ArticlePDF Available

Benzophenone C - and O -Glucosides from Cyclopia genistoides (Honeybush) Inhibit Mammalian α-Glucosidase

Authors:
  • Afriplex (Pty) Ltd. South Africa

Abstract and Figures

An enriched fraction of an aqueous extract prepared from the aerial parts of Cyclopia genistoides Vent. yielded a new benzophenone di-C,O-glucoside, 3-C-β-d-glucopyranosyl-4-O-β-d-glucopyranosyliriflophenone (1), together with small quantities of a known benzophenone C-glucoside, 3-C-β-d-glucopyranosylmaclurin (2). The isolated compounds showed α-glucosidase inhibitory activity against an enzyme mixture extracted from rat intestinal acetone powder. Compound 2 exhibited significantly (p < 0.05) higher inhibitory activity (54%) than 1 (43%) at 200 μM. In vitro tests in several cell models showed that 1 and its 3-C-monoglucosylated derivative (3-C-β-d-glucopyranosyliriflophenone) were marginally effective (p ≥ 0.05) in increasing glucose uptake.
Content may be subject to copyright.
Benzophenone C- and OGlucosides from Cyclopia genistoides
(Honeybush) Inhibit Mammalian αGlucosidase
Theresa Beelders,*
,,
D. Jacobus Brand,
§
Dalene de Beer,
Christiaan J. Malherbe,
Sithandiwe E. Mazibuko,
Christo J. F. Muller,
and Elizabeth Joubert
,
Department of Food Science, Stellenbosch University, Private Bag X1, Matieland 7602, South Africa
Post-Harvest and Wine Technology Division, Agricultural Research Council (ARC) Infruitec-Nietvoorbij, Private Bag X5026,
Stellenbosch 7599, South Africa
§
Central Analytical Facility (CAF), Nuclear Magnetic Resonance Unit, Private Bag X1, Matieland 7602, South Africa
Diabetes Discovery Platform, South African Medical Research Council, P.O. Box 19070, Tygerberg 7505, South Africa
*
SSupporting Information
ABSTRACT: An enriched fraction of an aqueous extract prepared from the aerial parts of Cyclopia genistoides Vent. yielded a
new benzophenone di-C,O-glucoside, 3-C-β-D-glucopyranosyl-4-O-β-D-glucopyranosyliriophenone (1), together with small
quantities of a known benzophenone C-glucoside, 3-C-β-D-glucopyranosylmaclurin (2). The isolated compounds showed α-
glucosidase inhibitory activity against an enzyme mixture extracted from rat intestinal acetone powder. Compound 2exhibited
signicantly (p< 0.05) higher inhibitory activity (54%) than 1(43%) at 200 μM. In vitro tests in several cell models showed that
1and its 3-C-monoglucosylated derivative (3-C-β-D-glucopyranosyliriophenone) were marginally eective (p0.05) in
increasing glucose uptake.
The global prevalence of diabetes is increasing at alarming
rates, leading to estimates of 439 million diabetics by
2030.
1
In Africa, South Africa tips the scale with a national
prevalence of over 7%.
2
It is projected that diabetes will rank as
the ninth leading cause of death in low-income countries in the
next few decades.
3
A sedentary lifestyle together with an
unhealthy diet, high in rened carbohydrates accompanied by a
low intake of fruits and vegetables, are considered contributing
factors. The search for natural and synthetic α-glucosidase
inhibitors
4,5
that delay the breakdown and absorption of
carbohydrates in the gut, thus mimicking the protective eect of
the drug acarbose, but without its side-eects,
6
is escalating.
Much prominence is given to polyphenols, with promising
results in vitro.
4
Synergistic eects between acarbose and
polyphenols suggest benets in terms of dose reduction of the
drug.
7
Subclasses of phenolic compounds that show promise as
α-glucosidase inhibitors include the polyhydroxyxanthones and
polyhydroxybenzophenones, together with their glycosylated
derivatives.
811
Extracts of Cyclopia genistoides Vent. (honey-
bush) are principally renowned for high levels of the
tetrahydroxyxanthone C-glucoside mangiferin,
12
a bioactive
compound
13
with potent α-glucosidase inhibitory activity.
9
Recent investigation of the phenolic constituents of C.
genistoides revealed the presence of several benzophenone C-
glucosides, including 3-C-β-D-glucopyranosyliriophenone and
3-C-β-D-glucopyranosylmaclurin.
1416
The major benzophe-
none glycoside present in hot water extracts of C. genistoides
has been tentatively identied as an iriophenone di-O,C-
hexoside thus far.
16
A compound with similar MS properties
has also been observed in extracts of another Cyclopia species,
C. subternata,
17
albeit at lower concentrations. In the present
paper the isolation and structural elucidation of this
benzophenone glucoside, identied as 3-C-β-D-glucopyrano-
syl-4-O-β-D-glucopyranosyliriophenone (1), is described.
During the isolation process, small quantities of the known
benzophenone C-glucoside 3-C-β-D-glucopyranosylmaclurin
(2) was also obtained. These compounds were isolated from
the aerial parts of C. genistoides using solid-phase extraction
(SPE) for extract enrichment, followed by purication by
semipreparative liquid chromatography (LC). The ability of 1
and 2to inhibit mammalian α-glucosidase was assessed and
Received: September 16, 2014
Article
pubs.acs.org/jnp
© XXXX American Chemical Society and
American Society of Pharmacognosy Adx.doi.org/10.1021/np5007247 |J. Nat. Prod. XXXX, XXX, XXXXXX
compared to that of 3-C-β-D-glucopyranosyliriophenone.
Included in the α-glucosidase assay was the benzophenone
aglucone maclurin, with acarbose employed as positive control.
To date, the α-glucosidase inhibitory activities of 1and 2as C-
and O-glucosylated polyhydroxybenzophenones have not been
assessed. In vitro tests in several cell models were conducted to
investigate the ability of 1and 3-C-β-D-glucopyranosylirio-
phenone to increase glucose uptake in vitro.
RESULTS AND DISCUSSION
Compound 1was obtained as a white, amorphous powder. Its
molecular formula was established as C25H30O15 from the
analyses of its HRESIMS and 13C NMR data. Initial LC-
ESIMSMS analyses of this compound in a C. genistoides sample
matrix
16
had pointed to an iriophenone di-O,C-hexoside. The
elucidation of the structure of 1and, in particular, the
identication of the hexoside moieties and their point of
attachment to the aglycone, were done via 1D and 2D NMR
experiments. The relative congurations of the glycoside units
can normally be determined by a 1D/2D NOESY NMR
experiment. However, NMR spectroscopic data for 1proved
inconclusive for unambiguous identication of the individual
glycoside units, due to overlapping proton resonances. Since
the NMR spectra of 1closely resembled those of 3-C-β-D-
glucopyranosyliriophenone, it was essential to rst selectively
hydrolyze and identify the terminal O-linked sugar moiety,
followed by comparison of the resulting iriophenone C-
hexoside to that of a commercial 3-C-β-D-glucopyranosyl-
iriophenone standard, also present in C. genistoides.
18
The terminal O-linked hexoside moiety was identied as D-
glucose by enzymatic hydrolysis and GC-MS analysis. The
resulting iriophenone C-hexoside was puried, and its 1H and
13C NMR data matched those of 3-C-β-D-glucopyranosyl-
iriophenone. Having identied the β-D-glucopyranosyl con-
stituent units, the rest of the molecule and the point of
attachment of the O-β-D-glucopyranosyl unit were dened by
additional 1D NOESY, COSY, HSQC, and HMBC NMR
experiments.
The aromatic region between 6.0 and 8.5 ppm of the 1H
NMR spectra showed two o-coupled doublets and a singlet, δ
7.61 (2H, d, J= 8.6 Hz), 6.78 (2H, d, J= 8.6 Hz), 6.24 (1H, s),
reminiscent of an AABBaromatic spin system with an
uncoupled singlet on a separate aromatic ring and supported by
the presence of aromatic carbons in the 13C NMR spectra of 1.
The 1H NMR spectra also presented the resonances of two β-
coupled (8 Hz anomeric coupling constant) glucoside units in
the characteristic 2.55.0 ppm region.
19
The presence of a
single carbon at δ193.1 in addition to the 12 aromatic carbons
in the 13C NMR spectrum was reminiscent of a benzophenone
aglycone unit.
19
The anomeric protons of the two glucopyr-
anosyl moieties at δ4.73 (1H, d, J= 7.8 Hz) and 4.66 (1H, d, J
= 9.8 Hz) were irradiated selectively utilizing a 1D NOESY
experiment to determine their position of attachment and other
proton resonances of the individual O-and C-linked
glucopyranosyl units. The anomeric proton of the glucopyr-
anosyl residue at δ4.73 (1H, d, J= 7.8 Hz, H-1) displayed a
strong NOE association with the H-5 singlet at δ6.24. This
assigned the O-linked glucosyl unit to C-4 of the
benzophenone A-ring. A weak NOE association of H-1with
H-4(δ3.24) of the neighboring glucosyl moiety indicated the
close proximity of the glucosyl entities.
An NOE association was also observed between the three
axial H-1, H-3, and H-5protons (Figure 1), arming the
glucose conguration of the O-linked sugar unit. The nature of
theC-linkedsugartotheaglyconewasconrmed by
comparison of the hydrolytic products to the NMR spectra
of the commercial 3-C-β-D-glucopyranosyliriophenone stand-
ard.
A COSY experiment conrmed the AABBaromatic spin
system of the B-ring and the connectivity of some of the
protons on the individual glucosyl moieties. This includes only
the protons that are suciently resolved to be assigned by the
COSY experiment and did not overlap with the glucosyl proton
resonances.
The HSQC NMR experiment was used to assign all the
directly bonded protons (1JHC) to their respective carbons. All
the carbons were resolved successfully by an HMBC experi-
ment showing the long-range (2JHC,3JHC) connectivity between
protons and carbons [up to four bonds (4JHC) in some cases].
Those structurally signicant HMBC correlations are shown in
Figure 2, enabling the assignment of the unprotonated carbons
in the molecule. Compound 1was thus identied as 3-C-β-D-
glucopyranosyl-4-O-β-D-glucopyranosyliriophenone.
Compound 2was obtained as a light yellow, amorphous
powder. Accurate measurement of the pseudomolecular ions in
the positive and negative HRESIMS data, in conjunction with
13C NMR data, allowed a molecular formula of C19H20O11 to be
assigned to 2. The structure of 2was conrmed as 3-C-β-D-
Figure 1. Relevant NOE associations in 1.
Figure 2. Relevant long-range HMBC correlations in 1.
Journal of Natural Products Article
dx.doi.org/10.1021/np5007247 |J. Nat. Prod. XXXX, XXX, XXXXXXB
glucopyranosylmaclurin by comparison of its observed and
reported 1H NMR and 13C NMR spectroscopic data.
14,20
Both benzophenone derivatives 1and 2, together with the
reference compounds, showed inhibitory activity against
mammalian α-glucosidase at various concentrations with all
values signicantly diering (p< 0.05) from all other values.
The observed inhibitory activities were both concentration- and
compound-specic (Figure 3), showing a clear doseresponse.
The activities of the benzophenones could thus be compared at
equimolar concentration levels, which gave insight into possible
structureactivity relations. Compound 2was the most active
inhibitor, followed by 3-C-β-D-glucopyranosyliriophenone and
1, while maclurin showed the weakest inhibitory activity. It
appears that C-monoglucosides tend to be more eective
inhibitors than their corresponding aglucones; that is, 2is
signicantly more active than maclurin (p< 0.05). This has also
been observed previously for 3-C-β-D-glucopyranosyl-
iriophenone and its aglucone, iriophenone.
9
Furthermore,
the higher activity of 2compared to that of 3-C-β-D-
glucopyranosyliriophenone was attributed to the additional
3-hydroxy group of 2. Previous studies on hydroxybenzophe-
nones
10
and hydroxyxanthones
8
have shown that increases in
the number of phenolic hydroxy groups on the basic diphenyl
ketone and dibenzo-γ-pyrone structures lead to signicant
increases in α-glucosidase inhibitory activities. Moreover, for
hydroxyavones it has been shown that a 3-hydroxy group in
particular leads to increased inhibitory activity.
21
The presence
of an additional O-glucopyranosyl moiety at C-4 on the
diphenyl ketone structure of 1signicantly (p< 0.05) lowered
α-glucosidase inhibitory activity compared to 3-C-β-D-
glucopyranosyliriophenone. A similar decrease in activity
due to an additional C-glucopyranosyl moiety has also been
reported for 3-C-β-D-glucopyranosyl-5-C-β-D-glucopyranosyl-
iriophenone compared to 3-C-β-D-glucopyranosyl-
iriophenone.
9
The eect of 1and 3-C-β-D-glucopyranosyliriophenone on
in vitro glucose uptake in L6 myocytes demonstrated marginal,
but not signicant, increases in glucose uptake relative to the
vehicle control at concentrations of 10 μMof1(ca. 22%
increase; Figure 4A) and 100 μMof3-C-β-D-glucopyranosyl-
iriophenone (ca. 27% increase; Figure 4B). Similar marginal
eects on glucose uptake were observed in 3T3-L1 adipocytes
(Figure 4C and D) and in C3A hepatocytes (data not shown).
It was anticipated that the isolated benzophenone glucoside (1)
and 3-C-β-D-glucopyranosyliriophenone could increase in
vitro glucose uptake activity by activating the cellular energy
regulator AMPK, as demonstrated for the latter compound
22,23
in mature 3T3-L1 adipocytes and diabetic KK-Aymice. The
assessed compounds were, however, less eective at increasing
cellular glucose uptake than the reference pharmacological
agent metformin, mechanistically known to be an activator of
AMPK.
24
EXPERIMENTAL SECTION
General Experimental Procedures. NMR spectra were recorded
on a Varian Unity Inova 600 NMR spectrometer with a 1H frequency
of 600 MHz and a 13C frequency of 150 MHz using a 5 mm inverse
detection PFG probe. The chemical shift frequencies are indicated on
the δscale. 1H and 13C NMR spectra were referenced to the residual
DMSO-d6peak at δ2.5 and 39.5, respectively. The spectra in acetone-
d6have the residual acetone peaks referenced at δ2.05 for 1H NMR
and at δ29.84 for the 13C NMR spectra. The spectra were recorded
using the standard VnmrJ instrument software and processed and
expanded further using the Mestrenova 9.0 software package.
HRESIMS analyses were conducted on an Acquity UPLC system,
tted with a photodiode-array detector and coupled to a Synapt G2 Q-
TOF equipped with an electrospray ionization source (Waters,
Milford, MA, USA). UV spectra were recorded online. Chromato-
graphic conditions and MS parameters were as described by
Beelders.
16
Semipreparative LC was performed on a LaChrom
HPLC system (Merck Hitachi, Hitachi High Technologies, Japan)
comprising a quaternary pump, autosampler, variable-wavelength
detector, and diode-array detector and tted with a Phenomenex
Gemini C18 column (5 μm, 110 Å, 150 ×10 mm) (Phenomenex,
Torrance, CA, USA). The column was protected by a guard column of
the same stationary phase (10 ×10 mm) and a high-pressure
semipreparative in-line lter (IDEX Health & Science, Oak Harbor,
WA, USA). Column temperature was maintained at 30 °C using an
external HPLC column oven (LKB Bromma, Sweden). Gas
chromatography was performed on an Agilent GC-MS instrument
(Agilent 6890N GC and Agilent 5975 MSD, Agilent Technologies,
Palo Alto, CA, USA) tted with a 30 m Zebron ZB-SemiVolatiles
column with 0.25 mm inner diameter and 0.25 μmlm thickness. The
GC chromatograms and mass spectra were evaluated using Agilent
MSD ChemStation (version D.02.00.237) software.
Plant Material. The leaves and ne stems from a selection of
Cyclopia genistoides bushes (Overberg type) were harvested from a
commercial plantation situated near Pearly Beach (Western Cape,
South Africa; GPS coor. 34.702, 19.618). Plant material was dried
without delay in a cross-ow drying tunnel at 40 °C for 16 h to a
moisture content of less than 10% and pulverized using a Retsch rotary
mill (1.4 mm sieve; Retsch GmbH, Haan, Germany).
Extraction and Isolation. Plant material (420 g) was extracted
with hot water (4.2 L, 93 °C) for 30 min using a ratio of 1:10 (w/v).
The crude extract was ltered through Whatman #4lter paper,
frozen, and freeze-dried using a VirTis Advantage Plus freeze-drier (SP
Scientic, Warminster, PA, USA). The freeze-dried hot water extract
(126.5 g) comprised 6.31% of 1and 0.53% of 2.
Enrichment of the crude extract in terms of 1and 2was performed
using C18 solid-phase extraction (Discovery DSC-18; 10 g/60 mL;
Sigma-Aldrich). The cartridge was conditioned sequentially with
MeOH and deionized water (60 mL). A solution of freeze-dried
extract (300 mg reconstituted in 50 mL of deionized water) was
applied to the cartridge, followed by ushing with deionized water
(100 mL). The target analytes were eluted with 5% aqueous MeOH
(300 mL). This process was repeated 48 times with new cartridges
(14.4 g of freeze-dried extract loaded), and the eluants were pooled,
vacuum-evaporated, and freeze-dried, yielding ca. 1.2 g of enriched
Figure 3. Percentage activity of rat α-glucosidase challenged with
various concentrations of 1,3-C-β-D-glucopyranosyliriophenone, 2,
and maclurin.
Journal of Natural Products Article
dx.doi.org/10.1021/np5007247 |J. Nat. Prod. XXXX, XXX, XXXXXXC
fraction. The enriched fraction comprised 56.5% of 1and 4.8% of 2.
The average recovery of target analytes after SPE was 75%.
The enriched fraction was reconstituted in deionized water (ca. 6
mg/mL), ltered, and subjected to semipreparative LC (MeCN2%
HOAc (aq), 4:96, v/v) using a ow rate of 4.8 mL/min. Aliquots (400
μL) of the reconstituted, enriched fraction were injected repeatedly,
equaling 880 mg. The fractions containing 1and 2were collected
based on retention times using a Gilson FC203B fraction collector
(Gilson, Middleton, WI, USA) and pooled, and the organic solvent
was evaporated under vacuum. The remaining aqueous solutions were
ltered, frozen, and freeze-dried, yielding 370 mg of 1(purity >99% by
HRESIMS; total yield of 74% from the enriched extract) and 30 mg of
2(purity 95% by HRESIMS; total yield of 71% from the enriched
extract).
3-C-β-D-Glucopyranosyl-4-O-β-D-glucopyranosyl-
iriophenone (1): white, amorphous powder; UV λmax online 234,
294 nm; 1H NMR 600 MHz (DMSO-d6, 298 K) δ7.61 (2H, d, J= 8.6
Hz, H-2,6), 6.78 (2H, d, J= 8.6 Hz, H-3,5), 6.24 (1H, s, H-5), 4.73
(1H, d, J= 7.8 Hz, H-1), 4.66 (1H, d, J= 9.8 Hz, H-1), 3.6 (3H, m,
H-6,2×H-6), 3.46 (2H, m, H-2, H-6), 3.25 (3H, m, H-3, H-4,
H-5), 3.18 (2H, m, H-3, H-5), 3.08 (1H, t, J= 9.2 Hz, H-4), 2.88
(1H, t, J= 8.4 Hz, H-2); 13C NMR 150 MHz (DMSO-d6, 298 K) δ
193.1 (C, CO), 161.8 (C, C-1), 157.7 (C, C-1), 155.2 (C, C-2),
155.2 (C, C-4), 131.8 (CH, C-3,5), 130.2 (CH, C-4), 114.8 (CH,
C-2,6), 110.0 (C, C-6), 106.2 (C, C-3), 100.5 (CH, C-1), 94.8
(CH, C-5), 81.0 (CH, C-3), 78.0 (CH, C-5), 77.1 (CH, C-5), 76.6
(CH, C-3), 74.9 (CH, C-1), 73.2 (CH, C-2), 72.3 (CH, C-2),
69.3 (CH, C-4), 69.3 (CH, C-4), 60.5 (CH2, C-6), 60.1 (CH2,C-
6); HRESIMS m/z569.1503 [M H](calcd for C25H29O15,
596.1506); ESIMS [m/z(%)] 569 (100) [M H], 1139 (20) [2M
H]; HRESIMS m/z571.1661 [M + H]+(calcd for C25H31O15,
571.1663); ESIMS [m/z(%)] 1141 (10) [2M + H]+, 571 (60) [M +
H]+, 409 (100) [M + H 162]+, 391 (60) [M + H 162 H2O]+,
373 (10) [M + H 162 2×H2O]+, 355 (10) [M + H 162 3×
H2O]+, 313 (10) [M + H 258]+, 289 (15) [M + H 162 120]+.
Acid Hydrolysis of 1. Aliquots of a solution of 1(105 mg in 70
mL of 1.1 M HCl) were heated in 5 mL glass Reacti-vials at 60 °Cina
Stuart heating block (Bibby Scientic Limited, Stone, UK) for 24 h.
The hydrolysis reaction was monitored using HPLC-DAD (86%
degradation at t= 24 h). The hydrolyzed mixture was cooled to room
temperature and adjusted to pH 4 using 1.1 M NaHCO3.
One half of this hydrolyzed mixture was vacuum-evaporated and
subjected to semipreparative LC (MeCN2% HOAc (aq), 8:92, v/v)
to obtain intact 1and iriophenone C-hexoside. The fraction
containing iriophenone C-hexoside was collected based on retention
time, pooled, vacuum-evaporated, and freeze-dried, followed by NMR
analysis.
1H NMR and 13C NMR Data for 3-C-β-D-glucopyranosyl-
iriophenone (hydrolyzed product of 1): 1H NMR 600 MHz
(acetone-d6, 298 K) δ7.64 (2H, d, J= 8.6 Hz, H-2,6), 6.85 (2H, d, J
= 8.6 Hz, H-3,5), 5.99 (1H, s, H-5), 4.92 (1H, d, J= 9.8 Hz, H-1),
3.85 (2H, d, J= 3.1 Hz, 2 ×H-6), 3.69 (H, t, J= 9.2 Hz, H-4), 3.63
(H, t, J= 9.2 Hz, H-2), 3.57 (H, t, J= 8.9 Hz, H-3), 3.49 (H, m, H-
5); 13C NMR 150 MHz (acetone-d6, 298 K) δ197.8 (CO), 162.6
(C-4), 161.9 (C-4), 161.4 (C-2), 160.5 (C-6), 133.3 (C-1), 132.5
(C-2,6), 115.3 (C-3,5), 106.6 (C-1), 104.7 (C-3), 96.9 (C-5), 82.1
(C-5), 79.3 (C-3), 76.6 (C-1), 74.6 (C-4), 70.6 (C-2), 61.7 (C-
6).
Identication of the O-Linked Hexose Residue in the
Hydrolyzed Product. The other half of the hydrolyzed product
was partially evaporated under vacuum, and a small volume of the
concentrated liquid was removed for an enzyme robot assay. This
assay was conducted on a Thermo Scientic Arena 20XT random
access chemistry analyzer (Thermo Fisher Scientic, Oy, Finland) with
the use of an Enytec uid D-glucose no. 5140 enzyme reagent kit
Figure 4. 3H-2-DOG uptake in L6 myocytes exposed to (A) 1and (B) 3-C-β-D-glucopyranosyliriophenone and 3T3-L1 adipocytes exposed to (C)
1and (D) 3-C-β-D-glucopyranosyliriophenone. Insulin (positive control) and metformin (reference drug control) were included at concentrations
of 1 μM. Mean activity is expressed relative to the vehicle control at 100% ±standard error of the means. Three independent experiments were
performed each with three technical repeats. **p< 0.01; ***p< 0.001.
Journal of Natural Products Article
dx.doi.org/10.1021/np5007247 |J. Nat. Prod. XXXX, XXX, XXXXXXD
(AEC-Amersham, Kayalami, South Africa) for the identication of
glucose according to the manufacturers instructions.
The remaining sample volume was freeze-dried, derivatized with
bis(trimethylsilyl)triuoroacetamide (BSTFA) using the procedure
described by Roessner,
25
and subjected to GC-MS analysis. D-Glucose
and D-galactose standards (Sigma-Aldrich) were also derivatized prior
to analysis. Sample volumes of 1 μL were injected with a split ratio of
1:10. The injection temperature was 280 °C, the interface set to 280
°C, and the ion source adjusted to 240 °C. The carrier gas was helium
set at a constant ow rate of 1.0 mL/min. The temperature program
comprised a 6 °C oven temperature ramp from 70 to 76 °C within 1
min, followed by a 32 °C/min ramp to 300 °C, and a nal 5 min
heating at 300 °C. Mass spectra were recorded over an m/zscanning
range of 40 to 650. Electron energy was 70 eV and solvent delay 8 min.
3-C-β-D-Glucopyranosylmaclurin (2): light yellow, amorphous
powder; UV λmax online 236, 290 (sh), 318 nm; 1H NMR 600 MHz
(DMSO-d6, 298 K) δ7.15 (1H, d, J= 1.93 Hz, H-2), 7.06 (1H, dd, J
= 1.93, 8.24 Hz, H-6), 6.74 (1H, d, J= 8.24 Hz, H-5), 5.94 (1H, s, H-
5), 4.60 (1H, d, J= 9.77 Hz, H-1), 3.61 (1H, d, 10.96 Hz, H-6a), 3.51
(2H, m, H-2,6), 3.20 (3H, H-3,4,5); 13C NMR 150 MHz
(DMSO-d6, 298 K) δ194.6 (C, CO), 158.4 (C, C-4), 156.7 (C, C-
6), 156.3 (C, C-2), 150.0 (C, C-4), 144.6 (C, C-3), 131.0 (C, C-1),
122.3 (CH, C-6), 116.2 (CH, C-2), 114.7 (CH, C-5), 107.6 (C, C-
3), 103.6 (C, C-1), 94.8 (CH, C-5), 81.1 (CH, C-5), 78.3 (CH, C-
3), 74.8 (CH, C-1), 72.1 (CH, C-4), 69.6 (CH, C-2), 60.4 (CH2,
C-6); HRESIMS m/z423.0933 [M H](calcd for C19H19O11,
423.0927); ESIMS [m/z(%)] 1271 (5) [3M H], 847 (70) [2M
H], 423 (100) [M H], 303 (20) [M H120]; HRESIMS m/
z425.1075 [M + H]+(calcd for C19H21O11, 425.1084); ESIMS [m/z
(%)] 425 (80) [M + H]+, 407 (100) [M + H H2O]+, 389 (10) [M +
H2×H2O]+, 371 (10) [M + H 3×H2O]+, 341 (10) [M + H
84]+, 329 (15) [M + H 96]+, 305 (20) [M + H 120]+, 287 (5) [M
+H120 H2O]+.
α-Glucosidase Inhibitory Activity. A method for the determi-
nation of α-glucosidase inhibitory eects
26
was adapted for use on a
BioTek SynergyHT microplate reader with dispenser (BioTek
Instruments, Winooski, VT, USA). A mixture containing α-glucosidase
was extracted from rat intestinal acetone powder (Sigma-Aldrich) by
suspending ca. 350 mg of powder in 10 mL of cold KH2PO4buer
(200 mM KH2PO4, pH 6.8 with KOH) followed by repeated
sonication on ice (sonication sequence repeated 12 times: 30 s
sonication with 25% amplitude, 1 min rest) using a model VCX600
high-intensity ultrasonic processor with a 3 mm stepped microtip
(Sonics & Materials Inc., Newton, CT, USA). The crude mixture was
centrifuged at 10000gfor 30 min at 4 °C in a Hettich Universal 320R
refrigerated centrifuge (Hettich Holding GmbH & Co. oHG,
Kirchlengern, Germany), and the supernatant was retrieved and
ltered using 0.45 μm pore size, 33 mm Millex HV PVDF lter
membranes (Merck Millipore). The supernatant was used as an
enzyme mixture after dilution to the standardized concentration based
on activity testing.
Activity determination of the enzyme mixture was performed daily
prior to each set of experiments, using the same procedure as for the
inhibition assays, but with H2O as sample controls and varying
dilutions of the enzyme mixture. Fluorescence measurements were
used to determine the correct concentration for optimal enzyme
activity, ca. 15 to 20 mg/mL of the original powder estimated as an
FL-value of 50 000 (λEX: 360 nm; λEM: 460 nm), 20 min after addition
of the substrate.
The inhibitory activities of 1,2,3-C-β-D-glucopyranosyl-
iriophenone (Fluka, Sigma-Aldrich, St. Louis, MO, USA), and
maclurin (Sigma-Aldrich) were assessed at three concentration levels
ranging between 50 and 400 μM. Acarbose (Sigma-Aldrich), a known
inhibitor of mammalian α-glucosidase, was used as positive control at
65 μM. The following test procedure was employed: 20 μL of the
assay control (H2O), positive control, or target analyte at selected
concentration was added to 125 μL of a 200 mM KH2PO4buer (pH
6.8) and 65 μL of the chosen enzyme dilution in 96-well black, at-
bottom microplates with a clear bottom (Greiner Bio-One GmbH,
Rainbach im Muhlkreis, Austria). The mixture was preincubated at 37
°C for 15 min, followed by the addition of 40 μL of the substrate, 1.2
mM 7-O-α-D-glucopyranosyl-4-methylumbelliferone, by dispenser.
Fluorescence (λEX: 360 nm; λEM: 460 nm) was monitored over 30
min, and the net uorescence (net FL) and percent enzyme activity
were calculated using the following formulas:
=−Net FL Fluorescence Fluorescence
30min 0mi
n
%
Enzyme Activity 100 Net FL
Net FL
sample
assay control
Statistical analysis was performed with GraphPad Prism version 5.00
for Windows (GraphPad Software, San Diego, CA, USA, www.
graphpad.com) using one-way ANOVA with Tukeysmultiple
comparison posthoc test to determine signicant dierences between
values at the 95% condence level (p< 0.05).
2-Deoxy-[3H]-D-glucose (3H-2-DOG) Uptake. To estimate in
vitro glucose uptake activity, cellular 3H-2-DOG uptake was assessed
by liquid scintillation counting, using the method described by
Mazibuko.
27
Briey, L6 myoblasts (2.5 ×104cells/mL) and 3T3-L1
broblasts (2.0 ×104cells/mL) were seeded into 24-well plates in
Dulbeccos modied Eagles medium (DMEM) supplemented with
10% fetal or normal calf serum, respectively. C3A hepatocytes were
seeded at 5.5 ×104cells/mL in Eagles minimal essential medium
(EMEM) supplemented with 10% fetal calf serum. All cells were
cultured at 37 °C in humidied air with 5% CO2. Cell culture media
DMEM, EMEM, and fetal and normal calf serum were obtained from
Lonza (Walkersville, MD, USA). The L6 myoblasts and 3T3-L1
broblasts were dierentiated into myotubule-forming myocytes and
adipocytes, respectively, while the C3A hepatocytes were used as
semiconuent cultures. For the 3H-2-DOG uptake experiments, cells
(1 h for L6 myocytes, and 3 h for 3T3-L1 adipocytes and C3A
hepatocytes) were exposed to 1and 3-C-β-D-glucopyranosyl-
iriophenone at concentrations ranging from 0.001 to 100 μM.
Compounds were dissolved in DMSO and diluted with Krebs-Ringer
bicarbonate HEPES buer (KRBH) containing 8 mM glucose (nal
DMSO concentrations <0.004%). For glucose uptake determination,
cells were pulse-labeled for 15 min with 0.5 μCi/mL 3H-2-DOG
(American Radiolabeled Chemicals, Inc., St. Louis, MO, USA) in
glucose- and serum-free KRBH containing 1and 3-C-β-D-
glucopyranosyliriophenone at the relevant concentrations. Insulin
and metformin (1,1-dimethylbiguanide hydrochloride) (Sigma-Al-
drich) both at 1 μM were included as positive and drug reference
controls, respectively. The amount of 3H-2-DOG taken up by cells was
measured using a liquid scintillation counter (2200CA Tricarb Series,
Packard Instrument Company, Downers Grove, IL, USA), and the
activity calculated as fmol (3H-2-DOG)/mg protein. Statistical analysis
was performed with GraphPad Prism version 5.00 for Windows using
one-way ANOVA with Dunnetts multiple comparison post hoc test to
determine signicant dierences between values at the 95% condence
level (p< 0.05).
ASSOCIATED CONTENT
*
SSupporting Information
S1 (1H), S2 (13C), S3 (COSY), S4 (1D NOESY), S5 (HSQC),
and S6 (HMBC) NMR spectra of 3-C-β-D-glucopyranosyl-4-O-
β-D-glucopyranosyliriophenone (1). S7: 1H NMR spectra for
3-C-β-D-glucopyranosyliriophenone (obtained by acid hydrol-
ysis of 1), with an overlay of corresponding spectra for the
commercial reference standard. S8: The unreacted product of 1
is also compared to the starting material of 1to assess the
stability of the sugar moiety during the acid hydrolysis reaction.
This material is available free of charge via the Internet at
http://pubs.acs.org.
Journal of Natural Products Article
dx.doi.org/10.1021/np5007247 |J. Nat. Prod. XXXX, XXX, XXXXXXE
AUTHOR INFORMATION
Corresponding Author
*E-mail: beelderst1@arc.agric.za (T. Beelders). Phone: +27-21-
809-3441. Fax: +27 21-809-3430.
Notes
The authors declare no competing nancial interest.
ACKNOWLEDGMENTS
This work is based on the research supported in part by the
South African Department of Science and Technology (DST/
CON 0133/2012). Other nancial support was received
through a grant from the Economic Competitive Support
Package for Agroprocessing to the ARC by the South African
Government. NRF grant holders (E.J. and C.J.M.) acknowledge
that opinions, ndings, and conclusions or recommendations
expressed in any publication generated by the NRF-supported
research (IFRR Grant 85277 and Thuthuka Grant 87849) are
those of the authors and that the NRF accepts no liability
whatsoever in this regard. An NRF-DST Professional Develop-
ment Program Doctoral Scholarship to T.B. is acknowledged.
Mr. F. Joubert (Koksrivier) is acknowledged for supplying plant
material. Ms. W. Kuhn (Water Analysis Division, CAF,
Stellenbosch University) is thanked for the enzyme robot
assay, and Mr. F. Hiten (Mass Spectrometry Unit, CAF,
Stellenbosch University) for GC-MS analyses.
REFERENCES
(1) Shaw, J. E.; Sicree, R. A.; Zimmet, P. Z. Diabetes Res. Clin. Pract.
2010,87,414.
(2) International Diabetes Federation. IDF Diabetes Atlas, 6th ed.;
International Diabetes Federation: Brussels, Belgium, 2013 (accessible
at http://www.idf.org/diabetesatlas).
(3) Mathers, C. D.; Loncar, D. PLoS Med. 2006,3, e442.
(4) Xiao, J.; Kai, G.; Yamamoto, K.; Chen, X. Crit. Rev. Food Sci. Nutr.
2013,53, 818836.
(5) Park, H.; Hwang, K. Y.; Oh, K. H.; Kim, Y. H.; Lee, J. Y.; Kim, K.
Bioorg. Med. Chem. 2008,16, 284292.
(6) Hollander, P. Diabetes Spectrum 2007,20, 159165.
(7) Boath, A. S.; Stewart, D.; McDougall, G. J. Food Chem. 2012,135,
929936.
(8) Liu, Y.; Zou, L.; Ma, L.; Chen, W.-H.; Wang, B.; Xu, Z.-L. Bioorg.
Med. Chem. 2006,14, 56835690.
(9) Feng, J.; Yang, X.-W.; Wang, R.-F. Phytochemistry 2011,72, 242
247.
(10) Hu, X.; Xiao, Y.; Wu, J.; Ma, L. Arch. Pharm. Chem. Life Sci.
2011,344,7177.
(11) Liu, Q.; Guo, T.; Li, W.; Li, D.; Feng, Z. Arch. Pharm. Chem. Life
Sci. 2012,345, 771783.
(12) Joubert, E.; Joubert, M. E.; Bester, C.; De Beer, D.; De Lange, J.
H. S. Afr. J. Bot. 2011,77, 887907.
(13) Vyas, A.; Syeda, K.; Ahmad, A.; Padhye, S.; Sarkar, F. H. Mini-
Rev. Med. Chem. 2012,12, 412425.
(14) Kokotkiewicz, A.; Luczkiewicz, M.; Pawlowska, J.; Luczkiewicz,
P.; Sowinski, P.; Witkowski, J.; Bryl, E.; Bucinski, A. Fitoterapia 2013,
90, 199208.
(15) Malherbe, C. J.; Willenburg, E.; De Beer, D.; Bonnet, S. L.; Van
der Westhuizen, J. H.; Joubert, E. J. Chromatogr. B 2014,951952C,
164171.
(16) Beelders, T.; De Beer, D.; Stander, M. A.; Joubert, L. Molecules
2014,19, 1176011790.
(17) De Beer, D.; Schulze, A. E.; Joubert, E.; De Villiers, A.;
Malherbe, C. J.; Stander, M. A. Molecules 2012,17, 1460214624.
(18) Kokotkiewicz, A.; Luczkiewicz, M.; Sowinski, P.; Glod, D.;
Gorynski, K.; Bucinski, A. Food Chem. 2012,133, 13731382.
(19) Jay, M. In The Flavonoids: Advances in Research since 1986;
Harborne, J. B., Ed.; Chapman and Hall: London, 1993; p 85.
(20) Tanaka, T.; Sueyasu, T.; Nonaka, G.-I.; Nishioka, I. Chem.
Pharm. Bull. 1984,32, 26762686.
(21) Li, H.; Song, F.; Xing, J.; Tsao, R.; Liu, Z.; Liua, S. J. Am. Soc.
Mass. Spectrom. 2009,20, 14961503.
(22) Zhang, Y.; Qian, Q.; Ge, D.; Li, Y.; Wang, X.; Chen, Q.; Gao, X.;
Wang, T. J. Agric. Food Chem. 2011,59, 1152611533.
(23) Zhang, Y.; Liu, X.; Han, L.; Gao, X.; Liu, E.; Wang, T. Food
Chem. 2013,141, 28962905.
(24) Zhou, G.; Myers, R.; Li, Y.; Chen, Y.; Shen, X.; Fenyk-Melody,
J.; Wu, M.; Ventre, J.; Doebber, T.; Fujii, N.; Musi, N.; Hirshman, M.
F.; Goodyear, L. J.; Moller, D. E. J. Clin. Invest. 2001,108, 11671174.
(25) Roessner, U.; Wagner, C.; Kopka, J.; Trethewey, R. N.;
Willmitzer, L. Plant J. 2000,23, 131142.
(26) Azuma, T.; Kayano, S.; Matsumura, Y.; Konishi, Y.; Tanaka, Y.;
Kikuzaki, H. Food Chem. 2011,125, 471475.
(27) Mazibuko, M. E. In Vitro and in Vivo Eect of Aspalathus
linearis and Its Major Polyphenols on Carbohydrate and Lipid
Metabolism in Insulin Resistant Models. Ph.D. Thesis, University of
Zululand, KwaDlangezwa, South Africa, 2013.
Journal of Natural Products Article
dx.doi.org/10.1021/np5007247 |J. Nat. Prod. XXXX, XXX, XXXXXXF
... Benzophenone derivatives has been reported to possess a wide range of bioactivities such as antimicrobial, antifungal, anti-carcinogenic, anti-HIV [19], antioxidant [20], anticancer [21], anesthetic [22], anti-inflammatory [23], and urease inhibitory [24] activities. In literature, benzophenone-O-glycosides are reported as α-glucosidase inhibitors [25,26] (Figure-1). ...
... A cubic box with a size of 8 Å was employed for the systems' solvation [24]. For system neutralization, Na + or Cl counter ions were supplied [25]. To reduce the energy of the systems, the two algorithms steepest descent and conjugate gradients were applied. ...
Article
Full-text available
Inhibiting α-glucosidase is a reliable method for reducing blood sugar levels in diabetic individuals. Bis(dimethylamino)benzophenone derivatives 1–27 were synthesized from bis(dimethylamino)benzophenone via two-step reaction. Different spectroscopic techniques, including EI-MS and 1H NMR, were employed to characterize all synthetic derivatives. The elemental composition of synthetic compounds was confirmed by elemental analysis and results were found in agreement with the calculated values. The synthetic compounds 1–27 were evaluated for α-glucosidase inhibitory activity, except five compounds all derivatives showed good to moderate inhibitory potential in the range of IC50 = 0.28 ± 2.65 - 0.94 ± 2.20 μM. Among them, the most active compounds were 5, 8, 9, and 12 with IC50 values of 0.29 ± 4.63, 0.29 ± 0.93, 0.28 ± 3.65, and 0.28 ± 2.65, respectively. Furthermore, all these compounds were found to be non-toxic on human fibroblast cell lines (BJ cell lines). Kinetics study of compounds 8 and 9 revealed competitive type of inhibition with Ki values 2.79 ± 0.011 and 3.64 ± 0.012 μM, respectively. The binding interactions of synthetic compounds were also confirmed through molecular docking studies that indicated that compounds fit well in the active site of enzyme. Furthermore, a total of 30ns MD simulation was carried out for the most potent complexes of the series. The molecular dynamics study revealed that compound-8 and compound-12 were stable during the MD simulation.
... This observation may be attributed to the higher reactivity and susceptibility of aromatic protons to undergo deuterium exchange, particularly in methanol-d 4 solvent where the deuteration rate is higher compared to DMSO-d 6 (Figures S7 and S8). This phenomenon is commonly observed in polyhydroxy diaryl ketoside compounds [12][13][14]. Excluding the carbon signals of the two glucose moieties, the 13 C NMR spectrum exhibited 14 carbon signals, which included one carbonyl carbon at δ C 187.6 and 13 aromatic carbon signals. ...
Article
Full-text available
‘Albedo bluing’ of fruits occurs in many varieties of citrus, resulting in a significant reduction in their commercial value. We first presented a breakthrough method for successfully extracting and purifying the ‘albedo bluing’ substance (ABS) from citrus fruits, resulting in the attainment of highly purified ABS. Then, HPLC and UPLC-QTOF-MS were used to prove that ABS in the fruits of three citrus varieties (Citrus reticulate Blanco cv. ‘Gonggan’, ‘Orah’, and ‘Mashuiju’) are identical. However, the chemical structure of ABS remains elusive for many reasons. Fortunately, a more stable derivative of ABS (ABS-D) was successfully obtained. Through various analytical techniques such as HRESIMS, 1D and 2D NMR, and chemical shift calculation, ABS-D was identified as 2,4-dihydroxy-6-(β-D-glucopyranosyloxy)phenyl(5,6-dihydroxy-7-(β-D-glucopyranosyloxy)benzo[d]thiazol-2-yl)methanone, indicating that both ABS and its derivative belong to a rare category of benzothiazole glucosides. Furthermore, both ABS and ABS-D demonstrated potent antioxidant abilities. These findings lay the groundwork for further elucidating the chemical structure of ABS and the causative mechanism of the ‘albedo bluing’ phenomenon in citrus fruits.
... Among the isolated compounds 2,4,4′,6-tetrahydroxy-3′-methoxybenzophenone 3-C-β-D-glucopyranoside (40), arjunolic acid (78), and actinidic acid (79) demonstrated the most potent α-glucosidase inhibitory activity with IC 50 values of 284.93 ± 20.29 μM, 239.60 ± 25.00 μM, and 297.37 ± 8.12 μM, respectively (Pan et al. 2016). The authors attributed the effectiveness of compound (40) to the number of phenolic hydroxyl groups on the basic diphenyl ketone and absence of methoxy groups at C-4 (Beelders et al. 2014). Another study reported the in vitro enzyme inhibitory activity of solvent-partitioned fractions of M. indica leaves. ...
Chapter
Mangifera indica L., known as mango, is an evergreen tree with characteristic edible fruits belonging to family Anacardiaceae. It is characterized by rich nutritional value and medicinal aspects for human health. Traditionally, it has been used an aphrodisiac, astringent, appetite stimulant, diuretic, anti-asthmatic, and to relieve gastrointestinal symptoms, sore throat, and dysmenorrhea. Several pharmacological activities such as anti-diabetic, antioxidant, neuroprotective, immunomodulatory, antimicrobial, anti-inflammatory and antioxidant have reported in recent years. Numerous bioactive compounds as xanthonoids, flavonoids, tannins, triterpenoids, and phenolic acids have been isolated from different types of mango extracts. Most of them exploited anti-diabetic properties acting on α-amylase, α-glucosidase, dipeptidyl peptidase-IV, pancreatic β cells, insulin sensitivity, and antioxidant defense mechanisms. This book chapter is designed to pile up all information regarding the potential role of M. indica in the management of diabetes mellitus. We discuss the in vitro and in vivo anti-diabetic studies of different extracts and isolated compounds. Interestingly, we focus on the molecular mechanism, pharmacological action, and docking investigations that have been performed on isolated compounds. Hopefully, this chapter presents a source for researchers to develop a new plant-derived anti-diabetic agent with a promising efficacy and low side effects.
... Te recently observed popularity and the high demand of honeybush have gained much attention in the feld of research in the discovery of its novel bioactive compounds as they are used as nutraceuticals. Even though the chemical characterization, antioxidant potentials, agerelated neurodegenerative disorder, and antihyperglycemic activities of diferent extracts of Cyclopia genistoides have been reported [11], Agapouda et al., 2020 [12], more studies are still required to further support its benefcial properties. Terefore, this study aimed at screening the phytochemical constituents of varying extracts of Cyclopia genistoides through GC-MS analysis, investigating the in vitro antidiabetic properties of the extracts on some key linked diabetic enzymes, as well as their antioxidant and cytotoxicity potentials. ...
Article
Full-text available
Background. This study evaluated in vitro antidiabetic and antioxidant properties of different extracts (n-hexane, dichloromethane (DCM), and 70% ethanol) of honeybush tea (Cyclopia genistoides). Over a period of 28 days, antiprotein glycation was evaluated and some antidiabetic indicators (α-amylase, α-glucosidase, and pancreatic lipase inhibitory effects) and antioxidant activities (DPPH, ABTS, hydroxyl radical, metal ion chelating, and reducing power) for each of the crude extracts were also investigated. The results showed that all of the tested C. genistoides extracts had strong α-amylase and lipase inhibitory activity in a concentration-dependent manner with IC50 values from 0.018 μg/ml (DCM extract) to 9.93 μg/ml (n-hexane extract), respectively. The extracts also displayed inhibitory effects on protein glycation between the 14th and 28th days. The DCM and ethanolic extracts further exhibited strong antioxidant activities as they effectively scavenged most of the radicals tested, with IC50 values ranging from 0.014–0.048 mg/ml to 0.019–0.043 mg/ml. Two hundred and seventy-four chemical constituents had been identified by GC-MS, with the n-hexane extract having the highest number of peaks (127) followed by the DCM extract (107). Six compounds were identified across all the following three extracts: decane (RT: 6.4), undecane (RT: 7.7), dodecane (RT: 9.00), phytol (RT: 21.32), heptadecanoic acid, 9-methyl, methyl ester (RT: 21.65), and 9-octadecenamide (RT: 24.30). The cytotoxicity of the extracts against C3A cell lines was determined using the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide) (MTT) assay, which demonstrated that honeybush tea had a toxicity effect ranging from 66.3–88.4 μg/ml on C3A cell lines. The results showed that honeybush has antioxidant and antidiabetic activities, which could be partially attributed to the phytochemical compounds identified within the extracts.
... A recent review has mentioned C. genistoides used as in the form of decoction or expectorant in the treatment of tuberculosis, chronic catarrh, and consumption [13]. Additionally, Beelders, Brand [21] reported the inhibition activity of C. genistoides isolated compounds on mammalian α-glucosidase, which further highlighted by works that demonstrated the synergistic effects of C. genistoides with acarbose in the inhibition activity of mammalian intestinal α-glucosidase in vitro [22]. One earlier paper published by Hubbe and Joubert [23] exhibited the in vitro antioxidant of C. genistoides. ...
Article
Red honeybush tea (RHT) is a tea product developed from Cyclopia spp. which is endemic to South Africa. Aside refreshment, RHT has over the years been used traditionally in the treatment of various diseases including type 2 diabetes. This study investigated the in vivo antioxidant and anti-diabetic activity of RHT concentrated hot water extract in type 2 diabetes (T2D) model of rats. T2D was induced starting with feeding 10% fructose solution ad libitum for 2 weeks followed by a single intraperitoneal injection of streptozotocin (40 mg/kg body weight (BW)). Five weeks of treatment of RHT led to significant (P < 0.05) elevation in serum insulin, pancreatic β-cell function, HDL-C levels with concomitant decrease in AST, ALT, ALP, urea, CK-MB, fructosamine, total cholesterol, triglycerides, LDL-C, and insulin resistance in diabetic rats. RHT also signifi cantly (P < 0.05) decreased MDA levels and enhanced level of GSH, activity of SOD, catalase, GR in most of organs (pancreas, liver, kidneys, and heart). Signifi cantly (P < 0.05) improved morphological changes in the islets and β-cells were observed in rats treated with RHT. The data of this study suggest that RHT demonstrates an outstanding antioxidant and anti-diabetic effects in STZ-induced T2D model of rats.
... Xanthone exhibits anti-inflammatory and anti-diabetic activities [127]. The occurrence of mangiferin with benzophenone α-glucosidase inhibitors in Cyclopia [128], and any other components that improve glucose uptake in vitro, such as iso mangiferin and scolymoside [129], makes Cyclopia extract a potential anti-diabetic nutraceutical [130]. ...
Article
Full-text available
South Africa is blessed with vast plant resources and unique vegetation types. Indigenous South African medicinal plants have been well-harnessed to generate income in rural communities. Many of these plants have been processed into natural products to heal a variety of diseases, making them valuable export commodities. South Africa has one of the most effective bio-conservation policies in Africa, which has protected the South African indigenous medicinal vegetation. However, there is a strong link between government policies for biodiversity conservation, the propagation of medicinal plants as a source of livelihood, and the development of propagation techniques by research scientists. Tertiary institutions nationwide have played a crucial role in the development of effective propagation protocols for valuable South African medicinal plants. The government-restricted harvest policies have also helped to nudge natural product companies and medicinal plant marketers to embrace the cultivated plants for their medicinal uses, and thus have helped support the South African economy and biodiversity conservation. Propagation methods used for the cultivation of the relevant medicinal plants vary according to plant family and vegetation type, among others. Plants from the Cape areas, such as the Karoo, are often resuscitated after bushfires, and propagation protocols mimicking these events have been established through seed propagation protocols with controlled temperatures and other conditions, to establish seedlings of such plants. Thus, this review highlights the role of the propagation of highly utilized and traded medicinal plants in the South African traditional medicinal system. Some valuable medicinal plants that sustain livelihoods and are highly sought-after as export raw materials are discussed. The effect of South African bio-conservation registration on the propagation of these plants and the roles of the communities and other stakeholders in the development of propagation protocols for highly utilized and endangered medicinal plants are also covered. The role of various propagation methods on the bioactive compounds’ composition of medicinal plants and issues of quality assurance are addressed. The available literature, media online news, newspapers, and other resources, such as published books and manuals, were scrutinized for information.
Article
Full-text available
Naturally occurring benzophenones represent a relatively small group of plant metabolites with narrow distribution, mainly in members of Clusiaceae, Gentianaceae, Hypericaceae, Polygalaceae, Myrtaceae, etc.; however, there were reports of several compounds derived from microorganisms belonging to the Aspergillaceae and Valsaceae families and propolis. Benzophenones exhibit many biological activities, such as antioxidant, anti-inflammatory, cytotoxic, antimicrobial, etc. Few reviews on benzophenones that have appeared in the literature were focused on their prenylated derivatives. Summarized information on structural diversity, distribution, and biological activities of simple oxygenated naturally occurring benzophenones and their glycosides has not been found in the literature. Until 2000, only benzophenone C-glycosides were known to occur in nature. Since then, many O-glycosides have been isolated, structurally, and biologically characterized. This review covers the years from 1850 to 2023 and was compiled using databases such as Chemical Abstracts, Scopus, Google Scholar, PubMed, and ResearchGate. Based on their degree of oxidation, 210 chemical structures of benzophenone derivatives and glycosides were grouped into six categories. In addition, in one group of 40 miscellaneous benzophenones, where one or several protons are replaced by a methyl, alcohol, carboxyl, or acyl group, glycosidic forms with such an aglycone and dimeric compounds with xanthone was included. Simple oxygenated benzophenones and their glycosides were found in 77 plant genera belonging to 44 families. The allergy-associated bezophenone-1, benzophenone-2 and benzophenone-3 have limited distribution across natural sources. A wide range of biological activities (antioxidant, anti-inflammatory, cytotoxic, antitumor, cytoprotective, antimicrobial, MAO-A, antiarthritic, anticholinesterase, anti-atherosclerotic, laxative, etc.) of simple oxygenated benzophenones and their glycosides that appeared in the literature were discussed.
Chapter
Cyclopia genistoides (L.) R.Br (Fabaceae), commonly referred to as ‘honeybush tea’, is one of 23 Cyclopia species endemic to South Africa. The natural habitat of this small fynbos shrub is restricted to very small areas in the Western Cape Province, spanning from the West Coast to Mossel Bay on the Southern Cape coast. Honeybush tea is mostly enjoyed as a hot beverage of the ‘fermented’ product. ‘Fermentation’ refers to the high-temperature oxidation process essential for the development of the sought-after sweet, floral aroma and flavour, and brown colour. Traditional medicinal uses include use as an expectorant in pulmonary tuberculosis, chronic catarrh, and a restorative with astringent properties. ‘Caspa Cyclopia Tea’ was the first branded product to appear on the market in the 1960s. The development of a formal industry in the 1990s gave rise to different branded honeybush products, and the production of extracts. Several studies investigating the anti-oxidant, anticancer, antidiabetic, anti-obesity, antimicrobial, anti-inflammatory and immunomodulating activities have been documented for different Cyclopia species (fermented and unfermented). Quality control protocols based on the chromatographic profiling of methanol extracts were developed using a semi-automated high-performance thin-layer chromatography (HPTLC) system. The HPTLC profiles of the extracts viewed under 366 nm radiation after derivatisation with vanillin-sulphuric acid reagent revealed the presence of mangiferin, isomangiferin and hesperidin in all samples, compounds ubiquitous to all Cyclopia species. Their presence in the methanol extracts was confirmed by ultra-performance liquid chromatography coupled to mass spectrometry (UPLC–MS).
Article
Full-text available
A high-performance liquid chromatographic (HPLC) method coupled with diode-array detection (DAD) was optimized for the qualitative analysis of aqueous extracts of Cyclopia genistoides. Comprehensive insight into the phenolic profile of unfermented and fermented sample extracts was achieved with the identification of ten compounds based on comparison with authentic reference standards and the tentative identification of 30 additional compounds by means of electrospray ionization mass spectrometry (ESI-MS) and tandem MS detection. Three iriflophenone-di-O,C-hexoside isomers, three xanthone-dihydrochalcone derivatives and one dihydrochalcone are herein tentatively identified for the first time in C. genistoides. Of special interest is one iriflophenone-di-O,C-hexoside present in large amounts. New compounds (tentatively) identified for the first time in this species, and also in the genus Cyclopia, include two aromatic amino acids, one flavone, an iriflophenone-di-C-hexoside, a maclurin-di-O,C-hexoside, two tetrahydroxyxanthone-C-hexoside isomers, a tetrahydroxyxanthone-di-O,C-hexoside, two symmetric tetrahydroxyxanthone-C-hexoside dimers, nine glycosylated flavanone derivatives and five glycosylated phenolic acid derivatives. The presence of new compound subclasses in Cyclopia, namely aromatic amino acids and glycosylated phenolic acids, was demonstrated. The HPLC-DAD method was successfully validated and applied to the quantitative analysis of the paired sample extracts. In-depth analysis of the chemical composition of C. genistoides hot water extracts gave a better understanding of the chemistry of this species that will guide further research into its medicinal properties and potential uses.
Article
Full-text available
The dietary polyphenols as α-glucosidases inhibitors have attracted great interest among researchers. The aim of this review is to give an overview of the research reports on the structure-activity relationship of dietary polyphenols inhibiting α-glucosidases. The molecular structures that influence the inhibition are the following: (1) The hydroxylation and galloylation of flavonoids including catechins improve the inhibitory activity. (2) The glycosylation of hyroxyl group and hydrogenation of the C2=C3 double bond on flavonoids weaken the inhibition. (3) However, cyaniding glycosides show higher inhibition against than cyanidin. Proanthocyanidins oligomers exhibit a stronger inhibitory activity than their polymers. (4) The hydroxylation on B ring and the glycosylation of stilbenes reduce the inhibitory activity. (5) Caffeoylquinic acids display strong inhibition against α-glucosidases. However, hydroxycinnamic acid, ferulic acid, and gallic acid hardly inhibited α-glucosidases. (6) The coupled galloyl structures attached to C-3 and C-6 of the 4C(1) glucose core of ellagitanin gave basic inhibitory activity. (7) The mono-glycosylation of chalcones slightly lowers the inhibition. However, the diglycosylation of chalcones significantly decreased the activity.
Article
Six new galloyl and p-hydroxybenzoyl esters (2-7) of benzophenone C-glucosides have been isolated, together with a new benzophenone C-glucoside (1), from the leaves of Mangifera indica L. (Anacardiaceae). On the basis of chemical and spectroscopic evidence, the structures of these compounds have been established as maclurin 3-C-β-D-glucoside (1), maclurin 3-C-(6"-O-p-hydroxybenzoyl)-β-D-glucoside (2), maclurin 3-C-(2"-O-galloyl-6"-O-p-hydroxybenzoyl)-β-D-glucoside (3), maclurin 3-C-(2"-O-p-hydroxybenzoyl-6"-O-galloyl)-β-D-glucoside (4), maclurin 3-C-(2", 3", 6"-tri-O-galloyl)-β-D-glucoside (5), iriflophenone 3-C-(2", 6"-di-O-galloyl)-β-D-glucoside (6) and iriflophenone 3-C-(2", 3", 6"-tri-O-galloyl)-β-D-glucoside (7). (-)-Epicatechin 3-O-gallate (9), mangiferin (10), isomangiferin (11) and a new xanthone C-glucoside gallate, mangiferin 6'-O-gallate (8), have also been isolated and their structures have been similarly characterized. Furthermore, the above plant source contained polygalloylglucoses which were characterized on the basis of chemical and high performance liquid chromatographic analyses as a mixture of penta-to undecagalloylglucoses based on a 1, 2, 3, 4, 6-penta-O-galloyl-β-D-glucose core. Maclurin 3-C-glucoside (1) has been transformed enzymatically to mangiferin (10) and isomangiferin (11), and it has become clear that 1 is a key intermediate in the biosynthesis of 10 and 11.
Article
A fast and efficient method for the isolation of the C-glucosidated xanthones mangiferin and isomangiferin from the South-African plant Cyclopia genistoides was developed for the first time. The procedure involved extraction, liquid-liquid partitioning with ethyl acetate and subsequent precipitation of mangiferin and isomangiferin from methanol and acetonitrile-water fractions, respectively. Additionally, two benzophenone derivatives: 3-C-β-glucosides of maclurin and iriflophenone, were isolated from C. genistoides extracts using semi-preparative HPLC. Apart from the above, the isolation procedure also yielded hesperidin and small amounts of luteolin. The structures of the compounds were determined by 1D and 2D NMR experiments and/or LC-DAD-ESI-MS. The selected Cyclopia constituents were screened for pro-apoptotic activity on TNF-α-stimulated synovial cells isolated from rheumatoid arthritis patients. The strongest effect, measured as percent of apoptotic cells, was recorded for isomangiferin (75 %), followed by iriflophenone 3-C-β-glucoside (71 %), hesperidin (67 %) and mangiferin (65 %). The results are encouraging for further studies on the use of the above compounds in the treatment of rheumatoid arthritis.
Article
Ethanolic extract of Mangifera indica (mango) dose-dependently decreased serum glucose and triglyceride in KK-A(y) mice. Our in vitro and in vivo investigations revealed that the effect of mango leave extract (ME) on glucose and lipid homeostasis is mediated, at least in part, through the PI3K/AKT and AMPK signaling pathway. ME up-regulated the expression of PI3K, AKT and GYS genes by 2.0-fold, 3.2-fold, and 2.7-fold, respectively, leading to a decrease in glucose level. On the other hand, ME up-regulated AMPK and altered lipid metabolism. ME also down-regulated ACC (2.8-fold), HSL (1.6-fold), FAS (1.8-fold) and PPAR-γ (4.0-fold). Finally, we determined that active metabolites of benzophenone C-glucosides, Iriflophenone 3-C-β-glucoside and Foliamangiferoside A from ME, may play a dominant role in this integrated regulation of sugar and lipid homeostasis.
Article
In Brief Choosing medications for people with diabetes involves consideration of a number of factors, including effects on weight. Improvements in glucose control are often linked to weight gain, but this does not have to be the inevitable result of diabetes treatment. Adding a drug that either promotes weight-loss or is weight neutral to one that promotes weight gain and providing medical nutrition therapy can be considered.
Article
In the presented work, an insight was made into the polyphenolic composition of intact plant material and in vitro cultures of indigenous South African plant Cyclopia subternata Vogel (honeybush). Ethyl acetate fractions of methanol extracts were separated by means of gravity column chromatography and/or semipreparative HPLC on two serially connected monolithic RP-18 columns. The structures of the isolated compounds were determined by means of 1D and 2D NMR techniques and additionally confirmed by LC-DAD-ESI-MS. Apart from the previously described honeybush components, that is mangiferin (1), scolymoside (2), hesperidin (3) and narirutin (4), three additional compounds: iriflophenone 3-C-beta-glucoside (benzophenone) (5), phloretin 3',S'-di-C-beta-glucoside (dihydrochalcone) (6), and isorhoifolin (flavone) (7) were identified for the first time in the herb of C. subternata. Additionally, three isoflavone glucosides, namely calycosin 7-O-beta-glucoside (8), rothindin (9) and ononin (10), which had not been previously reported in Cyclopia plants, were identified in the callus of the above species. As far as the authors are concerned, this is the first report on the presence of benzophenone and dihydrochalcone derivatives in Cyclopia genus.
Article
Honeybush tea (Cyclopia spp.), one of the traditional South African herbal teas with a long history of regional use, remained a cottage industry until the mid-1990s when researchers were instrumental in the development of a formal agricultural and agro-processing industry. It is one of the few indigenous South African plants that made the transition from the wild to a commercial product during the past 100 years. Research activities during the past 20 years included propagation, production, genetic improvement, processing, composition and the potential for value-adding. The present review provides an up-to-date and comprehensive record of the development of the South African honeybush industry, against the background of the historical highlights in the making of an industry. It provides a blueprint of the processes and actions involved in the development of a new agricultural and agro-processing industry from an herbal plant. Insight into challenges faced by the industry and future research needed to keep it competitive are provided.