activity, reduce the secretion of inflammatory factors such as IL-1, TNF, and play an anti-inflammatory role. [33] It was found that ginsenoside rh2 possesses pharmacological effects such as hypoglycemic, hypolipidemic, antioxidant, and anti-atherosclerotic. Hou et al [34] found that ginsenoside rh2 has the effect of increasing the activity of antioxidant enzymes such as SOD, reducing ROS, and decreasing oxidative stress. In addition, ginsenoside rh2 has anti-inflammatory activity, which can inhibit the release of pro-inflammatory cytokines and inhibit the inflammatory signaling pathway, effectively improving the inflammatory response. ...
Article
Full-text available
Diabetes mellitus (DM) is a chronic metabolic disease that predisposes to chronic damage and dysfunction of various organs, including leading to erectile dysfunction (ED) and asthenospermia. Literature suggests that ginseng plays an important role in the treatment and management of DM. Ginseng may have a therapeutic effect on the complications of DM-induced ED and asthenospermia. The study aimed to explore the mechanisms of ginseng in the treatment of DM-induced ED and asthenospermia following the Traditional Chinese Medicine (TCM) theory of “treating different diseases with the same treatment.” This study used network pharmacology and molecular docking to examine the potential targets and pharmacological mechanism of Ginseng for the treatment of DM-induced ED and asthenospermia. The chemical ingredients and targets of ginseng were acquired using the Traditional Chinese Medicine Systems Pharmacology database and analysis platform. The targets of DM, ED, and asthenospermia were extracted with the GeneCards and Online Mendelian Inheritance in Man databases. A protein–protein interaction network analysis was constructed. The Metascape platform was applied for analyzing the gene ontology and Kyoto Encyclopedia of Genes and Genomes pathways. AutoDock Vina was used to perform molecular docking. Network pharmacology revealed that the main active components of the target of action were kaempferol, beta-sitosterol, ginsenoside rh2, stigmasterol, and fumarine. Core targets of the protein–protein interaction network included TNF, IL-1β, AKT1, PTGS2, BCL2, and JUN. Kyoto Encyclopedia of Genes and Genomes enrichment analysis showed that they were mainly involved in AGE-RAGE signaling pathway in diabetic complications, TNF signaling pathway, Lipid and atherosclerosis. The interactions of core active components and targets were analyzed by molecular docking. Ginseng may play a comprehensive therapeutic role in the treatment of DM-induced ED and asthenospermia through “multicomponent, multi-target, and multi-pathway” biological mechanisms such as inflammation and oxidative stress.
... If we compare the diseases identified in this study with those reported by previous research, we can observe a great similarity with local populations using P. africana in other countries. Indeed, a number of ethnobotanical studies have found that not only local populations but also the scientific community are attracted to this species because of its effective active ingredients against benign prostatic hyperplasia and prostate hypertrophy (Abera 2014;Ibrahim et al. 2016;Jared Misonge et al. 2019;Qureshi et al. 2009;Tugume et al. 2016;Komakech and Kang 2019;Komakech et al. 2020;Komakech et al. 2022), an increasingly common problem in older men. Beyond the prostate, our ethnobotanical surveys showed that local people also use P. africana in the treatment of other diseases. ...
Article
Full-text available
Background: Prunus africana (Hook. f) Kalkman (Rosaceae) is a multi-purpose species with important utility value for the populations that depend on it. Indeed, local populations living in community forests use P. africana. The objective of this study is to document the different uses of P. africana by local populations in traditional medicine for a better vulgarization for a sustainable management of the resource. Methods: Ethnobotanical surveys were conducted in four P. africana distribution areas in North Kivu. Data were collected on the basis of a survey from 221 informants. Statistical analysis and calculation of ethnobotanical indices were performed using R 4.1.2 software. Results: Results of the investigations show that P. africana is by far used in medical practices (96.83%). It is also used as wood energy (60.18 %), charcoal (40.27 %), in handicrafts (7.24 %), construction (6.79 %), food (2.26 %) and traditional rites (0.45 %). In traditional pharmacopoeia, the characterization of the uses allowed us to identify 23 diseases for which P. africana extracts are used to treat them. The bark and leaves are the most used organs in the recipes. The decoction (99.5%), the macerated (10.41%) and the powders (7.69%) are the galenic or pharmaceutical forms in which the local populations prepare the remedies. The potions prepared are mainly administered orally (99.5%). Conclusion: The multiple forms of use of P. africana by the populations of North Kivu, both in medicine and in other categories of use, constitute a threat to its survival. For a sustainable management, the results of this study reveal that it is possible to substitute the species P. africana with other plant species with similar potential in order to limit its overexploitation
Article
The stem-bark of Prunus africana (Hook.f.) Kalkman (African cherry) is traditionally used for the treatment of stomach ache, chest pain, malaria, fever, diabetes and high blood pressure. A large number of constituents have been isolated from the stem-bark including β-sitosterol and ursolic acid, which were reported to have anti-inflammatory activity. The aim of the study was to establish chemotypic variation among Prunus africana samples collected from three African countries, Cameroon, Democratic Republic of Congo (DRC) and Zimbabwe. Ninety-five stem-bark samples were collected from a total of nineteen populations, 11 in Cameroon (n = 55), 4 in Democratic Republic of Congo (n = 20) and 4 in Zimbabwe (n = 20). The samples were extracted with ethyl acetate, dichloromethane and methanol (1 g in 10 mL, 1 g in 10 mL and 2 mg in 5 µL, respectively). Chemometric analysis of chromatographic data obtained from high-performance thin layer chromatography (HPTLC), ultra performance liquid chromatography coupled with mass spectrometry (UPLC–MS), one-dimensional gas chromatography-time-of-flight-mass spectrometry (1D GC–ToF–MS) and spectroscopic data obtained from proton nuclear magnetic resonance (1H NMR) were done using MetaboAnalyst 4.0 software to explore chemotypic variation. A sample from each country was analysed by two-dimensional gas chromatography-time-of-flight-mass spectrometry (2D GC–ToF–MS) to resolve compounds that co-eluted in the 1D GC analysis. Quantification of selected compounds (β-sitosterol and ursolic acid) were done using a validated ultra performance liquid chromatography-photodiode-array detection (UPLC–PDA) method. The chemical profiles obtained from each of the four techniques were very similar for the samples from each country. The HPTLC profiles for Zimbabwe samples were distinctly different from the other two countries with a unique band at Rf value of 0.80. Multivariate analysis of the HPTLC data (using rTLC software), UPLC–MS, 1D GC–ToF–MS and 1H–NMR data revealed three clusters that were country specific. Chemometric analysis through the construction of PCA and a PLS–DA scores plots was performed, followed by the construction of variable important in projection plots, whereby marker compounds contributing significantly to the separation of the three clusters were selected and tentatively identified. Quantitative analysis revealed that samples from Zimbabwe contained higher levels of β-sitosterol (29.2 – 119 µg/g dry weight (DW) of the extract) compared to DRC and Cameroon, while samples from Cameroon were found to contain the highest levels of ursolic acid (1.80 – 141 µg/g DW of the extract). HPTLC, UPLC–MS, 1D and 2D GC–ToF–MS and 1H–NMR are valid tools for quality control of P. africana plant materials after revealing three clusters of samples collected from the three countries. Cameroon samples had high content of ursolic acid, while Zimbabwe samples showed high levels of β-sitosterol. The number of compounds detected across the three countries by 1D GC–ToF–MS was less compared to 2D GC–ToF–MS, which enhanced separation and resolution.
Article
Full-text available
Prunus dulcis is one of the most widely cultivated species in the world. Its fruit (almond) is rich in various nutritious and bioactive compounds that exert several beneficial effects. The aim of this study was to determine the chemical profile and evaluate the biological potential in vitro of almond shell extracts. The chemical analysis of shell extracts led to the identification of 15 compounds by HPLC-DAD, of which 11 were first detected in the almond plant. Twenty-six volatile compounds were identified by the GC-MS technique; among them, seven were firstly detected in the studied plant. For the biological activities, the extracts demonstrated moderate inhibition potential against the antioxidant, antidiabetic, and cytotoxic activities. The methanol extract at 50 µg/mL showed the highest antioxidant (45%) and antidiabetic activities (45% against alpha-glucosidase and 31% against alpha-amylase extracts), while the cyclohexane and dichloromethane at 50 µg/mL showed the highest cytotoxic activity towards Hela (32.2% with cyclohexane) and RAW 264-7 (45% with dichloromethane). Overall, these findings demonstrate the potential of almond shell extracts as a source of bioactive compounds that could be applied in the pharmaceutical and medical fields.
Preprint
Full-text available
Prunus dulcis is one of the most widely cultivated species in the world. Its fruit (almond) is rich in various nutritious and bioactive compounds that exert several beneficial effects. The aim of this study was to determine the chemical profile and evaluate the biological potential in vitro of almond shell extracts. The chemical analysis of shell extracts led to the identification of 15 com-pounds by HPLC-DAD of which 11 were first detected in the almond plant. Twenty-six volatile compounds were identified by the GC-MS technique, among them; seven were firstly detected in the studied plant. For the biological activities, the extracts demonstrated moderate inhibition potential against the antioxidant, antidiabetic, and cytotoxic activities. The methanol extract at 50 µg/mL showed the highest antioxidant (45%) and antidiabetic activities (45% against alpha-glucosidase and 31% against alpha-amylase extracts), while the cyclohexane and dichloromethane at 50 µg/mL showed the highest cytotoxic activity towards Hela (32.2% with cyclohexane), and RAW 264-7 (45% with dichloromethane). Overall, these findings demonstrate the potential of almond shell extracts as a source of bioactive compounds that could be applied in the pharmaceutical and medical fields.
Article
Ethnopharmacological relevance: Prunus africana (Hook.f.) Kalkman (Rosaceae), commonly known as "Pygeum" or "African cherry", occurs in mainland montane forest "islands" scattered across sub-Saharan Africa, Madagascar, and some surrounding islands. Traditionally, decoctions of the stem-bark are taken orally for the treatment of a wide variety of conditions, such as benign prostatic hyperplasia (BPH), stomach ache, chest pain, malaria, heart conditions, and gonorrhoea, as well as urinary and kidney diseases. The timber is used to make axe handles and for other household needs. The dense wood is also sawn for timber. Aim: The fragmented information available on the ethnobotany, phytochemistry, and biological activities of the medicinally important P. africana was collated, organised, and analysed in this review, to highlight knowledge voids that can be addressed through future research. Materials and methods: A bibliometric analysis of research output on P. africana was conducted on literature retrieved, using the Scopus® database. The trend in the publications over time was assessed and a network analysis of collaborations between countries and authors was carried out. Furthermore, a detailed review of the literature over the period 1971 to 2021, acquired through Scopus, ScienceDirect, SciFinder, Pubmed, Scirp, DOAJ and Google Scholar, was conducted. All relevant abstracts, full-text articles and various book chapters on the botanical and ethnopharmacological aspects of P. africana, written in English and German, were consulted. Results: A total of 455 documents published from 1971 to 2021, were retrieved using the Scopus search. Analysis of the data showed that the majority of these documents were original research articles, followed by reviews and lastly a miscellaneous group comprising conference papers, book chapters, short surveys, editorials and letters. Data were analysed for annual output and areas of intense research focus, and countries with high research output, productive institutions and authors, and collaborative networks were identified. Prunus africana is reported to exhibit anti-inflammatory, analgesic, antimicrobial, anti-oxidant, antiviral, antimutagenic, anti-asthmatic, anti-androgenic, antiproliferative and apoptotic activities amongst others. Phytosterols and other secondary metabolites such as phenols, triterpenes, fatty acids, and linear alcohols have been the focus of phytochemical investigations. The biological activity has largely been ascribed to the phytosterols (mainly 3-β-sitosterol, 3-β-sitostenone, and 3-β-sitosterol-glucoside), which inhibit the production of prostaglandins in the prostate, thereby suppressing the inflammatory symptoms associated with BPH and chronic prostatitis. Conclusions: Many of the ethnobotanical assertions for the biological activity of P. africana have been confirmed through in vitro and in vivo studies. However, a disparity exists between the biological activity of the whole extract and that of single compounds isolated from the extract, which were reported to be less effective. This finding suggests that a different approach to biological activity studies should be encouraged that takes all secondary metabolites present into consideration. A robust technique, such as multivariate biochemometric data analysis, which allows for a holistic intervention to study the biological activity of a species is suggested. Furthermore, there is a need to develop rapid and efficient quality control methods for both raw materials and products to replace the time-consuming and laborious methods currently in use.
Article
Full-text available
Semen of Cuscuta chinensis has been reported to have an anti‐osteoporosis effect, however, the components which account for the anti‐osteoporosis effect have not been clarified. In this work we propose a biochemometrics strategy that integrates quantitation, anti‐osteoporosis evaluation in zebrafish, and grey relationship analysis for the identification of anti‐osteoporosis components from the semen of Cuscuta chinensis. In the beginning, a precise and accurate liquid chromatography‐tandem mass spectrometry method was established for simultaneous quantitation of seven major components in crude and salt‐processed Cuscuta chinensis. The mode of multiple reaction monitoring was used. Chloramphenicol was selected as the internal standard. The method showed good linearity and repeatability. The recovery rates of each component ranged from 95.4 to 103.9%. The precisions of intra‐day and inter‐day were all within 5.0%. The method was then applied for quantitation of the seven major components in 11 batches of crude and salt‐processed Cuscuta chinensis. Subsequently, the anti‐osteoporosis effects of crude and salt‐processed Cuscuta chinensis were evaluated in zebrafish. Principle component analysis, grey relationship analysis, and partial least squares regression were applied for deciphering the relationship between the contents of seven major components and the anti‐osteoporosis effects. Hyperin, p‐hydroxycinnamic acid, and astragalin were found to be the major anti‐osteoporosis components.
Article
Full-text available
Interleukin 6 (IL-6) is involved both in immune responses and in inflammation, hematopoiesis, bone metabolism and embryonic development. IL-6 plays roles in chronic inflammation (closely related to chronic inflammatory diseases, autoimmune diseases and cancer) and even in the cytokine storm of corona virus disease 2019 (COVID-19). Acute inflammation during the immune response and wound healing is a well-controlled response, whereas chronic inflammation and the cytokine storm are uncontrolled inflammatory responses. Non-immune cells and immune cells, cytokines such as IL-1β, IL-6 and tumor necrosis factor alpha (TNFα) and transcription factors nuclear factor-kappa B (NF-κB) and signal transducer and activator of transcription 3 (STAT3) play central roles in inflammation. Synergistic interactions between NF-κB and STAT3 induce the hyper-activation of NF-κB followed by the production of various inflammatory cytokines. Because IL-6 is a NF-κB target, simultaneous activation of NF-κB and STAT3 in non-immune cells triggers a positive feedback loop of NF-κB activation by the IL-6–STAT3 axis. This positive feedback loop is called the IL-6 amplifier (IL-6 Amp) and is a key player in the local initiation model, which states that local initiators, such as senescence, obesity, stressors, infection, injury and smoking, trigger diseases by promoting interactions between non-immune cells and immune cells. This model counters dogma that holds that autoimmunity and oncogenesis are triggered by the breakdown of tissue-specific immune tolerance and oncogenic mutations, respectively. The IL-6 Amp is activated by a variety of local initiators, demonstrating that the IL-6–STAT3 axis is a critical target for treating diseases.
Article
Full-text available
Osteoporosis is one of the main health problems in the world today characterized by low bone mass and deterioration in bone microarchitecture. In recent years, the use of natural products approach to treat it has been in the increase. In this study, in vitro antiosteoporosis activity and hepatotoxicity of P. jamasakura bark extracts were evaluated. Methods. Mouse bone marrow macrophage (BMM) cells were incubated with tartrate-resistant acid phosphate (TRAP) buffers and p-nitrophenyl phosphate and cultured with different P. jamasakura bark extracts at concentrations of 0, 6.25, 12.5, 25, and 50 μg/ml in the presence of the receptor activator of nuclear factor kappa-Β ligand (RANKL) for 6 days. The osteoclast TRAP activity and cell viability were measured. Nitric oxide (NO) assay was conducted using murine macrophage-like RAW 264.7 cells treated with P. jamasakura ethanolic and methanolic bark extracts at concentrations of 0, 6.25, 12.5, 25, 50, 100, and 200 μg/ml. For hepatotoxicity assessment, zebrafish larvae were exposed to P. jamasakura bark extracts, 0.05% dimethyl sulfoxide as a negative control, and 5 μM tamoxifen as a positive control. The surviving larvae were anesthetized and assessed for hepatocyte apoptosis. Results. TRAP activity was significantly inhibited ( < 0.001) at all concentrations of P. jamasakura extracts compared to the control treatment. At 50 μg/ml, both ethanolic and methanolic extracts of P. jamasakura exhibited significant ( < 0.01) BMM cell viability compared to the control treatment. P. jamasakura ethanolic and methanolic extracts had significant inhibitory ( < 0.01) effects on lipopolysaccharide (LPS)-induced NO production at 200 μg/ml and exhibited significant ( < 0.01) and ( < 0.05) stimulative effects, respectively, on RAW 264.7 cell viability. No overt hepatotoxicity was observed in the liver of zebrafish larvae in any of the treatments. Conclusion. The TRAP activity of P. jamasakura bark gives a foundation for further studies to enhance future development of antiosteoporosis drug. 1. Introduction Osteoporosis is a major global public health problem characterized by low bone mass and a deterioration of bone microarchitecture [1]. People suffering from an osteoporosis have increased risk of fractures [2, 3]. It is one of the major causes of morbidity in older people [4] due to imbalance between the bone formation and resorption rate [3]. Several factors have been associated with an increased risk of osteoporosis, including menopause, sex steroid deficiency, and aging [2, 3]. Chronic inflammation has long been associated with a broad range of noninfectious diseases [5], and recent studies suggest that inflammation is one of the key factors that influence bone turnover, leading to osteoporosis [4, 6]. In fact, proinflammatory cytokines have been implicated as primary mediators of accelerated bone loss during menopause [7]. Currently, the treatment of osteoporosis focuses on inhibition of bone resorption by osteoclasts and/or increase in bone formation by osteoblasts [1]. A number of conventional treatment options for osteoporosis are available such as bisphosphonates and estrogen but their adverse effects including burning sensation and gastrointestinal tract disturbances associated with these therapies limit their use [8]. Consequently, exploring the use of natural products in the treatment of osteoporosis may offer a better alternative to avoid the side effects of the conventional therapies [8]. Over the years, herbal medicines have been used to treat osteoporosis [1, 9] and as a crucial substitute of anti-inflammatory drugs [10]. The plants used in traditional medicine for the treatment of inflammation- and osteoporosis-related conditions are those of Prunus (family Rosaceae), including Prunus jamasakura f. hortensis (Maxim.) (Koidz) (Scientific synonym Prunus x lannesiana (Carrière) E. H. Wilson) [11]. P. jamasakura is native to Korea and Japan and has been used to treat several diseases in folk medicine including inflammatory diseases [12], cough, and food poisoning [11]. These medicinal activities have been attributed to several compounds including sakuranetin, sakuranin, naringenin, and genistein, found in its stem bark (Pruni cortex) [11]. Despite its myriad therapeutic uses, there are currently no studies on the antiosteoporosis activity of P. jamasakura. Hence, this study evaluated the in vitro antiosteoporosis activity of the ethanolic and methanolic bark extracts of P. jamasakura. In addition, we evaluated the hepatotoxicity of the extracts in zebrafish (Danio rerio) larvae and conducted the HPLC chemical profiling of the compounds in the extracts. This study may therefore provide the foundation for further studies regarding P. jamasakura for future drug development to treat and manage osteoporosis. 2. Materials and Methods 2.1. Chemicals All of the chemicals and solvents used in this study were of analytical grade. Acetonitrile (Fisher Scientific, UK) and trifluoroacetic acid (Sigma-Aldrich, USA) were of HPLC grade. Ultrapure water from a Milli-Q system (Millipore, USA) was used for the mobile phase preparation. Naringenin, genistein, and sakuranetin were purchased from ChemFaces (Wuhan, Hubei, China) and were used as the standard components. 2.2. Plant Material and Preparation of Extract The stem bark of P. jamasakura (Pruni cortex) was procured from Daejeon, South Korea. The voucher specimen number KIOM201501013821A was deposited in the Korean Herbarium of Standard Herbal Resources (Index Herbarium Code: KIOM) at the Korea Institute of Oriental Medicine (KIOM), South Korea. The stem bark of the sample was ground using a steel pulverizing machine (250G New Type Pulverizing Machine, Model RT-N04-2V, Taiwan) at 25,000 rpm to obtain a fine powder. The maceration and concentration process was done following the previous method [13]. 30 g of the fine powder was extracted via maceration using 600 ml of 100% methanol, 100% ethanol, and distilled water. The extracts were filtered using Whatman filter no. 1 after 24 h and concentrated under a vacuum reduced pressure at 40°C, 70 rpm, using an EYELA N-1200B (Tokyo Rikakikai Co. Ltd., Japan) efficient rotary evaporator. The concentrated extract was then vacuum dried. The resultant dried extract was used for subsequent HPLC phytochemical analysis, nitric oxide (NO) assay, tartrate-resistant acid phosphatase (TRAP) assay, and hepatotoxicity evaluation. 2.3. HPLC Chemical Profiles of Prunus jamasakura The method used was modified from that of the previous study [13]. The chemical standards naringenin, genistein, and sakuranetin used in this study were each dissolved in methanol at 1 mg/ml to make a stock solution and then further diluted to 20 μg/ml in methanol for the HPLC analysis. Similarly, P. jamasakura extracts were dissolved in methanol at 10 mg/ml and filtered using a 0.2 mm syringe membrane filter (Whatman Ltd., Maidstone, UK) for analysis. Separation was performed using an HPLC system (Dionex Ultimate 3000; Thermo Fisher Scientific, Sunnyvale, CA, USA) comprising a pump, an auto sampler, a column oven, and a diode array UV/VIS detector. The chromatograms were analyzed using the Chromeleon software system (version 7). The components of the P. jamasakura extracts were separated using a Gemini C18 column (4.6 × 250 mm, 5 μm) (Phenomenex, Torrance, CA, USA) at 40°C. An injection volume of 10 μl was used at a detection wavelength of 280 nm. The mobile phase, consisting of ultrapure water with 0.1% trifluoroacetic acid (A) and acetonitrile (B), was eluted at a flow rate of 1.0 ml/min. The gradient elution program used was as follows: 3% () B at 0–2 min; 3–35% (B) at 2–30 min; 35–50% (B) at 30–31 min; 50% (B) at 31–35 min; 50–100% (B) at 35–40 min; and 100% (B) at 40–45 min. 2.4. Inhibitory Effect of P. jamasakura on No Generation and Osteoclastogenesis 2.4.1. Cell Culture Murine macrophage-like RAW 264.7 cells (ATCC; Manassas, VA, USA) were cultured in Dulbecco’s modified Eagle medium supplemented with 10% fetal bovine serum (FBS) and 1% antibiotics following previously described method [13]. Mouse bone marrow macrophages (BMMs) were cultured in a proliferation medium (an α-MEM medium with 10% FBS and macrophage-colony stimulating factor (M-CSF) (60 ng/ml)) following the previously described method [14, 15]. To differentiate the osteoclasts, BMMs were cultured in a proliferation medium with RANKL (100 ng/ml) for 6 days. 2.4.2. NO Assay Murine macrophage-like RAW 264.7 cells were treated with the ethanolic and methanolic extracts of P. jamasakura samples at various concentrations of 0, 12.5, 25, 50, 100, and 200 μg/ml and cultured for 1 h prior to lipopolysaccharide (LPS) stimulation for 24 h following a previously described method [13]. The nitrite levels in the culture media were determined by incubation with Griess reagent (1% sulfanilamide, 0.1% naphthylethylenediamine dihydrochloride, and 2.5% phosphoric acid) for 5 min. The absorbance was measured at 570 nm using a microplate reader (VersaMax, Molecular Devices). The quantity of nitrite in the samples was calculated using the concentration of sodium nitrite as a standard. Cell viability was analyzed using a cell counting kit assay (Dojindo). Cells were plated in a 96-well plate and treated with different concentrations of P. jamasakura extracts at concentrations of 0, 12.5, 25, 50, 100, and 200 μg/ml for ethanolic and methanolic extracts for 24 h. After incubating with the cell counting kit (CCK) solutions and the cells for 1 h, the absorbance was measured at 450 nm using a microplate reader (VersaMax). The results are presented as a percentage of the control. 2.4.3. TRAP Assay and BMM Cell Viability The measurement of osteoclast TRAP activity was based on the generation of absorbance by incubating BMM cells with TRAP buffer (50 mM sodium tartrate, 0.12 M sodium acetate, and pH 5.2) and p-nitrophenyl phosphate (1 mg/ml) for 15 min based on method previously described [14]. For TRAP staining, the cells were incubated with TRAP buffer containing naphthol AS-MX phosphate (0.1 mg/ml) and Fast Red Violet (0.5 mg/ml). The BMM cells were then cultured with the different ethanolic and methanolic P. jamasakura extracts at different concentrations of 0, 6.25, 12.5, 25, and 50 μg/ml in the presence of RANKL for 6 days. The osteoclast TRAP activity was determined using a colorimetric assay with p-nitrophenyl phosphate as a substrate. The cell viability was determined using cell counting kit-8 (WST-8/CCK8; Dojindo), according to the manufacturer’s instructions. For the measurement of cell viability, cells were plated in 96-well plates and treated with ethanolic and methanolic P. jamasakura extracts at different concentrations of 0, 6.25, 12.5, 25, and 50 μg/ml for 24 h. After incubating with the CCK solutions and the cells for 1 h, the absorbance was measured at 450 nm using a microplate reader (Versa Max). The results are presented as a percentage of the control. 2.5. Hepatotoxicity Assay in Zebrafish (Danio rerio) Larvae Zebrafish larvae were bred under standard conditions as previously described [16] (Westerfield, 2000). At 90 h postfertilization (hpf), the larvae were transferred to a transparent 24-well plate (N = 10/well) with 1 ml of an embryonic medium. The larvae were then exposed to water, ethanolic, and methanolic P. jamasakura extracts at various concentrations of 50, 100, and 200 μg/ml for ethanolic and methanolic, and water extracts from 96 to 120 hpf. Dimethyl sulfoxide (DMSO) was used as a negative control while 5 μM of tamoxifen (Sigma-Aldrich, St. Louis, MO, USA) was used as a positive control. To obtain images, the larvae were anesthetized in tricaine (Sigma-Aldrich), mounted in 3% methyl cellulose (Sigma-Aldrich), and observed under a Leica MZ10F stereomicroscope equipped with a Leica DFC425 camera and Leica application Suite software (version 4.5). 2.6. Statistical Analysis Data are represented as the mean ± standard deviation. Statistical significance between groups was analyzed using Student’s t-test. values <0.05 were considered statistically significant. 3. Results and Discussion 3.1. HPLC Chemical Profiles of Prunus jamasakura HPLC is a versatile, reproducible chromatographic technique for the estimation and detection of secondary metabolites in plants [17]. In this study, the phytochemical components of P. jamasakura based on the HPLC fingerprinting of their methanol, ethanol, and water extracts at 203, 254, 280, and 320 nm UV wavelengths (data not shown) were conducted. Among the four types of UV wavelengths, good separation and selectivity were observed at 280 nm. The distinct profiling patterns of the components were confirmed regardless of the type of solvent used in Figure 1(a). (a)
Article
Full-text available
Osteoporosis is a metabolic bone disorder which causes bone loss in course of time leading to increasing the fracture risk. The disease is often silent and known only when fractures occur. Both men and women are equally affected. But women are more susceptible to osteoporosis. The main cause of fragility fractures in women is due to the estrogen deficiency. For the treatment of Osteoporosis the potential biological use of traditional medicines have been exposed to logical assessment since synthetic medications are considered to have a wide range of side effects and they lack efficacy. Hence broad research is to be done to examine the therapeutic plants to be utilized as an option for the treatment of Osteoporosis. The present survey work demonstrates the data on medicinal plants with their common name, phytochemical constituents, reported pharmacological activities and their Osteoporotic action.
Article
Full-text available
Momordica charantia Linn., commonly known as bitter gourd, has many protective roles due to its medicinal value as it contains bioactive components. However, this extract showed possible toxicity effect on zebrafish embryo. Thus this study was designed to differentiate the toxicity activities in two types of M. charantia sample which are Indian and Chinese M. charantia , as well as to compare between two different aqueous extraction methods, hot and cold aqueous method, using zebrafish embryo assay assessment. It was observed that the survival rate of zebrafish embryo decreased as the concentration of test extract increased for all samples of M. charantia . The LC 50 values of hot aqueous Chinese M. charantia , hot aqueous Indian M. charantia , and cold aqueous Chinese M. charantia were 144.54 μ g/ml, 199.53 μ g/ml, and 251.19 μ g/ml, respectively. However, cold aqueous Indian M. charantia has a higher LC 50 which was not in the range of the tested concentration. Hatchability of Danio rerio embryo reduced as the concentration of M. charantia extract increased while no hatching was observed in the highest concentration (1000 μ g/ml). Scoliosis of zebrafish larvae was only seen in higher concentrations (125-1000 μ g/ml) of extract. The heartbeat of zebrafish larvae treated with M. charantia extract was within the normal range, 120-180 bpm, but at higher concentrations (125-1000 μ g/ml) the heartbeat differed for all samples of test extract. Hence, although this plant extract was safe to be consumed due to its pharmaceutical effect, it still exhibited mild toxicity effect at higher concentration when it was evaluated on zebrafish embryo.
Article
Full-text available
Osteoporosis is a systemic disease with progressive bone loss. The bone loss is associated with an imbalance between bone resorption via osteoclasts and bone formation via osteoblasts. Other cells including T cells, B cells, macrophages, and osteocytes are also involved in the pathogenesis of osteoporosis. Different cytokines from activated macrophages can regulate or stimulate the development of osteoclastogenesis-associated bone loss. The fusion of macrophages can form multinucleated osteoclasts and, thus, cause bone resorption via the expression of IL-4 and IL-13. Different cytokines, endocrines, and chemokines are also expressed that may affect the presentation of macrophages in osteoporosis. Macrophages have an effect on bone formation during fracture-associated bone repair. However, activated macrophages may secrete proinflammatory cytokines that induce bone loss by osteoclastogenesis, and are associated with the activation of bone resorption. Targeting activated macrophages at an appropriate stage may help inhibit or slow the progression of bone loss in patients with osteoporosis.
Article
Full-text available
Astragalin (AG) is a biologically active flavonoid compound that can be extracted from a number of medicinal plants. However, the effects of AG on osteoblastic differentiation in mouse MC3T3-E1 cells and on bone formation in vivo have not been studied fully. In this study, we found that the activities of alkaline phosphatase (ALP) and mineralized nodules in MC3T3-E1 cells were both significantly increased after treatment with AG (5, 10, and 20 μM). Meanwhile, the mRNA and protein levels of osteoblastic marker genes in MC3T3-E1 cells after AG treatment were markedly increased compared with a control group. In addition, the levels of BMP-2, p-Smad1/5/9, and Runx2 were significantly elevated in AG-treated MC3T3-E1 cells. Moreover, we found that the protein levels of Erk1/2, p-Erk1/2, p38, p-p38, and p-JNK were also significantly increased in AG-treated MC3T3-E1 cells compared to those in the control group. Finally, in vivo experiments demonstrated that AG significantly promoted bone formation in an ovariectomized (OVX)-induced osteoporotic mouse model. This was evidenced by significant increases in the values of osteoblast-related parameters (BFR/BS, MAR, Ob.S/BS, and Ob.N/B.Pm) and bone histomorphometric parameters (BMD, BV/TV, Tb.Th, and Tb.N.) in OVX mice after AG treatment (5, 10, and 20 mg/kg). Collectively, these results demonstrated that AG may promote osteoblastic differentiation in MC3T3-E1 cells via the activation of the BMP and MAPK pathways and promote bone formation in vivo. These novel findings indicated that AG may be a useful bone anabolic agent for the prevention and treatment of osteoporosis.
Article
Osteoporosis is an important skeletal disease characterized by bone weakness and high risk of fracture in postmenopausal women. Tea consumption is known to play an important role in the prevention or alleviation of osteoporosis. However, the therapeutic effects of aqueous extracts of white tea (WT) have not been evaluated in osteoporosis rat models. The aim of this study was to investigate the potential anti-osteoporotic role of WT in ovariectomized (OVX) rats. WT was given orally at 0.5% w/v doses for 12 weeks in OVX rats. Biochemical parameters in blood samples, bone tartrate-resistant acid phosphatase (TRAP), C-terminal telopeptide of type 1 collagen (CTX) and estradiol levels were evaluated. Bone mineral density and bone mineral content values were measured in the left femur. In addition to histopathological examination, osteolcalcin, osteopontin and TUNEL levels were determined. OVX group data demonstrated that bone loss occurred by thinning of the metaphyseal growth plates of the femur. Similarly, the levels of TRAP and CTX, markers of osteoclastic activity, were found to be high concurrently with a decrease in femoral bone mineral density. In addition, increased osteolcalcin and osteopontin levels were present in the metaphyseal growth zones. On the other hand, while TRAP and CTX levels were suppressed in the OVX-WT group, bone mineral content increased. In ad-dition, TUNEL, osteocalcin and osteopontin positivity decreased in the right femoral metaphysis growth zones, proliferating zone and resting zone cells. These results showed that chronic WT consumption has a protective effect by reducing bone resorption in OVX-induced osteoporotic rats.
Article
In recent years, the number of patients with osteoporosis has increased as population grows older. Therefore, the chemoprevention of osteoporosis by better nutrition is important. White-rot fungi degrades milled wood lignin for growth and development. This degradation results in the formation of phenolic compounds such as syringic acid (SA) and vanillic acid (VA). In the artificial culture of edible mushrooms using a mushroom bed, the disposal of waste beds after mushroom cultivation is an important issue. The present study investigated the presence and amount of both SA and VA in the discarded waste beds after mushroom cultivation. The extracts from waste beds after cultivation of shiitake mushrooms, Lentinula edodes; buna shimeji, Hypsizygus marmoreus; maitake, Grifola frondosa; king trumpet mushrooms, Pleurotus eryngii; and butterscotch mushrooms, Pholiota microspora were analyzed using high performance liquid chromatography. Although the content of SA and VA was considerably different among the mushrooms, SA and VA were present in extracts obtained from all the waste beds. We also demonstrated that SA and VA exert their anti-osteoporotic effect independently of the estrogen receptor-mediated pathway using murine monocytic RAW264.7 cells, ovariectomized mice, and human breast cancer MCF-7 cells. Thus, these results suggest that the extracts are effective sources of SA and VA, which are effective in preventing osteoporosis.
Article
Medicinal plants have been an integral part of the healthcare system for centuries. Prunus africana, an evergreen plant of the family Rosaceae in sub-Saharan Africa, has been used to treat numerous diseases and disorders since time immemorial. Over the past decades, several ethnomedicinal surveys were conducted in many communities across Africa which validated the use of P. africana in traditional medicine. The increased interest in P. africana across the globe has led to several pharmacological studies that have further validated the continued use of this plant in traditional medicine. The aim of this study was therefore to review the various evidence based ethnopharmacological studies on the use of P. africana in traditional medicine in treatment and management of benign prostatic hyperplasia, prostate cancer, diabetes, malaria, chest pain, gastrointestinal conditions, wound healing, and skin infections based on peer-reviewed electronic databases. Scientific studies attributed the effectiveness of P. africana in the treatment and management of these diseases to the class of bioactive compounds contained in it, including tannins, saponins, alkaloids, flavonoids, terpenoids, phytosterols, and fatty acids. The anti-inflammatory, antimicrobial, antiandrogenic, antiangiogenic, antioxidant, anti-dipeptidyl peptidase-4, analgesic, and astringent effects of these phytochemicals makes P. africana very effective in the treatment and management of the above diseases. This review therefore provides the foundation of further preclinical and clinical studies on P. africana to enhance future production of high quality, safe, and efficacious medicines for treatment of benign prostatic hyperplasia, prostate cancer, diabetes, malaria, chest pain, gastrointestinal conditions, wound healing, and skin infections.