Download as pdf or txt
Download as pdf or txt
You are on page 1of 8

2016

NPC Natural Product Communications Vol. 11


No. 10
Natural Triterpenoids for the Treatment of Diabetes Mellitus: 1579 - 1586
A Review
Han Lyu, Jian Chen and Wei-lin Li*

Institute of Botany, Jiangsu Province and Chinese Academy of Sciences, Nanjing 210014, China

lwlcnbg@mail.cnbg.net

Received: January 28th, 2016; Accepted: July 18th, 2016

Triterpenoids, an important group of secondary metabolites, are widely distributed in nature. Many triterpenoids have been found with potential therapeutic
effect against diabetes mellitus. However, the use of triterpenoids for the treatment of diabetes has not been systematically discussed previously. This review
summarized the anti-diabetic activity of natural triterpenoids reported since the late 1980s with the emphasis on the molecular mechanisms.

Keywords: Natural triterpenoids, Diabetes mellitus.

Diabetes Mellitus (DM) is a multi-cause metabolic disease triterpenoid analogs or triterpenoids derived from animal and
characterized by hyperglycemia resulting from defects in insulin microbial species are not included), this review will highlight their
secretion, insulin action, or both. On the basis of etiology, diabetes anti-diabetic mechanisms, especially potential molecular
mellitus is categorized into two main types. Type 1 DM, also mechanisms. Given the diverse mechanistic insights of these
known as insulin-dependent diabetes mellitus (IDDM), arises from compounds, this review is structured according to the suggested
little or no endogenous insulin secretory capacity. Type 2 DM, or antidiabetic mechanisms of triterpenoids.
non-insulin-dependent diabetes mellitus (NIDDM), is a much more
prevalent category and often results from a combination of
resistance to insulin action and an inadequate compensatory insulin R2O
HO
secretory response [1]. H

R
Insulin and oral antidiabetic agents such as sulfonylureas,
biguanides, α-glucosidase inhibitors, and glinides are often used as HO R1 O
monotherapy or in combination to treat diabetes [2]. However,
many of the existing synthetic antidiabetic drugs have serious side 1. R1 = glc (1→2)-glc, R2 = H 5. R = CH3 (lupeol)
(ginsenoside Rg3) 6. R = COOH (betulinic acid)
effects. There is an increasing demand for more effective and safer 2. R1 = glc (1→2)-glc, R2 = glc (1→6)-glc Lupane-type triterpenoids
novel antidiabetic agents. Many natural products, such as (ginsenoside Rb1)
triterpenoids and flavonoids, have been extensively explored in the 3. R1 = glc (1→2)-glc, R2 = ara (1→6)-glc(p)
field of drug discovery and have led to remarkable successes [3-6]. (ginsenoside Rb2)
4. R = glc, R2 = H (ginsenoside Rh2)
Dammarane-type
Triterpenoids, a large class of natural products, are a rich reservoir
of candidate compounds for drug discovery. More than 20,000
triterpenoids exist in nature [7]. Triterpenoids are composed of six R2
isoprene (C5H8)6 units; they often occur as free triterpenoids, H COOH
triterpenic glycosides (saponins), or their esters. Triterpenoids are R1
R1
H COOH
structure-wise mainly classified into two chemical types, tetracyclic
HO
and pentacyclic. Triterpenoids can further be sub-classified into R2 HO
diverse groups including dammaranes, cycloartanes, lanostanes,
7. R1 = H, R2 = CH3 (oleanolic acid) 10. R1 = H, R2 = H (ursolic acid)
cucurbitanes (tetracyclic triterpenoids), lupanes, oleananes, ursanes, 8. R1 = OH, R2 = CH3 (maslinic acid) 11. R1 = OH, R2 = H (corosolic acid)
friedelanes (pentacyclic triterpenoids), and miscellaneous 9. R1 = OH, R2 = CH2OH (arjunolic acid) 12. R1 = OH, R2 = OH (tormentic acid)
compounds [8]. Oleanane-type triterpenoids 13. R1 = H, R2 = OH (pomolic acid)
Ursane-type triterpenoids
Triterpenoids have been proven to have a broad spectrum of Figure1: Structures of some active triterpenoids
pharmacological activities: anti-diabetic, anti-cancer, anti-
inflammatory, analgesic, antiviral, anti-HIV, antimicrobial, Antidiabetic activity of triterpenoids in vivo
antiplasmodial, cardioprotective and hepatoprotective effects Increasing studies have suggested that various triterpenoids
[4,5,7,8]. Among them, a large number of studies were focused on exhibited antidiabetic activity in normal or/and diabetic animal
their anti-diabetic properties. Pharmacological activities in diabetes models (Table 1). Triterpenoids were shown to reduce the plasma
and diabetic complications of some pentacyclic triterpenoids have glucose level and enhance glucose tolerance of experimental
been discussed earlier [4]. By collecting evidence in vitro and in animals [9-33]. These findings, which strongly suggest the
vivo bioassay of triterpenoids (tetracyclic triterpenoids and antidiabetic potential of triterpenoids, have initiated increasing
pentacyclic triterpenoids) of plant origin (semi-synthetic efforts in the field to explore the molecular mechanism responsible.
1580 Natural Product Communications Vol. 11 (10) 2016 Lyu et al.

Table 1: Antidiabetic activities of triterpenoids tested in animal models.


Compounds (type*) Model Antidiabetic activities Plant resource (family) References
ginsenoside Rg3(D) db/db diabetic mice ↓plasma glucose Panax ginseng C. A. Mey. (Araliaceae) [9]
↑insulin secretion
ginsenoside Rb2(D) STZ-induced rats ↓plasma glucose Panax ginseng C. A. Mey. (Araliaceae) [10]
↑hepatic glycogen
↓G6Pase
ginsenoside Rh2(D) fructose-rich-fed STZ-rats ↓plasma glucose Panax ginseng C. A. Mey. (Araliaceae) [11]
normal rats ↑response to exogenous insulin
↓plasma glucose
↑insulin secretion
ginsenoside Rg1(D) alloxan-diabetic mice ↓plasma glucose Panax ginseng C. A. Mey. (Araliaceae) [12]
normal mice ↑hepatic glycogen
ginsenoside Re(D) STZ-induced diabetic rats ↓plasma glucose Panax ginseng C. A. Mey. (Araliaceae) [13]
phanoside(D) normal rats ↓plasma glucose Gynostemma pentaphyllum (Thunb.) Mak. [14]
↑insulin secretion (Cucurbitaceae)
isoastragaloside I(Cy), db/db diabetic mice ↑adiponectin level Astragalus membranaceus (Fisch.) Bunge [15]
astragaloside II(Cy) ↑AMPK activity (Leguminosae)
↑insulin sensitivity
astragaloside IV(Cy) STZ-induced daibtetic rats ↓plasma glucose, HbA1C Astragalus membranaceus (Fisch.) Bubge [16]
↑insulin secretion (Leguminosae)
momordicoside S, T(Cu) normal and high-fat-fed mice ↑glucose tolerance Cucumis sativus L.( Cucurbitaceae) [17]
gymnemic acid II(O), STZ-diabetic mice ↓plasma glucose Gymnema sylvestre R. BR. [18]
gymnemic acid III(O), ↓sugar absorption in small intestinal (Asclepiadaceae)
gymnemasaponin V(O),
gymnemoside-f(O)
senegin II(O), normal mice ↓plasma glucose Polygala senega L. var. latifolia Torr. et Gray [19]
senegin III(O) KK-Ay diabetic mice (Polygalaceae)
kaikasaponin III(O) STZ-induced diabetic rats ↓plasma glucose Pueraria thunbergiana Benth. [20]
(Leguminosae)
arjunolic acid(O) STZ-induced diabetic rats ↓plasma glucose Terminalia arjuna (Roxb.) Wight & Arn. [21]
↓TNF-α,NO (Combretaceae)
↑antioxidant enzymes in pancreas
momordin Ic(O), normal rats ↓plasma glucose Kochia scoparia (L.) Schrad. [22,23]
oleanolic acid 3-O-glucuronide(O) ↓sugar absorption in small intestinal (Chenopodiaceae)
18 β-glycyrrhetinic acid(O) STZ-induced diabetic rats ↓plasma glucose,HbA1C Glycyrrhiza uralensis Fisch. [24]
↑insulin secretion (Leguminosae)
oleanolic acid(O) normal rats ↓plasma glucose Cornus officinalis Sieb. et Zucc. [25]
↑insulin secretion (Cornaceae)
maslinic acid(O), normal rats ↓plasma glucose Eriobotrya japonica (Thunb.) Lindl. [26]
pomolic acid(U) db/db/Ola diabetic mice ↓glycosuria (Rosaceae)
elatosides E(O) normal rats ↓plasma glucose Aralia elata (Miq.) Seem. (Araliaceae) [27,28]
escins-IIa(O), normal rats ↓plasma glucose Aesculus hippocastanum L. [29]
escins-IIb(O) (Hippocastanaceae)
tormentic acid(U) normal rats ↓plasma glucose Eriobotrya japonica (Thunb.) Lindl. [30]
↑insulin secretion (Rosaceae)
ursolic acid(U) high-fat diet mice ↓plasma glucose Cornus mas L. (Cornaceae) [31]
NA-STZ-induced diabetic mice ↑insulin secretion
↑hepatic glycogen
corosolic acid(U) normal mice ↓plasma glucose Lagerstroemia speciosa L. (Lythraceae) [32]
↓gluconeogenesis
euscaphic acid(U) normal rats ↓plasma glucose Eriobotrya japonica (Thunb.) Lindl. [33]
alloxan-diabetic mice (Rosaceae)
*In this review, the first appearance of a triterpenoid will be followed with the abbreviation of its chemical type in a parenthesis, D=dammarane-type, Cy=cycloartane-type,
Cu=cucurbitane-type, La=lanostane-type, Lu=lupane-type, O=oleanane-type, U=ursane-type, T=Taraxastane-type

Stimulation of insulin secretion with oleanolic acid increased total cellular insulin protein and
Insulin is a metabolic hormone secreted from pancreatic islet β- mRNA levels [36,37]. In spite of the well-documented activities, it
cells. The main function of insulin is to maintain normal glucose remains unclear how these triterpenoids stimulated insulin secretion.
homeostasis. Insulin stimulates hepatic glycogen synthesis,
decreases glycogenolysis and the release of glucose from hepatic Insulin secretion is modulated by several hormones and
gluconeogenesis. Insulin also promotes glucose uptake in the neurotransmitters, among which acetylcholine (ACh) is an
skeletal muscle and adipose tissue. Patients with Type 1 DM, who important mediator [38]. Ginsenoside Rh2 (1.0 mg/kg) and oleanolic
have little or no endogenous insulin secretory capacity, require acid (5, 10 and 20 mg/kg ) increased plasma insulin levels of Wistar
insulin therapy to maintain normoglycaemia. Insulin therapy has rats by releasing ACh from nerve terminals and then stimulating
also been used to prevent or slow the progression of diabetic muscarinic M3 receptors in pancreatic cells [11,25].
microvascular complications in patients with type 2 DM [34,35].
ATP-sensitive K+ channels (KATP channels) in the pancreatic β-
Several triterpenoids were reported to be able to increase the plasma cells take part in cell energy metabolism by coupling cell
insulin level in normal or diabetic animals (Table 1). These findings metabolism to electrical activity. The KATP channels in pancreatic
were also recapitulated in vitro studies. Phanoside at a dose up to β-cells are critical in the regulation of glucose-induced and
500 μM stimulated insulin secretion in isolated rat pancreatic islets sulfonylurea-induced insulin secretion. The opening of KATP
[14]. Oleanolic acid (50 µM) and oleanolic aldehyde (O) (6.25-50 channels will suppress β-cells hyperpolarization and insulin
mg/mL) enhanced insulin secretion in INS-1 832/13 pancreatic β- secretion [39]. 20(S)-Ginsenoside Rg3 (2-8 μM) enhanced the
cells. Oleanolic acid (30 µM) also enhanced acute glucose- insulin secretion in HIT-T15 cells in vitro, which was likely
stimulated insulin secretion in isolated rat islets. Chronic treatment associated with the closure of the KATP channels in β-cells [9].
Anti-diabetic natural triterpenoids Natural Product Communications Vol. 11 (10) 2016 1581

Table 2: Inhibitory activities of PTP1B.


Compdouds (type) IC50 (μM) Plant resource(family) References
ursolic acid 3.8 Symplocos paniculata (Thunb.) Miq. (Symplocaceae) [49]
2.3 Phoradendron reichenbachianum (Seem.) Oliv. (Santalaceae) [50]
3.1 Diospyros kaki Thunb. (Ebenaceae) [51]
corosolic acid 7.2 Symplocos paniculata (Thunb.) Miq. (Symplocaceae) [49]
7.0 Rhododendron brachycarpum G. Don (Ericaceae) [55]
rotungenic acid(U), 10.9 Diospyros kaki Thunb. (Ebenaceae) [51]
pomolic acid(U), 3.9
24-hydroxyursolic acid(U), 12.8
19α,24-dihydroxyurs-12-en-3-on-28-oic acid (U) 8.1
2a,3b-dihydroxy-24-nor-urs-4(23),11-dien-28,13b-olide (U), 29.1 Weigela subsessilis (Nakai) L.H.Bailey (Caprifoliaceae) [52]
2a,3b-dihydroxy-24-nor-urs-4(23),12-dien-28-oic acid (U) 5.3
3β-hydroxyurs-12-en-27-oic acid (U) 4.9 Astilbe koreana (Kom.) Nakai (Saxifragaceae) [54]
3-oxoolean-12-en-27-oic acid (O), 6.8 Astilbe koreana (Kom.) Nakai (Saxifragaceae) [54]
3β-hydroxyolean-12-en-27-oic acid (O), 5.2
3α, 24-dihydroxyolean-12-en-27-oic acid (O), 11.7
3β, 6β-dihydroxyolean-12-en-27-oic acid (O) 12.8
moronic acid (O), 13.2 Phoradendron reichenbachianum (Seem.) Oliv. (Santalaceae) [50]
morolic acid (O) 9.1
3β-acetoxyolean-12-en-28-acid (O), 7.8 Styrax japonica Sieb. et Zucc. (Styracaceae) [53]
3β-acetoxyolean-12-en-28-aldehyde (O) 9.3
oleanolic acid 9.5 Phoradendron reichenbachianum (Seem.) Oliv. (Santalaceae) [50]
7.6 Diospyros kaki Thunb. (Ebenaceae) [51]
spathodic acid (O) 18.8 Diospyros kaki Thunb. (Ebenaceae) [51]
rhododendric acid A (T) 6.3 Rhododendron brachycarpum G. Don (Ericaceae) [55]
lupeol (Lu), 13.7 Sorbus commixta Hedl. (Rosaceae) [56]
lupenone (Lu) 3.7
betulinic acid (Lu), 0.7 Saussurea lappa C.B.Clarke (Compositae) [57]
betulinic acid methyl ester (Lu) 0.9

Reversing insulin resistance mediated glucose uptake in 3T3-L1 adipocytes and C2C12
Insulin resistance is a pathologic state in which target cells fail to myotubes. Ginsenoside Rb1 (1 μM) promoted GLUT1 and GLUT4
respond to normal levels of circulating insulin. Reduced insulin translocations to the 3T3-L1 adipocytes cell surface and enhanced
sensitivity may result in inappropriate insulin levels, impaired translocation of GLUT4 in Chinese Hamster Ovary (CHO) cells.
fasting glucose or defective glucose tolerance. The resistance of Meanwhile, ginsenoside Rb1 increased the phosphorylation of
target tissues to insulin is the major pathophysiological event insulin receptor substrate-1 and PKB, and facilitated receptor [44].
resulting in the development of type 2 DM [40]. It was also suggested that ginsenoside Rb1 (at up to 10 μM)
enhanced basal and insulin-mediated glucose uptake accompanied
Studies showed that triterpenoids could reverse insulin resistance. In by the up-regulation of mRNA and protein level of GLUT4 in 3T3-
animal studies, ginsenoside Rh2 (1 mg/kg) could increase the L1 adipocytes [45]. These data together may suggest the compounds
responses to exogenous insulin and delay insulin resistance multiple functions in modulating the insulin signaling pathway.
induction by fructose-rich chow in streptozotocin (STZ)-induced
diabetic rats [41]. As the pathophysiological process accounting for Ursolic acid increased the activity of insulin on tyrosine
insulin resistance is still unclear, to fully understand the molecular phosphorylation of the insulin receptor β-subunit and the number of
basis of these compounds will rely on in-depth mechanistic studies. insulin-activated IRs in CHO/IR. Ursolic acid potentiated insulin-
Below describes that how insulin resistance is possibly reversed by mediated tyrosine phosphorylation of the IR β-subunit,
triterpenoids treatment, including the activation of insulin signaling, phosphorylation of Akt, glycogen synthase kinase-3β, and enhanced
increasing adiponectin level, inhibiting protein tyrosine phosphatase translocation of GLUT4 in 3T3-L1 adipocytes [46]. Likewise, 250
1B etc. nM of corosolic acid enhanced glucose uptake in L6 myotubes and
facilitated GLUT4 translocation in Chinese-hamster ovary cells
Activation of insulin signaling molecules: Insulin receptor expressing human IR cells. These actions could be blocked by PI3K
signaling is initiated by insulin binding to its cell surface receptor, inhibitor and regulated by insulin pathway activation [47].
followed by receptor autophosphorylation, and activation of
receptor tyrosine kinases, which result in tyrosine phosphorylation Inhibition of protein tyrosine phosphatase: Dephosphorylation of
of insulin receptor substrates and successive activations of signaling molecules by protein tyrosine phosphatases (PTPs) play a
phosphoinositide 3-kinase (PI3K), 3-phosphoinositidedependent critical negative regulatory role in insulin signal transduction. The
protein kinases (PDK-1 and PDK-2), Akt/protein kinase B (PKB) enhanced activity of one or more PTPs may lead to insulin
and atypical protein kinase C λ and ζ (PKCλ/ζ). These complex resistance. Current data indicate that inhibition of PTP1B, a member
actions stimulate insulin-mediated translocation of glucose of the PTP family, in peripheral tissues may be useful for treating
transporter type 4 (GLUT4) from intracellular vesicles to the plasma metabolic-related disorders such as obesity and type 2 DM [48].
membrane. Mitogen activated protein (MAP) kinase and the PI3K
signaling are two major pathways involved in insulin-receptor A large number of triterpenoids showed PTP1B inhibitory activities
signaling, and the metabolic response to insulin is primarily in vitro [49-57] (Table 2).These triterpenoids mainly belong to the
mediated via the PI3K pathway [42]. Defects in insulin receptor oleanane- and ursane-types. Structure-activity relationship findings
signaling may contribute to impaired GLUT4 translocation and suggest that a hydroxyl group at C-3 and a carbonyl group at either
insulin resistance [42,43]. C-28 or C-27 of the oleanane-and ursane-type triterpenoids may be
essential structural features to exert the PTP1B inhibitory activity.
Triterpenoids could act as an insulin sensitizer by influencing either Triterpenoids with a 3β-hydroxy group may show a better inhibitory
the upstream signaling pathway or the downstream mediators. activity than those with a 3α-hydroxy moiety [51].
Ginsenoside Rb1 (D) (0.001-1 μM) stimulated basal and insulin-
1582 Natural Product Communications Vol. 11 (10) 2016 Lyu et al.

Increasing adiponectin level: Adiponectin is an adipocyte-secreted, skeletal muscle glucose uptake. In liver, elevated FFA can impair
insulin-sensitizing hormone. Reduced circulating levels of insulin-mediated suppression of hepatic glucose output. TNFα is a
adiponectin may be exhibited in conditions of insulin resistance and cytokine largely expressed in adipose tissue, which may be linked
diabetes. Administration of recombinant adiponectin may increase with increased plasma FFA level and impaired insulin sensitivity in
glucose uptake, hepatic glucose production in liver and ameliorate obesity and type 2 DM [42,64].
whole body insulin resistance [58].
One hundred μM astragaloside IV antagonized TNFα-induced
Isoastragaloside I and astragaloside II (5 μg/mL each) increased insulin resistance and increased insulin stimulated 2-deoxy-D-[1-
3
adiponectin concentration in 3T3-L1 adipocytes. The same action H]-glucose uptake in 3T3-L1 adipocytes [65].
was found in mouse primary adipocytes after treatment with these
compounds. These actions were confirmed in animal tests: Promoting glycogen synthesis and inhibiting glycogen
Isoastragaloside I and astragaloside II induced significant increases degradation
in serum adiponectin, which resulted in alleviation of In most mammalian cells, glycogen is stored as a reserve for the
hyperglycemia, glucose tolerance and insulin resistance in db/db production of glucose 6-phosphate as a metabolic fuel for
mice and dietary mice [15]. glycolysis. Storage of available glucose to supply the tissues is
found principally in the liver. A defect in glycogen synthesis is a
Increasing the activity of AMP-activated protein kinase: potential factor contributing to postprandial hyperglycemia in
Adenosine 5'-monophosphate-activated protein kinase (AMPK) patients with type 2 DM [66]. Glycogen phosphorylase (GP) is a
plays an important role in mediating cell energy metabolism. key enzyme involved in glycogen breakdown to produce glucose
AMPK is involved in the stimulation of glucose uptake by muscle and related metabolites for energy supply. Pharmacological
contraction. AMPK also regulates insulin synthesis and secretion in inhibition of GP has been regarded as a promising therapeutic
pancreatic islet β-cells. Increased activity of AMPK may be approach for treating diabetes [67].
effective in correcting insulin resistance in patients with forms of
impaired glucose tolerance and Type 2 DM [59]. Administration of ginsenoside Rb2 (10 mg/rat/d) for six days
produced a significant decrease in blood glucose level in STZ-
Chronic treatment with (50 mg/kg) isoastragaloside I and induced rats. A moderate increase in the hepatic glycogen content
astragaloside II for 6 weeks resulted in a significant elevation in and a significant decrease in the activity of glucose-6-phosphatase, a
phosphorylation of AMPK in both liver tissue and soleus muscle of gluconeogenic enzyme in liver, were found at the same time [10].
db/db mice [15].
Ginsenoside Rg1 (200 mg/kg) promoted the synthesis of liver
Ten μM momordicoside Q, R, S, and T (Cu), and karaviloside XI glycogen in normal mice. In vitro, 0.1 µg/mL of ginsenoside Rg1
(Cu) isolated from Momordica charantia L. (Cucurbitaceae) (bitter stimulated the uptake of 3H-glucose in isolated rat hepatocytes [12].
melon), stimulated GLUT4 translocation to the cell membrane in
both L6 myotubes and 3T3-L1 adipocytes, associated with an Ursolic acid supplement (0.01, 0.05 g/100 g diet) increased hepatic
increased activity of AMPK [17]. glycogen content by decreasing hepatic glucose-6-phosphatase
activity, increasing glucokinase activity, and the
Ursolic acid, betulinic acid, oleanolic acid, and 30-norhederagenin glucokinase/glucose-6-phosphatase ratio in nicotinamide-
(T), isolated from the methanol extract of the bark of Paeonia streptozotocin-induced diabetic mice [68].
suffruticosa Andr. (Ranunculaceae) (Moutan Cortex), increased
glucose uptake and enhanced glycogen synthesis in HepG2 cells Hederagonic acid (O), gypsogenin (O), echinocystic acid (O),
under high glucose conditions. Ten μM concentrations of these oleanolic acid, hederagenin acid (O) and gypsogenic acid (O),
compounds significantly stimulated AMPK, glycogen synthase isolated from the ethanol extract of the roots of Gypsophila
kinase-3β and ACC phosphorylation [60]. oldhamiana Miq. (Caryophyllaceae), were found to be inhibitors of
GP in vitro; percent inhibition (10 μM) of these compounds were
Activation of peroxisome proliferator-activated receptors: 11.1, 45.1, 19.0, 73.1, 73.1 and 45.1%, respectively [69].
Peroxisome proliferator-activated receptors (PPARs) are a group of
nuclear receptor proteins that function as transcription factors. The Maslinic acid inhibited phosphorylase GP activity in homogenates
PPAR family consists of 3 subtypes of proteins encoded by separate of cultured astrocytes with an IC50 value of 5.7 μM. Moreover, pre-
genes: PPAR α (NR1C1), PPAR β/δ (NR1C3) and PPAR γ (also incubation with maslinic acid increased cellular glycogen content
known as γ or NR1C2). PPARs regulate lipid and lipoprotein and prevented norepinephrine-induced excessive glycogenolysis
metabolism, and glucose homeostasis. PPAR α and PPAR γ are [70].
essential for the actions of the many insulin sensitizers [61,62].
At a dose of 100 mg/kg, methyl-3β-hydroxylanosta-9,24-dien-21-
Ginsenoside Rb1 (10 μM) increased the expression of mRNA and oate (La), isolated from the chloroform extract of the stem bark of
the protein of PPARγ in 3T3-L1 adipocytes [45]. Protorhus longifolia (Bernh. ex C. Krauss) Engl. (Anacardiaceae),
increased hepatic glycogen content with increases both in
Ten μM 20(S)-protopanaxatriol (D) increased PPARγ- hexokinase and glucokinase activity and a decrease in glucose-6-
transactivation activity in 3T3-L1 adipocytes by increasing the phosphatase activity in STZ-induced diabetic rats [71].
expression of PPARγ target genes such as aP2, LPL and PEPCK, in
parallel with a significant increase in expression of GLUT4 [63]. Suppression of hydrolysis of starch and glucose transport in
small intestine
Decreasing free fatty acid: Increased plasma free fatty acid (FFA) Glycosidase enzymes, such as α-glucosidase and α-amylase, have
levels, a common symptom in individuals with obesity or type 2 the ability to degrade dietary starch and promote the rate of blood
DM, have been considered as an important factor associated with sugar absorption from the small intestine. The inhibition of α-
insulin resistance. Increased FFA can reduce insulin-stimulated glucosidase and α-amylase enzymes can significantly reduce the
Anti-diabetic natural triterpenoids Natural Product Communications Vol. 11 (10) 2016 1583

postprandial increase in blood glucose. Inhibition of α-glucosidase increased 11β-HSD1 activity may lead to metabolic syndrome,
and α-amylase can be used in patients with predominately obesity, insulin resistance, type 2 DM and cardiovascular
postprandial hyperglycemia. α-Glucosidase inhibitors, such as complications [85]. Selective inhibition or down-regulation of
acarbose, voglibose and miglitol, were developed in the 1990s and 11β-HSD1 results in a decrease of excessive hepatic glucose
commonly used in clinical therapy [72,73]. production in hyperglycemia and diabetes mellitus, and exerts a
positive effect on insulin sensitivity in diabetic subjects [86].
Corosolic acid (10 mg/kg) significantly reduced the hydrolysis of
sucrose in the small intestine of mice [74]. Ursolic acid, 3-epi-orosolic acid methyl ester (U), tormentic acid
methyl ester (U) and 2α-hydroxy-3-oxours-12-en-28-oic acid (U),
An oleanolic acid and ursolic acid (2:1) mixture was a potent isolated from leaves of Eriobotrya japonica (Thunb.) Lindl. (Rosaceae),
α-amylase inhibitor with an IC50 value of 4 μM in vitro. Oleanolic selectively inhibited 11β-HSD1 with IC50 values of 1.9, 5.2, 9.4 and
acid, ursolic acid (1, 2.5, 5 μg/mL) and lupeol (5 μg/mL) showed 17 μM, respectively [87].
inhibitory effects against α-amylase [75].
Masticadienonic acid (La) and isomasticadienonic acid (La),
Bartogenic acid (O), isolated from the seeds of Barringtonia isolated from the oleoresinous gum of Pistacia lentiscus var. chia
racemosa Roxb. (Lecythidaceae), showed both α-glucosidase (IC50 (L.) (Anacardiaceae), selectively inhibited 11β-HSD1 (IC50 values
= 168.1 μg/mL) and α-amylase inhibitory properties [76]. (2.51 and 1.94 μM) [88].
Cichoridiol (18α,19β-20(30)-taraxasten-3β,21α-diol) (T), isolated
from Cichorium intybus L. (Asteraceae), showed an α-glucosidase Inhibition of dipeptidyl peptidase-4
inhibitory activity (IC50 = 51.9 μM) [77]. Dipeptidyl peptidase 4 (DPP-4) is involved in various physiological
processes by cleaving dipeptides from various peptide hormones,
α-Amyrin-3-O-β-(5-hydroxy) ferulic acid (U) and lupine (Lu), neuropeptides and chemotactic agents. Glucagon-like peptide 1
isolated from the root bark of Euclea undulata Thunb. var. myrtina (GLP-1) and glucose dependent-insulinotropic polypeptide (GIP)
(Ebenaceae), inhibited α-glucosidase (IC50 = 7.76, 14.69 μM, are two incretin hormones released from enteroendocrine cells of
respectively) [78]. the intestine. GLP-1 and GIP are important insulin secretion
stimulators in postprandial blood glucose level control. DPP-4 could
2,3-Seco-20(29)-lupene-2,3-dioic acid (Lu), isolated from leaves inactivate the insulin-releasing effect of GLP-1 and GIP by removal
and twigs of Fagus hayatae Palib. ex Hayata (Fagaceae), showed an of His-Ala and Tyr-Ala dipeptides from the N-terminal end [89].
α-glucosidase inhibitory activity (IC50 = 62.1 μM) [79]. Several DPP-4 inhibitor drugs have been clinically used to treat
type 2 diabetes.
Pistagremic acid (D), isolated from galls of Pistacia integerrima
J.L. Stewart ex Brandis (Anacardiaceae), showed inhibitory activity Six triterpenoids isolated from the leaves of Cyclocarya paliurus
against both yeast α-glucosidase (IC50 = 89.1 μM), and rat intestinal (Batal.) Lljinsk. (Juglandaceae), cyclocariosides D, E, F, G and H
α-glucosidase (IC50 = 38.9 μM) [80]. (D), and cyclocarin A (D) (10 μM), exhibited inhibitory activities
against DPP-4 [90].
Inhibition of nutrient absorption is the basis of an agent used
clinically to inhibit intestinal catabolism of complex carbohydrates. Quinovic acid (U), quinovic acid-3β-O-β-D-glycopyranoside (U),
Agents that can delay or inhibit glucose absorption may have a quinovic acid-3β-O-β-D-glucopyranosyl-(28→1)-β-D-
promising impact in managing diabetes [81]. glucopyranosyl ester (U) {from Fagonia cretica L.
(Zygophyllaceae)} and lupeol {from Hedera nepalensis K. Koch
Gymnemic acids II, III, and IV (O) (0.5 mM), gymnemasaponin V (Araliaceae)} inhibited DPP-4 with IC50 values of 30.7, 57.9, 23.5
(O) and gymnemoside-f (O), isolated from the leaves of Gymnema and 31.6 µM, respectively [91].
sylvestre, showed an inhibitory effect on sugar absorption in rat
small intestinal fragments [82]. Conclusion and future directions
Natural products have been the most productive source of bioactive
Momordin Ic (25 and 50 mg/kg) and oleanolic acid 3-O-glucuronide molecules in drug discovery because of their diverse structures and
(50 mg/kg) suppressed gastric emptying in rats. An in vitro study functions in numerous physiological processes [92]. Triterpenoids
showed that these compounds inhibited glucose uptake in rat small are widely distributed in the plant kingdom. Currently, triterpenoids
intestine, concentration dependently, from 0.005-0.5 mM [22,23]. have become a focus in drug research and development for their
numerous biological and pharmacological properties.
Inhibition of gluconeogenesis
Gluconeogenesis is a metabolic pathway that results from the Many plants with triterpenoids as their major components are used
synthesis of glucose from other organic compounds. Individuals in various countries as traditional antidiabetic medicines. A large
with type 2 DM often show an increase in gluconeogenesis. number of investigations have confirmed their positive impact as
Gluconeogenesis inhibitors are potential therapeutic agents in the potential pharmaceutical agents in the treatment of diabetes. As
treating of diabetes [83]. Corosolic acid decreased gluconeogenesis demonstrated in this review, natural triterpenoids exhibit a wide
in perfused rat liver and in isolated hepatocytes in a dose range of spectrum of pharmacological activities against diabetes mellitus.
20 to 100 μM [84]. Some triterpenoids also showed multi-target pharmaceutical effects.
Ursolic acid could stimulate insulin secretion and reverse insulin
Inhibition of 11β-hydroxysteroid dehydrogenase type 1 resistance at the same time. Its properties involve multiple
11β-Hydroxysteroid dehydrogenase type 1 (11β-HSD1) is the mechanisms including activation of insulin signaling pathway;
enzyme that converts inactive 11-ketoglucocorticoids into active inhibition of PTP1B, GP, 11β-HSD1, α-glucosidase and α-amylase;
11β-hydroxyforms in metabolically relevant tissues such as the and activation of AMPK and PPARs.
liver, adipose tissue, skeletal muscles and pancreatic β-cells.
Prolonged exposure to elevated glucocorticoids as a result of
1584 Natural Product Communications Vol. 11 (10) 2016 Lyu et al.

All reported experimental results strongly suggest that triterpenoids mechanisms. Toxicity of these compounds needs further
are promising candidates for the development of novel therapies for examination before they can be used as potential sources for novel
diabetes mellitus. However, only very few compounds have been drug development.
evaluated in preclinical animal models. The complex pathology of
diabetes is a major challenge to develop suitable animal models. Acknowledgements - This work was funded by a project of the
Many studies showed that triterpenoids, especially some National Natural Science Foundation of China (21102058), Natural
triterpenoid glycosides, were metabolized and restructured into new Science Foundation of Jiangsu Province of China (BK20141387),
compounds by intestinal bacteria [93,94]. Further experimental and and a Jiangsu Province Science and Technology Infrastructure
clinical studies are required to elucidate and verify the active Construction plan (BM2011117).
compounds in vivo and detailed anti-diabetic molecular

References

[1] American Diabetes Association. (2009) Diagnosis and classification of diabetes mellitus. Diabetes Care, 33, (Supplement-1), S62–S67.
[2] Salim B. (2005) Diabetes mellitus and its treatment. International Journal of Diabetes and Metabolism, 13, 111-134.
[3] Li WL, Zheng HC, Bukuru J, De Kimpe N. (2004) Natural medicines used in the traditional Chinese medical system for therapy of diabetes
mellitus. Journal of Ethnopharmacology, 92, 1-21.
[4] Alqahtani A, Hamid K, Kam A, Wong KH, Abdelhak Z, Razmovski-Naumovski V, Chan K, Li KM, Groundwater PW, Li GQ. (2013) The
pentacyclic triterpenoids in herbal medicines and their pharmacological activities in diabetes and diabetic complications. Current Medicinal
Chemistry, 20, 908-931.
[5] Harlev E, Nevo E, Mirsky N, Ofir R. (2013) Antidiabetic attributes of desert and steppic plants: a review. Planta Medica, 79, 425-436.
[6] Chen J, Mangelinckx S, Adams A, Wang ZT, Li WL, De Kimpe N, Li WL. (2015) Natural flavonoids as potential herbal medication for the
treatment of diabetes mellitus and its complications. Natural Product Communications, 10, 187-200.
[7] Dzubak P, Hajduch M, Vydra D, Hustova A, Kvasnica M, Biedermann D, Markova L, Urban M, Sarek J. (2006) Pharmacological activities of
natural triterpenoids and their therapeutic implications. Natural Product Reports, 23, 394-411.
[8] Bis hayee A, Ahmed S, Brankov N, Perloff M. (2011) Triterpenoids as potential agents for the chemoprevention and therapy of breast cancer.
Frontiers in Bioscience, 16, 980-996.
[9] Park MW, Ha J, Chung SH. (2008) 20(S)-Ginsenoside Rg3 enhances glucose-stimulated insulin secretion and activates AMPK. Biological &
Pharmaceutical Bulletin, 31, 748-751.
[10] Yokozawa T, Kobayashi T, Oura H, Kawashima Y. (1985) Studies on the mechanism of the hypoglycemic activity of ginsenoside-Rb2 in
streptozotocin-diabetic rats. Chemical & Pharmaceutical Bulletin, 33, 869-872.
[11] Lee WK, Kao ST, Liu IM, Cheng JT. (2006) Increase of insulin secretion by ginsenoside Rh2 to lower plasma glucose in Wistar rats. Clinical and
Experimental Pharmacology and Physiology, 33, 27-32.
[12] Gong YH, Jiang JX, Li Z, Zhu LH, Zhang ZZ. (1991) Hypoglycemic effect of sanchinoside C1 in alloxan-diabetic mice. Yao Xue Xue Bao, 26, 81-
85
[13] Cho WC, Chung WS, Lee SK, Leung AW, Cheng CH, Yue KK. (2006) Ginsenoside Re of Panax ginseng possesses significant antioxidant and
antihyperlipidemic efficacies in streptozotocin-induced diabetic rats. European Journal of Pharmacology, 21, 173-179.
[14] Norberg A, Hoa NK, Liepinsh E, Van Phan D, Thuan ND, Jörnvall H, Sillard R, Ostenson CG. (2004) A novel insulin-releasing substance,
phanoside, from the plant Gynostemma pentaphyllum. The Journal of Biological Chemistry, 279, 41361-41367.
[15] Xu A, Wang H, Hoo RL, Sweeney G, Vanhoutte PM, Wang Y, Wu D, Chu W, Qin G, Lam KS. (2009) Selective elevation of adiponectin
production by the natural compounds derived from a medicinal herb alleviates insulin resistance and glucose intolerance in obese mice.
Endocrinology, 150, 625-633.
[16] Yu J, Zhang Y, Sun S, Shen J, Qiu J, Yin X, Yin H, Jiang S. (2006) Inhibitory effects of astragaloside IV on diabetic peripheral neuropathy in rats.
Canadian Journal of Physiology and Pharmacology, 84, 579-587.
[17] Tan MJ, Ye JM, Turner N, Hohnen-Behrens C, Ke CQ, Tang CP, Chen T, Weiss HC, Gesing ER, Rowland A, James DE, Ye Y. (2008)
Antidiabetic activities of triterpenoids isolated from bitter melon associated with activation of the AMPK pathway. Chemistry & Biology, 15,
263-273.
[18] Sugihara Y, Nojima H, Matsuda H, Murakami T, Yoshikawa M, Kimura I. (2000) Antihyperglycemic effects of gymnemic acid IV, a compound
derived from Gymnema sylvestre leaves in streptozotocin-diabetic mice. Journal of Asian Natural Products Research, 2, 321-327.
[19] Kako M, Miura T, Nishiyama Y, Ichimaru M, Moriyasu M, Kato A. (1997) Hypoglycemic activity of some triterpenoid glycosides. Journal of
Natural Products, 60, 604-605.
[20] Choi J, Shin MH, Park KY, Lee KT, Jung HJ, Lee MS, Park HJ. (2004) Effect of kaikasaponin III obtained from Pueraria thunbergiana flowers on
serum and hepatic lipid peroxides and tissue factor activity in the streptozotocin-induced diabetic rat. Journal of Medicinal Food, 7, 31-37.
[21] Manna P, Sinha M, Sil PC. (2009) Protective role of arjunolic acid in response to streptozotocin-induced type-I diabetes via the mitochondrial
dependent and independent pathways. Toxicology, 257, 3-63.
[22] Matsuda H, Li Y, Murakami T, Matsumura N, Yamahara J, Yoshikawa M. (1998) Antidiabetic principles of natural medicines. III. Structure-related
inhibitory activity and action mode of oleanolic acid glycosides on hypoglycemic activity. Chemical & Pharmaceutical Bulletin, 46, 1399-1403.
[23] Matsuda H, Murakami T, Shimada H, Matsumura N, Yoshikawa M, Yamahara J. (1997) Inhibitory mechanisms of oleanolic acid 3-O-
monodesmosides on glucose absorption in rats. Biological & Pharmaceutical Bulletin, 20, 717-719.
[24] Kalaiarasi P, Pugalendi KV. (2009) Antihyperglycemic effect of 18 beta-glycyrrhetinic acid, aglycone of glycyrrhizin, on streptozotocin-diabetic
rats. European Journal of Pharmacology, 606, 269-273.
[25] Hsu JH, Wu YC, Liu IM, Cheng JT. (2006) Release of acetylcholine to raise insulin secretion in Wistar rats by oleanolic acid, one of the active
principles contained in Cornus officinalis. Neuroscience Letters, 404, 112-116.
[26] De Tommasi N, De Simone F, Cirino G, Cicala C, Pizza C. (1991) Hypoglycemic effects of sesquiterpene glycosides and polyhydroxylated
triterpenoids of Eriobotrya japonica. Planta Medica, 57, 414-416.
[27] Yoshikawa M, Matsuda H, Harada E, Murakami T, Wariishi N, Yamahara J, Murakami N. (1994) Elatoside E, a new hypoglycemic principle from
the root cortex of Aralia elata Seem.: Structure-related hypoglycemic activity of oleanolic acid glycosides. Chemical & Pharmaceutical Bulletin,
42, 1354-13561.
[28] Yoshikawa M, Murakami T, Harada E, Murakami N, Yamahara J, Matsuda H. (1996) Bioactive saponins and glycosides. VII. On the hypoglycemic
principles from the root cortex of Aralia elata Seem.: Structure-related hypoglycemic activity of oleanolic acid oligoglycoside. Chemical &
Pharmaceutical Bulletin, 44, 1923-1927.
Anti-diabetic natural triterpenoids Natural Product Communications Vol. 11 (10) 2016 1585

[29] Yoshikawa M, Harada E, Murakami T, Matsuda H, Wariishi N, Yamahara J, Murakami N, Kitagawa I. (1994) Escins-Ia, Ib, IIa, IIb, and IIIa,
bioactive triterpene oligoglycosides from the seeds of Aesculus hippocastanum L.: their inhibitory effects on ethanol absorption and hypoglycemic
activity on glucose tolerance test. Chemical & Pharmaceutical Bulletin, 42, 1357-1359.
[30] Ivorra MD, Paya M, Villar A. (1988) Hypoglycemic and insulin release effects of tormentic acid: a new hypoglycemic natural product. Planta
Medica, 54, 282-285.
[31] Jayaprakasam B, Olson LK, Schutzki RE, Tai MH, Nair MG. (2006) Amelioration of obesity and glucose intolerance in high-fat-fed C57BL/6 mice
by anthocyanins and ursolic acid in Cornelian cherry (Cornus mas). Journal of Agricultural and Food Chemistry, 54, 243-248.
[32] Takagi S, Miura T, Ishibashi C, Kawata T, Ishihara E, Gu Y, Ishida T. (2008) Effect of corosolic acid on the hydrolysis of disaccharides. Journal of
Nutritional Science and Vitaminology, 54, 266-268.
[33] Chen J, Li WL, Wu JL, Ren BR, Zhang HQ. (2008) Euscaphic acid, a new hypoglycemic natural product from Folium Eriobotryae. Die Pharmazie,
63, 765-767.
[34] Tabatabaei-Malazy O, Larijani B, Abdollahi M. (2012) A systematic review of in vitro studies conducted on effect of herbal products on secretion
of insulin from Langerhans islets. Journal of Pharmaceutical Sciences, 15, 447-466..
[35] Bansal P, Wang Q. (2008) Insulin as a physiological modulator of glucagon secretion. American Journal of Physiology-Endocrinology and
Metabolism, 295, E751-761.
[36] Teodoro T, Zhang L, Alexander T, Yue J, Vranic M, Volchuk A. (2008) Oleanolic acid enhances insulin secretion in pancreatic beta-cells. FEBS
Letters, 582, 1375-1380.
[37] Zhang Y, Jayaprakasam B, Seeram NP, Olson LK, DeWitt D, Nair MG. (2004) Insulin secretion and cyclooxygenase enzyme inhibition by cabernet
sauvignon grape skin compounds. Journal of Agricultural and Food Chemistry, 52, 228-233.
[38] Gilon P, Henquin JC. (2001) Mechanisms and physiological significance of the cholinergic control of pancreatic beta-cell function. Endocrine
Reviews, 22, 565-604.
[39] Koster JC, Permutt MA, Nichols CG. (2005) Diabetes and insulin secretion: the ATP-sensitive k+ channel (k ATP) connection. Diabetes, 54, 3065-
3072.
[40] Reaven GM. (1993) Role of insulin resistance in human disease (syndrome X): an expanded definition. Annual Review of Medicine, 44, 121-131.
[41] Lee WK, Kao ST, Liu IM, Cheng JT. (2007) Ginsenoside Rh2 is one of the active principles of Panax ginseng root to improve insulin sensitivity in
fructose-rich chow-fed rats. Hormone and Metabolic Research, 39, 347-354.
[42] Le Roith D, Zick Y. (2001) Recent advances in our understanding of insulin action and insulin resistance. Diabetes Care, 24, 588-597.
[43] Choi K, Kim YB. (2010) Molecular mechanism of insulin resistance in obesity and type 2 diabetes. The Korean Journal of Internal Medicine, 25,
119-129.
[44] Shang W, Yang Y, Zhou L, Jiang B, Jin H, Chen M. (2008) Ginsenoside Rb1 stimulates glucose uptake through insulin-like signaling pathway in
3T3-L1 adipocytes. Journal of Endocrinology, 198, 561-569.
[45] Shang W,Yang Y, Jiang B, Jin H, Zhou L, Liu S, Chen M. (2007) Ginsenoside Rb1 promotes adipogenesis in 3T3-L1 cells by enhancing
PPARgamma2 and C/EBP alpha gene expression. Life Sciences, 80, 618-625.
[46] Jung SH, Ha YJ, Shim EK, Choi SY, Jin JL, Yun-Choi HS, Lee JR. (2007) Insulin-mimetic and insulin-sensitizing activities of a pentacyclic
triterpenoid insulin receptor activator. Biochemical Journal, 403, 243-250.
[47] Shi L, Zhang W, Zhou YY, Zhang YN, Li JY, Hu LH, Li J. (2008) Corosolic acid stimulates glucose uptake via enhancing insulin receptor
phosphorylation. European Journal of Pharmacology, 584, 21-29.
[48] Thareja S, Aggarwal S, Bhardwaj TR, Kumar M. (2012) Protein tyrosine phosphatase 1B inhibitors: a molecular level legitimate approach for the
management of diabetes mellitus. Medicinal Research Reviews, 32,459-517.
[49] Na M, Yang S, He L, Oh H, Kim BS, Oh WK, Kim BY, Ahn JS. (2006) Inhibition of protein tyrosine phosphatase 1B by ursane-type triterpenes
isolated from Symplocos paniculata. Planta Medica, 72, 261-263.
[50] Ramírez-Espinosa JJ, Rios MY, López-Martínez S, López-Vallejo F, Medina-Franco JL, Paoli P, Camici G, Navarrete-Vázquez G, Ortiz-Andrade
R, Estrada-Soto S. (2011) Antidiabetic activity of some pentacyclic acid triterpenoids, role of PTP-1B: in vitro, in silico, and in vivo approaches.
European Journal of Medicinal Chemistry, 46, 2243-2251.
[51] Thuong PT, Lee CH, Dao TT, Nguyen PH, Kim WG, Lee SJ, Oh WK. (2008) Triterpenoids from the leaves of Diospyros kaki (persimmon) and
their inhibitory effects on protein tyrosine phosphatase 1B. Journal of Natural Products, 10, 1775-1778.
[52] Na M, Thuong PT, Hwang IH, Bae K, Kim BY, Osada H, Ahn JS. (2010) Protein tyrosine phosphatase 1B inhibitory activity of 24-norursane
triterpenes isolated from Weigela subsessilis. Phytotherapy Research, 24, 1716-1719.
[53] Kwon JH, Chang MJ, Seo HW, Lee JH, Min BS, Na M, Kim JC, Woo MH, Choi JS, Lee HK, Bae K. (2008) Triterpenoids and a sterol from the
stem-bark of Styrax japonica and their protein tyrosine phosphatase 1B inhibitory activities. Phytotherapy Research, 22, 1303-1306.
[54] Na M, Cui L, Min BS, Bae K, Yoo JK, Kim BY, Oh WK, Ahn JS. (2006) Protein tyrosine phosphatase 1B inhibitory activity of triterpenes isolated
from Astilbe koreana. Bioorganic & Medicinal Chemistry Letters, 16, 3273-3276.
[55] Choi YH, Zhou W, Oh J, Choe S, Kim DW, Lee SH, Na M. (2012) Rhododendric acid A, a new ursane-type PTP1B inhibitor from the endangered
plant Rhododendron brachycarpum G. Don. Bioorganic & Medicinal Chemistry Letters, 22, 6116-6119.
[56] Na M, Kim BY, Osada H, Ahn JS. (2009) Inhibition of protein tyrosine phosphatase 1B by lupeol and lupenone isolated from Sorbus commixta.
Journal of Enzyme Inhibition and Medicinal Chemistry, 24, 1056-1059.
[57] Choi JY, Na M, Hyun Hwang I, Ho Lee S, Young Bae E, Yeon Kim B, Seog Ahn J. (2009) Isolation of betulinic acid, its methyl ester and guaiane
sesquiterpenoids with protein tyrosine phosphatase 1B inhibitory activity from the roots of Saussurea lappa C. B. Clarke. Molecules, 14, 266-272.
[58] Heilbronn LK, Smith SR, Ravussin E. (2003) The insulin-sensitizing role of the fat derived hormone adiponectin. Current Pharmaceutical Design,
9, 1411-1418.
[59] Winder WW, Hardie DG. (1999) AMP-activated protein kinase, a metabolic master switch: possible roles in type 2 diabetes. American Journal of
Physiology, 277, E1-10.
[60] Ha do T, Tuan DT, Thu NB, Nhiem NX, Ngoc TM, Yim N, Bae K. (2009) Palbinone and triterpenes from Moutan Cortex (Paeonia suffruticosa,
Paeoniaceae) stimulate glucose uptake and glycogen synthesis via activation of AMPK in insulin-resistant human HepG2 Cells. Bioorganic &
Medicinal Chemistry Letters, 19, 5556-5559.
[61] Terauchi Y, Kadowaki T. (2005) Peroxisome proliferator-activated receptors and insulin secretion. Endocrinology, 146, 3263-3265.
[62] Bermúdez V, Finol F, Parra N, Parra M, Pérez A, Peñaranda L, Vílchez D, Rojas J, Arráiz N, Velasco M. (2010) PPAR-gamma agonists and their
role in type 2 diabetes mellitus management. American Journal of Therapeutics, 17, 274-283.
[63] Han KL, Jung MH, Sohn JH, Hwang JK. (2006) Ginsenoside 20S-protopanaxatriol (PPT) activates peroxisome proliferator- activated receptor
gamma (PPARgamma) in 3T3-L1 adipocytes. Biological & Pharmaceutical Bulletin, 29, 110-113.
[64] Mensink M, Blaak EE, van Baak MA, Wagenmakers AJ, Saris WH. (2001) Plasma free fatty acid uptake and oxidation are already diminished in
subjects at high risk for developing type 2 diabetes. Diabetes, 50, 2548-2554
1586 Natural Product Communications Vol. 11 (10) 2016 Lyu et al.

[65] Jiang B, Yang Y, Jin H, Shang W, Zhou L, Qian L, Chen M. (2008) Astragaloside IV attenuates lipolysis and improves insulin resistance induced
by TNFalpha in 3T3-L1 adipocytes. Phytotherapy Research, 22, 1434-1439.
[66] Cline GW, Johnson K, Regittnig W, Perret P, Tozzo E, Xiao L, Damico C, Shulman GI. (2002) Effects of a novel glycogen synthase kinase-3
inhibitor on insulin-stimulated glucose metabolism in Zucker diabetic fatty (fa/fa) rats. Diabetes, 51, 2903-2910.
[67] Praly JP, Vidal S. (2010) Inhibition of glycogen phosphorylase in the context of type 2 diabetes, with focus on recent inhibitors bound at the active
site. Mini-Reviews in Medicinal Chemistry, 10, 1102-1126.
[68] Lee J, Lee HI, Seo KI, Cho HW, Kim MJ, Park EM, Lee MK. (2014) Effects of ursolic acid on glucose metabolism, the polyol pathway and
dyslipidemia in non-obese type 2 diabetic mice. Indian Journal of Experimental Biology, 52, 683-691.
[69] Luo JG, Liu J, Kong LY. (2008) New pentacyclic triterpenes from Gypsophila oldhamiana and their biological evaluation as glycogen
phosphorylase inhibitors. Chemistry & Biodiversity, 5, 751-757.
[70] Guan T, Li Y, Sun H, Tang X, Qian Y. (2009) Effects of maslinic acid, a natural triterpene, on glycogen metabolism in cultured cortical astrocytes.
Planta Medica, 75, 1141-1143.
[71] Mosa RA, Cele ND, Mabhida SE, Shabalala SC, Penduka D, Opoku AR. (2015) In vivo antihyperglycemic activity of a lanosteryl triterpene from
Protorhus longifolia. Molecules, 20, 13374-13383.
[72] Van de Laar FA. (2008) Alpha-glucosidase inhibitors in the early treatment of type 2 diabetes. Vascular Health and Risk Management, 4, 1189-
1195.
[73] Tundis R, Loizzo MR, Menichini F. (2010) Natural products as alpha-amylase and alpha-glucosidase inhibitors and their hypoglycaemic potential
in the treatment of diabetes: an update. Mini-Reviews in Medicinal Chemistry, 10, 315-331.
[74] Takagi S, Miura T, Ishibashi C, Kawata T, Ishihara E, Gu Y, Ishida T. (2008) Effect of corosolic acid on the hydrolysis of disaccharides. Journal of
Nutritional Science and Vitaminology, 54, 266-268.
[75] Ali H, Houghton PJ, Soumyanath A. (2006) alpha-Amylase inhibitory activity of some Malaysian plants used to treat diabetes; with particular
reference to Phyllanthus amarus. Journal of Ethnopharmacology, 107, 449-455.
[76] Gowri PM, Tiwari AK, Ali AZ, Rao JM. (2007) Inhibition of alpha-glucosidase and amylase by bartogenic acid isolated from Barringtonia
racemosa Roxb. seeds. Phytotherapy Research, 21,796-799.
[77] Atta-ur-Rahman Zareen S, Choudhary MI, Choudhary MI, Akhtar MN, Khan SN. (2008) alpha-Glucosidase inhibitory activity of triterpenoids from
Cichorium intybus. Journal of Natural Products, 71, 910-913.
[78] Deutschlӓndera MS, Lall N, Van de Venter M, Hussein AA. (2011) Hypoglycemic evaluation of a new triterpene and other compounds isolated
from Euclea undulata Thunb. var. myrtina (Ebenaceae) root bark. Journal of Ethnopharmacology, 133, 1091-1095
[79] Lai YC, Chen CK, Tsai SF, Lee SS. (2012) Triterpenes as α-glucosidase inhibitors from Fagus hayatae. Phytochemistry, 74, 206-211
[80] Uddin G, Rauf A, Al-Othman AM, Collina S, Arfan M, Ali A, Khan I. (2012) Pistagremic acid, a glucosidase inhibitor from Pistacia integerrima.
Fitoterapia, 83, 1648-1652.
[81] Kwon O, Eck P, Chen S, Corpe CP, Lee JH, Kruhlak M, Levine M. (2007) Inhibition of the intestinal glucose transporter GLUT2 by flavonoids.
Faseb Journal, 21, 366-377.
[82] Yoshikawa M, Murakami T, Matsuda H. (1997) Medicinal foodstuffs. X. Structures of new triterpene glycosides, gymnemosides-c, -d, -e, and -f,
from the leaves of Gymnema sylvestre R. Br.: influence of gymnema glycosides on glucose uptake in rat small intestinal fragments. Chemical &
Pharmaceutical Bulletin, 45, 2034-2038.
[83] Kashiwagi A. (1995) Rationale and hurdles of inhibitors of hepatic gluconeogenesis in treatment of diabetes mellitus. Diabetes Research and
Clinical Practice, 28 Suppl, S195-200.
[84] Yamada K, Hosokawa M, Fujimoto S, Fujiwara H, Fujita Y, Harada N, Yamada C, Fukushima M, Ueda N, Kaneko T, Matsuyama F, Yamada Y,
Seino Y, Inagaki N. (2008) Effect of corosolic acid on gluconeogenesis in rat liver. Diabetes Research and Clinical Practice, 80, 48-55.
[85] Wamil M, Seckl JR. (2007) Inhibition of 11β-hydroxysteroid dehydrogenase type 1 as a promising therapeutic target. Drug Discovery Today, 12,
504-520
[86] Stulnig TM, Waldhäusl W. (2004) 11β-Hydroxysteroid dehydrogenase type 1 in obesity and type 2 diabetes. Diabetologia, 47, 1-11.
[87] Rollinger JM, Kratschmar DV, Schuster D, Pfisterer PH, Gumy C, Aubry EM, Brandstötter S, Stuppner H, Wolber G, Odermatt A. (2010) 11β-
Hydroxysteroid dehydrogenase 1 inhibiting constituents from Eriobotrya japonica revealed by bioactivity-guided isolation and computational
approaches. Bioorganic & Medicinal Chemistry, 18, 1507-1515
[88] Vuorinen A, Seibert J, Papageorgiou VP, Rollinger JM, Odermatt A, Schuster D, Assimopoulou AN. (2015) Pistacia lentiscus oleoresin: virtual
screening and identification of masticadienonic and isomasticadienonic acids as inhibitors of 11β-hydroxysteroid dehydrogenase 1. Planta Medica,
81, 525-532.
[89] Deacon CF, Johnsen, AH, Holst JJ. (1995) Degradation of glucagon-like peptide-1 by human plasma in vitro yields an N-terminally truncated
peptide which is a major endogenous metabolite in vivo. Journal of Clinical Endocrinology and Metabolism, 80, 952-957.
[90] Li S, Cui B, Liu Q, Tang L, Yang Y, Jin X, Shen Z. (2012) New triterpenoids from the leaves of Cyclocarya paliurus. Planta Medica, 78, 290-296.
[91] Saleem S, Jafri L, ul Haq I, Chang LC, Calderwood D, Green BD, Mirza B. (2014) Plants Fagonia cretica L. and Hedera nepalensis K. Koch
contain natural compounds with potent dipeptidyl peptidase-4 (DPP-4) inhibitory activity. Journal of Ethnopharmacology, 156, 26-32.
[92] Harvey AL. (2008) Natural products in drug discovery. Drug Discovery Today, 13, 894-901.
[93] Bae EA, Park SY, Kim DH. (2000) Constitutive beta-glucosidases hydrolyzing ginsenoside Rb1 and Rb2 from human intestinal bacteria. Biological
& Pharmaceutical Bulletin, 23, 1481-1485
[94] Kim DH, Hong SW, Kim BT, Bae EA, Park HY, Han MJ. (2000) Biotransformation of glycyrrhizin by human intestinal bacteria and its relation to
biological activities. Archives of Pharmacal Research, 23, 172-177.

You might also like