ArticlePDF Available

Antidiabetic, Antihyperlipidemic, and Antioxidant Evaluation of Phytosteroids from Notholirion thomsonianum (Royle) Stapf

Authors:

Abstract

Abstract: Diabetes mellitus (DM) is a metabolic complication and can pose a serious challenge to human health. DM is the main cause of many life-threatening diseases. Researchers of natural products have been continuously engaged in treating vital diseases in an economical and efficient way. In this research, we extensively used phytosteroids from Notholirion thomsonianum (Royle) Stapf for the treatment of DM. The structures of phytosteroids NtSt01 and NtSt02 were confirmed with gas chromatography-mass spectrometry (GC-MS) and nuclear magnetic resonance (NMR) analyses. Through in vitro studies including α-glucosidase, α-amylase, and DPPH assays, compound NtSt01 was found to be comparatively potent. An elevated dose of compound NtSt01 was also found to be safe in an experimental study on rats. With a dose of 1.0 mg/kg of NtSt01, the effect on blood glucose levels in rats was observed to be 519 ± 3.98, 413 ± 1.87, 325 ± 1.62, 219 ± 2.87, and 116 ± 1.33 mg/dL on the 1st, 7th, 14th, 21st, and 28th, days, respectively. The in vivo results were compared with those of glibenclamide, which reduced the blood glucose level to 107 ± 2.33 mg/dL on the 28th day. On the 28th day of NtSt01 administration, the average weights of the rats and vital organs (liver, kidney, pancreas, and heart) remained healthy, with a slight increase. The biochemical parameters of the blood, i.e., serum creatinine, blood urea, serum bilirubin, SGPT (or ALT), and serum alkaline phosphatase, of rats treated with NtSt01 remained in the normal ranges. Similarly, the serum cholesterol, triglycerides, high-density lipoprotein (HDL), and low-density lipoprotein (LDL) levels also remained within the standard ranges. It is obvious from our overall results that the phytosteroids (specifically NtSt01) had an efficient therapeutic effect on the blood glucose level, protection of vital organs, and blood biochemistry.
Citation: Huneif, M.A.; Fahad, S.;
Abdulwahab, A.; Alqahtani, S.M.;
Mahnashi, M.H.; Nawaz, A.; Hussain,
F.; Sadiq, A. Antidiabetic,
Antihyperlipidemic, and Antioxidant
Evaluation of Phytosteroids from
Notholirion thomsonianum (Royle)
Stapf. Plants 2023,12, 3591. https://
doi.org/10.3390/plants12203591
Academic Editors: Natália
Cruz-Martins and Christophe Hano
Received: 15 August 2023
Revised: 3 October 2023
Accepted: 7 October 2023
Published: 17 October 2023
Copyright: © 2023 by the authors.
Licensee MDPI, Basel, Switzerland.
This article is an open access article
distributed under the terms and
conditions of the Creative Commons
Attribution (CC BY) license (https://
creativecommons.org/licenses/by/
4.0/).
plants
Article
Antidiabetic, Antihyperlipidemic, and Antioxidant Evaluation
of Phytosteroids from Notholirion thomsonianum (Royle) Stapf
Mohammad A. Huneif 1, Shah Fahad 2, Alqahtani Abdulwahab 1, Seham M. Alqahtani 1,
Mater H. Mahnashi 3, * , Asif Nawaz 4, Fida Hussain 5and Abdul Sadiq 4, *
1Pediatric Department, Medical College, Najran University, Najran 61441, Saudi Arabia;
maalhuneif@nu.edu.sa (M.A.H.); aaalsharih@nu.edu.sa (A.A.); drseham2015@gmail.com (S.M.A.)
2Department of Agronomy, Abdul Wali Khan University Mardan, Mardan 23200, KP, Pakistan;
shahfahad@awkum.edu.pk
3Department of Pharmaceutical Chemistry, College of Pharmacy, Najran University, Najran 61441, Saudi Arabia
4Department of Pharmacy, Faculty of Biological Sciences, University of Malakand,
Chakdara 18000, KP, Pakistan; asifnawaz2446@gmail.com
5Department of Pharmacy, University of Swabi, Swabi 23561, KP, Pakistan; fida2k9@yahoo.com
*Correspondence: matermaha@gmail.com (M.H.M.); sadiquom@yahoo.com (A.S.);
Tel.: +966-508734539 (M.H.M.)
Abstract:
Diabetes mellitus (DM) is a metabolic complication and can pose a serious challenge to
human health. DM is the main cause of many life-threatening diseases. Researchers of natural
products have been continuously engaged in treating vital diseases in an economical and efficient
way. In this research, we extensively used phytosteroids from Notholirion thomsonianum (Royle)
Stapf for the treatment of DM. The structures of phytosteroids
NtSt01
and
NtSt02
were confirmed
with gas chromatography–mass spectrometry (GC-MS) and nuclear magnetic resonance (NMR)
analyses. Through
in vitro
studies including
α
-glucosidase,
α
-amylase, and DPPH assays, compound
NtSt01
was found to be comparatively potent. An elevated dose of compound
NtSt01
was also
found to be safe in an experimental study on rats. With a dose of 1.0 mg/kg of NtSt01, the effect on
blood glucose levels in rats was observed to be 519
±
3.98, 413
±
1.87, 325
±
1.62, 219
±
2.87, and
116 ±1.33 mg/dL
on the 1st, 7th, 14th, 21st, and 28th, days, respectively. The
in vivo
results were
compared with those of glibenclamide, which reduced the blood glucose level to 107
±
2.33 mg/dL
on the 28th day. On the 28th day of
NtSt01
administration, the average weights of the rats and vital
organs (liver, kidney, pancreas, and heart) remained healthy, with a slight increase. The biochemical
parameters of the blood, i.e., serum creatinine, blood urea, serum bilirubin, SGPT (or ALT), and
serum alkaline phosphatase, of rats treated with
NtSt01
remained in the normal ranges. Similarly,
the serum cholesterol, triglycerides, high-density lipoprotein (HDL), and low-density lipoprotein
(LDL) levels also remained within the standard ranges. It is obvious from our overall results that
the phytosteroids (specifically
NtSt01
) had an efficient therapeutic effect on the blood glucose level,
protection of vital organs, and blood biochemistry.
Keywords:
Notholirion thomsonianum; diabetes;
in vivo
and
in vitro
studies; phytosteroids; blood
biochemistry
1. Introduction
As a chronic disease, DM is a metabolic disorder in which insulin production by the
pancreas is reduced or the produced insulin may be ineffective, leading to hyperglycemia,
which can cause further damage to other body systems like the circulatory and nervous
systems. Some of the abnormalities that account for DM include defects in the production
of insulin or in its action or secretion and dysfunction in the metabolism of fat, protein, and
carbohydrates [
1
3
]. Polyphagia, polyuria, and polydipsia are the some of the symptoms
associated with the state of hyperglycemia [
4
]. There are about 450 million diabetic patients
Plants 2023,12, 3591. https://doi.org/10.3390/plants12203591 https://www.mdpi.com/journal/plants
Plants 2023,12, 3591 2 of 14
around the globe, with this figure expected to reach 690 million by the year 2044 [
5
].
Currently, DM prevalence has been reported as 8.5% of adults globally, and a rapid increase
has been observed in countries with a low or middle income [
6
]. DM is a serious metabolic
disease, and chronic hyperglycemia can cause numerous complications and the dysfunction
of several organs like the kidneys, nerves, eyes, heart, blood vessels, and liver [2,7,8].
There are different types of DM; the common types are type 1 and type 2 DM. Type
1 DM is insulin-dependent and is due to insulin deficiency, along with an impairment
of the
β
cells of the pancreas [
9
], whereas type 2 DM is non-insulin-dependent and is
due to insulin resistance or decreased insulin secretion [
10
]: 95% of diabetic patients have
type 2 DM [
11
]. DM development in people with an impaired tolerance of glucose can be
prevented or managed with the use of antidiabetic drugs or through changing their lifestyle
via exercise, diet control, and/or weight loss [
12
]. The use of antidiabetic drugs can have
number of adverse effects, including hypoglycemia, retention of fluids, osteoporosis, and
heart failure, due to which their use is limited [
13
15
]. Hence, the development of new,
effective drugs that have fewer adverse effects is needed to control and manage diabetes. In
the development of antidiabetic drugs that are specific for type 2 DM, several biochemical
approaches can be used. Among the important biochemical pathways, the inhibition of
α
-amylase and
α
-glucosidase is common. Both of these enzymes break down starch and
oligosaccharides into glucose, leading to an increase in the concentration of glucose; hence,
their inhibition is important for decreasing glucose absorption in the intestine [16].
For thousands of years, medicinal plants and natural products have been reported
for the treatment of many diseases, including diabetes, especially type 2 DM [
17
19
]. The
crude extracts of medicinal plants and their bioactive compounds have been found to be
useful in many pharmacological activities [
20
22
]. Approximately 400 plants have been
demonstrated to have antidiabetic activity, but only some of them have been evaluated for
their efficacy [
23
]. A number of natural products of plant origin have been shown to have an
antidiabetic activity. The most important reported phytochemicals include alkaloids, carbo-
hydrates, peptidoglycan, amino acids, glycosides, steroids, glycopeptides, galactomannan
gum, terpenoids, hypoglycans, guanidine, and inorganic ions [
24
]. Different plants and
microorganisms produce
α
-glucosidase and
α
-amylase inhibitors for the regulation of such
enzyme activities [
25
]. Synthetic compounds are also being developed in parallel in order
to create
α
-glucosidase and
α
-amylase inhibitors [
26
,
27
]. Inhibitors of
α
-amylase enzymes
reduce the conversion of starch into glucose usually after eating a meal, which results in a
decrease in the level of glucose in the blood. Therefore,
α
-amylase inhibitors are needed to
control the glucose levels of diabetic patients.
Notholirion thomsonianum is a small bulbous plant of the family Liliaceae. This small
lilium-like medicinal plant has been studied for various pharmacological effects [
28
]. The
plant can be used to improve the digestive system and in the management of microbial
infections [
29
]. Our group has been exploring the medicinal aspects of this species for a
decade. We have previously explored its crude extract for antibacterial, antifungal, and
analgesic effects [
29
,
30
]. In recent years, we explored the potential of this plant for the
management of diabetes mellitus using its various fractions and some bio-guided bioactive
compounds following multitarget
in vitro
and in silico approaches [
31
]. Based on our
previous experience with this plant, its hydroalcoholic extract contains phytosteroids,
which have been identified. This current study is extensive compared with our previously
published work. In this study, we extensively used these phytosteroids for investigating
in vitro
and
in vivo
antidiabetic targets. Furthermore, the beneficial effects of the identified
phytosteroids were also explored on the vital organs of the body like the liver, kidney,
pancreas, and heart and on blood biochemistry.
2. Results
2.1. Phytochemistry
In this research, we initially purified and identified two different phytosteroids (
NtSt01
and
NtSt02
, as shown in Figure 1). The isolated amount of
NtSt01
was 830 mg as a white
Plants 2023,12, 3591 3 of 14
powder, while 375 mg of
NtSt02
was isolated as a yellowish-brown solid. The structures
of these two isolated compounds were initially confirmed with GCMS analysis. The re-
tention time of compound
NtSt01
was 56.964 min, with a base peak value of 55.1 (Table
S1 Supporting Information). The fragmentation pattern of compound
NtSt01
is shown
in the Figure S1 of the Supporting Information. The spectrum and its fragmentation pat-
tern were compared with the library spectrum and the difference spectrum, as shown in
Figures S2 and S3 of the Supporting Information, respectively. The chemical name of the
identified compound
NtSt01
is 3-
β
-Acetoxystigmasta-4,6,22-triene with an IUPAC name
of (E)-17-(5-ethyl-6-methylhept-3-en-2-yl)-10,13-dimethyl-2,3,8,9,10,11,12,13,14,15,16,17-
dodecahydro-1H-cyclopenta[a]phenanthren-3-yl acetate. Similarly, from the same GCMS
analysis, the compound
NtSt02
was observed at a retention time of 57.812 min, with a major
peak at m/z135 (Table S2 of Supporting Information). The fragmentation pattern of the com-
pound
NtSt02
is shown in Figure S4 of the Supporting Information. This spectrum and its
fragmentation pattern were compared with the library spectrum and difference spectrum,
as shown in Figures S5 and S6 of the Supporting Information, respectively. The chemi-
cal name of the identified compound
NtSt02
is 4,6-cholestadien-3beta-ol, benzoate, and
the IUPAC name is 10,13-dimethyl-17-(6-methylheptan-2-yl)-2,3,8,9,10,11,12,13,14,15,16,17-
dodecahydro-1H-cyclopenta[a]phenanthren-3-yl benzoate.
Plants 2023, 12, x FOR PEER REVIEW 3 of 15
2. Results
2.1. Phytochemistry
In this research, we initially puried and identied two dierent phytosteroids
(NtSt01 and NtSt02, as shown in Figure 1). The isolated amount of NtSt01 was 830 mg as
a white powder, while 375 mg of NtSt02 was isolated as a yellowish-brown solid. The
structures of these two isolated compounds were initially conrmed with GCMS analysis.
The retention time of compound NtSt01 was 56.964 min, with a base peak value of 55.1
(Table S1 Supporting Information). The fragmentation paern of compound NtSt01 is
shown in the Figure S1 of the Supporting Information. The spectrum and its fragmenta-
tion paern were compared with the library spectrum and the dierence spectrum, as
shown in Figures S2 and S3 of the Supporting Information, respectively. The chemical
name of the identied compound NtSt01 is 3-β-Acetoxystigmasta-4,6,22-triene with an
IUPAC name of (E)-17-(5-ethyl-6-methylhept-3-en-2-yl)-10,13-dimethyl-
2,3,8,9,10,11,12,13,14,15,16,17-dodecahydro-1H-cyclopenta[a]phenanthren-3-yl acetate.
Similarly, from the same GCMS analysis, the compound NtSt02 was observed at a reten-
tion time of 57.812 min, with a major peak at m/z 135 (Table S2 of Supporting Information).
The fragmentation paern of the compound NtSt02 is shown in Figure S4 of the Support-
ing Information. This spectrum and its fragmentation paern were compared with the
library spectrum and dierence spectrum, as shown in Figures S5 and S6 of the Support-
ing Information, respectively. The chemical name of the identied compound NtSt02 is
4,6-cholestadien-3beta-ol, benzoate, and the IUPAC name is 10,13-dimethyl-17-(6-
methylheptan-2-yl)-2,3,8,9,10,11,12,13,14,15,16,17-dodecahydro-1H-cyclopenta[a]phe-
nanthren-3-yl benzoate.
Figure 1. Chemical structures of the identied phytosteroids in Notholirion thomsonianum.
2.2. Alpha Glucosidase Inhibition
The in vitro α-glucosidase inhibitory results of both phytosteroids of N. thomsonia-
num (NtSt01 and NtSt02) are shown in Table 1. The percent inhibitions were recorded on
all concentrations in triplicate. NtSt01 was three times more potent than NtSt02, as ob-
served from their respective IC50 values. NtSt01 exhibited inhibitions of 85.00 ± 1.52, 81.52
± 1.85, 77.63 ± 1.56, 68.78 ± 1.02, and 61.22 ± 0.85% at experimental concentrations of 500,
250, 125, 62.50, and 31.25 μg/mL, respectively. The IC50 values of NtSt01 and NtSt02 were
7.34 and 22.87 μg/mL, respectively, in comparison to the standard drug, acarbose, with an
IC50 value of 2.14 μg/mL.
Table 1. Alpha-glucosidase inhibitions of the phytosteroids.
Comp/Standard
Conc (μg/mL)
Percent Inhibition
(Mean ± SEM)
IC50 (μg/mL)
NtSt01
500
250
85.00 ± 1.52 ***
81.52 ± 1.85 ***
7.34
Figure 1. Chemical structures of the identified phytosteroids in Notholirion thomsonianum.
2.2. Alpha Glucosidase Inhibition
The
in vitro α
-glucosidase inhibitory results of both phytosteroids of N. thomsonianum
(
NtSt01
and
NtSt02
) are shown in Table 1. The percent inhibitions were recorded on all
concentrations in triplicate.
NtSt01
was three times more potent than
NtSt02
, as observed
from their respective IC
50
values.
NtSt01
exhibited inhibitions of 85.00
±
1.52, 81.52
±
1.85,
77.63
±
1.56, 68.78
±
1.02, and 61.22
±
0.85% at experimental concentrations of 500, 250,
125, 62.50, and 31.25
µ
g/mL, respectively. The IC
50
values of
NtSt01
and
NtSt02
were 7.34
and 22.87
µ
g/mL, respectively, in comparison to the standard drug, acarbose, with an IC
50
value of 2.14 µg/mL.
2.3. Alpha Amylase Inhibition
The
in vitro α
-amylase activities of
NtSt01
and
NtSt02
were also analyzed in compari-
son to the standard acarbose, as shown in Table 2. Likewise,
NtSt01
was found with very
practical result which was eleven folds more potent than
NtSt02
. The IC
50
value of
NtSt01
and
NtSt02
were 4.17 and 46.73
µ
g/mL respectively in comparison to the standard drug
acarbose with the IC
50
value of 1.96
µ
g/mL. The
NtSt01
demonstrated percent inhibitions
of 80.03
±
2.11, 75.52
±
0.96, 71.63
±
0.92, 67.63
±
2.51 and 62.35
±
1.78% at experimental
concentrations of 500, 250, 125, 62.50 and 31.25 µg/mL respectively.
Plants 2023,12, 3591 4 of 14
Table 1. Alpha-glucosidase inhibitions of the phytosteroids.
Comp/Standard Conc (µg/mL) Percent Inhibition
(Mean ±SEM) IC50 (µg/mL)
NtSt01
500
250
125
62.50
31.25
85.00 ±1.52 ***
81.52 ±1.85 ***
77.63 ±1.56 ***
68.78 ±1.02 ***
61.22 ±0.85 ***
7.34
NtSt02
500
250
125
62.50
31.25
77.56 ±3.22 ***
70.63 ±2.45 ***
64.52 ±3.15 ***
59.98 ±1.88 ***
52.63 ±1.52 ***
22.87
Standard Drug
500
250
125
62.50
31.25
92.65 ±0.55
89.53 ±1.45
83.89 ±2.65
78.63 ±1.98
70.52 ±2.63
2.14
All the values are expressed as mean
±
SEM compared with the standard. Two-way ANOVA followed by
Dunnett’s test was applied. ***, significantly different (p< 0.001) compared with standard drug.
Table 2. Alpha amylase inhibitions of the phytosteroids.
Comp/Standard Conc (µg/mL) Percent Inhibition
(Mean ±SEM) IC50 (µg/mL)
NtSt01
500
250
125
62.50
31.25
80.03 ±2.11 **
75.52 ±0.96 **
71.63 ±0.92 **
67.63 ±2.51 **
62.35 ±1.78 **
4.17
NtSt02
500
250
125
62.50
31.25
74.99 ±1.53 ***
64.32 ±1.85 ***
60.04 ±0.86 ***
53.10 ±2.05 ***
46.84 ±0.67 ***
46.73
Standard Drug
500
250
125
62.50
31.25
91.01 ±1.36
87.79 ±1.27
82.33 ±1.00
75.63 ±0.86
71.07 ±1.82
1.96
All the values are expressed as mean
±
SEM compared with the standard. Two-way ANOVA followed by
Dunnett’s test was applied. ** = p< 0.01, *** = p< 0.001 compared with standard drug.
2.4. Antioxidant Assay
Antioxidant activity is a very important supplementary activity in antidiabetic studies.
Antioxidants combat excessive free radicals within the body. This concept applies to the
reduction of free radicals in the pancreas, which partially protects the pancreas from damage
and helps with diabetic control. Keeping this concept in mind, we also determined the
antioxidant activity of the phytosteroids. Though both of our phytosteroids demonstrated
low antioxidant activity profiles, they were both notable as supplementary targets. The
observed IC
50
values for
NtSt01
and
NtSt02
were 142.76 and 223.43
µ
g/mL, respectively,
as shown in Table 3.
Plants 2023,12, 3591 5 of 14
Table 3. Antioxidant potential of the phytosteroids.
Comp/Standard Conc (µg/mL) Percent Inhibition
(Mean ±SEM) IC50 (µg/mL)
NtSt01
500
250
125
62.50
31.25
61.05 ±2.05 ***
55.79 ±0.92 ***
47.08 ±2.41 ***
42.53 ±0.69 ***
37.25 ±2.66 ***
142.76
NtSt02
500
250
125
62.50
31.25
59.12 ±1.96 ***
52.01 ±1.07 ***
42.99 ±2.70 ***
33.08 ±1.17 ***
28.52 ±2.63 ***
223.43
Standard Drug
500
250
125
62.50
31.25
91.52 ±2.52
85.04 ±0.63
81.54 ±1.28
77.87 ±1.49
75.37 ±1.69
0.74
All the values are expressed as mean
±
SEM compared with the standard. Two-way ANOVA followed by
Dunnett’s test was applied. *** = p< 0.001 compared with the standard drug.
2.5. In Vivo Results
Based on the enzymatic assays, we observed that
NtSt01
is a potential inhibitor of
α
-glucosidase and
α
-amylase. With these results, we further analyzed the compound
NtSt01
in
in vivo
studies using experimental rats following ethical guidelines. The dose
of
NtSt01
was started at 200 and was increased up to 2000 mg/kg body weight. The rats
remained healthy; no morbidity, mortality or irritation was observed for the tested doses
during the acute toxicity studies.
Phytosteroid
NtSt01
(1.0 mg/kg) was administered to alloxan-induced fasting diabetic
rats, and the blood glucose levels were observed until 28 days, as shown in Table 4.
The blood glucose level in the normal control group remained within the normal range
during the observational time. In contrast, the blood glucose level of the diabetic control
group remained elevated throughout the experiment. At 1.0 mg/kg of
NtSt01
, the effect
on the blood glucose levels in rats were 519
±
3.98, 413
±
1.87, 325
±
1.62, 219
±
2.87
and
116 ±1.33 mg/dL
on days 1, 7, 14, 21, and 28, respectively. In comparison, in the
glibenclamide control group (0.5 mg/kg), the blood glucose level dropped from 517
±
0.77
(day 1) to 103 ±1.70 mg/dL (day 28).
Table 4.
Observational changes in blood glucose level in alloxan-induced fasting diabetic rats in
mg/dL.
Groups Day 1 Day 7 Day 14 Day 21 Day 28
Normal control 102 ±2.36 106 ±0.98 105 ±1.77 110 ±0.91 109 ±1.07
Diabetic control 521 ±1.28 517 ±2.06 526 ±1.00 524 ±2.66 529 ±2.80
Glibenclamide (0.5 mg/kg) 517 ±0.77 398 ±2.08 302 ±1.38 207 ±1.91 103 ±1.70
NtSt01 (1.0 mg/kg) 519 ±3.98 413 ±1.87 325 ±1.62 219 ±2.87 116 ±1.33
During the observational time period, the effect on the body weight (in grams) of
the rats was also closely observed (as shown in Table 5). In the normal control group, the
body weight of the rats remained the same throughout the four weeks. In the diabetic
control group, we observed a body weight loss of 18 g of rats at four weeks. With the
administration of phytosteroid
NtSt01
at a concentration of 1.0 mg/kg, the rats became
healthy, and there was a gain in body weight. The body weight changed from 238
±
0.33 g
Plants 2023,12, 3591 6 of 14
(day 1) to 246
±
0.11 g (day 28) in our sample. In comparison, the weight of the rats in the
glibenclamide control group also increased slightly.
Table 5. The effect of NtSt01 on body weight in grams of fasting rats.
Groups Day 1 Day 7 Day 14 Day 21 Day 28
Normal control 226 ±0.98 227 ±1.27 228 ±0.63 229 ±1.67 233 ±0.48
Diabetic control 232 ±2.69 229 ±1.98 225 ±1.39 217 ±2.69 214 ±3.09
Glibenclamide (0.5 mg/kg) 235 ±1.36 236 ±1.37 239 ±0.67 244 ±2.38 249 ±3.18
NtSt01 (1.0 mg/kg) 238 ±0.33 240 ±0.49 242 ±1.04 245 ±1.11 246 ±0.11
Diabetes mellitus is a perilous disease, and it affect all the vital organs of the body.
After the
in vivo
experiments, the rats from the different groups were euthanized following
the ethical procedure. The vital organs were isolated, and the weights were recorded, as
shown in Table 6. The weights of the liver, kidney, pancreas, and heart were
9.83 ±0.92
,
1.01
±
0.15, 0.92
±
0.12, and 1.04
±
0.13 g, respectively, in the
NtSt01
-administered group.
In diabetic control group, we observed a drastic effect on the average weights of the
vital organs.
Table 6. The effect of NtSt01 on vital organ weights in grams of fasting rats.
Groups Liver Kidney Pancreas Heart
Normal control 9.16 ±0.45 0.97 ±0.13 0.88 ±0.02 1.02 ±0.11
Diabetic control 6.38 ±0.10 1.26 ±0.37 0.71 ±0.13 0.83 ±0.08
Glibenclamide (0.5 mg/kg) 9.21 ±0.66 0.99 ±0.16 0.84 ±0.21 0.90 ±0.02
NtSt01 (1.0 mg/kg) 9.83 ±0.92 1.01 ±0.15 0.92 ±0.12 1.04 ±0.13
The results of the renal function and liver function tests are summarized in Table 7.
The serum creatinine, blood urea, serum bilirubin, ALT, and serum alkaline phosphatase of
the normal, diabetic, and standard (glibenclamide) groups were compared with the results
of phytosteroid
NtSt01
. In the normal group, all the parameters were within the normal
range. In the diabetic group, elevated blood urea, ALT, and serum alkaline phosphatase
levels were observed. In a pattern like that of the standard group, the phytosteroid
NtSt01
group exhibited normal serum creatinine, blood urea, and serum bilirubin values. However,
the ALT and serum alkaline phosphatase values were near the upper normal limit. These
overall results showed that compound
NtSt01
was effective in protecting the vital organs
from damage over the four weeks of the experiment.
Table 7. Biochemical profile of blood in alloxan-induced diabetic rats.
Test N.Control D.Control Standard NtSt01
(1.0 mg/kg) Unit Reference
Range
S.Creatinine 0.42 ±0.02 0.96 ±0.10 0.43 ±0.01 0.6 ±0.02 mg/dL 0.4–0.8
Blood Urea 19.1 ±0.17 187 ±3.65 20.2 ±0.33 19.1 ±2.36 mg/dL 15–22
S.bilurubin 0.71 ±0.24 0.96 ±0.01 0.81 ±0.03 0.72 ±0.77 mg/dL Up to 1.0
SGPT(ALT) 29.4 ±1.07 268 ±1.37 37 ±1.11 32 ±0.25 U/L 17–30
S.ALK.Phosphatase 114.7 ±0.39 159 ±1.07 125 ±1.22 134 ±2.33 U/L 30–130
The lipid profile is a major concern in the evaluation of serum triglycerides, total
cholesterol, high-density cholesterol (HDL), and low-density cholesterol (LDL). The results
Plants 2023,12, 3591 7 of 14
of lipid profile of all the experimental groups are summarized in Table 8. Except for the
serum cholesterol, all other values were within the normal ranges.
Table 8. Antilipidemic effects of NtSt01 on alloxan-induced diabetic rats.
Groups S.Cholesterol S.Triglycerides HDL LDL
Normal control 52.06 ±1.04 93.54 ±1.37 43.50 ±2.35 23.88 ±0.82
Diabetic control 280.63 ±2.33 456 ±3.21 40.76 ±1.49 165.5 ±3.65
Glibenclamide (0.5 mg/kg) 59.98 ±0.99 112 ±0.99 32.55 ±2.35 34.16 ±2.66
NtSt01 (1.0 mg/kg) 77.45 ±1.72 125 ±3.65 38.10 ±1.64 48.85 ±1.08
References Range 10–54 26–145 Up to 50 10–54
2.6. Molecular Docking Studies
For the determination of the pharmacological parameters of the ligand with targeted
protein moieties, using the fit model theory with ligand–enzyme interactions, docking
studies were performed. These docking results were examined to understand the different
interaction parameters.
NtSt01
was docked with the targeted macromolecules to analyze
the binding affinity. It was found to have excellent binding affinity with an energy of
8.84 Kcal/mol against
α
-glucosidase 7K9Q and -8.9 Kcal/mol against
α
-amylase 4W93.
The results of the interaction with
α
-glucosidase are depicted in Figure 2. The prominent
interactions are Arg 500 and Asn 551, respectively.
Plants2023,12,xFORPEERREVIEW8of16
Tab l e8.AntilipidemiceectsofNtSt01onalloxan-induceddiabeticrats.
GroupsS.CholesterolS.TriglyceridesHDLLDL
Normalcontrol52.06±1.0493.54±1.3743.50±2.3523.88±0.82
Diabeticcontrol280.63±2.33456±3.2140.76±1.49165.5±3.65
Glibenclamide(0.5mg/kg)59.98±0.99112±0.9932.55±2.3534.16±2.66
NtSt01(1.0mg/kg)77.45±1.72125±3.6538.10±1.6448.85±1.08
ReferencesRange10–5426–145Upto5010–54
2.6.MolecularDockingStudies
Forthedeterminationofthepharmacologicalparametersoftheligandwithtargeted
proteinmoieties,usingthetmodeltheorywithligand–enzymeinteractions,docking
studieswereperformed.Thesedockingresultswereexaminedtounderstandthedierent
interactionparameters.NtSt01wasdockedwiththetargetedmacromoleculestoanalyze
thebindinganity.Itwasfoundtohaveexcellentbindinganitywithanenergyof−8.84
Kcal/molagainstα-glucosidase7K9Qand-8.9Kcal/molagainstα-amylase4W93.There-
sultsoftheinteractionwithα-glucosidasearedepictedinFigure2.Theprominentinter-
actionsareArg500andAsn551,respectively.
Figure2. The(a)3Dand(b)2DvisualizationsofNtSt01withα-glucosidase.
NtSt01alsodisplayedexcellentresults,withgreatbindinganitiesagainstα-amyl-
ase.TheresultsaredepictedinFigure3.Theprominentinteractionswerefoundtobewith
Trp59,His101,Leu162,His201,Ala198,andIle235,respectively.
Figure 2. The (a) 3D and (b) 2D visualizations of NtSt01 with α-glucosidase.
NtSt01
also displayed excellent results, with great binding affinities against
α
-amylase.
The results are depicted in Figure 3. The prominent interactions were found to be with Trp
59, His 101, Leu 162, His 201, Ala 198, and Ile 235, respectively.
Plants 2023,12, 3591 8 of 14
Plants2023,12,xFORPEERREVIEW9of16
Figure3. The(a)3Dand(b)2DvisualizationofNtSt01withα-amylase.
3.MaterialsandMethods
3.1.Phytochemistry
WecollectedrhizomesofNotholirionthomsonianum,whichwerealsousedinourpre-
viousresearch[29–31].Inthisstudy,wesubjectedthehydroalcoholicextractofN.thom
sonianumtoisolation.Initially,weusedalarge-diametergravitycolumntopartiallypurify
thesample.Thesolventsystemusedinthischromatographyincludedn-hexaneandethyl
acetate.Initially,thecolumnwasstartedwithpuren-hexanefora100mLelution.Then,
thepolarityofsolventwasgraduallyincreasedbyadding5%ethylacetateeachtime,and
thefractionswerecollected.Thefractionswerepreliminarycheckedviathin-layerchro-
matography(TLC)analysis.Oneofthefractionscontainingtwomajorandafewminor
spotswasconcentratedandsubjectedtoasmallcolumnforfurtherpurication.Theelu-
tionfractionsweremonitoredcloselyandcollectedinseparatedvials.Thevialswithma-
jorcoelutedspotswerecombinedanddriedviaarotaryevaporator.Thetwomajorspots
weresubjectedtoGCMSanalysisfortheidenticationofthecomponentsasperthepre-
viouslydescribedprocedure[32].
3.2.AlphaGlucosidaseInhibition
Thealpha-glucosidaseinhibitoryeectsofourcompoundsweredeterminedviaa
reportedprotocol[31].Sampledilutions(50µL)and100µLofα-glucosidasesolution(0.5
U/mL)weremixed.Then,600µLofphosphatebuer(0.1M,pH6.9)wasadded,andthe
mixturewasincubatedat37°Cfor15min.Afterthis,5mMsubstrate(p-nitrophenylα-D-
glucopyranoside)solution(20µL)in0.1Mphosphatebuer(pH6.9)wasaddedandagain
incubatedunderthesameconditions.Sodiumcarbonatewasaddedtostopthereaction;
at405nm,theabsorbancewasnotedusingaspectrophotometer.Amixturewithoutα-
glucosidaseservedastheblank,andamixturewithoutthetestcompoundservedasthe
control.Thepercentenzymeinhibitionwascalculatedas:
%Alphaglucosidaseinhibition  .  .
 . 100(1)
3.3.AlphaAmylaseInhibition
Figure 3. The (a) 3D and (b) 2D visualization of NtSt01 with α-amylase.
3. Materials and Methods
3.1. Phytochemistry
We collected rhizomes of Notholirion thomsonianum, which were also used in our
previous research [
29
31
]. In this study, we subjected the hydroalcoholic extract of N.
thomsonianum to isolation. Initially, we used a large-diameter gravity column to partially
purify the sample. The solvent system used in this chromatography included n-hexane and
ethyl acetate. Initially, the column was started with pure n-hexane for a 100 mL elution.
Then, the polarity of solvent was gradually increased by adding 5% ethyl acetate each time,
and the fractions were collected. The fractions were preliminary checked via thin-layer
chromatography (TLC) analysis. One of the fractions containing two major and a few
minor spots was concentrated and subjected to a small column for further purification. The
elution fractions were monitored closely and collected in separated vials. The vials with
major coeluted spots were combined and dried via a rotary evaporator. The two major
spots were subjected to GCMS analysis for the identification of the components as per the
previously described procedure [32].
3.2. Alpha-Glucosidase Inhibition
The alpha-glucosidase inhibitory effects of our compounds were determined via a
reported protocol [
31
]. Sample dilutions (50
µ
L) and 100
µ
L of
α
-glucosidase solution
(
0.5 U/mL
) were mixed. Then, 600
µ
L of phosphate buffer (0.1 M, pH 6.9) was added, and
the mixture was incubated at 37
C for 15 min. After this, 5 mM substrate (p-nitrophenyl
α
-D-glucopyranoside) solution (20
µ
L) in 0.1 M phosphate buffer (pH 6.9) was added
and again incubated under the same conditions. Sodium carbonate was added to stop
the reaction; at 405 nm, the absorbance was noted using a spectrophotometer. A mixture
without
α
-glucosidase served as the blank, and a mixture without the test compound
served as the control. The percent enzyme inhibition was calculated as:
% Alpha glucosidase inhibition =
Control abs.Sample abs.
Control abs.×100 (1)
3.3. Alpha-Amylase Inhibition
The inhibitory activity of our compounds against alpha-amylase enzymes was deter-
mined as per the previously reported protocols [
31
]. Test samples of 100
µ
L were mixed
with an enzyme solution (200
µ
L) and 100
µ
L of phosphate buffer (pH 6.9, 2 mM). The
mixture was then incubated at 25
C for 20 min, followed by the addition of 100
µ
L of
Plants 2023,12, 3591 9 of 14
starch solution (1%). The same procedure was followed for positive controls, where phos-
phate buffer was added instead of enzyme solution (200
µ
L). After 5 min of incubation,
3,5-dinitrosalicylic acid reagent (500
µ
L) was added to the test samples and control group.
The mixtures were incubated again for 10 min; at 580 nm, the absorbance was recorded
using a spectrophotometer. The alpha-amylase percent inhibition was calculated as:
% Amylase inhibition =1Test sample abs.
Control abs.×100 (2)
3.4. Antioxidant Assay
To evaluate the antioxidant potential of our compounds, the DPPH free radical scav-
enging assay developed by Brand William was used [
33
]. The DPPH solution was made
by dissolving 20 mg of DPPH in 100 mL of methanol, and its absorbance at 517 nm was
adjusted to 0.75. Then, 2 mL of DPPH solution was added to 2 mL of sample dilutions
ranging from 31.25 to 500 µg/mL, and the mixture was incubated at room temperature in
the dark for 15 min. The absorbance was noted at 517 nm, and the following formula was
used to determine the % inhibition of DPPH free radicals.
% Inhibition =
Control abs.Sample abs.
Control abs.×100 (3)
3.5. Molecular Docking Studies
The docking studies were performed using Auto Dock Vina 1.2.2. PyRx. The three-
dimensional structure of ligand
NtSt01
was drawn in ChemDraw 20.0 software and saved
as a Mol.file. The structure was modified by adding polar hydrogen using Discovery
Studio Visualizer and saved in PDB format. The three-dimensional structures of both
targeted proteins,
α
-glucosidase and
α
-amylase, were acquired from the RCSB protein data
bank (http://www.rcsb.org accessed on 22 September 2023) as PDB id 4W93 and 7K9Q,
respectively, and saved in PDB format. Before starting the computational studies on the
ligand, the docking process protocols were validated through a redocking process. These
ligands and targeted protein structures were permitted for energy minimization through
the Charm force field factor, which detached the unwanted crystallographic observations.
The ligand and targeted protein structures were opened in Autodock Vina and converted
to ligands and macromolecules as Pdbqt molecules. The grid box was adjusted as center X:
10.112, Y: 68.356, and Z: 32.853, with dimensions (Angstrom) X: 75.2435, Y: 105.3254, and Z:
103.4758. The results were visualized through Discovery Studio Visualizer software 2017
R2 [22].
3.6. In Vivo Experiments
3.6.1. Experimental Animals and Ethical Approval
The experimental rats were purchased from the Breeding House of National Institute
Health, Islamabad, Pakistan. The average weights of these rats ranged from 220 to 250 g.
The animals were handled as per the guidelines of the University of Malakand animals
By-Laws 2008 (Scientific Procedure Issue I) under the approval of the ethical committee via
letter No. DREC-140/B. The food, water, and light/dark cycles provided to animals were
observed by the ethical committee [
34
]. The animals were euthanized as per the AVMA
guidelines version 2020. The animals were subjected to slow exposure of halothane vapors
to induce anesthesia. There was a gradual increase in the dosage of halothane vapors,
which eventually euthanized the animals [35].
3.6.2. Acute Toxicity
In experimental animals, acute toxicity studies were performed to determine the safe
dose of our test compounds for
in vivo
studies. Test compounds were administered at
increasing doses of up to 2000 mg/kg, and rats were observed for any aberrant behavior or
lethality [34].
Plants 2023,12, 3591 10 of 14
3.6.3. Induction of Diabetes
Alloxan monohydrate (Sigma Aldrich; Steinhein, Switzerland) was used to induce
diabetes at a dose of 160 mg/kg in the experimental animals [
36
]. The experimental animals
were kept in fasting mode for 8–12 h but allowed water only before being subjected to the
bioassay. The level of blood glucose was checked after 48 h of administration of alloxan
using a glucometer, and only the animals that were diabetic were considered for the study,
having a blood glucose level of more than 200 mg/dL [34].
3.6.4. Experimental Design
The antidiabetic activity of our test compounds was studied in alloxan-induced dia-
betic animals. All the experimental animals were placed into four different groups, with
6 animals in each group.
Group 1:
Only normal saline i.p was given to this nondiabetic group throughout the
experimental period.
Group 2:
Alloxan was administered i.p to this group for diabetes induction and rats
were under observation without any treatment throughout the experiment.
Group 3:
This group was the alloxan-induced diabetic group, which was treated with
0.5 mg/kg of the standard drug, glibenclamide.
Group 4:
This group was the alloxan-induced diabetic group, which was treated with
1.0 mg/kg of NtSt01 intraperitoneally.
3.6.5. Lipid Profile
Standard methods were used for the analysis of total cholesterol, serum triglycerides,
low-density lipoprotein (LDL), and high-density lipoprotein (HDL). Briefly, 10 mL of serum
sample was added to 1000 mL solution of triglyceride. The mixture was incubated for
10 min
at 37
C and the absorbance was recorded at 546 nm. To monitor blood cholesterol,
diagnostic kits were utilized following the specifications of the manufacturer. A total
of 10
µ
L of serum sample was added to 1000
µ
L of cholesterol solution, followed by
incubation for 10 min at 37
C; the absorbance against blank was noted at 546 nm. For
HDL concentration determination, 200
µ
L of serum sample and 500
µ
L of HDL solution
were combined, and the mixture was allowed to stand at room temperature for 5 min.
Again, the mixture was mixed and subjected to further centrifugation for 5 min, followed
by supernatant collection. Then, 50
µ
L of supernatant of HDL was added to 500
µ
L of
solution of cholesterol; the mixture was incubated at 37
C for 5 min. Then, at 546 nm, the
sample absorbance was measured. Similarly, LDL was determined in all groups following
the manufacturer’s specifications [37].
LDL = Total cholesterol + HDL Triglycerides/5 (4)
3.6.6. Renal Functions Tests
Renal functions tests were performed to determine blood urea and serum creati-
nine [
38
]. To determine blood urea, enzyme reagent 1 (1000
µ
L) and 10
µ
L of serum were
mixed together and incubated for 5 min at 25
C, followed by the addition of 1000
µ
L of
reagent 2 to the mixture. After 5 min, the absorbance was recorded at 578 nm. The serum
creatinine was measured very carefully, as it is highly sensitive reaction to temperature.
Then, 500
µ
L of reagent and 50
µ
L of serum were mixed, followed by incubation for 1 min
at 37 C, and the absorbance was noted at 500 nm.
3.6.7. Liver Functions Tests
Serum glutamate pyruvate transaminase (SGPT/ALT), alkaline phosphatase (ALP),
and bilirubin tests were performed following standard protocols using micro lab 300 and
tecno plus biochemistry analyzers [
39
]. To perform the SGPT/ALT test, 50
µ
L of serum
sample was added to 500 µL of reagent (400 µL of reagent 1 and 100 µL of reagent 2). The
mixture was incubated at 37
C for 30 s, and the absorbance was noted at 340 nm. To
Plants 2023,12, 3591 11 of 14
determine ALP, a kit was used following the manufacturer’s specifications: 10
µ
L of serum
sample was added to 500 µL of reagent (400 µL of reagent 1 and 100 µL of reagent 2). The
mixture was then incubated at 37
C for 30 s, and the absorbance was noted at 405 nm.
Similarly, the concentration of bilirubin was evaluated using standard protocols. Four
types of reagents, R1, R2, R3, and R4, were used in this test. R2 (25
µ
L) was added to R1
(
100 µL
). After this,
100 µL
of serum sample and 500
µ
L of R3 were added and allowed to
stand at
25 C
for 5 min. Then, 500
µ
L of R4 was added; at 25
C for 5 min, the mixture
was incubated. The absorbance of the sample was recorded at 546 nm.
3.6.8. Statistical Analysis
Two-way ANOVA followed by Dunnett’s post-test were applied for the comparison of
the positive control with the test groups using GraphPad prism 8.0.1 software. pvalues less
than or equal to 0.05 were considered statistically significant. The findings of the statistical
analysis are shown as mean ±SEM.
4. Discussion
For thousands of years, number of medicinal plants and natural products have been re-
ported for the treatment of many diseases including diabetes [
40
42
]. Metformin, obtained
from Galega officinalis, has been the first-line drug used for 60 years for the treatment of type
2 diabetes [
43
]. Plant-based drugs have minimal or no side effects compared with synthetic
drugs; therefore, researchers have mainly focused on natural products for developing new
drugs. In DM, the main treatment strategy involves controlling high glucose levels in the
blood. Apart from this,
α
-amylase and
α
-glucosidase enzymes inhibitions are important
strategies for controlling hyperglycemia, as these enzymes convert starch into glucose,
resulting in increased levels of blood glucose [
44
]. For Notholirion thomsonianum, the
in vitro
antidiabetic properties of the
in vitro
targets of
α
-amylase,
α
-glucosidase, and tyrosine
phosphatase 1B, and the antioxidant potential have been studied using free radical assays
of ABTS, DPPH, and H
2
O
2
[
1
]. In this study, we determined the antidiabetic potential of
Notholirion thomsonianum through
in vitro
(
α
-amylase and
α
-glucosidase inhibition) and
in vivo studies.
The
in vitro
antidiabetic properties of Notholirion thomsonianum were determined
against
α
-amylase and
α
-glucosidase.
α
-Amylase converts starch into disaccharides and
oligosaccharides, whereas
α
-glucosidase hydrolyzes disaccharides into glucose [
45
].
α
-
Amylase and
α
-glucosidase enzymes, if inhibited, decrease the glucose level via the break-
down of starch in GIT, which is retarded via the inhibition of these enzymes, which
ameliorates hyperglycemia in diabetic patients. In the
α
-glucosidase inhibition assay, the
compounds
NtSt01
and
NtSt02
exhibited inhibitions of 85.00
±
1.52 and 77.56
±
3.22%
at the highest concentration (500
µ
g/mL), with IC
50
values of 7.34 and 22.87
µ
g/mL,
respectively. Compounds
NtSt01
and
NtSt02
exhibited 80.03
±
2.11 and 74.99
±
1.53%
inhibition at 500
µ
g/mL against
α
-amylase with IC
50
values of 4.17 and 46.73
µ
g/mL,
respectively. However, at the same tested concentration (500
µ
g/mL), the standard drug
acarbose demonstrated 92.65
±
0.55% inhibition against
α
-glucosidase and 91.01
±
1.36%
inhibition against α-amylase with IC50 values of 2.14 and 1.96 µg/mL, respectively.
In the blood glucose test, diabetes induction with alloxan was confirmed, and the blood
glucose levels in the diabetic group were observed as 521
±
1.28, 517
±
2.06,
526 ±1.00
,
524
±
2.66, and 529
±
2.80 mg/dL on days 1, 7, 14, 21 and 28, respectively. Treatment of
diabetic animals with
NtSt01
at a dose of 1.0 mg/kg produced a decline in blood glucose
levels, i.e., 519
±
3.98, 413
±
1.87, 325
±
1.62, 219
±
2.87, and 116
±
1.33 mg/dL on the 1st,
7th, 21st, and 28th days of the treatment, respectively.
No statistical differences in the weights of the liver, kidney, pancreas, and heart
were observed among all groups after completion of the experiments. The kidney func-
tion profile, lipid profile, and biochemical profile of the test animals in all groups were
also determined. In the biochemical tests, blood urea, serum bilirubin, serum creatinine,
SGPT(ALT), and serum alkaline phosphatase levels were high in the diabetic disease group
Plants 2023,12, 3591 12 of 14
and decreased after treatment with 1.0 mg/kg
NtSt01
, as shown in Table 7. In the lipid tests,
considering serum cholesterol, serum triglycerides, LDL, and HDL, a significant decline
was demonstrated in our sample (
NtSt01
1.0 mg/kg) in comparison with the diabetic
animals, as shown in Table 8. Both the
in vitro
and
in vivo
antidiabetic results for
NtSt01
revealed the potential of this compound for the management of DM and as a multitarget
antidiabetic agent.
Molecular docking is an important approach to determine the binding energies and
interactions of a drug molecule for its target. We docked our isolated compounds against
α
-
glucosidase and
α
-amylase enzymes. The molecular docking studies revealed encouraging
binding interactions with the target proteins for our compounds.
Though the whole of our work is based on the development of an antidiabetic drug
from medicinal plant source, significant efforts have also been made by synthetic organic
chemists for the discovery of new antidiabetic agents [
46
]. Among the synthetic approaches,
the modification of immunosugar has been identified to develop potential inhibitors of
alpha-glucosidase [4749].
5. Conclusions
Herein, we explored the antidiabetic potential of phytosteroids from Notholirion thom-
sonianum. In an attempt to combat diabetes and its consequences, weave used phytosteroid
NtSt01
as a natural drug. In the
in vitro α
-glucosidase,
α
-amylase, and DPPH assays,
compound
NtSt01
was found to be potent enough to be tested in experimental animals.
The compound was initially found safe in experimental rats and was then found effective in
combating induced diabetes over the four weeks of the experiment. After the experiment,
we also observed that the weights of the liver, kidney, pancreases, and heart and their
functions were within the allowed limits. Our overall results show that phytosteroids
(specifically
NtSt01
) have an efficient therapeutic effect on the blood glucose level and
other protective effects on organs like the liver, kidney, pancreases, and heart, so may serve
as a multitarget antidiabetic drug.
Supplementary Materials:
The following supporting information can be downloaded at: https:
//www.mdpi.com/article/10.3390/plants12203591/s1, Table S1: GCMS results of compound
NtSt01
.
Figure S1: MS spectrum and fragmentation pattern of
NtSt01
. Figure S2: Library spectrum and
fragmentation pattern of
NtSt01
. Figure S3: Difference spectrum of
NtSt01
. Figure S4:
1
H NMR
spectrum of
NtSt01
. Table S2: GCMS results of compound
NtSt02
. Figure S5: MS spectrum and
fragmentation pattern of
NtSt02
. Figure S6: Library spectrum and fragmentation pattern of
NtSt02
.
Figure S7: Difference spectrum of NtSt02. Figure S8: 1H NMR spectrum of NtSt02.
Author Contributions:
M.A.H., S.F., A.A., S.M.A., A.N. and F.H.: conceptualization, methodology,
software, validation, formal analysis, investigation, resources, data curation, writing—original draft
preparation, and writing—review and editing; M.H.M. and A.S.: Conceptualization, methodology,
validation, investigation, resources, writing—original draft preparation, supervision, writing—review
and editing, visualization, and funding acquisition. All authors have read and agreed to the published
version of the manuscript.
Funding:
This research obtained funding under the National Research Priorities and Najran Area
Research funding program grant code number NU/NRP/MRC/12/40.
Institutional Review Board Statement:
The animals were handled as per the guidelines of the
University of Malakand animals By-Laws 2008 (Scientific Procedure Issue I) under the approval of
the ethical committee via letter No. DREC-140/B.
Data Availability Statement:
The data are presented in the manuscript and Supporting Information.
Acknowledgments:
The authors acknowledge support from the Deanship of Scientific Research, Najran
University, Kingdom of Saudi Arabia, for funding this work under the National Research Priorities and
Najran Area Research funding program grant code number NU/NRP/MRC/12/4). We are also grateful
to the Department of Pharmacy, University of Malakand, Pakistan, for the laboratory facilities.
Conflicts of Interest: The authors declare no conflict of interest.
Plants 2023,12, 3591 13 of 14
References
1.
Booth, G.; Lipscombe, L.; Butalia, S.; Dasgupta, K.; Eurich, D.; Goldenberg, R.; Khan, N.; MacCallum, L.; Shah, B.; Simpson,
S.; et al. Pharmacologic Management of Type 2 Diabetes: 2016 Interim Update. Can. J. Diabetes
2016
,40, 484–486. [CrossRef]
[PubMed]
2.
Huneif, M.A.; Alqahtani, S.M.; Abdulwahab, A.; Almedhesh, S.A.; Mahnashi, M.H.; Riaz, M.; Ur-Rahman, N.; Jan, M.S.; Ullah, F.;
Aasim, M.; et al.
α
-glucosidase,
α
-amylase and antioxidant evaluations of isolated bioactives from wild strawberry. Molecules
2022,27, 3444. [CrossRef] [PubMed]
3. Lin, Y.; Sun, Z. Current views on type 2 diabetes. J. Endocrinol. 2010,204, 1–11. [CrossRef]
4.
Ahmed, F.; Urooj, A. Antihyperglycemic activity of Ficus glomerata stem bark in streptozotocin-induced diabetic rats. Global J.
Pharmacol. 2008,2, 41–45.
5.
Cho, N.H.; Shaw, J.E.; Karuranga, S.; Huang, Y.; da Rocha Fernandes, J.D.; Ohlrogge, A.W.; Malanda, B. IDF Diabetes Atlas:
Global estimates of diabetes prevalence for 2017 and projections for 2045. Diabetes Res. Clin. Pract.
2018
,138, 271–281. [CrossRef]
[PubMed]
6.
Mathers, C.D.; Loncar, D. Projections of global mortality and burden of disease from 2002 to 2030. PLoS Med.
2006
,3, e442.
[CrossRef] [PubMed]
7.
Labazi, H.; Trask, A.J. Coronary microvascular disease as an early culprit in the pathophysiology of diabetes and metabolic
syndrome. Pharmacol. Res. 2017,123, 114–121. [CrossRef]
8.
Konig, M.; Lamos, E.M.; Stein, S.A.; Davis, S.N. An insight into the recent diabetes trials: What is the best approach to prevent
macrovascular and microvascular complications? Curr. Diabetes Rev. 2013,9, 371–381. [CrossRef]
9.
Chillarón, J.J.; Le-Roux, J.A.; Benaiges, D.; Pedro-Botet, J. Type 1 diabetes, metabolic syndrome and cardiovascular risk. Metabolism
2014,63, 181–187. [CrossRef]
10. DeFronzo, R.A. Pathogenesis of type 2 diabetes mellitus. Med. Clin. 2004,88, 787–835. [CrossRef]
11. Thomas, C.C.; Philipson, L.H. Update on Diabetes Classification. Med. Clin. N. Am. 2015,99, 1–16. [CrossRef] [PubMed]
12.
Franz, M.J.; Boucher, J.L.; Rutten-Ramos, S.; VanWormer, J.J. Lifestyle Weight-Loss Intervention Outcomes in Overweight and
Obese Adults with Type 2 Diabetes: A Systematic Review and Meta-Analysis of Randomized Clinical Trials. J. Acad. Nutr. Diet.
2015,115, 1447–1463. [CrossRef] [PubMed]
13.
Ghosh, D.; Parida, P. Drug Discovery and Development of Type 2 Diabetes Mellitus: Modern-Integrative Medicinal Approach.
Curr. Cancer Drug Targets 2016,13, 60–67. [CrossRef] [PubMed]
14.
Phung, O.J.; Scholle, J.M.; Talwar, M.; Coleman, C.I. Effect of noninsulin antidiabetic drugs added to metformin therapy on
glycemic control, weight gain, and hypoglycemia in type 2 diabetes. JAMA 2010,303, 1410–1418. [CrossRef]
15.
Van Staa, T.; Abenhaim, L.; Monette, J. Rates of hypoglycemia in users of sulfonylureas. J. Clin. Epidemiol.
1997
,50, 735–741.
[CrossRef]
16.
Gin, H.; Rigalleau, V. Post-prandial hyperglycemia. post-prandial hyperglycemia and diabetes. Diabetes Metab.
2000
,26, 265–272.
17.
He, L.; Wang, H.; Gu, C.; He, X.; Zhao, L.; Tong, X. Administration of traditional Chinese blood circulation activating drugs for
microvascular complications in patients with type 2 diabetes mellitus. J. Diabetes Res. 2016,2016, 1081657. [CrossRef]
18.
Pang, B.; Zhou, Q.; Li, J.L.; Zhao, L.H.; Tong, X.L. Treatment of refractory diabetic gastroparesis: Western medicine and traditional
Chinese medicine therapies. World J. Gastroenterol. 2014,20, 6504. [CrossRef]
19.
Xie, W.; Du, L. Diabetes is an inflammatory disease: Evidence from traditional Chinese medicines. Diabetes Obes. Metab.
2011
,
13, 289–301. [CrossRef]
20.
Shah, S.M.M.; Sadiq, A.; Ullah, F. Antioxidant, total phenolic contents and antinociceptive potential of Teucrium stocksianum
methanolic extract in different animal models. BMC Complement. Altern. Med. 2014,14, 181. [CrossRef]
21.
Mahnashi, M.H.; Alyami, B.A.; Alqahtani, Y.S.; Jan, M.S.; Rashid, U.; Sadiq, A.; Alqarni, A.O. Phytochemical profiling of bioactive
compounds, anti-inflammatory and analgesic potentials of Habenaria digitata Lindl.: Molecular docking based synergistic effect
of the identified compounds. J. Ethnopharmacol. 2021,273, 113976. [CrossRef] [PubMed]
22.
Huneif, M.A.; Alshehri, D.B.; Alshaibari, K.S.; Dammaj, M.Z.; Mahnashi, M.H.; Majid, S.U.; Javed, M.A.; Ahmad, S.; Rashid,
U.; Sadiq, A. Design, synthesis and bioevaluation of new vanillin hybrid as multitarget inhibitor of
α
-glucosidase,
α
-amylase,
PTP-1B and DPP4 for the treatment of type-II diabetes. Biomed. Pharmacother. 2022,150, 113038. [CrossRef] [PubMed]
23.
Bailey, C.J.; Day, C. Traditional plant medicines as treatments for diabetes. Diabetes Care
1989
,12, 553–564. [CrossRef] [PubMed]
24.
Prabhakar, P.K.; Doble, M. A target based therapeutic approach towards diabetes mellitus using medicinal plants. Curr. Diabetes
Rev. 2008,4, 291–308. [CrossRef] [PubMed]
25.
Choudhury, A.; Maeda, K.; Murayama, R.; DiMagno, E. Character of a wheat amylase inhibitor preparation and effects on fasting
human pancreaticobiliary secretions and hormones. Gastroenterology 1996,111, 1313–1320. [CrossRef]
26.
Sadiq, A.; Mahnashi, M.H.; Rashid, U.; Jan, M.S.; Alshahrani, M.A.; Huneif, M.A. 3-(((1 S, 3 S)-3-((R)-Hydroxy (4-(trifluoromethyl)
phenyl) methyl)-4-oxocyclohexyl) methyl) pentane-2, 4-dione: Design and Synthesis of New Stereopure Multi-Target Antidiabetic
Agent. Molecules 2022,27, 3265. [CrossRef]
27.
Mahnashi, M.H.; Alam, W.; Huneif, M.A.; Abdulwahab, A.; Alzahrani, M.J.; Alshaibari, K.S.; Rashid, U.; Sadiq, A.; Jan, M.S.
Exploration of Succinimide Derivative as a Multi-Target, Anti-Diabetic Agent: In Vitro and In Vivo Approaches. Molecules
2023
,
28, 1589. [CrossRef]
Plants 2023,12, 3591 14 of 14
28.
Ajaib, M.; Ali, S.; Khan, Z. Antioxidant and antimicrobial activities of an ethnobotanically important plant Notholirion thomsoni-
anum from district Kotli, Azad Jammu & Kashmir. J. Anim. Plant Sci. 2014,24, 774–780.
29.
Sadiq, A.; Ahmad, S.; Ali, R.; Ahmad, F.; Ahmad, S.; Zeb, A.; Ayaz, M.; Ullah, F.; Siddique, A.N. Antibacterial and antifungal
potentials of the solvents extracts from Eryngium caeruleum, Notholirion thomsonianum and Allium consanguineum. BMC
Complement. Altern. Med. 2016,16, 478. [CrossRef]
30.
Mahmood, F.; Ali, R.; Jan, M.S.; Chishti, K.A.; Ahmad, S.; Zeb, A.; Ayaz, M.; Ullah, F.; Aasim, M.; Khan, N.Z.; et al. Chemical
characterization and analgesic potential of Notholirion thomsonianum extract. Lat. Am. J. Pharm. 2019,38, 807–812.
31.
Mahnashi, M.H.; Alqahtani, Y.S.; Alqarni, A.O.; Alyami, B.A.; Jan, M.S.; Ayaz, M.; Ullah, F.; Rashid, U.; Sadiq, A. Crude extract
and isolated bioactive compounds from Notholirion thomsonianum (Royale) Stapf as multitargets antidiabetic agents: In-vitro
and molecular docking approaches. BMC Complement. Med. Ther. 2021,21, 270. [CrossRef] [PubMed]
32.
Sadiq, A.; Zeb, A.; Ullah, F.; Ahmad, S.; Ayaz, M.; Rashid, U.; Muhammad, N. Chemical Characterization, Analgesic, Antioxidant,
and Anticholinesterase potentials of essential oils from Isodon rugosus Wall. ex. Benth. Front. Pharmacol.
2018
,9, 623. [CrossRef]
[PubMed]
33.
Shah, S.M.M.; Ahmad, Z.; Yaseen, M.; Shah, R.; Khan, S.; Khan, B. Phytochemicals,
in vitro
antioxidant, total phenolic contents
and phytotoxic activity of Cornus macrophylla Wall bark collected from the North-West of Pakistan. Pak. J. Pharm. Sci.
2015
,28.
34.
Huneif, M.A.; Mahnashi, M.H.; Jan, M.S.; Shah, M.; Almedhesh, S.A.; Alqahtani, S.M.; Alzahrani, M.J.; Ayaz, M.; Ullah, F.; Rashid,
U.; et al. New Succinimide–Thiazolidinedione Hybrids as Multitarget Antidiabetic Agents: Design, Synthesis, Bioevaluation, and
Molecular Modelling Studies. Molecules 2023,28, 1207. [CrossRef] [PubMed]
35.
Underwood, W.; Anthony, R. AVMA Guidelines for the Euthanasia of Animals: 2020 Edition; American Veterinary Medical Association:
Schaumburg, IL, USA, 2020.
36.
Sadiq, A.; Rashid, U.; Ahmad, S.; Zahoor, M.; AlAjmi, M.F.; Ullah, R.; Noman, O.M.; Ullah, F.; Ayaz, M.; Khan, I.; et al. Treating
hyperglycemia from Eryngium caeruleum M. Bieb: In-vitro
α
-glucosidase, antioxidant, in-vivo antidiabetic and molecular
docking-based approaches. Front. Chem. 2020,8. [CrossRef]
37.
Ponnulakshmi, R.; Shyamaladevi, B.; Vijayalakshmi, P.; Selvaraj, J. In silico and
in vivo
analysis to identify the antidiabetic activity
of beta sitosterol in adipose tissue of high fat diet and sucrose induced type-2 diabetic experimental rats. Toxicol. Mech. Methods
2019,29, 276–290. [CrossRef]
38.
Hasan, M.; Mohieldein, A.
In vivo
evaluation of anti diabetic, hypolipidemic, antioxidative activities of Saudi date seed extract
on streptozotocin induced diabetic rats. J. Clin. Diagn. Res. 2016,10, FF06. [CrossRef]
39.
Galli, A.; Crabb, D.W.; Ceni, E.; Salzano, R.; Mello, T.; Svegliati–Baroni, G.; Ridolfi, F.; Trozzi, L.; Surrenti, C.; Casini, A.
Antidiabetic thiazolidinediones inhibit collagen synthesis and hepatic stellate cell activation
in vivo
and
in vitro
.Gastroenterology
2002,122, 1924–1940. [CrossRef]
40.
Aslam, H.; Khan, A.-U.; Naureen, H.; Ali, F.; Ullah, F.; Sadiq, A. Potential application of Conyza canadensis (L) Cronquist in the
management of diabetes: In vitro and in vivo evaluation. Trop. J. Pharm. Res. 2018,17, 1287–1293. [CrossRef]
41.
Farooq, U.; Naz, S.; Shams, A.; Raza, Y.; Ahmed, A.; Rashid, U.; Sadiq, A. Isolation of dihydrobenzofuran derivatives from
ethnomedicinal species Polygonum barbatum as anticancer compounds. Biol. Res. 2019,52, 1. [CrossRef]
42.
Mahnashi, M.H.; Alyami, B.A.; Alqahtani, Y.S.; Alqarni, A.O.; Jan, M.S.; Hussain, F.; Zafar, R.; Rashid, U.; Abbas, M.; Tariq, M.;
et al. Antioxidant molecules isolated from edible prostrate knotweed: Rational derivatization to produce more potent molecules.
Oxidative Med. Cell. Longev. 2022,2022, 3127480. [CrossRef] [PubMed]
43. Bailey, C.; Day, C. Metformin: Its botanical background. Pract. Diabetes Int. 2004,21, 115–117. [CrossRef]
44.
Buttermore, E.; Campanella, V.; Priefer, R. The increasing trend of Type 2 diabetes in youth: An overview. Diabetes Metab. Syndr.
Clin. Res. Rev. 2021,15, 102253. [CrossRef] [PubMed]
45.
Mahnashi, M.H.; Alqahtani, Y.S.; Alqarni, A.O.; Alyami, B.A.; Alqahtani, O.S.; Jan, M.S.; Hussain, F.; Islam, Z.U.; Ullah, F.; Ayaz,
M.; et al. Phytochemistry, anti-diabetic and antioxidant potentials of Allium consanguineum Kunth. BMC Complement. Med. Ther.
2022,22, 154. [CrossRef] [PubMed]
46.
Hussain, F.; Khan, Z.; Jan, M.S.; Ahmad, S.; Ahmad, A.; Rashid, U.; Ullah, F.; Ayaz, M.; Sadiq, A. Synthesis, in-vitro
α
-glucosidase
inhibition, antioxidant, in-vivo antidiabetic and molecular docking studies of pyrrolidine-2,5-dione and thiazolidine-2,4-dione
derivatives. Bioorganic Chem. 2019,91, 103128. [CrossRef] [PubMed]
47.
Tseng, P.S.; Ande, C.; Moremen, K.W.; Crich, D. Influence of side chain conformation on the activity of glycosidase inhibitors.
Angew. Chem. 2023,135, e202217809. [CrossRef]
48.
Compain, P.; Martin, O.R. (Eds.) Iminosugars: From Synthesis to Therapeutic Applications; John Wiley & Sons: New York, NY, USA, 2007.
49.
Rajasekaran, P.; Ande, C.; Vankar, Y.D. Synthesis of (5,6 & 6,6)-oxa-oxa annulated sugars as glycosidase inhibitors from 2-formyl
galactal using iodocyclization as a key step. Arkivoc 2022,2022, 5–23. [CrossRef]
Disclaimer/Publisher’s Note:
The statements, opinions and data contained in all publications are solely those of the individual
author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to
people or property resulting from any ideas, methods, instructions or products referred to in the content.
... For the DPPH assay on our samples, we used previously published procedure (Huneif et al., 2023). The solution of 2,2 Diphenyl 1 picrylhydrazyl was prepared by dissolving the DPPH (20 mg) in methanol (100 mL). ...
Article
Background Favism is a metabolic disease and this study evaluates the effectiveness of palm oil and its triacylglycerol constituent in favism-induced female rats to restore serum female hormones, ovarian antioxidants, inflammatory markers, and DNA fragmentation. Methods Animals were 36 female albino rats. They classified to two equal (normal and favism) groups. The normal group was divided into three equal subgroups: the control, palm oil, and triacylglycerol subgroups. The normal rats were given 1 mL of saline, 1 mL of palm oil, and 1 mL of triacylglycerol orally, respectively. The Favism group was classified also into three equal subgroups: the favism group, the favism + palm oil, the Favism + triacylglycerol. The favism rats were given 1 mL of saline, 1 mL of palm oil, and 1 mL of triacylglycerol orally. For four weeks, all treatments were administered orally via oral gavage once daily. Results The hemoglobin, hematocrite, the blood cells, glucose and glucose-6-phosphate dehydrogenase, and liver function were decreased in favism. Female hormones such as serum luteinizing hormone, follicle stimulating hormone, Estrone, Estriol, 17α-Estradiol, 17β-Estradiol, and Estradiol-17-β-stearate were decreased in favism. Ovarian antioxidants were decreased while ovarian inflammatory markers were increased in favism. Favism induced ovarian DNA apoptosis. Furthermore, oral administration with palm oil or its triacylglycerol constituent in favism-induced female rats restored all these parameters to be approached the control levels. Conclusions Palm oil restored serum female hormones, ovarian antioxidants, inflammatory markers, and DNA fragmentation in favism-induced female rats and this effect related to oil triacylglycerol constituent.
Article
Full-text available
Diabetes mellitus (DM) is counted among one of the leading challenges in the recent era, and it is a life-threatening disorder. Compound 4-hydroxy 3-methoxy phenylacetone (compound 1) was previously isolated from Polygonum aviculare. This compound was reacted with N-benzylmaleimide to synthesize the targeted compound 3. The purpose of this research is to exhibit our developed compound 3’s ability to concurrently inhibit many targets that are responsible for hyperglycemia. Compound 3 was capable of inhibiting α-amylase, α-glucosidase, and protein tyrosine phosphatase 1 B. Even so, outstanding in vitro inhibition was shown by the compound against dipeptidyl peptidase-4 (DPP-4) with an IC50 value of 0.07 µM. Additionally, by using DPPH in the antioxidant activity, it exhibited good antioxidant potential. Similarly, in the in vivo activity, the experimental mice proved to be safe by treatment with compound 3. After 21 days of examination, the compound 3 activity pattern was found to be effective in experimental mice. Compound 3 decreased the excess peak of total triglycerides, total cholesterol, AST, ALT, ALP, LDL, BUN, and creatinine in the STZ-induced diabetic mice. Likewise, the histopathology of the kidneys, liver, and pancreas of the treated animals was also evaluated. Overall, the succinimde moiety, such as compound 3, can affect several targets simultaneously, and, finally, we were successful in synthesizing a multi-targeted preclinical therapy.
Article
Full-text available
Diabetes mellitus (DM) is a metabolic disorder majorly arising from the pathophysiology of the pancreas manifested as a decline in the insulin production or the tissue’s resistance to the insulin. In this research, we have rationally designed and synthesized new succinimide–thiazolidinedione hybrids for the management of DM. In a multistep reaction, we were able to synthesize five new derivatives (10a–e). All the compounds were new containing a different substitution pattern on the N-atom of the succinimide ring. Initially, all the compounds were tested against the in vitro α-glucosidase, α-amylase, PTP1B, and DPP4 targets. In all of these targets, the compound 10d was observed to be the most potential antidiabetic agent. Based on this, the antidiabetic activity of the compound 10d was further investigated in experimental animals, which overall gave us encouraging results. The molecular docking studies of the compound 10d was also performed against the target enzymes α-glucosidase, α-amylase, PTP1B, and DPP4 using MOE. Overall, we observed that we have explored a new class of compounds as potential antidiabetic agents.
Article
Full-text available
Substrate side chain conformation impacts reactivity during glycosylation and glycoside hydrolysis and is restricted by many glycosidases and glycosyltransferases during catalysis. We show that the side chains of gluco and manno iminosugars can be restricted to predominant conformations by strategic installation of a methyl group. Glycosidase inhibition studies reveal that iminosugars with the gauche,gauche side chain conformations are 6‐ to 10‐fold more potent than isosteric compounds with the gauche,trans conformation; a manno‐configured iminosugar with the gauche,gauche conformation is a 27‐fold better inhibitor than 1‐deoxymannojirimycin. The results are discussed in terms of the energetic benefits of preorganization, particularly when in synergy with favorable hydrophobic interactions. The demonstration that inhibitor side chain preorganization can favorably impact glycosidase inhibition paves the way for improved inhibitor design through conformational preorganization.
Article
Full-text available
Aim The study was planned to investigate the phytochemicals, antidiabetic and antioxidant studies of A. consanguineum . Methods The preliminary studies were performed on crude extract and different solvent fractions. Based on the potency, the chloroform fraction was semi-purified to phyto-fractions CHF-1 – 5. Furthermore, CHF-3 was subjected to isolation of pure compounds using column chromatography. The α-glucosidase, α-amylase and antioxidant assays (DPPH, ABTS, H 2 O 2 ) were performed on all samples. The in-vivo experiments on compounds 1 and 2 were also performed using oral glucose tolerance test. Docking studies were performed on α-glucosidase and α-amylase targets. Results Among all fractions, the chloroform fraction exhibited excellent activities profile giving IC 50 values of 824, 55, 117, 58 and 85 μg/ml against α-glucosidase, α-amylase, DPPH, ABTS and H 2 O 2 targets respectively. Among the five semi-purified chloroform phyto-fractions (CHF-1-5), CHF-3 was the leading fraction in activities giving IC 50 values of 85.54, 61.19 and 26.58 μg/ml against α-glucosidase, α-amylase and DPPH respectively. Based on the overall potency and physical amount of CHF-3, it was subjected to purification to get compounds 1 and 2. The two compounds were also found potent in in-vitro activities. The observed IC 50 values for compound 1 were 7.93, 28.01 and 6.19 μg/ml against α-glucosidase, α-amylase and DPPH respectively. Similarly, the compound 2 exhibited IC 50 of 14.63, 24.82 and 7.654 μg/ml against α-glucosidase, α-amylase and DPPH respectively. Compounds 1 and 2 were potent in decreasing the blood glucose levels in experimental animals. Compounds 1 and 2 also showed interactions with the respective enzymes with molecular docking. Conclusions We can conclude that A. Consanguineum is a rich source of natural antidiabetic agents. Bioguided isolation of compound 1 and 2 showed potential inhibitions in all tested in-vitro antidiabetic targets. Further, both the compounds were also able to decrease the blood glucose levels in experimental animals.
Article
Full-text available
Diabetes mellitus is a metabolic disorder and is a global challenge to the current medicinal chemists and pharmacologists. This research has been designed to isolate and evaluate antidiabetic bioactives from Fragaria indica. The crude extracts, semi-purified and pure bioactives have been used in all in vitro assays. The in vitro α-glucosidase, α-amylase and DPPH free radical activities have been performed on all plant samples. The initial activities showed that ethyl acetate (Fi.EtAc) was the potent fraction in all the assays. This fraction was initially semi-purified to obtain Fi.EtAc 1–3. Among the semi-purified fractions, Fi.EtAc 2 was dominant, exhibiting potent IC50 values in all the in vitro assays. Based on the potency and availability of materials, Fi.EtAc 2 was subjected to further purification to obtain compounds 1 (2,4-dichloro-6-hydroxy-3,5-dimethoxytoluene) and 2 (2-methyl-6-(4-methylphenyl)-2-hepten-4-one). The two isolated compounds were characterized by mass and NMR analyses. The compounds 1 and 2 showed excellent inhibitions against α-glucosidase (21.45 for 1 and 15.03 for 2 μg/mL), α-amylase (17.65 and 16.56 μg/mL) and DPPH free radicals (7.62 and 14.30 μg/mL). Our study provides baseline research for the antidiabetic bioactives exploration from Fragaria indica. The bioactive compounds can be evaluated in animals-based antidiabetic activity in future.
Article
Full-text available
The chiral drug candidates have more effective binding affinities for their specific protein or receptor site for the onset of pharmacological action. Achieving all carbon stereopure compounds is not trivial in chemical synthesis. However, with the development of asymmetric organocatalysis, the synthesis of certain vital chiral drug candidates is now possible. In this research, we have synthesized 3-(((1S,3S)-3-((R)-hydroxy(4-(trifluoromethyl)phenyl)methyl)-4-oxocyclohexyl)methyl)pentane-2,4-dione (S,S,R-5) and have evaluated it potential as multi-target antidiabetic agent. The stereopure compound S,S,R-5 was synthesized with a 99:1 enantiomeric ratio. The synthesized compound gave encouraging results against all in vitro antidiabetic targets, exhibiting IC50 values of 6.28, 4.58, 0.91, and 2.36 in α-glucosidase, α-amylase, PTP1B, and DPPH targets, respectively. The molecular docking shows the binding of the compound in homology models of the respective enzymes. In conclusion, we have synthesized a new chiral molecule (S,S,R-5). The compound proved to be a potential inhibitor of the tested antidiabetic targets. With the observed results and molecular docking, it is evident that S,S,R-5 is a potential multitarget antidiabetic agent. Our study laid the baseline for the animal-based studies of this compound in antidiabetic confirmation.
Article
Full-text available
Prostrate knotweed also called Polygonum aviculare is an important edible plant. The polygonum is majorly known for the phenolics and antioxidants. The antioxidants combat the excessive free radicals within the body. The excessive free radicals are implicated in various other diseases like diabetes, Alzheimer’s, and inflammation. This study was aimed at exploring the antioxidant bioactives and their derivatizations to produce new molecules with advanced pharmacological features. We have isolated six compounds (1–6) from Polygonum aviculare. Furthermore, rational-based chemical derivatives for compound 5 have been formed for the management of diabetes, Alzheimer’s, and inflammation. In preliminary antioxidant studies, all the isolated compounds (1–6) showed potential results against DPPH and ABTS free radicals. Based on the IC50 and chemical nature of the compounds, compound 5 was subjected to derivatization. Keeping the phenolic part of compound 5 unaffected, hydroxy succinimide (5A) and thiazolidinedione (5B) were synthesized. The compound 5A was found to be a potent inhibitor of AChE, BChE, COX-1, COX-2, 5-LOX, and DPPH giving IC50 values of 10.60, 15.10, 13.91, 1.08, 0.71, and 1.05 μM, respectively. The COX-2 selectivity of compound 5A was found at 12.9. The compound 5B was found to be a potent multitarget antidiabetic agent giving IC50 values of 15.34, 21.83, 53.28, and 1.94 μM against α-glucosidase, α-amylase, protein tyrosine phosphatase 1B, and DPPH. Docking studies were performed to manipulate the binding interactions. The docking pose of all the tested compounds was found to have increased binding affinity against all tested targets that supported the in vitro results. Our results showed that Polygonum aviculare is a rich source of antioxidant compounds. The two new derivatives have enhanced pharmacological features to treat diabetes, inflammation, and Alzheimer’s disease.
Article
Substrate side chain conformation impacts reactivity during glycosylation and glycoside hydrolysis and is restricted by many glycosidases and glycosyltransferases during catalysis. We show that the side chains of gluco and manno iminosugars can be restricted to predominant conformations by strategic installation of a methyl group. Glycosidase inhibition studies reveal that iminosugars with the gauche,gauche side chain conformations are 6‐ to 10‐fold more potent than isosteric compounds with the gauche,trans conformation; a manno‐configured iminosugar with the gauche,gauche conformation is a 27‐fold better inhibitor than 1‐deoxymannojirimycin. The results are discussed in terms of the energetic benefits of preorganization, particularly when in synergy with favorable hydrophobic interactions. The demonstration that inhibitor side chain preorganization can favorably impact glycosidase inhibition paves the way for improved inhibitor design through conformational preorganization.
Article
Diabetes mellitus (DM) is a real challenge to the recent era and is one of the major diseases for initiating life-threatening disorders. In current research, a compound was designed by combining vanillin, thiazolidinedione and morpholine. The goal of our designed work is to demonstrate the ability of our design compound (9) to modulate more than one target responsible for hyperglycemia at the same time. The synthesized compound was able to show good to moderate inhibition potential against α-glucosidase, α-amylase and protein tyrosine phosphatase 1B. However, it exhibited excellent in-vitro inhibition of Dipeptidyl peptidase-4 (DPP-4) with IC50 value of 0.09 µM. Antioxidant activity by using DPPH assay also showed its good antioxidant potential. In in-vivo experiments, the compound 9 was proved to be safe in experimental mice. The activity profile of the compound was observed for 21 days which showed that the compound was also effective in experimental mice. Binding orientations and Interactions with key amino acid residues of the selected targets were also studied by using docking studies. Overall, we were successful in synthesizing multitarget preclinical therapeutic by combining three pharmacophoric moieties into a single chemical entity that can modulate more than one target at the same time.