ArticlePDF AvailableLiterature Review

Bioactive Constituents and Toxicological Evaluation of Selected Antidiabetic Medicinal Plants of Saudi Arabia

Authors:

Abstract

The purpose of this review is to summarize the available antidiabetic medicinal plants in the Kingdom of Saudi Arabia with its phytoconstituents and toxicological findings supporting by the latest literature. Required data about medicinal plants having antidiabetic activities and growing in the Kingdom of Saudi Arabia were searched/collected from the online databases including Wiley, Google, PubMed, Google Scholar, ScienceDirect, and Scopus. Keywords used in search are in vivo antidiabetic activities, flora of Saudi Arabia, active ingredients, toxicological evaluations, and medicinal plants. A total of 50 plant species belonging to 27 families were found in the flora of Saudi Arabia. Dominant family was found Lamiaceae with 5 species (highest) followed by Moraceae with 4 species. β-Amyrin, β-sitosterol, stigmasterol, oleanolic acid, ursolic acid, rutin, chlorogenic acid, quercetin, and kaempferol are the very common bioactive constituents of these selected plant species. This paper has presented a list of antidiabetic plants used in the treatment of diabetes mellitus. Bioactive antidiabetic phytoconstituents which showed that these plants have hypoglycemic effects and highly recommended for further pharmacological purposes and to isolate/identify antidiabetes mellitus (anti-DM) active agents also need to investigate the side effects of active ingredients.
Review Article
Bioactive Constituents and Toxicological Evaluation of Selected
Antidiabetic Medicinal Plants of Saudi Arabia
Ali S. Alqahtani ,
1
,
2
Riaz Ullah ,
1
,
2
and Abdelaaty A. Shahat
1
,
2
1
Department of Pharmacognosy, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
2
Medicinal Aromatic and Poisonous Plants Research Centre, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
Correspondence should be addressed to Riaz Ullah; rullah@ksu.edu.sa
Received 13 December 2021; Accepted 30 December 2021; Published 17 January 2022
Academic Editor: Rajadurai Murugan
Copyright ©2022 Ali S. Alqahtani et al. is is an open access article distributed under the Creative Commons Attribution
License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is
properly cited.
e purpose of this review is to summarize the available antidiabetic medicinal plants in the Kingdom of Saudi Arabia with its
phytoconstituents and toxicological findings supporting by the latest literature. Required data about medicinal plants having
antidiabetic activities and growing in the Kingdom of Saudi Arabia were searched/collected from the online databases including
Wiley, Google, PubMed, Google Scholar, ScienceDirect, and Scopus. Keywords used in search are in vivo antidiabetic activities,
flora of Saudi Arabia, active ingredients, toxicological evaluations, and medicinal plants. A total of 50 plant species belonging to 27
families were found in the flora of Saudi Arabia. Dominant family was found Lamiaceae with 5 species (highest) followed by
Moraceae with 4 species. β-Amyrin, β-sitosterol, stigmasterol, oleanolic acid, ursolic acid, rutin, chlorogenic acid, quercetin, and
kaempferol are the very common bioactive constituents of these selected plant species. is paper has presented a list of an-
tidiabetic plants used in the treatment of diabetes mellitus. Bioactive antidiabetic phytoconstituents which showed that these
plants have hypoglycemic effects and highly recommended for further pharmacological purposes and to isolate/identify anti-
diabetes mellitus (anti-DM) active agents also need to investigate the side effects of active ingredients.
1. Introduction
Medicinal plants are used for the treatment of different in-
fections [1, 2]. ese plants contributed as a source of inspi-
ration for novel therapeutic compounds [3]. e medicinal
value of plants is due to the presence of a wide variety of
secondary metabolites including alkaloids, glycosides, tannins,
volatile oil, and terpenoids [4, 5]. Medicinal plants and their
extracts represent a rich source of crude medications that
possess therapeutic properties. Indeed, the World Health Or-
ganization reports that various plant fractions and their dy-
namic constituents are utilized as traditional medicines by 80%
of the world population [6]. Plants are the primary source for
different pharmaceutical, perfumery, flavor, and cosmetics in-
dustries; the use of modern drugs dramatically resulted into
resistant microorganisms toward different modern drugs; the
researchers are now in search for alternate source of treatment
of various disorders [7, 8]. For this purpose, the medicinal herbs
are the best alternate to various drugs. Most of natural products
possess interesting biological activities and medicinal potential.
Various herbs, fruits, and grains have been found to have
different important biological activities such as antioxidant, [9]
antitumor, antimutagenic, antidiabetes, antianalgesic, [10]
antidementia, inflammation inhibitory effect, [9] antitumor,
[11] anticancer, [12] antimicrobial, antileishmanial, and anti-
malarial properties [13, 14]. e consumption of natural an-
tioxidants will reduce risk of many diseases including cancer,
cardiovascular disease, diabetes, and other diseases allied with
aging [15]. For natural antioxidants, a larger number of me-
dicinal herbs have been evaluated by applying laboratories’
developed procedures. Plants derived substances, collectively
called phytonutrients or phytochemicals, been recognized as
good source of natural antioxidants [16, 17].
e Kingdom of Saudi Arabia is a huge arid land with an
area of about 2,250,000 km
2
covering the major part of the
Arabian Peninsula, characterized by different ecosystems
Hindawi
Evidence-Based Complementary and Alternative Medicine
Volume 2022, Article ID 7123521, 23 pages
https://doi.org/10.1155/2022/7123521
and diversity of plant species. e climate in Saudi Arabia
differs greatly between the coast and the interior. High
humidity coupled with more moderate temperatures is
prevalent along the coast, whereas aridity and extreme
temperatures characterize the interior. e flora of Saudi
Arabia is one of the richest biodiversities in the Arabian
Peninsula and comprises very important genetic resources of
crops and medicinal plants. Saudi Arabia contains 97 trees,
564 shrubs, and about 1620 herbs, which cover, respectively,
4.25%, 24.73%, and 71.02% of higher plant diversity of the
country [18].
Diabetes mellitus is one of the most prevalent diseases in
endocrine gland system with an increasing incidence in
human community [19]. Type I diabetes is caused by insulin
secretion deficit, while type II diabetes is accompanied with
progressive rate of insulin resistance in liver and peripheral
tissues, reducing β-cell mass, and deficient insulin secretion
[20, 21]. is disease brings about acute metabolic side ef-
fects including ketoacidosis, hyperosmolar coma accom-
panied with chronic disorders, and long term, adverse side
effects such as retinopathy, renal failure, neuropathy, skin
complications, as well as increasing cardiovascular com-
plication risks [22, 23]. Also, common symptoms of diabetes
are frequent urine, thirsty, and overeating [24]. Diabetes
inflicts 100 million people yearly and is recognized as the
seventh cause of death in the world [25]. It has been esti-
mated that the number of diabetic people will increase from
150 million individuals in 2003 to 300 million by 2025 [26].
e essential and effective drugs for diabetes mellitus are
insulin injection and hypoglycemic agents, but these com-
pounds possess several adverse effects and have no effects on
diabetes complications in long term. erefore, it is im-
portant to find effective compounds with lower side effects in
treating diabetes [27]. Medicinal plants are good sources as
alternative or complementary treatments for this and other
diseases [28–30]. Although various plants have been tra-
ditionally used throughout history to reduce blood glucose
and improve diabetes complications, there is not enough
scientific information about some of them. Herbal medicines
are commonly prescribed throughout the world because of
low side effects, availability, roughly low cost, and also its
effectiveness [31, 32].
In Saudi Arabia, the number of people who suffer from
DM increased from 890,000 in 2000 to a staggering pro-
jection of 2,523,000 in 2030. In 2011, Saudi Arabia reported a
prevalence of DM at 30% of the total population, with a rate
of 27.6% in women and 34.1% in men [33]. According to
2010 data from several sources (WHO, World Bank,
UNESCO, CIA, and individual country databases), DM is
the number three disease-related cause of death in Saudi
Arabia [34].
In the present situation, herbal medicines’ usage has
significantly increased and published studies from developed
countries emphasize that a paramount proportion of
medicines supplied by them have herbal origins, so growing
and producing the herbal medicines could be helpful to both
economic development and community’s health [35].
Keeping in mind the importance of medicinal plants, in the
current review various medicinal plants used for antidiabetic
treatment around the world, native to or cultivated in Saudi
Arabia, are documented for the purpose to provide up-to-
date insight on medicinal plant used for DM, so that re-
searcher easily selects plant for bioscreening and active
constituents’ identification purposes. erefore, we invite
researchers’ attention to carry out detailed ethno-
pharmacological and toxicological studies on unexplored
antidiabetic plants in order to provide reliable knowledge to
the patients and develop novel antidiabetic drugs.
2. Methods
Required data about medicinal plants having antidiabetic
activities and growing in the Kingdom of Saudi Arabia were
searched/collected from the online databases including
Wiley, Google, PubMed, Google Scholar, ScienceDirect, and
Scopus. Keywords used in search are in vivo antidiabetic
activities, flora of Saudi Arabia, active ingredients, toxico-
logical evaluations, and medicinal plants. Latest published
data approximately in the last ten years with the key outcome
of change in blood glucose level in animal model were in-
cluded. One or two articles are selected as references for each
plant’s species on priority basis from the journals found in
web of science and latest years.
3. Results
e names, families, used parts, location, and antidiabetic
properties in animal model of the native/cultivated Saudi
medicinal plants are summarized in Table 1. e active
ingredients and toxicological effect of these plants in animal
model are given in Table 2. A total of 50 plant species belong
from 27 families were found in the flora of Saudi Arabia.
Dominant family was found Lamiaceae with 5 species
(highest) followed by Moraceae with 4 species.
4. Discussion
e majority of the experiments confirmed the benefits of
medicinal plants with hypoglycemic effects in the man-
agement of diabetes mellitus. From Table 1, it can be
concluded that among the plants used for the treatment of
diabetes, H. salicornicum, T. oliverianum, A. cepa, A. herba-
alba, Teucrium polium, Sesamum indicum, Z. spina-christi,
and U. dioica seem to be most common plants used to treat
diabetes and are available everywhere in the world. e
leaves were most commonly used plant part, and other parts
(root, stem, bark, flower, seed, and whole plant) were also
useful for curing. e most common diabetic model that was
used was the streptozotocin and alloxan-induced diabetic
mouse or rat as diabetic models. e most commonly in-
volved active constituents are flavonoid, alkaloid, saponin,
carbohydrate, vitamins, amino acid and its derivatives,
phenol and its derivatives, and benzoic acid derivatives. e
very common phytoconstituents, targeted metabolic path-
ways, and its structure are given in Table 3 [194, 195]. e
native to or cultivated plant species of the kingdom given in
Table 1 are selected from the published literature about
ethnobotanical value and antidiabetic potential of medicinal
2Evidence-Based Complementary and Alternative Medicine
Table 1: Antidiabetic medicinal plants growing in Saudi Arabia.
S.
no. Names of plants Family Part used location Antidiabetes Activities
1. Allium cepa Liliaceae Bulb Central Saudi Arabia
[36]
Ethanol extract of A. cepa in STZ-induced diabetic
rats causes 66% decreased at 200 mg/kg after 24 h
in blood glucose level [37].
0.4 g/100gbw of A. cepa reduced 50% the fasting
glucose levels of diabetic rats [38]. Similar results
reported by other researchers [39].
2. Anthemis herba-
alba
Compositae/
Asteraceae Aerial parts Farasan Island of Red
Sea [40]
72% plasma glucose levels decreased in albino
mice by ethyl alcohol extract of Artemisia herba-
alba [41]
3. Cichorium intybus Asteraceae Seeds Qassim region [42]
C. intybus leaf powder, ethanol, aqueous seed
extracts, and hexane extracts led to a decrease in
blood glucose levels to near normal value.
Hypoglycemic effects of C. intybus were observed
in diabetic rats, and a dose of 125 mg of plant
extract/kg body weight exhibited the most potent
hypoglycemic effect [43–45]
4. Clitoria ternatea Fabaceae Aerial parts Cultivated throughout
Saudi Arabia [46]
e aqueous extract of Clitoria ternatea leaves and
flower administered for 84 days to diabetic rats
significantly decreased blood glucose [46–48]
5. Ficus carica Moraceae Leaves Southwest of Saudi
Arabia [49]
Different extracts and fractions of F. carica showed
a clear hypoglycemic effect in diabetic rats.
F. carica leaves exerted significant effect on
carbohydrate metabolism enzymes with promising
hypoglycemic and hypolipidemic activities in type
2 diabetic rats [50, 51]
6. Ficus
benghalensis Moraceae Bark Riyadh [52]
In streptozotocin-induced diabetic rats, bark
aqueous extract, and an isolated compound,
α-amyrin acetate exhibited antidiabetic activity by
decreasing the blood glucose level and increasing
the HDL level [53]
7. Ficus religiosa Moraceae
Root bark,
stem bark,
aerial roots
Riyadh [52]
e aqueous extract of bark and ethanol extract of
leaves and fruits had a promising antidiabetic
effect in streptozotocin-induced diabetic rats by
decreasing the blood glucose, serum triglyceride,
and total cholesterol levels and increasing serum
insulin, body weight, and glycogen content in the
liver and skeletal muscle [53]
8. Ficus microcarpa Moraceae Leaves Riyadh [52]
F. microcarpa leaves showed protective effect
against alloxan-induced diabetic rats by reducing
blood glucose, cholesterol and triglyceride levels,
and increased insulin level [53]
9. Hypericum
perforatum Hypericaceae Leaves Western Saudi Arabia
[54]
H. perforatum ethyl acetate extract possesses
potent antihyperglycemic activity in STZ-induced
diabetic rats [55].
10 Anethum
graveolens Apiaceae Seeds Makka [56]
Different extracts and tablets of Anethum
graveolens possess potent antihyperglycemic
activity in alloxan-induced diabetic mice [57]
11 Cuminum
cyminum L.
Apiaceae or
Umbelliferae Seeds Makka [56]
Oral administration of cumin seeds crude ethanol
extract and glibenclamide to diabetic rats
significantly and progressively restored toward
normal. Cumin seeds crude ethanol extract and
glibenclamide reduced plasma glucose levels by
38.34 and 37.73%, respectively, compared with
diabetic control [58]. Other studies also reported
similar results [59].
12 Marrubium
vulgare Lamiaceae Whole plant Widely distributed in
Saudi Arabia [60]
M. vulgare extracts lower blood glucose level 30 to
60% in dose-dependent manner in streptozotocin-
induced diabetic rats [60].
Evidence-Based Complementary and Alternative Medicine 3
Table 1: Continued.
S.
no. Names of plants Family Part used location Antidiabetes Activities
13 Mentha longifolia Lamiaceae Whole plant Madinah [61]
Remarkable antidiabetic, anticholinesterase, and
antityrosinase effects were recorded for the mint
oil [61, 62]. Still need to investigate in vivo
antidiabetic potential.
14 Origanum
syriacum Lamiaceae Leaves Saudi Desert [63]
e whole plant extract of O. syriacum at 100 and
400 mg/kg significantly lowers glucose level in
diabetic induced rats [64].
15 Teucrium
oliverianum Lamiaceae Aerial parts roughout Saudi
Arabia [65,66]
Aqueous and ethanol extract of Teucrium
oliverianum were tested for antidiabetic activity in
alloxan-induced diabetic mice. Both extracts
significantly reduced blood sugar levels [65]
16 Teucrium polium Lamiaceae Leaves Madinah [67]
Infusion orally (64% decrease glucose level) and
intraperitoneal of different extracts of T. polium
caused significant reductions in blood glucose
concentration in STZ hyperglycemic rats [68]
17 Achyranthes
aspera Amaranthaceae Whole plant Al Hada Road Taif [69]
e methanolic and ethanolic extract of A. aspera
exhibited significant hypoglycemic activity in
streptozotocin-induced diabetic rats [70]
18 Aerva lanata Amaranthaceae Leaves Southwest region of
Saudi Arabia [71, 72]
Extracts of Aerva lanata and glibenclamide were
found to significantly (P<0.01 and P<0.05)
reduce the blood glucose level and lipid profile in
streptozotocin-induced diabetic rats [73]
19 Alternanthera
sessilis Amaranthaceae Whole plant Hail region, Saudi
Arabia [74]
In diabetic mice at doses of 50, 100, 200, and
400 mg per kg body weight, the extract reduced
blood sugar levels by 22.9, 30.7, 45.4, and 46.1%,
respectively, compared to control animals. By
comparison, a standard antihyperglycemic drug,
glibenclamide, when administered at a dose of
10 mg per kg body weight, reduced blood glucose
level by 48.9% [75]
20 Carissa edulis Apocynaceae Leaves Southern region of
Saudi Arabia [76]
Oral administration of C. edulis extracts of the
leaves significantly reduced the blood glucose level
in STZ diabetic rats [77].
21 Catharanthus
roseus Apocynaceae
Flower,
leaves, stem,
and root
Western Saudi Arabia
[78]
C. roseus (100 mg/kg BW) lowered the glucose
level more than metformin-treated group (100 mg/
kg BW) in STZ-induced hyperglycemia rats.
C. roseus 200 mg/kg dose was found to be more
effective in reducing fasting blood glucose levels
[79]
22 Rhazya stricta Apocynaceae Leaves, seeds
Middle and western
region of Saudi Arabia
[80]
Extracts Rhazya stricta lowered 37.9% blood
glucose level in the streptozotocin-induced
diabetic rats. Serum cholesterol and triglyceride
levels were significantly (P<0.05) reduced in the
treated diabetic group compared to the untreated
diabetic group [81]
23 Calotropis procera Asclepiadaceae Latex Al-Kharj [82]
Different extracts of C. procera at dose of 250 mg/
kg were orally administered as single dose per day
to diabetes-induced rats for the period of 15 days
significantly decreases blood glucose level to the
level of standard drug glibenclamide [83]
24 Opuntia dillenii Cactaceae Fruit Jazan Region [84]
Researcher observed the significant hypoglycemic
activity of Opuntia dillenii extract in
streptozotocin-induced diabetic mice and rabbits
[85]
4Evidence-Based Complementary and Alternative Medicine
Table 1: Continued.
S.
no. Names of plants Family Part used location Antidiabetes Activities
25 Opuntia ficus-
indica Cactaceae Stem Jazan Region [84]
Powder and water extract of O. ficus-indica
significantly (in comparison with control group)
returned blood glucose level to the initial level,
180 min after administration in STZ-induced
diabetic rats [86]. Many studies confirmed the
hypoglycemic activities of O. ficus-indica [87]
26 Capparis decidua Capparaceae Fruits, seeds Jazan Region [84]
C. decidua extracts at dose level of 200 and 800 mg/
kg significantly reduce sugar level (in a dose-
dependent manner) compared to standard drug in
STZ-induced diabetic and normal rats [88].
27 Beta vulgaris Chenopodiaceae Root bark
North Hejaz and
Eastern Najd region of
Saudi Arabia [89]
Extract of B. vulgaris at does level 50, 100, and
200 mg/kg of significantly reduced sugar level and
increased in insulin level (in a dose-dependent
manner) in streptozotocin or alloxan-induced
diabetic mice [90]. Other researchers also
concluded similar finding in STZ-induced diabetic
rats [91].
28
Haloxylon
salicornicum
Bunge
Chenopodiaceae Whole plant Wadi-Hafr-Al-Batin,
Saudi Arabia [92]
Ethanol extract (100 and 200 mg/kg of bw) of
H. salicornicum (oral administration) exhibited
persistent hypoglycemic effects in STZ-induced
diabetic rats [93]
29 Evolvulus
alsinoides Convolvulaceae Whole plant Jazan Region [84]
E. alsinoides ethanol extract at dose level (150 mg/
kg bw) in normal and streptozotocin-induced
diabetic rats leads to hyperglycemia in
experimental diabetic rats that decreased
utilization of glucose by insulin-dependent
pathways [94, 95]
30 Ipomea aquatica Convolvulaceae Whole plant Jazan Region [96]
I. aquatica ethanol extract at dose level (10, 100,
and 1000 µg/ml in streptozotocin-induced diabetic
rats significantly (P<.05) exhibited the ability to
enhance insulin-mediated glucose uptake into
3T3F442A adipocytes cells compared to insulin
alone [97]. Another study confirmed that doses
(200 mg/kg and 400 mg/kg) reduced blood glucose
level, and it was statistically highly significant
(P<0.001) in comparison with control group [98].
31 Citrullus
colocynthis Cucurbitaceae Fruits Jazan Region [84]
1 ml/kg and 2 ml/kg of C. colocynthis extract
(orally administered) stabilized animal body
weight and ameliorated hyperglycemia in a dose-
and time-dependent manner in alloxan-induced
diabetic rats [99]
32 Citrullus lanatus Cucurbitaceae Seed Wadi Lajab, Saudi
Arabia [100]
C. lanatus seed extract (2, 4 g/kg) treatment
significantly lowers glucose level which suggested
that C. lanatus had antidiabetic property in STZ-
induced diabetes mice [101]. Other researcher also
concluded similar finding in STZ-induced diabetic
rats [102]
33 Coccinia grandis Cucurbitaceae Whole plant Jazan Region [84]
e C. grandis extract (0.75 mg/kg, orally) showed
remarkable glycemic effect which confirmed
antidiabetic potential in streptozotocin-induced
diabetic rats [103].
34 Jatropha curcas Euphorbiaceae Leaves Jazan Region [84]
Ethanolic extract of J. curcas leaves at doses of (250
and 500 mg ml
1
bw by administered orally)
reduced glucose level from 219.5 to 116.5 and 237
to 98.8, respectively, in alloxan-induced diabetic
rats. e results were comparable to reduction in
rats treated with the standard glibenclamide
232–94.5 at 600 μg kg
1
[104].
Evidence-Based Complementary and Alternative Medicine 5
Table 1: Continued.
S.
no. Names of plants Family Part used location Antidiabetes Activities
35 Ricinus communis Euphorbiaceae Leaves Jazan Region [84]
R. communis extracts at doses of 300 and 600 mg/
kg/BW administered orally caused hyperglycemia
in a dose-dependent manner in streptozotocin-
induced diabetic rats [105].
36 Ficus carica Moraceae Leaves Jazan Region [84]
A review article focusing on antidiabetic potential
of F. carica confirmed that different extracts and
fractions of F. carica and different doses
significantly reducing hyperglycemia in
streptozotocin-induced diabetic rats compared to
standard drug [106].
37 Ficus sycomorus Moraceae Leaves Jazan Region [84]
Alloxan-induced type 2 diabetic albino Wistar rats
treated with 250, 500, and 1000 mg/kg (body
weight) of the extract of F. sycomorus
intraperitoneally reduced glucose level in diabetic
rats almost to the normal as compared to diabetic
control [107]
38 Sesamum indicum Pedaliaceae Seeds Jazan Region [84]
Alloxan-induced diabetic rats treated with 5% and
10% of Sesamum indicum seed powder
significantly decreased blood glucose and
increased insulin levels as compared with the
positive (diabetic) control group [108]
39 Plantago ovata Plantaginaceae Husk
Northern border
region of Saudi Arabia
[18]
In intravenous administration of alloxan-induced
diabetic rabbits glucose level lowering effect
observed (time dependent manner) with P. ovata
husk extract of dose level (300 mg/kg, orally
administered) [109]
40 Polygala erioptera Polygalaceae Aerial part Jazan Region [84]
0.7 g/kg of P. erioptera extract showed significant
antidiabetic effect compared to standard drug
metformin and glibenclamide in normal and
alloxan-induced diabetic rats [110]
41 Polygonum
aviculare LPolygonaceae Aerial parts Taif Region [111, 112]
Many ethnopharmacological investigations
reported its antidiabetic potential but still need to
study its in vivo and in vitro antidiabetic potential
[113, 114]
42 Ziziphus spina-
christi Rhamnaceae Leaves Eastern region of Saudi
Arabia [84, 112]
e strongest (P<0.001) antidiabetic activity
(25.59 and 39.48% after 7 and 15 days,
respectively) was found following treatment with
dose level of 500 mg/kg of Z. spina-christi extract
in streptozotocin-induced diabetes mice [115].
43 Bacopa monnieri Scrophulariaceae Aerial parts Jazan Region [84]
B. monnieri extract at dose level of 50, 100, 200,
and 400 mg/kg significantly inhibited (33.3, 34.2,
42.1, and 44.2%, respectively) the increase in
serum glucose concentration in a dose-dependent
manner compared to standard drug [116].
44 Lycium shawii Solanaceae Aerial parts Taif Region [112]
e strongest (P<0.001) antidiabetic activity of
L. shawii extract of 250 and 500 mg/kg bw was
found in a dose-dependent manner in
streptozotocin-induced diabetes rats [117].
45 Solanum nigrum Solanaceae Whole plant Jazan Region [84]
S. nigrum extract was given orally in the dose level
of 200 and 400 mg/kg/day (7 days) significantly
lowering the blood glucose level in fasting
compared to standard drug in alloxan-induced
diabetic albino Wistar rats [118].
6Evidence-Based Complementary and Alternative Medicine
plants around the world. e ethnobotanical information
reports about 800 plants that may possess antidiabetic po-
tential [196, 197]. Jeeva and Anlin also reported 177 plants
belonging to 156 genera and 76 families used traditionally
for antidiabetic treatment [198]. In the Middle East coun-
tries, there are 129 plant species still in use in traditional
Arabic medicine. is indicates that the medicinal plant
species require preservation as well as the ethnobotanical
and ethnopharmacological knowledge. e preservation of
the herbs is an essential requirement for maintaining tra-
ditional Arabic medicine as a medicinal and cultural re-
source [199]. e selected plant species H. salicornicum,
T. oliverianum, A. cepa, A. herba-alba, Teucrium polium,
Sesamum indicum, Z. spina-christi, and F. religiosa are the
native Saudi medicinal plants traditionally used for the
treatment of DM [200]. Similarly published data showed that
20 medicinal plants are traditionally used in Tabuk region of
Saudi Arabia [201]. Anisotes trisulcus, Artemisia judaica, and
Moringa peregrine are used in Al Khobah village, Saudi
Arabia, for DM treatment [202]. O. europaea is used in Al
Bahah region of KSA for DM treatment [203]. C. roseus,
A. cepa, U. dioica, A. aspera, C. intybus, C. cyminum,
F. bengalensis, C. colocynthis, and T. polium are the highly
investigated medicinal plants for antidiabetic potential
[204–206].
Desiring to contribute to the conservation priorities of
traditional medicine knowledge of various medicinal plants
native to or cultivated in Saudi Arabia and to make it easy
and familiarized with disease treatment, the present com-
pilation was conducted. According to the International
Union for Conservation of Nature and the World Wildlife
Fund, there about 15,000 medicinal plant species are
threatened with extinction from overharvesting and habitat
destruction and 20% of their wild resources have already
been nearly exhausted with the increasing human pop-
ulation and plant consumption [207]. Each plant species lost
due to extinction phenomena could represent not only the
loss of healthcare saving cures for special diseases but also
the loss of probable primary metabolite liker protein- or
vitamin-rich foods [208]. Medicinal plants have been cited as
a potential source of heavy metal toxicity to both man and
animals. e most common heavy metals implicated in
human toxicity include lead, mercury, arsenic, and cad-
mium, although aluminum and cobalt may also cause
Table 1: Continued.
S.
no. Names of plants Family Part used location Antidiabetes Activities
46 Withania
somnifera Solanaceae Leaves Jazan Region [84]
W. somnifera extract oral administration at two
doses (200 and 400 mg/kg) reduced the blood
glucose level significantly (P<0.001) in a dose-
dependent manner in streptozotocin-induced
diabetes rats. Only WS treatment did not register
any significant change in the blood glucose level
when compared to citrate control rats [119].
Another study also confirmed similar results in
alloxan-induced diabetic rats [120]
47 Lantana camara Verbenaceae Leaves Jazan Region [84]
Literature survey showed that L. camara leaf
extract oral administration (200, 250, and 500 mg/
kg of bw) showed antidiabetic potential in alloxan-
induced diabetic rats [121]
48 Peganum harmala Zygophyllaceae Seeds Taif Region [112]
P. harmala seed extract at dose level of (30, 60, and
120 mg/kg, orally administered for four weeks)
significantly decreases in blood glucose (in all
doses, P<0.001), in comparison with diabetic
group [122].
49 Tribulus terrestris Zygophyllaceae Stem, leaves Jazan Region [84]
Taif Region [112]
T. terrestris extract at (2 g/kg body weight)
produced protective effect in streptozotocin-
induced diabetic rats by inhibiting oxidative stress
[123].
T. terrestris L. extract (250 mg/kg of bw orally
administered) significantly lowers glucose level to
normal compared to standard drug in glucose-
loaded normal rabbits [124]
50 Urtica dioica Urticaceae Leaves Wild plant, Tanhat,
Saudi Arabia [125]
Urtica dioica extract at 100 mg/kg (P<0.01) and
200 mg/kg (P<0.001) significantly decreased
serum glucose fructose-induced insulin resistance
rats [126].e aqueous extract
of U. dioica significantly (P<0.001; 67.92%)
reduced the blood glucose level at dose of 300 mg/
kg, IP) in streptozotocin-induced diabetes rats
[127]
Evidence-Based Complementary and Alternative Medicine 7
Table 2: Active ingredients and toxicological evaluation of the medicinal plants given in Table 1.
S.
No Names Active ingredients Toxicological evaluation
1Allium cepa
Quercetin, N-acetylcysteine, alliuocide, cycloalliin, S-
methyl-L-cysteine, S-propyl-L-cysteine, sulfoxide, dimethyl
trisulfide, S-methyl-L-cysteine sulfoxide [128]
e animals tested were found healthy with no sign
of toxicity up to the dose of 2 500 mg/kg. However,
at 5 000 mg/kg, animals were weak and had intense
extreme tachycardia and disorientation but no
death was recorded. us, LD
50
was more than
5 000 mg/kg [129].
2Anthemis herba
alba
Guainalides, eudesmanolide, pseudogua inolides,
xanthonolides, flavone, flavonol glycosides, hispidulin,
cirsilineol, vicenin-2, schaftoside, isoschaftoside, 5,4-
dihydroxy-6,7,3-trimethoxyflavone, quercetin-3-
rutinoside, patuletin 3-rutinoside, patuletin 3-glucoside
[130]
e available toxicological investigations have
shown generally that Anthemis herba-alba is free
from toxic effects at the different doses used in the
studies [130]
3Cichorium intybus
Chicoric acid, inulin, cichoralexin, cichoriin, esculetin,
isochlorogenic acid, chlorogenic acid, caffeic acid,
dicaffeoylquinic acid, aesculin, arginine, histidine,
isoleucine, leucine, lysine, methionine, cysteine,
phenylalanine, tyrosine, threonine, valine, serine, glutamic
acid, glycine, alanine, aspartic acid, and proline [44, 45]
ere were no treatment-related toxic effects from
chicory extract administered orally at 70, 350, or
1000 mg/kg/day. ere were no observed adverse
effects of chicory extract in these studies [45]
4Clitoria ternatea
Kaempferol, quercetin, myricetin, taxaxerol, tannic acid, 3-
monoglucoside, β-sitosterol, delphinidin-3,5-diglucoside,
anthoxanthin glucoside, p-hydroxycinnamic acid,
kaempferol 3-neohesperidoside, myricetin 3-rutinoside,
hexacosanol [48]
Ethanolic extract of aerial parts and root of CT led
to lethargy in mice at the doses of 1500 mg/kg and
above, orally [31]. Ptosis was seen above 2000 mg/
kg dose in mice. rough intraperitoneal route,
2900 mg/kg dose was lethal within 6 hr due to
severe CNS depression [47].
5Ficus carica
Over 100 bioactive compounds have been identified in fig
such as rutin, arabinose, chlorogenic acid, β-amyrins,
syringic acid, β-carotenes, glycosides, β-sitosterols, and
xanthotoxol [131]
e rats tested were found healthy with no sign of
toxicity up to the dose of 5000, 5500, and 6000 mg/
kg. However, at 5 000 mg/kg, animals were weak
and had intense extreme tachycardia
and disorientation but no death was recorded.
us, LD
50
was more than 6000 mg/kg [132]
6Ficus
benghalensis
Leucopelargonidin-3-0-α-L rhamnoside, eucodelphinidin,
leucoanthocyanidia, leucoanthocyanin, α-amyrin acetate
[53]
In acute toxicity studies, no mortality and signs of
toxicity were observed at the dose of 2000 and
5000 mg/kg body weight for aqueous and ethanol
extracts, respectively [53]
7Ficus religiosa
Lupeol, β-sitosterol, β-sitosterol-d-glucoside, stigmasterol,
lanosterol, campesterol, octacosanol, methyl oleonate,
lupen-3-one, bergapten, and bergaptol [53]
Acute toxicity reported up to dose level 2000 mg/kg
showed no mortality [53]
8Ficus microcarpa
Polyphenols, organic acids, alkaloids, polysaccharides,
megastigmanes, pheophytins, catechin, epicatechin,
isovitexin, phenolic acids [53]
e oral administration of a single dose of 2000 mg/
kg ethanol or methanol extract of leaves showed no
mortality or behavioral alterations in the tested
animals [53]
9Hypericum
perforatum
Quercitrin, rutin, hypericin, kaempferol, biapigenin,
hyperforin [133]
Acute toxicity studies revealed the nontoxic nature
of the H. perforatum [55]
10 Anethum
graveolens
Carvone, α-phellandrene, limonene, dill ether, myristicin
coumarins, flavonoids, phenolic acids, steroids [134]
e mice treated with AG of different doses of 1000,
2000, 3000, 4000, and 5000 mg/kg of body showed
no toxicity [135]
11 Cuminum
cyminum
Cuminaldehyde, limonene, α- and β-pinene, 1, 8-cineole, o-
and p-cymene, α- and c-terpinene, safranal, and linalool
[58, 59]
e acute lethal toxicity test revealed that cumin
crude extract was very safe [58]
12 Marrubium
vulgare
Furanic labdane diterpenes, marrubenol, marrubiin,
ladanein [60]
An acute toxicity study of M. vulgare (1 g/kg)
extract orally administered at a dose of 1 g/kg body
weight to the mice and treated mice showed
tachycardia 1 h after intake of the infusion and loss
of appetite 3 h after intake of the infusion. In
another experiment, a single dose of 2000 mg/kg
extract of M. vulgare for an acute toxicity study
showed no toxicity [60].
8Evidence-Based Complementary and Alternative Medicine
Table 2: Continued.
S.
No Names Active ingredients Toxicological evaluation
13 Mentha longifolia
Lucenin-1, lucenin-2, camphelinone, camphene, carveol,
carvone, carvone oxide, limonene, linalool, menthatriene,
menthofuran, menthol, menthone, myrcene, p-cymene,
piperitenone, piperitone, sabinene, α-pinene, α-terpinene,
α-terpineol, longifone, pulegone, longifoamide-A,
longifoamide-B, longiside-A, longiside-B, eugenol,
salvianolic acid, eriodictyol-7-rutinoside, apigenin-7-O-
glucoside, hypolaetin, longitin, luteolin, etc. [136]
M. longifolia extract was safe, and no toxicity or
mortality was observed in both the oral (3200 mg/
kg) and intraperitoneal (1730 mg/kg)
administration in rats. Fourteen days of oral
administration of the essential oil (125, 250, 375,
and 500 µL/kg) resulted in the reduction of red
blood cells and lymphocytes and elevation of
neutrophils and monocytes compared with normal
animals [136].
14 Origanum
syriacum Carvacrol, thymol, thymoquinone [137] Not available
15 Teucrium
oliverianum
8-O-acetylharpagide, 12-O-methylteucrolin A, teucrolivin
A, eupatorin, teucrolivin B, μ24(S)-stigmasta-5,22,25-trin-
3β-ol [66]
Not available
16 Teucrium polium
Apigenin, luteolin, rutin, cirsiliol, cirsimaritin, salvigenin,
and eupatorin in the roots, aerial parts, and inflorescences,
teucardoside, b-sitosterol, stigmasterol, campesterol,
brassicasterol, and clerosterol [68]
All rats treated with different concentrations of the
total extract of TP were alive during the 14 days of
observation. e animals did not show visible signs
of acute toxicity. It suggested that the LD50 of the
total extract was higher than 8 g/kg [138]
17 Achyranthes
aspera
Aliphatic acid, betaine, achyranthine, β-ecdysterone,
achyranthes saponins A, B, C, D, oleonolic acid, glycosides,
triacontanol, E-sitosterol and spinasterol, triacontanol,
hydroquinone, eugenol [70]
In acute oral toxicity studies, there was no increase
or decrease in any of the parameters studied, in
comparison with control animals [139]
18 Aerva lanata
Quercetin, betulin, aervine, ervoside, methylervine, aervine,
lupeol, kaempferol, aervolanine, aervolanine, ervoside,
methylaervine, persinosides A and B, tannic acid, lupeol
acetate, benzoic acid, methyl grevillate [140]
e LD
50
of the extract of AL for oral and IP acute
toxicity tests were 22.62 g/kg and 0.432 g/kg,
respectively. e extract produced apparent
changes in body weights of both male and female
rats and increased the weights of lung, brain, and
pancreas of female rats while reducing the weight of
testes in male rats. Hematological parameters were
also altered [72]
19 Alternanthera
sessilis
Stigmasterol, β-sitosterol, β-carotene, ricinoleic acid,
myristic, palmitic, stearic, oleic, and linoleic acids,
α-spiraterol, uronic acid, cyclo eucalenol, choline, oleanolic
acid, lupeol [141]
e crude extract did not show any toxicity in mice
even at the highest dose tested [75]
20 Carissa edulis
Β-Amyrin, (+)-carissone, 2α-carissanol, 6α-carissanol,
dehydrocarissone, pinene, myrcene, limonene, sabanene,
rutin, epicatechin gallate, carinol, lariciresinol, β-sitosterol,
sitosterol glucoside, stigmasterol glucoside, scopoletin,
isofraxidin, pinitol [77]
Lethal effects were not observed after the oral
administration of the standardized ethanol extract
at doses of 1600, 2900, and 5000 mg/kg. No
behavioral changes were observed during the
observation period. e oral LD50 of the extract
was estimated to be greater than 5000 mg/kg. [142].
21 Catharanthus
roseus
Vinblastine, vincristine, vindesine, vindeline tabersonine,
ajmalicine, vinceine, vineamine, raubasin, reserpine,
catharanthine, rosindin [143]
No mortality, but dose level higher than 300 mg of
C. roseus extract can produce signs of biochemical
and histopathological toxicity in liver, kidney, and
heart. It is recommended that lower doses than the
studied ones should be used for treatment [144]
22 Rhazya stricta
Polyneuridine, stemmadenine, strictanol, rhazimine,
rhazinilam, rhazimanine, sewarine, vallesiachotamine,
tetrahydrosecamine [145]
Daily oral dosing of R. stricta extract (0.25 g/kg) for
42 days was not fatal to sheep [145].
Evidence-Based Complementary and Alternative Medicine 9
Table 2: Continued.
S.
No Names Active ingredients Toxicological evaluation
23 Calotropis procera
Calotropin, calotoxin, calactin, uscharin, voruscharin,
uzarigenin, syriogenin, proceroside, calotropagenin,
calotropain enzymes, α-amyrin, β-amyrin, lupeol,
β-sitosterol, ursolic acid, calotropin, gigantin, giganteol
[146]
2000 mg/kg body weight in single oral
administration of aqueous and hydroalcoholic
extract did not cause any death after 72 h post-
treatment in male and female mice. Daily
administration of aqueous extract to male and
female Wistar rats during 3 and 6 weeks at the dose
of 20 mg/kg/day induced no mortality in either sex
[147]. Whoever C. procera is a toxic plant that is
avoided by grazing animals. Its latex is used by
tribes to poison arrows used for hunting. If in
contact with human eye, it could cause ocular
toxicity, causing loss of vision and photophobia
[146]
24 Opuntia dillenii
Betanin, betanidin, kaempferol, kaempferide, quercetin,
isorhamnetin, β-sitosterol, C29-5β-sterols, taraxerol,
friedelin, methyl linoleate, 7-oxositosterol, 6β-
hydroxystigmast-4-ene-3-one, daucosterol, methyl
eucomate, eucomic acid [85]
During the oral toxicity study of the crude drug in
rats, given doses up to 50 ml/kg exhibited no
symptoms of toxicity [85]
25 Opuntia ficus
indica
Quercetin, isorhamnetin, kaempferol, luteolin,
isorhamnetin, isorhamnetin glycosides, gallic acid,
coumaric, narcissin, rutin, nicotiflorin, isoquercetin, ferulic
acid [87].
In vivo toxicity study suggests that the oral
administration of Opuntia ficus indica extract at
levels up to 2000 mg/kg/day does not cause adverse
effects in male and female rats [148].
26 Capparis decidua
n-Triacontane, n-pentacosane, β-carotene, n-triacontanol,
kaempferol, quercetin, isodulcite, nanocosane, capric acid,
glucocapparin, capparine, capparinine, cappariline,
codonocarpin, β-sitosterol [88]
e oral administration of C. decidua extract (500,
1000, 2000, and 4000 mg/kg) did not provoke any
gross behavioral changes or manifestations of toxic
symptoms in male rats [149].
27 Beta vulgaris Betaine, betacyanins, betaxanthins, oxalic acid, and
ascorbic acid [89]
In acute oral toxicity studies, the BVBF did not
show any sign and symptoms of toxicity and
mortality up to 2000 mg/kg dose, considered
relatively safe [150]
28
Haloxylon
salicorlicum
Bunge
Kaempferol, quercetin, betaine chloride, piperidine,
anabasine, aldotripiperideine, haloxine, halosaline,
oxedrine, tyramine, N-methyltyramine, scopoletin,
scopolin, umbelliferone, xanthotoxol, isooxyimperatorin,
esculetin, β-sitosterol, ursolic acid, β-amyrin [93].
H. salicorlicum extract at doses 0.1, 0.2, 0.3, 0.4, and
0.5 mL/kg orally administered in rats was safe and
showed no mortality or adverse effect [92].
29 Evolvulus
alsinoides
β-Sitosterol, betaine, shankpushpin, evolvine, caffeic acid,
6-methoxy-7-O-β-glucopyranoside coumarin, 2-C-methyl
erythritol, kaempferol-7-O-β-glucopyranoside,
kaempferol-3-O-β-glucopyranoside, quercetin-3-O-
β-glucopyranoside, scopoletin, scopolin [151]
e Evolvulus alsinoides extract did not cause any
mortality up to a dose of 1500 mg/kg body weight
and no behavioral, neurological, and autonomic
profiles and was found to be safe [151].
30 Ipomea aquatica
Caffeic acid, chlorogenic acid, quercetin glucoside,
quercetin malonyl glucoside, quercetin diglucoside,
catechin, isochlorogenic acid A, C, aspartic acid, glycine,
alanine and leucine, 7-O-β-D-
glucopyranosyldihydroquercetin-3-O-α-D-
glucopyranoside [97, 98]
In acute toxicity studies, I. aquatica extract was
found to be safe up to 2g to 5 g/kg in mice. No
mortality or toxic symptoms were observed during
the entire duration of the study [152, 153].
31 Citrullus
colocynthis
Cucurbitane, gallic acid, kaempferol, cucurbitacin A-E, I-L,
chlorogenic acid, caffeic acid, colocynthoside A,B,C;
choline, almitic acid, stearic acid, linoleic acid, oleic acids,
catechin, myricetin, α-tocopherol, c-tocopherol, β-carotene
[153]
C. colocynthis plant is safe to use. Studies showed
that lethal dose (LD
50
) to be 200 mg/kg, which
indicate that the studied plant is not toxic when
comparing the LD
50
values of most bioactive
pharmaceuticals currently used in therapeutics
[154]
32 Citrullus lanatus
Lycopene, vitamin A, cucurbitacin E, citrulline arginine,
glutamine and aspartic acid, pectin, vitamin b-complex and
minerals [155, 156]
In acute toxicity study, there was no mortality
observed up to the maximum dose level of
2000 mg/kg body weight of the extract after
administered orally [102]
10 Evidence-Based Complementary and Alternative Medicine
Table 2: Continued.
S.
No Names Active ingredients Toxicological evaluation
33 Coccinia grandis
Cephalandrol, β-sitosterol, cephalandrins A and B,
β-amyrin acetate, lupeol, cucurbitacin B, taraxerone,
taraxerol, β-carotene, lycopene, cryptoxanthin, xyloglucan,
carotenoids, β-sitosterol, stigma-7-en-3-one, lupeol,
β-amyrin, β-sitosterol, taraxerol [157]
e acute toxicity study indicated that treatment of
C. grandis is safe up to 2 g/kg tested on animal
[158].
34 Jatropha curcas
Jatrophol, jatropha factor C1, C2, C3,C4, C5, C6,
jatropholones A, B,palmarumycin CP1, JC1,JCV2,curcin,
curcacycline A, curcain, β-amyrin, β-sitosterol,
stigmasterol, friedelin, taraxasterol, diamide, pyrimidine-
2,4-dione, nobiletin, tomentin [103]
Many researchers have confirmed that J. curcas is
highly toxic for animal as well as human. All parts
of J. curcas are toxic and toxic compound reported
from this plant like lectins, curcin, phorbol esters,
phytate, protease inhibitors [103]
35 Ricinus communis
Rutin, quercetin, gallic acid, ricin, ricin A, kaempferol-3-O-
βrutinoside, gentistic acid, linolenic acid, α-pinene,
α-thujone, stigmasterol, ricinine, β-sitosterol, lupeol, castor
oil [159, 160]
R. communis extracts given by oral route were safe
up to a dose of 2,000 mg/kg/BW and did not show
any mortality and toxic effects in the behavior of
the treated animals [104].
36 Ficus carica
Ferulic acid, quercetin-3-O-glucoside, quercetin-3-O-
rutinoside, psoralen, bergapten, coumarin, oleanolic acid,
eugenol, angelicin, germacrene D, menthol, α-pinene,
β-pinene [161]
Toxicity of 70% methanolic extract of Ficus carica
leaves showed LD
50
value of brine shrimp assay was
0.158 mg/ml [162]
37 Ficus sycomorus Tannins, saponins, flavones, aglycones, anthraquinone
glycosides, and flavonoid glycosides [53]
F. sycomorus methanol extract of stem bark is
nontoxic up to the dose of 5000 mg/kg [53]
38 Sesamum indicum
elignans, sesamolin, sesamin, pinoresinol, lariciresinol,
α-globulin, β-globulin, triacylglycerols, oleic, linoleic acids,
sesamol, c-tocopherol, 2-furfurylthiol, 2-phenylethylthiol,
2-methoxyphenol, 2-pentylpyridine, vitamin E, quinone,
sesangolin [163, 164]
S. indicum ethanol extract is safe, up to dose level of
2000 mg/kg in acute toxicity studies in tested
animals [165]
39 Plantago ovata
Hemicellulose,D-xylose, L-arabinose, D-glucose, D-
galactose, and L-rhamnose, 5, 6,8-epiloganic acid,
gardoside, plantamajoside [166]
4,5 Gram seed husk one to four times a day soaked
in 150 ml of warm water recommend by WHO.
Studies confirmed its side effect like bloating, gas,
and allergy. No mortality reported [167]
40 Polygala erioptera
Helioxanthin [168]. No literature available. Recommended
for natural products, isolation, biological and toxicological
evaluation.
No literature available. Recommended for
pharmacological and toxicological evaluation.
41 Polygonum
aviculare L
Protocatechuic acid, catechin, myricitrin, epicatechin-3-O-
gallate, avicularin, quercetin, juglanin, kaempferol,
myricetin 3-0-(3-0-galloyl-rhamnopyranoside, cinaroside,
liquiritin, rutin [169, 170]
No data available
42 Ziziphus spina-
christi
Jujuboside B1, christinin A, christinin A1 and A2, lotoside
II, catechin, epicatechin, kaempferol 3-O-(6-O-rhamnosyl-
galactoside), quercetin 7-O-(6-O-rhamnosyl-glucoside),
quercetin 3-O-glucoside, kaempferol 3-O-glucoside
[171, 172]
Butanol and water extract of Ziziphus spina-christi
up to 100 mg/kg and 5g/kg, respectively, in animal
model produced no functional or structural
disturbances in liver and kidney and no
hematological changes [173, 174]
43 Bacopa monnieri
Bromine, β-sitosterol, betulinic acid, stigmasterol
nicotinine, herpestine, bacosides A, bacopasides (I, II, III,
IV, V), pseudojujubogenin glycoside, saponins (A, B, C)
[175]
B. monnieri extract at the dose of 5,000 mg/kg did
not cause any side effects. Similarly doses of 30, 60,
300, and 1,500 mg/kg given for 270 days did not
produce any toxicity in rats [176].
44 Lycium shawii
Lyciumate, cyclopentapyrrolidine, imidazole, piperidine,
nortropane, tropane, pyrrole, spermine, costunolide,
catechin, lyciumaside, emodin, betulinic acid, β-sitosterol
glucopyranoside, quercetin, gallic acid, rutin, ρ-coumaric
acid, ferulic acid [177, 178]
Reported data revealed that LD
50
of the L. shawii
extract was greater than 2000 mg/kg b.w in animal
model [179].
45 Solanum nigrum
Chlorogenic acid, quercetin, naringenin, solasodine,
solamargine, solasonine, α-solanigrine, β-solanigrine,
ascorbic acid, nigrumnins I and II [180]
S. nigrum extract at a dose of 2000 mg/kg p.o. was
safe and showed no changes/alteration in normal
behavior in animal model. No mortality was
observed [181]
Evidence-Based Complementary and Alternative Medicine 11
Table 2: Continued.
S.
No Names Active ingredients Toxicological evaluation
46 Withania
somnifera
Withanolides, withaferin, withaferin A, withanone,
withanolide A, withanolide IV, withanolide V, withanolide
D [182].
LD
50
value of W. somnifera extract in rates was
greater than 2000 mg/kg body weight. Compared to
the control group in subacute toxicity study,
administration of extract did not show any
toxicologically significant treatment-related
changes in clinical observations, and the
toxicological studies revealed that the reasonable
doses of W. somnifera are nontoxic and safe
[182, 183]
47 Lantana camara
Eicosane, squalene, β-ionone, caryophyllene oxide,
β-caryophyllene, hexanoic acid, tiglic acid, lantanilic acid,
camaric acid, lantadene B, oleanolic acid, lantadene A,
lantaninilic acid, lantoic acid, ursolic acid, betulinic acid
[184]
L. camara extracts at dose level of 2000 mg/kg and
5000 mg.kg body weight in mice and rats,
respectively, showed no significant toxic signs or
mortality [185, 186]
48 Peganum harmala
Harmine, harmaline, harmalol, harman, vasicine and
vasicinone, pegamine, acacetin 7-O-rhamnoside, 7-O-6-
O-glucosyl-2 -O-(3-acetylrhamnosyl) glucoside, 7-0-
(2-0-rhamnosyl-2-O-glucosylglucoside), peganone 1
and 2, p-cymene, limonene, eugenol, thujico acid,
β-cubebene [187–189]
P. harmala different doses of different extracts in
animal and human clinical studies confirmed that
this plant showed side effect like intoxications,
abdominal writhing, body tremors, and toxic at
high does level causing paralysis, liver
degeneration, euphoria, convulsions, nausea,
vomiting, hypothermia. However, therapeutic
doses have been reported to be safe in a rodent
model [189]
49 Tribulus terrestris
Naringin, rutin, hyperoside, quercitrin, naringenin,
quercetin, hesperetin, kaempferol, apigenin, pyrogallol,
gallic acid, catechin, catechol, chlorogenic acid, caffeic acid,
vanillic acid, ferulic acid, salicylic acid, ellagic acid,
coumaric acid, cinnamic acid [190]
T. terrestris extract showed no mortality/or toxicity
at a dose up to 1 g kg
1
of bw in mice [190]
50 Urtica dioica
β-Amyrin, β-sitosterol, stigmasterol, oleanolic acid, ursolic
acid, quercetin, rutin, chlorogenic, and 2-O-caffeoyl malic
acid expressed as caffeic acid, isoquercetin, kaempferol 3-
O-rutinoside [191–193].
U. dioica extracts up to dose level of 2000 mg/kg
body weight in animal model showed no mortality
or changes/alteration in normal behavior [127].
Table 3: Selected antidiabetic phytoconstituents and its targeted metabolic pathway.
Phytoconstituents Targeted metabolic pathways Structures
Catharanthine
Free radical; our body has a defense system containing
several enzymes, which are catalase, superoxide
dismutase, and glutathione-S transferases and reduced
glutathione. Catharanthine activates these free radical
scavenging enzymes and prevents our body from their
adverse effects.
NH
O
O
H
Harmine Insulin secretion and β-cell regeneration
O
N
H
N
Betaine
Achyranthine
β-ecdysone
Carbohydrate digestion and absorption
H3C
H3C
H3C
N
O
O
O
OH
N
HO
HO
H
H
H
O
OH
OH
OH
12 Evidence-Based Complementary and Alternative Medicine
Table 3: Continued.
Phytoconstituents Targeted metabolic pathways Structures
Apigenin Cholesterol synthesis, glycogen synthesis
OH
O
O
OH
HO
Betaine choline Regeneration of pancreatic βcells and insulin secretion
OH
N+X-
Ferulic acid Free radical scavenging activity, insulin secretion
O
OH
H3CO
HO
Leucine, isoleucine, alanine Insulin secretion
Chlorogenic acid Krebs cycle
HO
HO
OH
O
O
OH
OH
CO2H
Emodin, cinnamic acid Insulin secretion
OH OH
OH
OH
O
O
O
H3C
Eugenol, α-pinene, limonene, p-
cymene thujone Insulin secretion, regeneration of pancreatic βcells
O
HO
H3C
H3C
CH3
H3CCH3
CH3
CH3
CH3
H3C
HO
Pectin Glucose transport, carbohydrate metabolism, stabilizing
agents
O
OH
HO
OH
OH
CO2H
Evidence-Based Complementary and Alternative Medicine 13
Table 3: Continued.
Phytoconstituents Targeted metabolic pathways Structures
Inulin Glucose transport, carbohydrate digestion and
absorption
HO
O
HO
OH OH
OH
HO
OH
O
O
O
O
O
HO
n
OH
6
4
HO 3
5
2
OH
1
1
2
3
O
HO
4
5
6
OH
OH
Cucurbitacin B Insulin secretion, glycogen synthesis
CH3
CH3
CH3
CH3
CH3
CH3
CH3
O
O
O
Cucurbitacin-B
OH
HH
HO
HO
O
H3C
H3C
H
O
Catechin
Epicatechin
Scavenging activity
Insulinomematic
HO O
OH
OH
OH
OH
Cucurbitacin-B
OH
OH
OH
OH
HO O
Naringin Glycogen synthesis, glycolysis, gluconeogenesis
HO
HO
OH
OH
OH
OH
OH
OO
O
O
O
HO
H3C
O
Flavones Insulin secretion
O
O
Quercetin, quercitrin, apigenin,
rutin, apigenin-7-O-glucoside Insulin secretion
OOO
O
OH
OH
OH
HO
HO
OH
HO O
OO
O
O
OOH
OH
HO
OH OH
OH
OH
HO
HO
H3C
HO
OH O
O
OH
HO
OH O
O
HO
OH
OH
OH
OH
O
O
O
O
OH
OH
OH
HO
HO
Naringenin Insulin secretion
O
O
HO
OH
OH
14 Evidence-Based Complementary and Alternative Medicine
toxicity. From the study, the levels of these metals differed in
the same plant collected from different geographical loca-
tions. A study conducted showed that the levels of lead in
Cassia alata varied from 17.7 to 4.45 μg/g for the 5 collection
sites. Similarly for Cassia occidentalis and Rauvolfia vomi-
toria, the level is varied between 7.85-4.35 and 9.25–1.55 μg/
g, respectively. Similarly, that of aluminum varied between
105.53 and 23.3 for Rauvolfia vomitoria and 104.25–12.4 μg/
Table 3: Continued.
Phytoconstituents Targeted metabolic pathways Structures
α-Terpineol Insulin secretion
OH
Kaempferol, isorhamnetin,
caffeic acid, p-coumaric acid
Free radical scavenging activity
Carbohydrate digestion and absorption, insulin
secretion
HO
HO
O
OH
HO
O
OH
HO O
O
OH
OH
OH
OCH3
O
O
OH
OH
OH
HO
Vitamin A, E Free radical scavenging activity
OH
HO
H3C
CH3
CH3
CH3
CH3CH3
CH3
CH3
O
HH
Ellagic acid Carbohydrate digestion and absorption, insulin
secretion
OH
OH
O
O
O
O
HO
HO
Carvacrol, linalool Insulin secretion, carbohydrate digestion and
absorption
OH
HO
Stigmasterol Regeneration of pancreatic βcells, insulin secretion
HO
H
H
H
H
Ursolic acid Regeneration of pancreatic βcells and insulin secretion
O
H
H
H
OH
HO
β-Sitosterol Insulin receptor (IR) and glucose transporter 4
H
HH
H
HO
Evidence-Based Complementary and Alternative Medicine 15
g for Paullinia pinnata. e levels of heavy metals also varied
for different plants collected from the same location. Uptake
of metals by plants is influenced by a number of factors
including metal concentrations in soils, cation exchange
capacity, soil pH, organic matter content, types and varieties
of plants, and plant age. However, the prevailing factor is the
concentration of the metal in the soil and thus the existing
environmental conditions [209]. Another study conducted
on onion bulb showed that the concentrations of Cr in onion
bulb and Fe in onion leaf were above the permissible level
(2.3 mg/kg, 425.5 mg/kg) set by FAO/WHO at Mojo
(4.87 mg/kg, 1090.40 mg/kg), Meki (4.13 mg/kg, 1836.47 mg/
kg), and Ziway (3.33 mg/kg, 764.33 mg/kg), respectively. e
results generally indicate that the consumption of these
onion bulbs could be the health risk respective to Cr [210].
erefore, it is suggested that the medicinal plant source for
the treatment of diabetes must not be taken from heavy
metal contaminated areas to avoid their uptake by the plants
because migration of these contaminants into non-
contaminated areas (or leaching through the soil and
spreading of heavy metal contaminated sewage sludge) are a
few examples of events contributing to contamination of the
ecosystem.
5. Conclusion and Recommendations
e present review provides a picture of medicinal plants
that have been studied as anti-DM drugs, which can be
grown either in combination with other medicinal plants or
alone as treatment for diabetes and drawbacks should be
properly addressed so that medicinal plants can be effectively
utilized as anti-DM drugs. Diabetes is a metabolic disorder
which can be considered as a major cause of high economic
loss which can in turn impede the development of nations.
Moreover, uncontrolled diabetes leads to many chronic
complications such as blindness, heart failure, and renal
failure. In order to prevent this alarming health problem, the
development of research into new hypoglycemic and po-
tentially antidiabetic agents is of great interest. In conclu-
sion, this paper has presented a list of anti-DM plants used in
the treatment of diabetes mellitus. Bioactive antidiabetic
phytoconstituents which showed that these plants have
hypoglycemic effects and highly recommended for further
pharmacological purposes and to isolate/identify anti-DM
active agents also need to investigate the side effects of active
ingredients.
Data Availability
is is a review article. All data are taken from published
research papers and available online.
Conflicts of Interest
e authors declare no conflicts of interest.
Authors’ Contributions
All three authors contributed equally.
Acknowledgments
e authors wish to thank Research Center College of
Pharmacy at King Saud University, Riyadh, Saudi Arabia for
their financial support and for providing free access to digital
library and laboratory.
References
[1] B. Mahesh and S. Satish, “Antimicrobial activity of some
important medicinal plant against plant and human path-
ogens,” World Journal of Agricultural Sciences, vol. 4,
pp. 839–843, 2008.
[2] B.-E. van Wyk and J. van Staden, “A review of ethnobotanical
research in southern Africa,” South African Journal of
Botany, vol. 68, no. 1, pp. 1–13, 2002.
[3] P. Bhojane, S. Damle, A. ite, and V. Dabholkar, “Anti-
microbial effects of some leafy vegetables—a comparative
analysis,” International Research Journal of Biological Sci-
ences, vol. 3, pp. 26–32, 2014.
[4] W. Talib and A. Mahasneh, “Antimicrobial, cytotoxicity and
phytochemical screening of Jordanian plants used in tra-
ditional medicine,” Molecules, vol. 15, no. 3, pp. 1811–1824,
2010.
[5] H. Rahman, I. Khan, A. Hussain et al., “Glycyrrhiza glabra
HPLC fractions: identification of Aldehydo Iso-
ophiopogonone and Liquirtigenin having activity against
multidrug resistant bacteria,” BMC Complementary and
Alternative Medicine, vol. 18, no. 1, p. 140, 2018.
[6] A. A. Shahat, R. Ullah, A. S. Alqahtani, M. S. Alsaid,
H. A. Husseiny, and O. T. R. Meanazel, “Hepatoprotective
effect of Eriobotrya japonica leaf extract and its various
fractions against carbon tetra chloride induced hepatotox-
icity in rats,” Evidence-based Complementary and Alternative
Medicine, vol. 2018, Article ID 3782768, 8 pages, 2018.
[7] N. A. A. Ali, M. Wurster, N. Arnold, U. Lindequist, and
L. Wessjohan, “Chemical composition of the essential oil of
Teucrium yemense Deflers,” Records of Natural Products,
vol. 2, no. 2, p. 25, 2012.
[8] D. M. Livermore, “Has the era of untreatable infections
arrived?” Journal of Antimicrobial Chemotherapy, vol. 64,
no. 1, pp. 29–36, 2009.
[9] N. Mohd Azman, M. Gallego, F. Segovia, S. Abdullah,
S. Shaarani, and M. Almajano Pablos, “Study of the prop-
erties of bearberry leaf extract as a natural antioxidant in
model foods,” Antioxidants, vol. 5, no. 2, p. 11, 2016.
[10] R. Radhakrishnan, M. N. M. Zakaria, M. W. Islam et al.,
“Analgesic and anti-inflammatory activities of Teucrium
stocksianum,” Pharmaceutical Biology, vol. 39, no. 6,
pp. 455–459, 2001.
[11] S. Kanwal, N. Ullah, I. U. Haq, I. Afzal, and B. Mirza,
“Antioxidant, antitumor activities and phytochemical in-
vestigation of Hedera nepalensis K. Koch, an important
medicinal plant from Pakistan,” Pakistan Journal of Botany,
vol. 43, no. 8, pp. 85–89, 2011.
[12] S. Madhuri and G. Pandey, “Some anticancer medicinal
plants of foreign origin,” Current Science, vol. 96, no. 6,
pp. 779–783, 2009.
[13] P. Gauniyal and U. V. S. Teotia, “Antimicrobial activity and
phytochemical medicinal plants against oral microorgan-
ism,” International Journal of Pharmaceutical and Medicinal
Research, vol. 2, no. 1, pp. 21–27, 2014.
[14] A. C. Goren, G. Bilsel, M. Bilsel, H. Demir, and
E. E. Kocabas¸, “Analysis of essential oil of Coridothymus
16 Evidence-Based Complementary and Alternative Medicine
capitatus (L.) and its antibacterial and antifungal activity,”
Zeitschrift fur Naturforschung. C, Journal of biosciences,
vol. 58, no. 9-10, pp. 687–690, 2003.
[15] A. J. Afolayan, “Extracts from the shoots of Arctotis arcto-
toides inhibit the growth of bacteria and fungi,” Pharma-
ceutical Biology, vol. 41, no. 1, pp. 22–25, 2003.
[16] V. P. Veerapur, K. R. Prabhakar, V. K. Parihar et al., “Ficus
racemosaStem bark extract: a potent antioxidant and a
probable natural radioprotector,” Evidence-based Comple-
mentary and Alternative Medicine, vol. 6, no. 3, pp. 317–324,
2009.
[17] C. Manach, A. Scalbert, C. Morand, C. R´
em´
esy, and
L. Jim´
enez, “Polyphenols: food sources and bioavailability,”
e American journal of clinical nutrition, vol. 79, no. 5,
pp. 727–747, 2004.
[18] A. K. Osman, F. Al-Ghamdi, and A. Bawadekji, “Floristic
diversity and vegetation analysis of wadi arar: a typical desert
wadi of the northern border region of Saudi Arabia,” Saudi
Journal of Biological Sciences, vol. 21, no. 6, pp. 554–565,
2014.
[19] S. C. Morey, “ADA recommends a lower fasting glucose
value in the diagnosis of diabetes mellitus,” American Family
Physician, vol. 56, no. 8, pp. 2128–2130, 1997.
[20] K. Srinivasan and P. Ramarao, “Animal models in type 2
diabetes research: an overview,” Indian Journal of Medical
Research, vol. 125, no. 3, pp. 451–472, 2007.
[21] S. Kazemi, S. Asgary, J. Moshtaghian, M. Rafieian,
A. Adelnia, and F. Shamsi, “Liver-protective effects of
hydroalcoholic extract of Allium hirtifolium Boiss. In rats
with alloxan-induced diabetes mellitus,” ARYA atheroscle-
rosis, vol. 6, no. 1, pp. 11–15, 2010.
[22] R. Gleckman and J. Morr, “Diabetes-related foot infections,”
Contemporary Internal Medicine, vol. 6, no. 8, pp. 57–64,
1994.
[23] S. Behradmanesh, F. Derees, and M. Rafieian-kopaei, “Effect
of Salvia officinalis on diabetic patients,” Journal of Renal
Injury Prevention, vol. 2, no. 2, pp. 51–54, 2013.
[24] J. B. Meigs, “Epidemiology of the metabolic syndrome,”
American Journal of Managed Care, vol. 8, no. 11,
pp. S283–S292, 2002.
[25] A. J. Boulton, L. Vileikyte, G. Ragnarson-Tennvall, and
J. Apelqvist, “e global burden of diabetic foot disease,” e
Lancet, vol. 366, no. 9498, pp. 1719–1724, 2005.
[26] R. H. Williams and P. R. Larsen, Textbook of Endocrinology,
Saunders, Philadelphia, PA, USA, 10th edition, 2003.
[27] S. K. Swanston-Flatt, C. Day, C. J. Bailey, and P. R. Flatt,
“Evaluation of traditional plant treatments for diabetes:
studies in streptozotocin diabetic mice,” Acta Diabetologica
Latina, vol. 26, no. 1, pp. 51–55, 1989.
[28] H. Nasri and M. Rafieian-Kopaei, “Herbal medicine and
diabetic kidney disease,” Journal of Nephropharmacology,
vol. 2, no. 1, pp. 1-2, 2013.
[29] M. Eddouks, D. Chattopadhyay, V. De Feo, and
W. C.-s. Cho, “Medicinal plants in the prevention and
treatment of chronic diseases 2013,” Evidence-based Com-
plementary and Alternative Medicine, vol. 2014, Article ID
180981, 3 pages, 2014.
[30] F. Jamila and E. Mostafa, “Ethnobotanical survey of me-
dicinal plants used by people in Oriental Morocco to manage
various ailments,” Journal of Ethnopharmacology, vol. 154,
no. 1, pp. 76–87, 2014.
[31] S. Venkatesh, G. D. Reddy, B. M. Reddy, M. Ramesh, and
A. V. N. A. Rao, “Antihyperglycemic activity of Caralluma
attenuata,” Fitoterapia, vol. 74, no. 3, pp. 274–279, 2003.
[32] H. Nasri and H. Shirzad, “Toxicity and safety of medicinal
plants,” Journal of Herbmed Pharmacology, vol. 2, no. 2,
pp. 21-22, 2013.
[33] M. El-Bab, N. Shawky, A. Al-Sisi, and M. Akhtar, “Reti-
nopathy and risk factors in diabetic patients from Al-
Madinah Al-Munawarah in the Kingdom of Saudi Arabia,”
Clinical Ophthalmology, vol. 6, no. 1, pp. 269–276, 2012.
[34] S. N. Alqahtani, S. Omar Alkholy, and M. Pontes Ferreira,
“Antidiabetic and anticancer potential of native medicinal
plants from Saudi Arabia,” Polyphenols in Human Health
and Disease, vol. 1, pp. 119–132, 2014.
[35] M. Behradmanesh, M. Ahmadi, and M. Rafieian-kopaei,
“Effect of glycogol on blood glucose level of patients with
type II diabetes,” Iranian Journal of Endocrinology and
Metabolism, vol. 14, no. 2, pp. 163–168, 2012.
[36] M. Abdulrahman, “Al-moshileh effects of planting date and
irrigation water level on onion (Allium cepa L.) production
under central Saudi arabian conditions,” Scientific Journal of
King Faisal University (Basic and Applied Sciences), vol. 8,
no. 1, pp. 75–85, 2007.
[37] A. E. Baragob, N. A. Al-Wabel, N. A. Ahmed, M. F. Babiker,
A. S. Abdalkarim, and M. I. Elboshra, “Study to investigate
the pancreatic regeneration and evaluation of the antidia-
betic and antihyperlipidemic potential of aerial parts of
Allium cepa,” Biochemistry and Biotechnology Research,
vol. 3, pp. 19–29, 2015.
[38] A. E. Ojieh, E. C. Adegor, A. C. Okolo, O. L. Ewhre,
I. P. Njoku, and C. U. Onyekpe, “Hypoglycaemic and
hypolipidaemic effect of Allium cepa in streptozotocin-in-
duced diabetes,” International Journal of Scientific Engi-
neering and Research, vol. 6, pp. 23–29, 2015.
[39] M. Imad, T. Eldin, E. M. Ahmed, and H. M. Abd Elwahab,
“Preliminary study of the clinical hypoglycemic effects of
Allium cepa (red onion) in type 1 and type 2 diabetic pa-
tients,” Environmental Health Insights, vol. 4, pp. 71–77,
2010.
[40] A. Mohammed, S. Syed, M. Y. Syed, and A. D Ali, “Use of
herbal extract from Artemisia herba-alba (Shih) in phar-
maceutical preparations for dental hygiene,” Saudi Phar-
maceutical Journal: the Official Publication of the Saudi
Pharmaceutical Society, vol. 26, pp. 822–828, 2018.
[41] N. E. Awad, A. A. Seida, Z. El-Khayat, N. Shaffie, M. Ahmed,
and Abd El-Aziz, “Hypoglycemic activity of Artemisia
herba-alba (asso.) used in Egyptian traditional medicine as
hypoglycemic remedy,” Journal of Applied Pharmaceutical
Science, vol. 2, no. 3, pp. 30–39, 2012.
[42] E. P´
erez-L´
opez, A. F. Omar, K. M. Al-Jamhan, and
T. J. Dumonceaux, “Molecular identification and charac-
terization of the new 16SrIX-J and cpn60 UT IX-J phyto-
plasma subgroup associated with chicory bushy stunt disease
in Saudi Arabia,” International Journal of Systematic and
Evolutionary Microbiology, vol. 68, no. 2, pp. 518–522, 2018.
[43] R. A. Street, J. Sidana, and G. Prinsloo, “Cichorium intybus:
traditional uses, phytochemistry, pharmacology, and toxi-
cology,” Evidence-Based Complementary and Alternative
Medicine, vol. 2013, Article ID 579319, 13 pages, 2013.
[44] D. Kanj, K. Raafat, A. El-Lakany, S. Baydoun, and M. Aboul-
Ela, “Phytochemical compounds of cichorium intybus by
exploring its antioxidant and antidiabetic activities,” Phar-
macognosy Journal, vol. 11, no. 2, pp. 248–257, 2019.
[45] S. Das, N. Vasudeva, and S. Sharma, “Cichorium intybus: a
concise report on its ethnomedicinal, botanical, and phy-
topharmacological aspects,” Drug Design, Development and
erapy, vol. 7, pp. 1–12, 2016.
Evidence-Based Complementary and Alternative Medicine 17
[46] A. E. Al-Snafi, “Pharmacological importance of Clitoria
ternatea—a review,” IOSR Journal of Pharmacy, vol. 6, no. 3,
pp. 68–83, 2016.
[47] B. Gollen, J. Mehla, and P. Gupta, “Clitoria ternatea linn: a
herb with potential pharmacological activities: future
prospects as therapeutic herbal medicine,” Journal of Reports
in Pharmaceutical Sciences, vol. 3, no. 141, pp. 1–8, 2018.
[48] M. B. Lijon, N. S. Meghla, E. Jahedi, M. A. Rahman, and
I. Hossain, “Phytochemistry and pharmacological activities
of Clitoria ternatea,” International Journal of Natural and
Social Sciences, vol. 4, no. 1, pp. 1–10, 2017.
[49] H. Khemira and M. Mars, “Fig production in subtropical
south-western Saudi Arabia,” Acta Horticulturae, vol. 1173,
no. 1173, pp. 169–172, 2017.
[50] K. Y. Khan, M. A. Khan, M. Ahmad et al., “Hypoglycemic
potential of genus Ficus L.: a review of ten years of plant
based medicine used to cure diabetes (2000–2010),” Journal
of Applied Pharmaceutical Science, vol. 1, no. 6, pp. 223–227,
2011.
[51] S. Stephen Irudayaraj, S. Christudas, S. Antony,
V. Duraipandiyan, A.-D. N. Abdullah, and
A.-D. Ignacimuthu, “Protective effects of Ficus carica leaves
on glucose and lipids levels, carbohydrate metabolism en-
zymes and β-cells in type 2 diabetic rats,” Pharmaceutical
Biology, vol. 55, no. 1, pp. 1074–1081, 2017.
[52] L. I. El-Juhany, A. Abdullah, and Al-Harby, “Status and
diversity of ornamental plants in king Saud university
campus at Riyadh, Saudi Arabia American-eurasian,”
Journal of Agriculture and Environmental Sciences, vol. 13,
no. 4, pp. 471–478, 2013.
[53] P. Deepa, K. Sowndhararajan, S. Kim, and S. J. Park, “A role
of Ficus species in the management of diabetes mellitus: a
review,” Journal of Ethnopharmacology, vol. 215, pp. 210–
232, 2018.
[54] F. Scotti, K. L¨
obel, A. Booker, and M. Heinrich, “St. John’s
wort (Hypericum perforatum) products—how variable is the
primary material?” Frontiers of Plant Science, vol. 9, p. 1973,
2019.
[55] S. Arokiyaraj, R. Balamurugan, and P. Augustian, “Anti-
hyperglycemic effect of Hypericum perforatum ethyl acetate
extract on streptozotocin-induced diabetic rats,” Asian Pa-
cific Journal of Tropical Biomedicine, vol. 1, no. 5, pp. 386–
390, 2011.
[56] A. Alqethami, J. A. Hawkins, and I. Teixidor-Toneu, “Me-
dicinal plants used by women in Mecca: urban, Muslim and
gendered knowledge,” Journal of Ethnobiology and Ethno-
medicine, vol. 13, no. 62, pp. 62–24, 2017.
[57] M. T. Goodarzi, I. Khodadadi, H. Tavilani, and E. Abbasi
Oshaghi, “e role of Anethum graveolens L. (Dill) in the
management of diabetes,” Journal of Tropical Medicine,
vol. 2016, Article ID 1098916, 11 pages, 2016.
[58] D. A. Mohamed, I. Mohamed Hamed, and K. Aly Fouda,
“Antioxidant and anti-diabetic effects of cumin seeds crude
ethanol,” Extract Journal of Biological Sciences, vol. 18, no. 5,
pp. 251–259, 2018.
[59] R. P. Singh, H. V. Gangadharappa, and K. Mruthunjaya,
“Cuminum cyminum—a popular spice: an updated review,”
Pharmacognosy Journal, vol. 9, no. 3, pp. 292–301, 2017.
[60] S. Lodhi, G. Vadnere, V. K. Sharma, and M. Rageeb Usman,
“Marrubium vulgare L.: a review on phytochemical and
pharmacological aspects,” Journal of Intercultural Ethno-
pharmacology, vol. 6, no. 4, pp. 429–452, 2017.
[61] H. A. S. Murad, H. M. Abdallah, and S. S. Ali, “Mentha
longifolia protects against acetic-acid induced colitis in rats,”
Journal of Ethnopharmacology, vol. 190, pp. 354–361, 2016.
[62] B. Asgharia, G. Zenginb, M. Babak Bahadoric, and
M. Abbas-Mohammadid, “Leila Dinparast Amylase, glu-
cosidase, tyrosinase, and cholinesterases inhibitory, anti-
oxidant effects, and GC-MS analysis of wild mint (Mentha
longifolia var. calliantha) essential oil: a natural remedy,”
European Journal of Integrative Medicine, vol. 22, pp. 44–49,
2018.
[63] M. Eldesouky Zain, Amani Shafeek Awaad, and M. R. Al-
Outhman, “Reham mostafa el-meligy antimicrobial activities
of Saudi arabian desert plants,” Phytopharmacology, vol. 2,
no. 1, pp. 106–113, 2012.
[64] A.-A. A. Rasheed, A.-Q. T. Musbah, A. M. Sulaiman,
S. M. Saed, and B. A. Mohammad, “Short-term feeding
effects of origanum syriacum crude extract on blood con-
stituents in rats,” International Journal of Research in
Ayurveda and Pharmacy, vol. 8, no. 1, pp. 118–120, 2017.
[65] N. Fatima, “A Review on Teucrium oliveranum, a plant found
abundantly in Saudi Arabia,” Science International, vol. 28,
no. 2, pp. 1229–1231, 2016.
[66] A. A. Shahat, M. S. Alsaid, J. A. Khan, M. Higgins, and
A. T. Dinkova-Kostova, “Chemical constituents and
NAD(P)H:quinone oxidoreductase 1 (NQO1) inducer ac-
tivity of Teucrium oliverianum Ging. ex Benth,” Indian
Journal of Traditional Knowledge, vol. 15, no. 2, pp. 232–236,
2016.
[67] M. Hossam, G. A. M. Abdallah, M. A. Farag, K. Z. Alshali,
E. A. Alsherif, and S. A. Ross, “Volatile oil profile of some
lamiaceous plants growing in Saudi Arabia and their bio-
logical activities,” Zeitschrift f¨
ur Naturforschung C, vol. 72,
no. 1-2, pp. 35–41, 2017.
[68] S. Bahramikia and R. Yazdanparast, “phytochemistry and
medicinal properties of Teucrium polium L. (Lamiaceae),”
Phytotherapy Research, vol. 26, no. 11, pp. 1581–1593, 2012.
[69] H. M. Abdallah, A. B. Abdel-Naimc, O. M. Ashourd,
I. A. Shehata, and E. A. Abdel-Sattar, “Anti-infl ammatory
activity of selected plants from Saudi Arabia Z,” Natur-
forscher, vol. 69c, pp. 1–9, 2014.
[70] R. Vijayaraj and R. Vidhya, “Biological activity of Achyr-
anthes aspera linn.—a review,” Asian Journal of Biochemical
and Pharmaceutical Research, vol. 69, no. 1, pp. 2231–2560,
2016.
[71] M. A. Rahman, S. M. Jaber, M. S. Al-Said, and M. A. Al-
Yahya, “Medicinal plant diversity in the flora of Saudi Arabia
1: a report on seven plant families,” Fitoterapia, vol. 75,
pp. 149–161, 2004.
[72] K. S. Omotoso, F. R. Aigbe, O. A. Salako, M. C. Chijioke, and
O. O. Adeyem, “Toxicological evaluation of the aqueous
whole plant extract of Aerva lanata (l.) Juss. ex Schult
(Amaranthaceae),” Journal of Ethnopharmacology, vol. 208,
pp. 174–184, 2017.
[73] R. Rajesh, K. Chitra, and P. M. Paarakh, “Anti hyperglycemic
and antihyperlipidemic activity of aerial parts of Aerva lanata
Linn Juss in streptozotocin induced diabetic rats,” Asian
Pacific Journal of Tropical Biomedicine, vol. 2, no. 2,
pp. S924–S929, 2012.
[74] W. M. El-Ghanim, L. M. Hassan, T. M. Galal, and A. Badr,
“Floristic composition and vegetation analysis in Hail region
north of central Saudi Arabia,” Saudi Journal of Biological
Sciences, vol. 17, no. 2, pp. 119–128, 2010.
[75] A. I. Hossain, M. Faisal, S. Rahman, R. Jahan, and
M. Rahmatullah, “A preliminary evaluation of
18 Evidence-Based Complementary and Alternative Medicine
antihyperglycemic and analgesic activity of Alternanthera
sessilis aerial parts,” BMC Complementary and Alternative
Medicine, vol. 14, no. 169, pp. 1–5, 2014.
[76] H. Al-Youssef and W. Hassan, “Chemical constituents of
carissa edulis vahl,” Arabian Journal of Chemistry, vol. 10,
pp. 109–113, 2017.
[77] J. S. Kaunda and Y.-J. Zhang, “e genus carissa: an eth-
nopharmacological, phytochemical and pharmacological
review,” Natural Products and Bioprospecting, vol. 7,
pp. 181–199, 2017.
[78] E. K. F. Elbeshehy, H. S. M. AL-Zahrani, A. Y. Aldhebiani,
and T. Elbeaino, “Viruses infecting periwinkle (catharanthus
roseus L.) in western Saudi Arabia phytopathologia medi-
terranea,” vol. 56, no. 3, pp. 479–485, 2017.
[79] W. M. Al-Shaqha, M. Khan, N. Salam, A. Azzi, and
A. A. Chaudhary, “Anti-diabetic potential of Catharanthus
roseus Linn. and its effect on the glucose transport gene
(GLUT-2 and GLUT-4) in streptozotocin induced diabetic
wistar rats,” BMC Complementary and Alternative Medicine,
vol. 15, no. 379, pp. 1–8, 2015.
[80] N. A. Bukhari, R. A. Al-Otaibi, and M. M. Ibhrahim,
“Phytochemical and taxonomic evaluation of Rhazya stricta
in Saudi Arabia,” Saudi Journal of Biological Sciences, vol. 24,
no. 7, pp. 1513–1521, 2017.
[81] N. Menon, J. Sparks, and F. Omoruyi, “Hypoglycemic and
hypocholesterolemic activities of the aqueous preparation of
Kalanchoe pinnata leaves in streptozotocin-induced diabetic
rats,” Asian Pacific Journal of Tropical Biomedicine, vol. 5,
no. 1, pp. 3–9, 2015.
[82] A. A. Awaad, H. F. Alkanhal, R. M. El-Meligy et al., “Anti-
ulcerative colitis activity of Calotropis procera Linn,” Saudi
Pharmaceutical Journal, vol. 26, no. 1, pp. 75–78, 2018.
[83] K. Bhagour, D. Arya, and R. S. Gupta, “A review: anti-
hyperglycemic plant medicines in management of diabetes,”
Acupuncture and Related erapies, vol. 4, no. 4, pp. 7–16,
2016.
[84] A. H. Alfarhan, T. A. Al-Turki and A. Y. Basahy, Flora of
jizan region,” Final Report Volume-1, 2005.
[85] C. Sharma, S. Rani, B. Kumar, A. Kumar, and V. Raj, “Plant
opuntia dillenii: a review on its traditional uses, phyto-
chemical and pharmacological properties,” EC Pharma-
ceutical Science, vol. 1, no. 1, pp. 29–43, 2015.
[86] S. H. Hwang, I. J. Kang, and S. S. Lim, “Antidiabetic effect of
fresh nopal (Opuntia ficus-indica) in low-dose streptozo-
tocin-induced diabetic rats fed a high-fat diet,” Evidence-
based Complementary and Alternative Medicine, vol. 2017,
Article ID 4380721, 8 pages, 2017.
[87] K. El-Mostafa, Y. El Kharrassi, A. Badreddine et al., “Nopal
cactus (Opuntia ficus-indica) as a source of bioactive
compounds for nutrition, health and disease,” Molecules,
vol. 19, no. 9, pp. 14879–14901, 2014.
[88] T. Gull, F. Anwar, B. Sultana, M. A. C. Alcayde, and
W. Nouman, “Capparis species: a potential source of bio-
actives and high-value components: a review,” Industrial
Crops and Products, vol. 67, pp. 81–96, 2015.
[89] A. K. Al-Asmari, A. M. Al-Elaiwi, M. T. Athar, M. Tariq,
A. Al Eid, and S. M. Al-Asmary, “A review of hep-
atoprotective plants used in Saudi traditional medicine,”
Evidence-based Complementary and Alternative Medicine,
vol. 2014, Article ID 890842, 22 pages, 2014.
[90] A. U. Kabir, M. B. Samad, A. Ahmed et al., “Aqueous fraction
of Beta vulgaris ameliorates hyperglycemia in diabetic mice
due to enhanced glucose stimulated insulin secretion, me-
diated by acetylcholine and GLP-1, and elevated glucose
uptake via increased membrane bound GLUT4 trans-
porters,” PLoS One, vol. 10, no. 2, Article ID e0116546, 2015.
[91] S. Gezginci-Oktayoglu, O. Sacan, S. Bolkent et al., “Chard
(Beta vulgaris L. var. cicla) extract ameliorates hyperglyce-
mia by increasing GLUT2 through Akt2 and antioxidant
defense in the liver of rats,” Acta Histochemica, vol. 116,
no. 1, pp. 32–39, 2014.
[92] R. Ullah, M. S. Alsaid, A. A. Shahat et al., “Antioxidant and
hepatoprotective effects of methanolic extracts of Zilla spi-
nosa and Hammada elegans against carbon tetra-
chlorideinduced hepatotoxicity in rats,” Open Chemistry,
vol. 16, no. 1, pp. 133–140, 2018.
[93] J. P. Singh, V. S. Rathore, and M. M. Roy, “Notes about
Haloxylon salicornicum (Moq.) Bunge ex Boiss., a promising
shrub for arid regions,” Genetic Resources and Crop Evo-
lution, vol. 62, no. 3, pp. 451–463, 2015.
[94] G. Duraisamy, R. Ganesan, K. Manokaran,
D. Kanakasabapathi, and U. Chandrasekar, “Protective effect
of the whole plant extract of Evolvulus alsinoides on gly-
coprotein alterations in streptozotocin induced diabetic
rats,” Journal of Acute Disease, vol. 2, no. 2, pp. 148–150,
2013.
[95] P. Sundaramurthi and K. K. Packiam, “A review on phar-
macognosy and pharmacology of Evolvulus alsinoides (l.) L,”
International Research Journal of Pharmacy, vol. 8, no. 7,
pp. 1–4, 2017.
[96] Y. M. Al-Sodany, “A new record to the flora of Saudi Arabia:
Ipomoea carneaJacq., Convolvulaceae,” World Journal of
Research and Review, vol. 3, no. 4, pp. 25–30, 2016.
[97] A. A. Sajak, A. Mediani, N. S. Dom et al., “Effect of Ipomoea
aquatica ethanolic extract in streptozotocin (STZ) induced
diabetic rats via1H NMR-based metabolomics approach,”
Phytomedicine, vol. 36, pp. 201–209, 2017.
[98] K. Hamid, M. O. Ullah, S. Sultana et al., “Evaluation of the
leaves of Ipomoea aquatica for its hypoglycemic and anti-
oxidant activity,” Journal of Pharmaceutical Sciences and
Research, vol. 3, no. 7, pp. 1330–1333, 2011.
[99] A. Amin, M. Tahir, and K. P. Lone, “Effect of Citrullus
colocynthis aqueous seed extract on beta cell regeneration
and intra-islet vasculature in alloxan induced diabetic male
albino rats,” Journal of Pakistan Medical Association, vol. 67,
no. 5, pp. 715–721, 2017.
[100] Y. S. Masrahi, M. Remesh, and O. H. Sayed, “Enumeration of
the flora of wadi lajab Saudi Arabia,” Ecology Environment
and Conservation, vol. 23, no. 1, pp. 115–123, 2017.
[101] F. Wang, H. Li, H. Zhao et al., “Antidiabetic activity and
chemical composition of sanbai melon seed oil,” Evidence-
based Complementary and Alternative Medicine, vol. 2018,
Article ID 5434156, 14 pages, 2018.
[102] S. Varghese, R. Narmadha, D. Gomathi, M. Kalaiselvi, and
K. Devaki, “Evaluation of hypoglycemic effect of ethanolic
seed extracts of Citrullus lanatus,” Journal of Phyto-
pharmacology, vol. 2, pp. 31–40, 2013.
[103] A. P. Attanayake, K. A. Jayatilaka, C. Pathirana, and
L. K. Mudduwa, “Antihyperglycemic activity of Coccinia
grandis (L.) Voigt in streptozotocin induced diabetic rats,”
Indian journal of traditonal knowledge, vol. 14, no. 3,
pp. 376–381, 2015.
[104] H. A. Abdelgadir and J. Van Staden, “Ethnobotany, eth-
nopharmacology and toxicity of Jatropha curcas
L.(Euphorbiaceae): a review,” South African Journal of
Botany, vol. 88, pp. 204–218, 2013.
[105] M. A. Gad-Elkareem, E. H. Abdelgadir, O. M. Badawy, and
A. Kadri, “Potential antidiabetic effect of ethanolic and
Evidence-Based Complementary and Alternative Medicine 19
aqueous-ethanolic extracts of Ricinus communis leaves on
streptozotocin-induced diabetes in rats,” PeerJ, vol. 7,
pp. 1–15, Article ID e6441, 2019.
[106] K. Y. Khan, M. A. Khan, M. Ahmad et al., “Hypoglycemic
potential of genus Ficus L.: a review of ten years of plant
based medicine used to cure diabetes (2000–2010),” Journal
of Applied Pharmaceutical Science, vol. 1, no. 6, pp. 223–227,
2011.
[107] O. A. Adoum, B. O. Micheal, and I. S. Mohammad, “Phy-
tochemicals and hypoglycaemic effect of methanol stem-
bark extract of Ficus sycomorus Linn (Moraceae) on alloxan
induced diabetic Wistar albino rats,” African Journal of
Biotechnology, vol. 11, no. 17, pp. 4095–4097, 2012.
[108] T. A. Ibrahiem, “Beneficial effects of diet supplementation
with Nigella sativa (Black Seed) and sesame seeds in Alloxan-
Diabetic Rats,” International Journal of Current Microbiology
and Applied Sciences, vol. 5, pp. 411–423, 2016.
[109] R. Diez, J. J. Garc´
ıa, M. J. Diez, M. Sierra, A. M. Sahagun, and
N. Fernandez, “Influence of Plantago ovata husk (dietary
fiber) on the bioavailability and other pharmacokinetic
parameters of metformin in diabetic rabbits,” BMC Com-
plementary and Alternative Medicine, vol. 17, no. 1, p. 298,
2017.
[110] G. Sammaiah and R. S. Srivastava, “Hypoglycemic activity of
Polygala erioptera (Whole plant) in normal and alloxan
induced diabetic rats,” Asian Journal of Chemistry, vol. 20,
no. 1, pp. 107–112, 2008.
[111] H. M. Al-Hazimi and S. N. Haque, “A new naphthoquinone
from Polygonum aviculare,” Natural Product Letters, vol. 16,
no. 2, pp. 115–118, 2002.
[112] Y. M. Al-Sodany, A. B. Salih, and H. A. Mosallam, “Me-
dicinal plants in Saudi Arabia: I. Sarrwat mountains at taif,
KSA,” Academic Journal of Plant Sciences, vol. 6, pp. 134–
145, 2013.
[113] B. Baharvand-Ahmadi, M. Bahmani, P. Tajeddini,
N. Naghdi, and M. Rafieian-Kopaei, “An ethno-medicinal
study of medicinal plants used for the treatment of diabetes,”
Journal of nephropathology, vol. 5, no. 1, pp. 44–50, 2015.
[114] M. Bahmani, A. Zargaran, M. Rafieian-Kopaei, and K. Saki,
“Ethnobotanical study of medicinal plants used in the
management of diabetes mellitus in the Urmia, Northwest
Iran,” Asian Pacific journal of tropical medicine, vol. 7,
pp. S348–S354, 2014.
[115] A. A. Al-Ghamdi and A. A. Shahat, “Antioxidant, hypo-
glycemic and anti-diabetic activities of Ziziphus spina-christi
(L) Willd (Rhamnacae) leaf extract,” Tropical Journal of
Pharmaceutical Research, vol. 16, no.11, pp. 2601–2610, 2017.
[116] T. Inin, M. Mohsina, and R. Mohammed, “Bacopa monnieri:
an evaluation of antihyperglycemic and antinociceptive
potential of methanolic extract of whole plants,” Pakistan
journal of pharmaceutical sciences, vol. 28, no. 6, pp. 2135–
2139, 2015.
[117] H. Sher and M. N. Alyemeni, “Evaluation of anti-diabetic
activity and toxic potential of Lycium shawii in animal
models,” Journal of Medicinal Plants Research, vol. 5, no. 15,
pp. 3387–3395, 2011.
[118] M. S. Umamageswari, T. M. Karthikeyan, and Y. A. Maniyar,
“Antidiabetic activity of aqueous extract of Solanum nigrum
linn berries in alloxan induced diabetic wistar albino rats,”
Journal of Clinical and Diagnostic Research: Journal of
Clinical and Diagnostic Research, vol. 11, no. 7, pp. FC16–
FC19, 2017.
[119] T. Anwer, M. Sharma, K. K. Pillai, and M. Iqbal, “Effect of
withania somnifera on insulin sensitivity in non-insulin-
dependent diabetes mellitus rats,” Basic and Clinical Phar-
macology and Toxicology, vol. 102, no. 6, pp. 498–503, 2008.
[120] R. Udayakumar, S. Kasthurirengan, T. S. Mariashibu et al.,
“Hypoglycaemic and hypolipidaemic effects of Withania
somnifera root and leaf extracts on alloxan-induced diabetic
rats,” International Journal of Molecular Sciences, vol. 10,
no. 5, pp. 2367–2382, 2009.
[121] M. Saxena, J. Saxena, and S. Khare, “A brief review on:
therapeutical values of Lantana camara plant,” International
Journal of Pharmacy & Life Sciences, vol. 3, no. 3,
pp. 1551–1554, 2012.
[122] G. Komeili, M. Hashemi, and M. Bameri-Niafar, “Evaluation
of antidiabetic and antihyperlipidemic effects of peganum
harmala seeds in diabetic rats,” Cholesterol, vol. 2016, Article
ID 7389864, 6 pages, 2016.
[123] S. Chhatre, T. Nesari, G. Somani, D. Kanchan, and
S. Sathaye, “Phytopharmacological overview of Tribulus
terrestris,” Pharmacognosy Reviews, vol. 8, no. 15, p. 45, 2014.
[124] A. El-Shaibany, A. H. Molham, B. Al-Tahami, and S. Al-
Massarani, “Anti-hyperglycaemic activity of Tribulus ter-
restris L aerial part extract in glucose-loaded normal rab-
bits,” Tropical Journal of Pharmaceutical Research, vol. 14,
no. 12, pp. 2263–2268, 2015.
[125] A. A. Shahat, A. Y. Ibrahim, E. Ezzeldin, and M. S. Alsaid,
“Acetylcholinesterase inhibition and antioxidant activity of
some medicinal plants for treating neuro degenarative dis-
ease,” African Journal of Traditional, Complementary and
Alternative Medicines, vol. 12, no. 3, pp. 97–103, 2015.
[126] A. Ahangarpour, M. Mohammadian, and M. Dianat, “An-
tidiabetic effect of hydroalcholic Urtica dioica leaf extract in
male rats with fructose-induced insulin resistance,” Iranian
Journal of Medical Sciences, vol. 37, no. 3, pp. 181–186, 2012.
[127] S. A. Dar, F. A. Ganai, A. R. Yousuf, M. U. Balkhi, T. M. Bhat,
and P. Sharma, “Pharmacological and toxicological evalu-
ation of Urtica dioica,” Pharmaceutical Biology, vol. 51, no. 2,
pp. 170–180, 2013.
[128] M. Marrelli, V. Amodeo, G. Statti, and F. Conforti, “Bio-
logical properties and bioactive components of Allium cepa
L.: focus on potential benefits in the treatment of obesity and
related comorbidities,” Molecules, vol. 24, no. 119, pp. 1–18,
2019.
[129] O. BashirLawal, K. Shittu, F. I. Oibiokpa, H. Mohammed,
S. I. Umar, and G. M. Haruna, “Antimicrobial evaluation,
acute and sub-acute toxicity studies of Allium sativum,”
Journal of Acute Disease, vol. 5, no. 4, pp. 296–301, 2016.
[130] A. Moufid and M. Eddouks, “Artemisia herba alba: A
popular plant with potential medicinal propertie,” Pakistan
Journal of Biological Sciences, vol. 15, no. 24, pp. 1152–1159,
2012.
[131] G. Gani, T. Fatima, T. Qadri, N. J. Beenish, and O. Bashir,
“Phytochemistry and pharmacological activities of fig (Ficus
carica),” A review nternational Journal of Research in
Pharmacy and Pharmaceutical Sciences, vol. 3, no. 2,
pp. 80–82, 2018.
[132] G. E. Odo, J. E. Agwu, N. Newze et al., “Toxicity and effects of
fig (Ficus carica) leaf aqueous extract on haematology and
some biochemical indices of wistar albino rats (Rattus
norvegicus),” Journal of Medicinal Plants Research, vol. 10,
no. 22, pp. 298–305, 2016.
[133] A. I. Oliveira, C. Pinho, B. Sarmento, and A. C. P. Dias,
“Neuroprotective activity of Hypericum perforatum and its
major components,” Frontiers of Plant Science, vol. 7, p. 1004,
2016.
20 Evidence-Based Complementary and Alternative Medicine
[134] S. Jana and G. S. Shekhawat, “Anethum graveolens: an In-
dian traditional medicinal herb and spice,” Pharmacognosy
Reviews, vol. 4, no. 8, pp. 179–184, 2010.
[135] N. D. Swati and K. P. Dipak, “Acute and sub chronic tox-
icoloigcal evaluation of anethum graveolens seed extract,”
Journal of Global bioseciences, vol. 5, no. 6, pp. 4213–4220,
2016.
[136] M. H. Farzaei, R. Bahramsoltani, G. Ali, and F. Farzaei,
“Fariba najafi pharmacological activity of mentha longifolia
and its phytoconstituents,” Journal of Traditional Chinese
Medicine, vol. 37, no. 5, pp. 710–720, 2017.
[137] B. Lukas, C. Schmiderer, C. Franz, and J. Novak, “Com-
position of essential oil compounds from different Syrian
populations of origanum syriacum L. (Lamiaceae),” Journal
of Agricultural and Food Chemistry, vol. 57, pp. 1362–1365,
2009.
[138] H. R. Rasekh, H. Yazdanpanah, and L. Hosseinzadeh,
“Narges bazmohammadi and mohammad kamalinejad acute
and subchronic toxicity of Teucrium polium total extract in
rats,” Iranian Journal of Pharmaceutical Research, vol. 4,
pp. 245–249, 2005.
[139] C. C. Barua, A. Talukdar, S. Ara Begum et al., “In vivo
wound-healing efficacy and antioxidant activity of Achyr-
anthes aspera in experimental burns,” Pharmaceutical Bi-
ology, vol. 50, no. 7, pp. 892–899, 2012.
[140] B. M. Dinnimath, S. S. Jalalpure, and U. K. Patil, “Anti-
urolithiatic activity of natural constituents isolated from
Aerva lanata,” Journal of Ayurveda and Integrative Medicine,
vol. 8, pp. 226–233, 2017.
[141] R. Sivakumar and D. Sunmathi, “Phytochemical screening
and antimicrobial activity of ethanolic leaf extract of alter-
nanthera sessilis (l.) r.br. ex dc and alternanthera philox-
eroides (MART.) griseb,” European Journal of
Pharmaceutical and Medical Research, vol. 3, no. 3,
pp. 409–412, 2016.
[142] J. Ya’u, B. A. Chindo, A. H. Yaro, S. E. Okhale, J. A. Anuka,
and I. M. Hussaini, “Safety assessment of the standardized
extract of Carissa edulis root bark in rats,” Journal of Eth-
nopharmacology, vol. 147, no. 3, pp. 653–661, 2013.
[143] S. Das and A. B. Sharangi, “Madagascar periwinkle
(Catharanthus roseus L.): diverse medicinal and therapeutic
benefits to humankind,” Journal of Pharmacognosy and
Phytochemistry, vol. 6, no. 5, pp. 1695–1701, 2017.
[144] V. R. Vutukuri, M. C. Das, M. Reddy, S. Prabodh, and
P. Sunethri, “Evaluation of acute oral toxicity of ethanol
leaves extract of Catharanthus roseus in wistar albino rats,”
Journal of Clinical and Diagnostic Research, vol. 11, no. 3,
pp. FF01–FF04, 2017.
[145] S. A. Gilani, A. Kikuchi, Z. Khan Shinwari, Z. I. Khattak, and
N. Kazuo, “Watanabe phytochemical, pharmacological and
ethnobotanical studies of rhazya stricta decne,” Phytotherapy
Research, vol. 21, pp. 301–307, 2007.
[146] G. Parihar and N. Balekar, “Calotropis procera: a phyto-
chemical and pharmacological review,” ai Journal of
Pharmaceutical Sciences, vol. 40, no. 3, 2016.
[147] G. G. Ouedraogo, M. Ouedraogo, A. Lamien-Sanou,
M. Lompo, O. M. Goumbri-Lompo, and P. I. Guissou,
“Acute and subchronic toxicity studies of roots barks ex-
tracts of “calotropis procera” (ait.) R. Br used in the treat-
ment of sickle cell disease in Burkina Faso,” British Journal of
Pharmacology and Toxicology, vol. 4, no. 5, pp. 194–200,
2013.
[148] E. H. Han, M. K. Lim, S. H. Lee, M. M. Rahman, and
Y. H. Lim, “An oral toxicity test in rats and a genotoxicity
study of extracts from the stems of Opuntia ficus-indica var.
saboten,” BMC Complementary and Alternative Medicine,
vol. 19, no. 1, p. 31, 2019.
[149] P. K. Dhakad, P. K. Sharma, and S. Kumar, “Effect of
hydroalcoholic extract of Capparis decidua (forssk.) edgew
on sexual behavior of male rats,” Advances in Pharmacology
and Pharmacy, vol. 6, no. 2, pp. 50–56, 2018.
[150] N. K. Jain and A. K. Singhai, “Protective role of Beta vulgaris
L. leaves extract and fractions on ethanol-mediated hepatic
toxicity,” Acta Poloniae Pharmaceutica, vol. 69, no. 5,
pp. 945–950, 2012.
[151] T. R. Chander and Y. N. Reddy, “Evaluation of hep-
atoprotective and antihepatotoxic activity of ethanolic ex-
tract of evolvulus alsinoides linn on CCL4 induced rats,”
Asian Journal of Pharmacology and Toxicology, vol. 2, no. 4,
pp. 1–6, 2014.
[152] S. S. Alkiyumi, M. A. Abdullah, A. S. Alrashdi, S. M. Salama,
S. I. Abdelwahab, and A. H. Hadi, “Ipomoea aquatica extract
shows protective action against thioacetamide-induced
hepatotoxicity,” Molecules, vol. 17, no. 5, pp. 6146–6155,
2012.
[153] M. J. Khan, V. Saini, V. S. Bhati, M. S. Karchuli, and
M. B. Kasturi, “Anxiolytic activity of Ipomoea aquatica
leaves,” European Journal of Experimental Biology, vol. 1,
pp. 63–70, 2011.
[154] A. I. Hussain, H. A. Rathore, M. Z. Sattar, S. A. Chatha,
S. D. Sarker, and A. H. Gilani, “Citrullus colocynthis (L.)
Schrad (bitter apple fruit): a review of its phytochemistry,
pharmacology, traditional uses and nutritional potential,”
Journal of Ethnopharmacology, vol. 155, no. 1, pp. 54–66,
2014.
[155] R. S. Rajasree, P. I. Sibi, F. Francis, and H. William, “Phy-
tochemicals of Cucurbitaceae family—a review,” Interna-
tional Journal of Pharmacognosy and Phytochemical
Research, vol. 8, pp. 113–123, 2016.
[156] C. D. Deshmukh, A. Jain, and M. S. Tambe, “Phytochemical
and pharmacological profile of Citrullus lanatus (THUNB),”
e Biologist, vol. 3, no. 2, pp. 483–488, 2015.
[157] S. Pekamwar, T. Kalyankar, and S. Kokate, “Pharmacological
activities of Coccinia grandis,” Journal of Applied Pharma-
ceutical Science, vol. 3, pp. 114–119, 2013.
[158] A. P. Attanayake, K. A. Jayatilaka, C. Pathirana, and
L. K. Mudduwa, “Efficacy and toxicological evaluation of
Coccinia grandis (Cucurbitaceae) extract in male Wistar
rats,” Asian Pacific Journal of Tropical Disease, vol. 3, no. 6,
pp. 460–466, 2013.
[159] S. Khan Marwat, E. A. Khan, M. S. Baloch et al., “Ricinus
cmmunis: ethnomedicinal uses and pharmacological activ-
ities,” Pakistan Journal of Pharmaceutical Sciences, vol. 30,
no. 5, pp. 1815–1827, 2017.
[160] W. M. Abdul, N. H. Hajrah, J. S. Sabir et al., “erapeutic role
of Ricinus communis L. and its bioactive compounds in
disease prevention and treatment,” Asian Pacific Journal of
Tropical Medicine, vol. 11, no. 3, pp. 177–185, 2018.
[161] S. Mawa, K. Husain, and I. Jantan, “Ficus carica L.(Mor-
aceae): phytochemistry, traditional uses and biological ac-
tivities,” Evidence-based Complementary and Alternative
Medicine, vol. 2013, Article ID 974256, 8 pages, 2013.
[162] S. Allahyari, A. Delazar, and M. Najafi, “Evaluation of
general toxicity, anti-oxidant activity and effects of Ficus
carica leaves extract on ischemia/reperfusion injuries in
isolated heart of rat,” Advanced Pharmaceutical Bulletin,
vol. 4, no. 2, pp. 577–582, 2014.
Evidence-Based Complementary and Alternative Medicine 21
[163] H. Raeisi-Dehkordi, M. Mohammadi, F. Moghtaderi, and
A. Salehi-Abargouei, “Do sesame seed and its products affect
body weight and composition? A systematic review and
meta-analysis of controlled clinical trials,” Journal of
Functional Foods, vol. 49, pp. 324–332, 2018.
[164] S. Miraj and S. Kiani, “Bioactivity of Sesamum indicum: a
review study,” Der Pharmacia Lettre, vol. 8, no. 6,
pp. 328–334, 2016.
[165] A. Ruckmani, V. Meti, R. Vijayashree et al., “Anti-rheu-
matoid activity of ethanolic extract of Sesamum indicum
seed extract in Freund’s complete adjuvant induced arthritis
in Wistar albino rats,” Journal of Traditional and Comple-
mentary Medicine, vol. 8, no. 3, pp. 377–386, 2018.
[166] D. Tewari and N. Anjum, “Tripathi phytochemistry and
pharmacology of plantago ovata: a natural source of laxative
medicine,” World Journal of Pharmaceutical Research, vol. 3,
no. 9, pp. 361–372, 2014.
[167] K. Haddadian, K. Haddadian, and M. Zahmatkash, “A re-
view of Plantago plant,” Indian Journal of Tradational
knowledge, vol. 13, no. 4, pp. 681–685, 2014.
[168] G. Sammaiah, N. Narsaiah, G. Dayakar et al., “Constituents
and cytotoxicity of polygala erioptera,” International Journal
of Chemical Sciences, vol. 6, no. 2, pp. 643–646, 2008.
[169] H. Shin, H. Chung, B. Park, and K. Y. Lee, “Identification of
antioxidative constituents from Polygonum aviculare using
LC-MS coupled with DPPH assay,” Natural Product Sciences,
vol. 22, no. 1, pp. 64–69, 2016.
[170] B. B. Shen, Y. P. Yang, S. Yasamin et al., “Analysis of the
phytochemistry and bioactivity of the genus polygonum of
polygonaceae,” Digital Chinese Medicine, vol. 1, no. 1,
pp. 19–36, 2018.
[171] A. Bozicevic, M. De Mieri, A. Di Benedetto, F. Gafner, and
M. Hamburger, “Dammarane-type saponins from leaves of
Ziziphus spina-christi,” Phytochemistry, vol. 138, pp. 134–
144, 2017.
[172] M. E. Karar, L. Quiet, A. Rezk et al., “Phenolic profile and in
vitro assessment of cytotoxicity and antibacterial activity of
Ziziphus spina-christi Leaf Extracts,” Medicinal Chemistry,
vol. 6, no. 3, pp. 143–156, 2016.
[173] A. O. Abdel-Zaher, S. Y. Salim, M. H. Assaf, and R. H. Abdel-
Hady, “Antidiabetic activity and toxicity of Zizyphus spina-
christi leaves,” Journal of Ethnopharmacology, vol. 101,
no. 1–3, pp. 129–138, 2005.
[174] S. Sireeratawong, S. Vannasiri, U. Nanna, T. Singhalak, and
K. Jaijoy, “Evaluation of acute and chronic toxicities of the
water extract from Ziziphus attopensis pierre,” ISRN phar-
macology, vol. 2012, Article ID 789651, 7 pages, 2012.
[175] A. E. Al-Snafi, “e pharmacology of Bacopa monniera. A
review,” International Journal of Pharma Sciences and Re-
search, vol. 4, no. 12, pp. 154–159, 2013.
[176] S. Sireeratawong, K. Jaijoy, P. Khonsung, N. Lertprasertsuk,
and K. Ingkaninan, “Acute and chronic toxicities of Bacopa
monnieri extract in Sprague-Dawley rats,” BMC Comple-
mentary and Alternative Medicine, vol. 16, no. 1, pp. 1–10,
2016.
[177] N. U. Rehman, H. Hussain, S. A. Al-Riyami, I. R. Green, and
A. Al-Harrasi, “Chemical constituents isolated from lycium
shawii and their chemotaxonomic significance,” Records of
Natural Products, vol. 12, no. 4, pp. 380–384, 2018.
[178] F. A. Ahmed, N. M. Abdallah, M. K. Ezz, and M. M. El-Azab,
“Seasonal variations and identification of biologically active
constituents of lycium shawii plant roem. & shult. (family
solanaceae),” International Journal of Innovative Science, Engi-
neering & Technology, vol. 4, no. 11, pp. 23487968, 2017.
[179] U. Saleem, S. Amin, B. Ahmad, H. Azeem, F. Anwar, and
S. Mary, “Acute oral toxicity evaluation of aqueous ethanolic
extract of Saccharum munja Roxb. roots in albino mice as
per OECD 425 TG,” Toxicology Reports, vol. 4, pp. 580–585,
2017.
[180] T. M. Saleem, C. M. Chetty, S. Ramkanth et al., “Solanum
nigrum Linn.—a review,” Pharmacognosy Reviews, vol. 3,
no. 6, pp. 342–345, 2009.
[181] A. Patel, S. Biswas, M. H. Shoja, G. V. Ramalingayya, and
K. Nandakumar, “Protective effects of aqueous extract of
Solanum nigrum Linn. leaves in rat models of oral muco-
sitis,” e Scientific World Journal, vol. 2014, Article ID
345939, 10 pages, 2014.
[182] M. Rai, P. S. Jogee, G. Agarkar, and C. A. Santos, “Anticancer
activities of Withania somnifera: current research, formu-
lations, and future perspectives,” Pharmaceutical Biology,
vol. 54, no. 2, pp. 189–197, 2016.
[183] S. B. Patel, N. J. Rao, and L. L. Hingorani, “Safety assessment
of Withania somnifera extract standardized for Withaferin
A: acute and sub-acute toxicity study,” Journal of Ayurveda
and Integrative Medicine, vol. 7, no. 1, pp. 30–37, 2016.
[184] R. Delgado-Altamirano, R. I. L´
opez-Palma, L. Monzote et al.,
“Chemical constituents with leishmanicidal activity from a
pink-yellow cultivar of Lantana camara var. aculeata (L.)
collected in Central Mexico,” International Journal of Mo-
lecular Sciences, vol. 20, no. 4, pp. 1–17, 2019.
[185] B. M. Pour, L. Y. Latha, and S. Sasidharan, “Cytotoxicity and
oral acute toxicity studies of Lantana camara leaf extract,”
Molecules, vol. 16, no. 5, pp. 3663–3674, 2011.
[186] I. Jawonisi and G. Adoga, “Acute toxicity, haematological
and histopathological assessment of the ethanolic extract of
Lantana camara leaf,” Journal of Pharmacy & Bioresources,
vol. 14, no. 1, pp. 45–50, 2017.
[187] M. T. Moradi, A. Karimi, F. Fotouhi, S. Kheiri, and A. Torabi,
“In vitro and in vivo effects of Peganum harmala L. seeds
extract against influenza A virus,” Avicenna Journal of
Phytomedicine, vol. 7, no. 6, pp. 519–530, 2017.
[188] X. Shang, X. Guo, B. Li et al., “Microwave-assisted extraction
of three bioactive alkaloids from Peganum harmala L. and
their acaricidal activity against Psoroptes cuniculi in vitro,”
Journal of Ethnopharmacology, vol. 192, pp. 350–361, 2016.
[189] A. Majid, “A review study of the chemical constituents and
therapeutic effects of Peganum harmala L,” Global Journal of
Pure and Applied Chemistry Research, vol. 6, no. 2, pp. 12–19,
2018.
[190] N. M. Ammar, S. S. EL-Hawary, D. A. Mohamed, M. S. Afifi,
D. M. Ghanem, and G. Awad, “Phytochemical and biological
studies of Tribulus terrestris L. growing in Egypt,” Inter-
national Journal of Pharmacology, vol. 14, no. 2, pp. 248–259,
2018.
[191] R. Ullah, I. Hussain, and S. Ahmad, “Diocanol; one new
phenol derivative isolated and characterized from Urtica
dioica,” Arabian Journal of Chemistry, vol. 10, pp. S1284–
S1286, 2017.
[192] C. Bourgeois, ´
E. A. Leclerc, C. Corbin et al., “Nettle (Urtica
dioica L.) as a source of antioxidant and anti-aging phyto-
chemicals for cosmetic applications,” Comptes Rendus
Chimie, vol. 19, no. 9, pp. 1090–1100, 2016.
[193] U. J. Vajic, J. Grujic-Milanovic, Z. Miloradovic et al., “Urtica
dioica L. leaf extract modulates blood pressure and oxidative
stress in spontaneously hypertensive rats,” Phytomedicine,
vol. 46, pp. 39–45, 2018.
[194] S. K. Bharti, S. Krishnan, A. Kumar, and A. Kumar, “An-
tidiabetic phytoconstituents and their mode of action on
22 Evidence-Based Complementary and Alternative Medicine
metabolic pathways,” erapeutic Advances in Endocrinology
and Metabolism, vol. 9, no. 3, pp. 81–100, 2018.
[195] R. Ponnulakshmi, B. Shyamaladevi, P. Vijayalakshmi, and
J. Selvaraj, “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 experi-
mental rats,” Toxicology Mechanisms and Methods, vol. 29,
no. 4, pp. 276–290, 2019.
[196] G. Arumugam, P. Manjula, and N. Paari, “A review: anti
diabetic medicinal plants used for diabetes mellitus,” Journal
of Acute Disease, vol. 2, no. 3, pp. 196–200, 2013.
[197] P. P. Jaya, “Herbal medicine for diabetes mellitus,” A Review
International Journal of Phytopharmacy, vol. 3, no. 1,
pp. 1–22, 2013.
[198] S. Jeeva and S. Y. Anlin, “A review of antidiabetic potential of
ethnomedicinal plants,” Medicinal & Aromatic Plants, vol. 3,
no. 4, pp. 1–8, 2014.
[199] H. Azaizeh, S. Fulder, K. Khalil, and O. Said, “Ethnobotanical
knowledge of local Arab practitioners in the Middle Eastern
region,” Fitoterapia, vol. 74, no. 1-2, pp. 98–108, 2003.
[200] G. E. El-Ghazali, K. S. Al-Khalifa, G. A. Saleem, and
E. M. Abdallah, “Traditional medicinal plants indigenous to
Al-Rass province, Saudi Arabia,” Journal of Medicinal Plants
Research, vol. 4, no. 24, pp. 2680–2683, 2010.
[201] M. F. Amal and M. M. Masarrat, “Migahid ethnobotanical
survey of plants used in the treatment of diabetes mellitus in
Tabuk region,” Saudi Arabia International Journal of Current
Microbiology and Applied Sciences, vol. 5, no. 6, pp. 258–270,
2016.
[202] M. S. Abdel-Kader, A. M. Hazazi, O. A. Elmakki, and
S. I. Alqasoumi, “A survey on traditional plants used in Al
Khobah village,” Saudi Pharmaceutical Journal, vol. 26, no. 6,
pp. 817–821, 2018.
[203] N. A. Ali, S. S. Al Sokari, A. Gushash, S. Anwar, K. Al-Karani,
and A. Al-Khulaidi, “Ethnopharmacological survey of me-
dicinal plants in Albaha region, Saudi Arabia,” Pharma-
cognosy Research, vol. 9, no. 4, p. 401, 2017.
[204] R. J. Marlesa and N. R. Farnsworth, “Antidiabetic plants and
their active constituents,” Phytomedicine, vol. 2, no. 2,
pp. 137–189, 1995.
[205] S. Hasani-Ranjbar, B. Larijani, and M. Abdollahi, “A sys-
tematic review of Iranian medicinal plants useful in diabetes
mellitus,” Archives of Medical Science, vol. 4, no. 3,
pp. 285–292, 2008.
[206] G. B. Kavishankar, N. Lakshmidevi, S. M. Murthy,
H. S. Prakash, and S. R. Niranjana, “Diabetes and medicinal
plants—a review,” International Journal of Pharma Bio
Sciences, vol. 2, no. 3, pp. 65–80, 2011.
[207] S. L. Chen, H. Yu, H. M. Luo, Q. Wu, C. F. Li, and
A. Steinmetz, “Conservation and sustainable use of me-
dicinal plants: problems, progress, and prospects,” Chinese
Medicine, vol. 11, p. 37, 2016.
[208] E. Robertson, Medicinal Plants at Risk. Nature’s Pharmacy,
Our Treasure Chest: Why We Must Conserve Our Natural
Heritage, Cent. Biol. Divers., Tucson, AZ, USA, 2008.
[209] K. Annan, R. A. Dickson, I. K. Amponsah, and I. K. Nooni,
“e heavy metal contents of some selected medicinal plants
sampled from different geographical locations,” Pharma-
cognosy Research, vol. 5, no. 2, p. 103, 2013.
[210] M. Bedassa, A. Abebaw, and T. Desalegn, “Assessment of
selected heavy metals in onion bulb and onion leaf (Allium
cepa L.), in selected areas of central rift valley of Oromia
region Ethiopia,” Journal of Horticulture and Forestry, vol. 4,
p. 217, 2017.
Evidence-Based Complementary and Alternative Medicine 23
... Te World Health Organization reports that various plant fractions and their dynamic constituents are utilized as traditional medicines by 80% of the world population. Te medicinal potential of plant species is because of the occurrence of secondary phytoconstituents, which have numerous functions, such as antioxidant, antimicrobial, cytotoxic, anticancer, and antiviral [14,15]. ...
Article
Full-text available
Background. Helicobacter pylori are the principal causative factor in the etiological factors of chronic, active, or type B gastritis; peptic and duodenal ulcers; stomach carcinoma; and epithelial tissue lymphoid malignancies. It infects more than half of the population worldwide. To reduce H. pylori production, pharmacological therapy of H. pylori diseases typically involves using threefold treatment methods. However, as a result of such therapy, antimicrobial resistance is commonly developed. Alternative therapeutics for H. pylori diseases are thus of particular interest. Methods. Thyme essential oils (EOs) obtained from T. caramanicus Jalas plants in Iran were tested for antibacterial activity against H. pylori obtained from 320 poultry specimens in this investigation. Antibacterial activity was measured using inhibition zones, minimum inhibitory concentrations (MICs), and minimum bactericidal concentrations (MBCs). The impact of T. caramanicus Jalas essential oils on H. pylori isolate cagA, vacA, and babA2 gene expression was evaluated using a quantitative real-time PCR method p<0.05. Results. The chemical content of these EOs varied significantly according to chromatographic examination. Thymol, carvacrol, and terpinene-4-ol are the most abundant components in these EOs. H. pylori was recognized as a Helicobacter species with a 175-bp PCR product of 16S rRNA in 20/20 (100%). According to PCR results, all 20 (100%) isolates belonged to H. pylori. The EOs inhibited H. pylori in a dose-dependent manner, with T. caramanicus Jalas being the most effective, followed by pterygium EOs in decreasing order. At 8 mg/mL of T. caramanicus Jalas EOs, IZs against H. pylori were 27.4 ± 0.42 mm, and at 8 mg/mL of pterygium, IZs against H. pylori were 1 ± 0.02. T. caramanicus Jalas essential oils were used to treat all bacteria, and the findings showed that T. caramanicus Jalas had a substantial inhibitory impact on the expression of cagA, vacA, and babA2 virulence-related genes p<0.05. Conclusions. In a dose-dependent manner, the EOs of T. caramanicus Jalas EO demonstrated a high degree of antimicrobial property against H. pylori bacteria. The most efficient EOs were those from T. caramanicus Jalas with relative concentrations of thymol and carvacrol, followed by the coumarin-dominated pterygium EO with reduced antibacterial activity.
... For example, most people in Saudi Arabia used medicinal plants and also preferred their use in the management of diabetes and obesity [9][10][11][12][13]. In addition, the reports listed several medicinal plants used in the treatment of diabetes all over the world [14][15][16][17]. Several species of antidiabetic plants have been phytochemically, biologically, and clinically investigated in numerous articles [18][19][20]. ...
Article
Full-text available
Ajuga iva (L.) Schreb. is a well-known antidia-betic medicinal plant used for several traditional medicine aspects in different areas of the world, including Libya. This study includes phytochemical analysis, antidiabetic evaluation, and in silico studies of the plant, A. iva, growing in Libya. The constituents of the plant were profiled using LC-MS/MS-QTOF analysis, and a total of 28 compounds were tentatively identified, including engeletin, pyrocatechol, eriodyctiol-7-hexoside, and 3,4-dihydroxybenzaldehyde, as major constituents. In addition, the steroidal compounds, i.e., 20-hydroxyecdysone, 24-dehydroprecyasterone, makis-terone A, and ajugasterone D, which are considered chemo-markers for the plant, were also annotated by LC-MS analysis. The plant extract induced inhibition of α-amylase and α-glu-cosidase enzymes at IC 50 values of 0.18 and 0.12 mg/mL, compared to the IC 50 of the standard acarbose at 0.11 and 0.09 mg/mL, respectively. Fasting blood glucose (FBG, 360.7 mg/dL) levels were significantly reduced by the treatment of streptozotocin (STZ)-diabetic animals with 400 mg/kg (140.5 mg/dl) and 500 mg/kg (112.3 mg/dL) doses of the plant extract. The plant extract also induced a significant (p < 0.01) increase in insulin serum level compared to the untreated diabetic rats; however, the higher dose of the plant induced similar insulin induction compared to glibenclamide. Histopathological examination of the pancreatic and liver tissues indicated that A. iva extract induced regeneration in the islets of Langerhans and liver cells compared to the untreated diabetic rats. Docking analysis demonstrated that eriodyctiol-7-hexoside, echinacoside, and 2″-galloylhyperin showed the lowest binding energies to the target sites of α-amylase and α-glucosidase enzymes, indicating their potential role in A. iva antidiabetic bioactivities. The results support the recorded traditional bioactivity of A. iva as an antidiabetic herb, whereas its contents of polyphe-nols play a major role in the plant's antidiabetic effect.  Mosleh M. Abomughaid:
... While pharmacological therapy remains the primary choice, some medications can cause unwanted adverse effects [3]. Herbal treatments have been recently proposed as an alternative option for DM patients [4]. One of the herbal plants with a potential antidiabetic effect is Gynura procumbens (Lour.) ...
... These plants and their derivatives offer a wealth of potential therapeutic agents. Indeed, the World Health Organization (WHO) reports that around 80% of the global population relies on plant-based components and their active constituents for traditional remedies (Shahat et al. 2018;Alqahtani et al. 2022). Rhodiola rosea L. (Crassulaceae) (RR), also known as 'golden root' or 'roseroot' , is a traditional Chinese medicine (TCM) with various biological functions, such as antidiabetes, anticancer, anti-aging, antioxidation, anti-inflammation, and immune regulation (Tao et al. 2019;Pu et al. 2020 1080/13880209.2024.2319117 ...
Article
Full-text available
Context Sepsis-induced acute lung injury (ALI) is associated with high morbidity and mortality. Rhodiola rosea L. (Crassulaceae) (RR) and its extracts have shown anti-inflammatory, antioxidant, immunomodulatory, and lung-protective effects. Objective This study elucidates the molecular mechanisms of RR against sepsis-induced ALI. Materials and methods The pivotal targets of RR against sepsis-induced ALI and underlying mechanisms were revealed by network pharmacology and molecular docking. Human umbilical vein endothelial cells (HUVECs) were stimulated by 1 μg/mL lipopolysaccharide for 0.5 h and treated with 6.3, 12.5, 25, 50, 100, and 200 μg/mL RR for 24 h. Then, the lipopolysaccharide-stimulated HUVECs were subjected to cell counting kit-8 (CCK-8), enzyme-linked immunosorbent, apoptosis, and Western blot analyses. C57BL/6 mice were divided into sham, model, low-dose (40 mg/kg), mid-dose (80 mg/kg), and high-dose (160 mg/kg) RR groups. The mouse model was constructed through caecal ligation and puncture, and histological, apoptosis, and Western blot analyses were performed for further validation. Results We identified six hub targets (MPO, HRAS, PPARG, FGF2, JUN, and IL6), and the PI3K-AKT pathway was the core pathway. CCK-8 assays showed that RR promoted the viability of the lipopolysaccharide-stimulated HUVECs [median effective dose (ED50) = 18.98 μg/mL]. Furthermore, RR inhibited inflammation, oxidative stress, cell apoptosis, and PI3K-AKT activation in lipopolysaccharide-stimulated HUVECs and ALI mice, which was consistent with the network pharmacology results. Discussion and conclusion This study provides foundational knowledge of the effective components, potential targets, and molecular mechanisms of RR against ALI, which could be critical for developing targeted therapeutic strategies for sepsis-induced ALI.
... This dual role applies to the large number of free OH-groups, especially 3-OH and the higher reactivity of flavonoid is observed in hydroxyl groups with oxidants (65,72,73). Currently, it is considered that phenolics and flavonoids function to promote health as anti-inflammatory and antioxidant agents, thus protecting the body against chronic diseases (74)(75)(76). In addition, these secondary metabolites also provide protection to plants against abiotic and biotic stresses (77,78). ...
Article
Full-text available
Artocarpus lacucha is an endemic plant to North Sumatera, Indonesia. This plant has pharmacological activities, including acting as an antioxidant and antibacterial. The aim of the present study was to analyze the antibacterial and antioxidant activities, and determine the flavonoid compounds from four parts of A. lachuca, namely leaves, barks, twigs and fruits. Antioxidant activity was investigated using the 2,2-diphenyl 1-picrylhydrazyl (DPPH) and cupric reducing antioxidant capacity (CUPRAC) methods. Antibacterial activity was analyzed using disk diffusion and microdilution methods. Several flavonoids, such as luteolin-7-O-glucoside, rutin, quercetin, kaempferol and apigenin, were determined using high performance liquid chromatography. Based on the antioxidant activity test results using the DPPH method, the bark ethanolic extract provided the highest antioxidant capacity, while the CUPRAC method indicated that the twig ethanolic extract had the highest antioxidant capacity. The antibacterial activity test results demonstrated that at a low concentration of 750 µg/disk the bark ethanolic extract obtained the highest inhibition zone and minimum inhibitory concentration level against six of nine pathogenic bacteria. Therefore, A. lachuca bark ethanolic extract could be potentially developed as antioxidant and antibacterial agents.
... It is distinguished by its ovoid bulb of 15 to 30 cm long and its long lanceolate leaves pointed at their tips [1], is a species polymorphic, with several forms and variants. Many plants, including D. maritima, are renowned for their medicinal potential and interesting therapeutic powers [2]. In Algeria, the red variety has been the subject of a severe collection to hunt for novel bioactive chemicals; it is a key player much like many other conventionally used therapeutic herbs. ...
Article
Based on the numerous medicinal benefits of the plant Drimia maritima (L.) Baker (Asparagaceae) and its use in the environment, this study examined the risk of this use on non-target species by examining the phytochemical and toxicological profile of the aqueous extract of D. maritima bulb and leaves on the liver and kidney function of Wistar rats. Seven groups of rats (n = 6) were utilized for subacute studies , including a control, three doses of an aqueous extract of D. maritima bulb (DBAE), and three doses of an aqueous extract of D. maritima leaves (DLAE), each at a dose of 98, 49, and 24.5 mg/ kg. The findings showed that both plant extracts include a large amount of phenolic chemicals, which provide them great antioxidant capability. Additionally, only the high oral dosage of DBAE showed a modest rise in hepatic and renal enzymes, even though the antioxi-dant enzyme levels (GST, GPx, and GSH) in the liver and kidney remained steady and hematological parameters did not change. However, none of the measures under study significantly changed because of the three DLAE dosages involved. The antioxi-dant status of both D. maritima extracts is raised. After subacute administration at levels below 98 mg/ kg body weight, DBAE was mostly non-toxic. But at the studied levels, DLAE was safe.
... MPs offer a wide range of biological activities and medicinal potentials [4]. Various herbs, fruits and grains have been found to possess pharmacological activities, undergo toxicity analysis, exhibit anti-inflammatory effects and display anti-aging properties [5]. These natural products, enriched with antioxidants, play a significant role in reducing the risk of diseases, including cancer, cardiovascular disorders, diabetes and age-related ailments. ...
Article
The present study aims to investigate the therapeutic potential of phytocompounds derived from Annona reticulata leaves for the treatment of hypertension, utilizing computational methodologies. Gaining a comprehensive understanding of the molecular interactions between neophytadiene and γ-sitosterol holds significant importance in the advancement of innovative therapeutic approaches. This study aims to examine the inhibitory effects of neophytadiene and γ-sitosterol using molecular docking and dynamics simulations. Additionally, we will evaluate their stability and predict their drug-like properties as well as their ADME/toxicity profiles. Neophytadiene and γ-sitosterol have a substantial binding affinity with 1O8A, as shown by the docking study. The stability of the complexes was confirmed through molecular dynamics simulations, while distinct clusters were identified using PCA. These findings suggest the presence of potential stabilizers. The drug-likeness and ADME/toxicity predictions revealed positive characteristics, such as efficient absorption rates, limited distribution volume and non-hazardous profiles. The neophytadiene and γ-sitosterol exhibit potential as hypertension medication options. Computational investigations reveal that these compounds exhibit high affinity for binding, stability and favourable pharmacokinetic properties. The results of this study lay the groundwork for additional experimental verification and highlight the promising prospects of utilizing natural compounds in the field of pharmaceutical research. Target proteins (1O8A) were used to perform molecular docking with representative molecules. Stability, conformational changes and binding energies were assessed through molecular dynamics simulations lasting 100 ns. Principal component analysis (PCA) was utilized to analyze molecular dynamics (MD) simulation data, to identify potential compounds that could stabilize the main protease. The safety and pharmacokinetic profiles of the compounds were evaluated through drug-likeness and ADME/toxicity predictions.
Article
Full-text available
Traditional herbal medicines (THM) are used by almost 75%-80% of the people in Bangladesh to treat various types of skin diseases. This review includes ethnobotanical data as well as relevant scientific studies on plants used in traditional medicine in Bangladesh to treat various types of skin disorders like inflammatory skin disorders, viral infections, bacterial infections, fungal infections, parasitic infections, pigmentation disorders, skin cancers, trauma and other skin disease conditions. Using specified key phrases, information on the traditional herbal medicinal plants of Bangladesh that are effective against various skin ailments was collated from several electronic databases such as PubMed, Google Scholar, Web of Science, Scopus, ScienceDirect, Springer and others. A total of 45 relevant publications were investigated, providing complete information on 221 distinct herbal plant species belonging to 73 families utilised by the diverse peoples of Bangladesh for the treatment of skin problems. The most often utilised plant organ in this study was leaves, followed by roots, fruits, entire plants, barks, seeds, stems, rhizomes and flowers. The Asteraceae family has the most species, whereas in terms of plant habit, herbs have the most species. Azadirachta indica, Curcuma longa, Vitex negundo, Aegle marmelos, Allium sativum and Lawsonia inermis are the most commonly utilised species. This research might be used as a beginning point for a systematic search for natural herbal remedies to treat skin diseases in Bangladesh's flora.
Article
Full-text available
Diabetes is a metabolic disorder in which blood sugar is high. Symptoms linked with high blood sugar include frequent urination, increased thirst and hunger. Complication in diabetes can be acute or severe. Acute complications include diabetic ketoacidosis, hyperosmolar, hyperglycemic or death; while serious long-term complications include cardiovascular disease, stroke, chronic kidney disease, foot ulcers, and damage to the eyes. Medications used to treat diabetes do so by lowering blood sugar levels. Glucose levels in normal range result in fewer complications like kidney problems and diabetic retinopathy. There are number of different classes of anti-diabetic medications; some of them include metformin, insulin etc. Alternate or herbal medications are also available which include plants like Trigonellafoenumgraecum, Nigella sativum, Azadirachtaindica, and Cichoriumintybus. Trigonella seeds are antidiabetic and antihyperlipedemic, while Nigella sativa, also called as Kalongi, can significantly reduce fasting, post prandial and glycosylated haemoglobin. Neem, which is a common herb found on Indian soil, has very good medicinal value, it blocks inhibitory effect of serotonin on insulin secretion mediated by glucose. Cichoriumintybus, also called as Kasini, shows antidiabetic effect, it reduces serum glucose by 20%, triglycerides by 91% and cholesterol by 16%. This shows that the mentioned herbs have antidiabetic effect i.e. they help in lowering blood sugar, so they can be used in day to day life or can be used in herbal formulation. It can be used as alternate medication besides allopathic treatment, as these do not have any side or adverse effect on diabetic cases.
Article
Full-text available
Lantana camara (L.) is employed by several ethnical groups to treat numerous diseases. Although there are no ethnomedical reports on its use against leishmaniasis, organic extracts prepared from L. camara were shown to display leishmanicidal activity. In the present study, we carried out a bioassay-guided fractionation of the dichloromethane extract from Mexican L. camara in order to identify the compounds responsible for the leishmanicidal activity. Eighteen chromatographic fractions (FI–FXVIII) were evaluated in vitro against Leishmania mexicana and L. amazonensis. FII, FX, FXI, FXV, and FXVI showed significant activity against both Leishmania strains, the most potent of which was FXV. Eicosane (1), squalene (2), β-ionone (3), caryophyllene oxide (4), β-caryophyllene (5), hexanoic acid (6), tiglic acid (7), a mixture of lantanilic (8) and camaric (9) acids, and lantadene B (10) were identified and obtained from the active fractions and evaluated for their leishmanicidal activity. The mixture of lantanilic (8) and camaric (9) acids (79%/21%) was the most potent one (half maximal inhibitory concentration (IC50) = 12.02 ± 0.36 μM). This study indicates that this cultivar of L. camara has high potential for the development of phytomedicines or as a source of natural products, which might represent lead compounds for the design of new drugs against leishmaniasis.
Article
Full-text available
Recently, herbal drugs and their bioactive compounds have gained popularity in the management of diabetes mellitus (DM), which has become an epidemic disease all over the world and is especially prevalent in the Kingdom of Saudi Arabia (KSA). This study aimed to investigate the antidiabetic effect of ethanolic and aqueous-ethanolic extracts of wild Ricinus communis (R. communis) leaves in streptozotocin (STZ) induced diabetic rats. Diabetic rats were administered orally with the mentioned extracts at doses of 300 and 600 mg/kg/BW for 14 days, and the obtained results of different biochemical parameters were compared with normal control, diabetic control and standard drug glibenclamide (5 mg/kg/BW). The obtained results revealed a remarkable and significantly (P < 0.05) reverse effect of the body weight loss, observed when diabetic rats were treated with ethanol and aqueous-ethanol extracts at 300 mg/kg/BW. Administration of the ethanol extract at 600 mg/kg/BW significantly (P < 0.05) reduced the blood glucose level. A significant increase in the AST, ALT and ALP levels (P < 0.05) was observed in the diabetic control and in the experimental groups with glibenclamide which was also significantly (P < 0.05) lowered after treatment with extracts at special doses. Total proteins, albumin, total bilirubin, direct bilirubin, creatinine and urea were also investigated and compared to the corresponding controls. We showed that administration of R. communis extract generally significantly (P < 0.05) ameliorated the biochemical parameters of diabetic rats. Also, the changes in serum electrolyte profile were assessed and the results demonstrate that administration of extracts at concentration of 600 mg/kg/BW generally inhibits the alteration maintain their levels. The obtained data imply the hypoglycemic effects of this plant, which may be used as a good alternative for managing DM and therefore validating its traditional usage in KSA.
Article
Full-text available
The main chemical constituents of the genus Polygonum (Polygonaceae) are flavonoids, quinones, phenylpropanoids, and terpenoids, which show anticancer, antitumor, anti-oxidative, anti-inflammatory, analgesic, antibacterial, insecticidal, and other pharmacological effects. This paper summarizes research on the chemical constituents and pharmacological effects of compounds from the genus Polygonum in last 15 years. Keywords: Polygonum, Chemical constituents, Bioactivity, Flavonoid, Anticancer
Article
Full-text available
Background Opuntia ficus-indica var. saboten (OFIS) is used widely in Korea to treat constipation due to its diuretic effects and its enhancement of bowel function and appetite. However, its safety has not yet been established. The aim of this study was to evaluate the repeated oral toxicity and genotoxicity of OFIS extract (OE). Methods White female and male Sprague Dawley rats (n = 6) were divided into 4 groups, and OE was administered to them orally (0, 500, 1000, and 2000 mg/kg/day, respectively) for one week. The Ames test, the chromosomal aberration assay, and the mammalian micronucleus test were performed to determine the OE genotoxicity. The Ames test was conducted using Salmonella typhimurium (S. typhimurium) strains TA100, TA1535, TA98, and TA153 and Escherichia coli (E. coli) WP2 urvA, and Chinese hamster lung (CHL) cells were used for the chromosomal aberration assay. The mammalian micronucleus test was performed using mouse bone marrow cells. Results This study revealed that OE administration did not alter the normal rat behavior, body weight gain, and food and water consumption with respect to the normal controls. In addition, there were no toxic effects observed during the ophthalmological test. The biochemical hematological and serum values as well as urinalysis parameters and organ weights were all similar to those of the normal control group. In addition, no mutagenicity effects from the OE were found in S. typhimurium or E. coli with or without S9 activation according to the Ames test. The OE did not significantly alter the number of structural aberrations in the CHL cells in the presence or absence of S9 activation. The oral administration of OE also caused no significant increase in the number of micronucleated polychromatic erythrocytes or in the mean ratio of polychromatic to total erythrocytes. Conclusions In conclusion, OE could be considered as a reliable and safe herbal medicine or functional food since no toxicity was found under the conditions of this study.
Article
Background: Diabetes is one of the most serious, chronic diseases characterized by hyperglycemia resulting from defects in insulin secretion, insulin action, or both. It is becoming the third “killer” of the health of mankind after cancer and cardiovascular diseases. Diabetes is a metabolic disorder affecting about 220 million people worldwide. A number of plants have been described in Ayurveda for the management of diabetes. The present study was aimed to find out the hypoglycemic and non-toxic effect of Citrullus lanatus seed extracts. Materials and Methods: In acute toxicity study, there was no mortality observed up to the maximum dose level of 2000 mg/kg body weight of the extract after administered orally. After toxicity studies the various extracts of plant were used for hypoglycaemic activity in which the ethanolic extract showed very good reduction in blood glucose level. From that three doses were selected for oral glucose tolerance test and 400 mg/kg of ethanolic seed extract showed better glycaemic control. Hence antidiabetic studies were conducted with 400 mg/kg dose level for 30 days and the tissues antioxidant levels and histopathological studies were carried out by using standard protocols. Results: The ethanolic extract of Citrullus lanatus helps to maintain the antioxidant level in various organs and also helps to protect the organs from oxidative damage. Conclusion: From our study, the ethanolic seed extract of Citrullus lanatus controls the blood glucose level and also helps to prevent the organ from oxidative damage. Hence it can be used in the management of diabetes mellitus.
Article
This study was carried out to investigate the effect of the whole crude extract of Origanum syriacum on different biochemical parameters in blood using animal model. The aerial parts of Origanum syriacum were extracted with petroleum ether, chloroform, methanol and water, all these extracts were combined to prepare the whole extract then administrated orally in two doses 100mg/kg and 400mg/kg. The results obtained indicate that the extract showed a significant decreasing effect on low density lipoprotein (LDL‐cholesterol), triglycerides and glucose in both doses, while a non significant increase of urea and high density lipoprotein (HDL‐cholesterol) was observed as compared to normal saline treated controls. This study showed that the whole extract of Origanum syriacum possess a potential pharmacological activity in lowering the major lipid profile parameters in which we have reported for the first time. This work is sponsored by Al‐Zaytoonah University of Jordan.
Article
Introduction: The current study aims to evaluate the therapeutic effect of Cichorium intybus n-hexane extract on diabetes mellitus and its antioxidant effect in vivo in alloxinated animals. Diabetic neuropathy improvement was also tested as well as insulin levels and histology of the pancreas were performed. Methods: The chromatographic standardization of C. intybus extract was performed using isocratic HPLC, which indicated the presence of numerous phyto-constituents. The hexane extract was studied for its effect on blood glucose levels and painful Diabetic neuropathy (DN) in diabetic mice. Hyperalgesia and mechanical-allodynia were evaluated using thermal stimuli, pain response to radiant energy experiments and a mechanical sensitivity test respectively. Subsequently, after eight weeks of being alloxinated, BGL, body weight, antioxidant activity, insulin levels and glycated hemoglobin were recorded to evaluate antidiabetic potential and the DN. Results: The administration of Cichorium intybus extract (50, 75 and 100 mg/kg) and a combination of Cichorium intybus extract and Camellia sinensis (50 + 200 mg/kg, respectively) have revealed an acute hypoglycemic effect ranging from 14.15% and 42.4%. The sub-chronic anti-diabetic effect ranged from 23.41% and 44.8%. They diminished hyperalgesia and tangible allodynia significantly (p<0.05), (n=7 per group). The powerful neuroprotective properties might serve as potential lead-compounds for further analysis. Conclusion: The histological study and the potent antioxidant effect showed that they could assist in the management of diabetes mellitus and DN by amelioration of insulin levels and regeneration of pancreatic beta cells.