Perez-Gutierrez 2012

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 9

1516 Regular Article Biol. Pharm. Bull. 35(9) 1516–1524 (2012) Vol. 35, No.

Meliacinolin: A Potent α-Glucosidase and α-Amylase Inhibitor Isolated


from Azadirachta indica Leaves and in Vivo Antidiabetic Property in
Streptozotocin-Nicotinamide-Induced Type 2 Diabetes in Mice
Rosa Martha Perez-Gutierrez*,a and Monica Damian-Guzmanb
a
Laboratorio de Investigación de Productos Naturales, Escuela Superior de Ingeniería Química e Industrias
Extractivas IPN; Av Instituto Politecnico S/N, Col Zacatenco, CP 07758, México D.F.: and b Departamento de
Tecnologia de Alimentos, Escuela Nacional de Ciencias Biologicas IPN; Carpio/Plan de Ayala S/N, Casco Santo
Tomas, CP 0734, México D.F. Received March 13, 2012; accepted June 7, 2012

In India, Azadirachta indica is typically known as ‘neem tree’ and its leaves has long been used in
the ayurvedic medical tradition as a treatment for diabetes mellitus. In-depth chromatographic investiga-
tion on chloroform extract resulted in identification of one new tetranortriterpenoid. Structural elucida-
tion was established on the basis of spectral data as 24,25,26,27-tetranor-apotirucalla-(apoeupha)-1α-
senecioyloxy-3α,7α-dihydroxy-14,20,22-trien-21,23-epoxy named by us as meliacinolin (1). The present study
investigated the effect hypoglycaemic, hypolipidemic, oxidative stress, insulin resistance, α-glucosidase and
α-amylase of 1 from A. indica. Diabetic rats were treated with 1 for 28 d and a set of biochemical parameters
were studied including: glucose level, total cholesterol, triglycerides, lipid peroxidation, liver and muscle gly-
cogen, superoxide dismutase, catalase, glutathione peroxidase and glutathione reductase. We also looked into
liver function by determining glucose-6-phosphatase, glucokinase and hexokinase activities, and the effect on
insulin level. While in vitro inhibition of α-glucosidase and α-amylase enzyme activities were used as indices
of effect on glucose absorption. As a result we found that blood glucose level, serum biochemical parameters,
hepatic enzymes, thiobarbituric acid reactive substances, and insulin level were restored in streptozotocin
(STZ)-diabetic mice to normal levels with 1. Meliacinolin inhibited α-glucosidase and α-amylase activities.
We conclude that meliacinolin can efficiently inhibit insulin resistance, improvement of renal function, lipid
abnormalities, and oxidative stress, indicating that its therapeutic properties may be due to the interaction
of meliacinolin with multiple targets involved in diabetes pathogenesis. α-Glucosidase and α-amylase inhibi-
tors offer an effective strategy to lower the levels of post prandial hyperglycemia prevents the digestion of
carbohydrates.
Key words hypoglycaemic; hypolipidemic; insulin; neem

In India, Azadirachta indica is typically known as ‘neem headache.13) Oral administration of nimbidin demonstrated
tree’ and its leaves has long been used in the ayurvedic medi- significant hypoglycaemic effect in fasting rabbits.14) Azadi-
cal tradition as a treatment for diabetes mellitus and peptic ul- rachtolide possesses α-amylase and α-glucosidase inhibition,
cers. Recalling the insecticidal properties of neem, researchers anti-hyperglycemic and anti-lipidemic effect at dose of 50
began programs in the early 60’s to identify the active princi- and 100 mg/kg.15) In fact, this two tetranortriterpenoids are
ples and screen them against major crop pests. In the last two known as active ingredients in A. indica that are in partly re-
decades research on neem has been intensified and many agri- sponsible for the decrease in blood glucose in animal models
cultural and medical properties of the tree were rediscovered.1) of diabetes. The possible mechanisms underlying the hypo-
There has been a tremendous interest in this plant as can be glycaemic activity of the aqueous leaf extract have also been
evidenced by the voluminous work on its different parts have discussed indicated that A. indica leaf extract, in itself, was
been carried out by various groups, leading to the isolation found to have no action on peripheral utilization of glucose
and structure elucidation of more than 135 compounds.2) A. in- or on hepatic glycogen however, increase insulin release in
dica, belongs to the family of meliaceae.3) It is one of the most rat pancreas.16) In this study we took advantage of the partial
useful medicinal plants.4) Blood glucose lowering activity of protection exerted by suitable dosage of nicotinamide against
A. indica seed oil and leaf extracts have been reported in vari- the beta-cytotoxic effect of streptozotocin (STZ) to create a
ous models of diabetic animals.5–9) Ethanol extracts of neem new experimental diabetic syndrome in adult mice that ap-
leaves have also been shown to demonstrate anti-lipid per- pears closer to non-insulin-dependent diabetes mellitus type
oxidative, antihyperglycaemic and anti-hypercholesterolaemic 2 than other available animal models with regard to insulin
activities as well as reduce serum triglyceride level in diabetic responsiveness to glucose and meliacinolin. In conclusion, this
rat model.10) Chloroform extract produced attenuation of anti- novel diabetes type 2 syndrome with reduced pancreatic insu-
glycation non-enzimatic, increase level antioxidant enzymes, lin stores, which is similar to human diabetes type 2 in that it
glucose-6-phosphatase, glucokinase, hepatic glucogen and has a significant response to glucose may provide a particu-
insulin plasma levels.11) In other study aqueous extract of this larly advantageous. Streptozotocin-induced severely diabetic
plant increased body weight gain and decreased blood glucose rats and streptozotocin-nicotinamide-induced mildly diabetic
in rats12) whereas supplementation with leaf powder (1 g daily) mice were established to compare their characteristics and to
in diabetic reduced symtoms of polydypsia, polyphagia and investigate the hypoglycaemic effects of antidiabetic drugs.17)
α-Glucosidase is an enzyme that plays a central role
The authors declare no conflict of interest. in carbohydrate metabolism by hydrolysing the terminal
* To whom correspondence should be addressed. e-mail: rmpg@prodigy.net.mx © 2012 The Pharmaceutical Society of Japan
September 2012 1517

glycosidic bonds at the non-reducing end of saccharide poly- pooled together according to their similarities provides by thin
mers to release α-glucose. Much attention has been given layer chromatography analysis. The fraction F5 was the frac-
to α-glucosidase in the pharmaceutical community because tion that showed hypoglycemic properties. F5 was subjected
inhibition of its catalytic activity leads to impaired glucose to silica gel column chromatography eluted with chloroform–
absorption and a decrease in postprandial blood glucose ethyl acetate (4.5 : 1) to produce nine fractions (F4-1 to F4-9).
levels. Therefore, α-glucosidase is currently the preferred The active fraction F4-2 was subjected to chromatographed
target for the development of new antidiabetes agents and over silica gel column using acetone–hexane 1 : 3 to yield five
some α-glucosidase inhibitors have been developed as clini- subfractions (F42-1 to F42-5). The F42-5 fraction was further
cal drugs, such as acarbose and miglitol, these hypoglycemic purified by preparative plate using petroleum ether–ethyl ac-
agents have their limitations and are known to produce serious etate 1 : 4 and visualized with UV at 254 nm. Fraction F425-4
side effects.18) Pancreatic α-amylase is a key enzyme in the was separated by Sephadex LH-20 using a gradient of CHCl3–
digestive system and catalyses the initial step in hydrolysis MeOH (from 10 : 1 to 5 : 1) to yield 1 (687 mg).
of starch to maltose and finally to glucose. Degradation of Animals The study was conducted in male mice, weigh-
this dietary starch proceeds rapidly and leads to elevated post ing about 20–25 g. Before and during the experiment, animals
prandial hyperglycemia. It has been shown that activity of were fed a standard laboratory diet (Mouse Chow 5015, Pu-
human pancreatic α-amylase in the small intestine correlates rina) with free access to water. Mice were procured from the
to an increase in post-prandial glucose levels, the control of bioterio of Escuela Nacional de Ciencias Biologicas, and were
which is therefore an important aspect in treatment of diabe- housed in microloan boxes in a controlled environment (tem-
tes.19) Hence retardation of starch digestion by inhibition of perature 25±2°C). Animals were acclimatized for a period of
enzymes such as α-amylase would play a key role in the three days in their new environment before the initiation of
control of diabetes. However, the discovery of specific high- experiment. Litter in cages was renewed three times a week
affinity inhibitors of pancreatic α-amylase for the development to ensure hygiene and maximum comfort for animals. The
of therapeutics has remained elusive. In the present study, we experiments reported in this study were can out following
extended the research on the antidiabetic effects of A. indica the guidelines stated in Principles of Laboratory Animal Care
to examine the possible actions of the new tetranortriterpe- (NIH publication 85-23, revised 1985 and the Mexican Official
noid isolated from chloroform extract of A. indica on glucose Normativity (Norma oficial Mexicana NOM-062-Z00-1999).
levels, lipid metabolism, oxidative stress, insulin resistance, Food consumption and weight gain were measured daily.
α-glucosidase, α-amylase. Experimental Induction of Diabetes Type-2 diabetes
mellitus was induced in overnight fasted mice by administer-
MATERIALS AND METHODS ing a single dose of freshly prepared solution of streptozotocin
(STZ), 60 mg/kg body weight (b.w.) intraperitoneally (i.p.)) in
General Experimental Procedures IR spectra were 0.1 mol/L cold citrate buffer (pH 4.5), 15 min after the intra-
recorded on a Perkin-Elmer FTRI 1720X. 1H- and 13C-NMR peritoneal administration of 120 mg/kg nicotinamide. The STZ
spectra were taken at a Bruker DRX-300 NMR spectrometer, treated animals were allowed to drink 5% glucose solution
with UXNMR software package, was used for NMR experi- over night to overcome drug induced hypoglycemia. After 10 d
ments; chemical shifts are reported in δ (ppm), downfield rela- of development of diabetes, mice with moderate diabetes hav-
tive to tetramethylsilane (TMS) as an internal standard. The ing persistent glycosuria and hyperglycaemia (blood glucose
NMR experiments were carried out using the conventional >250 mg/dL) were used for further experimentation.20)
pulse sequences as described in the literature. High resolution- Experimental Design Mice were divided into seven
electron ionization (HR-EI)-MS were measured on a JEOL groups, of six animals in each: (i) Group I normoglucemic
HX 110 mass spectrometer. Precoated TLC silica gel 60 F254 served as control and received 2% w/v Tween 80; (ii) Group
aluminum sheets and Sephadex LH-20 from Sigma-Aldrich II served as diabetic control; (III) Groups III–V normal mice
(St. Louis, U.S.A.) were used. Column chromatography was for single dose of 1 at 10, 20 and 30 mg/kg; (IV) Groups
carried on Silica gel 60 (230–400 mesh, Merck Co., New VI–VIII diabetic mice for single dose of 1 at 10, 20 and
Jersey (U.S.A.), solvents used as eluents from Fermont (Cali- 30 mg/kg; Group IX diabetic mice administered with gliben-
fornia, U.S.A.). clamide 4 mg/kg. Ten days after the STZ injection, animals of
Plant Material The aerial part of the plant A. indica was groups III to VIII received 1 and glibenclamide, respectively.
collected in the month of August 2010 from Colima State Animals were fasted for 5 h prior to drug administration and
Colima. The plant material was taxonomically identified and blood samples were obtained 0, 2 h, 4 h, 6 h, 8 h and 12 h af-
the voucher specimen was deposited in Herbarium of Escuela ter administration of test substances. In a parallel study four
Nacional de Ciencias Biologicas, IPN for further reference groups (n=6) of diabetic rats were used to determine the
(No. 7216). chronic effect of 1. After 4 weeks, the animals were fasted
Extraction, Isolation and Characterization of the A. in- overnight and autopsied under light ether anesthesia. All the
dica Tetranortriterpenoid-Derived Plant material was air drugs solutions or vehicle were administered orally by gastric
dried and the ground leaves (25 kg) was extracted twice with intubations once daily at 9:00 a.m. for 28 d. At the end of the
chloroform each for 3 h. The plant extracts were combined and experiment the effect each group was investigated by serum
evaporated in vacuo to generate a residue (1972 g residue). The determination of total cholesterol (TC), triglycerides (TG) and
resulting extract was loaded onto a silica gel column chroma- high density lipoprotein (HDL)-cholesterol, using a commer-
tography and eluted with chloroform–ethylic ether 4 : 0.5 and cial Diagnostic Kit (Genzyme Diagnostics) and low density
7 fractions (F1–F7) have been obtained. These fractions were lipoprotein (LDL)-cholesterol was calculated as the remaining
then tested for hypoglycemic activity. Active fractions were difference of total cholesterol and HDL. Blood glucose levels
1518 Vol. 35, No. 9

were measured employing the glucose oxidase–peroxidase (mU) was defined as mmol of substrate molecules converted
(GOD–POD) method.21) Lipid peroxidation, that is, thiobarbi- by 1 mg protein per minute. GK activity was estimated as
turic acid reactive substances (TBARS) was estimated by the the difference in activity when samples were assayed at
method of Fraga et al.,22) and expressed as µM /g of liver and 100 mmol/L (GK plus hexokinase activity) and 0.5 mmol/L
kidney tissue. glucose (hexokinase activity).
Oral Glucose Tolerance Test Mice of each group were Measurements of Glutathione Reductase (GSH), Super-
orally administered meliacinolin at dose of 25 mg/kg body oxide Dismutase (SOD), Catalase Activity (CAT), Glutathi-
weight on a daily basis for 28 d. At the end of the experiment, one Peroxidase (GPx) and Lipid Peroxidation After 28 d
an oral glucose tolerance test (OGTT) was performed to as- of treatment, mice were fasted overnight and were euthanized
sess the animals sensitivity to a high glucose load. Overnight- by anesthesia. Antioxidant enzyme activities in the liver,
fasted mice were fed orally 2 g glucose/kg b.w. Blood samples pancreas and kidney were assayed using commercial kits:
were collected from the caudal vein from a small incision at SOD assay kit Bioxytech SOD-525 for SOD activity (Oxis
the end of the tail at 0 min (immediately after glucose load), International), catalase assay kit for CAT (Cayman Chemi-
30, 60, 90 and 120 min after glucose administration. cal), and GSH assay kit Bioxytech GR-340 for GR activity,
Assay of Glycogen Content in Liver and Skeletal Muscle (Oxis International), GPx assay kit GPx-340 for GPx (Oxis
and Glucose-6-phosphatase (G6Pase) and Glucokinase International) and lipid peroxidation using malondialdehyde
(GK) Activities in Liver Mice were sacrificed by decapita- level by commercial kit (thiobarbituric acid-reactive substance
tion, livers and kidneys were extracted. Frozen livers were (TBARS) Assay Kit). In the pancreas the protein concentra-
thawed, weighed and homogenized in Tris–HCl (5 mmol/L, tion was determined by the Bradford method as described in
pH 7.4) buffer containing 2 mmol/L ethylenediaminetetra- the Bio-Rad protein assay kit.
acetic acid (EDTA). Homogenates were centrifuged at 1000×g Determination of Serum Insulin Level, and Pancreatic
for 15 min at 4°C. The supernatant was mixed with glucose- Insulin Content Diabetic mice serum and pancreatic insulin
6-phosphate dehydrogenase and reduced nicotinamide ad- were measured by a Glazyme Insulin-EIA Test according to
enine dinucleotide phosphate (NADPH) and the activity of the manufacturer’s instructions.29) Blood samples and pancreas
hexokinase was determined at 37°C for 3 min at 30 s intervals were taken for insulin determination. The level of insulin in
at 340 nm.23) The hepatic glycogen content was measured ac- serum was expressed in µIU/mL.
cording to the anthrone–H2SO4 method.24) Briefly, liver tissue α-Amylase Inhibition The known porcine pancre-
(64–144 mg) was homogenized in five volumes of an ice-cold atic α-amylase (PPA) and acarbose were used for preliminary
30% KOH solution and the homogenate was placed in a boil- screening of α-amylase inhibitors from the compound. The
ing water-bath (l00°C) for 20 min. The glycogen was re-dis- inhibition assay was performed using the chromogenic DSA
solved in distilled water and treated with an anthrone reagent method.30) The total assay mixture composed of 500 µL of
(2 g anthrone/L of 95% (v/v) H 2SO4) and the absorbance was 0.02 M sodium phosphate buffer (pH 6.9 containing 6 m M so-
measured at 620 nm. dium chloride), 0.04 units of PPA solution, and 1 at concentra-
The hepatic G6Pase activity was assayed by the method of tion from 10–100 µg/mL were incubated at 37°C for 10 min.
Baginski et al.25) Shortly, the glucose-6-phosphate in the liver After pre-incubation, 500 µL of 1% (v/v) starch solution in the
extract was converted into glucose and inorganic phosphate. above buffer was added to each tube and incubated at 37°C
The inorganic phosphate liberated was determined with am- for 15 min. The reaction was terminated with 1.0 mL DNSA
monium molybdate; ascorbic acid was used as the reducing reagent, placed in boiling water bath for 5 min, cooled to room
agent. Excess molybdate was removed by the arsenite–citrate temperature, diluted, and the absorbance measured at 540 nm.
reagent, so that it could no longer react with other phosphate The control reaction representing 100% enzyme activity did
esters or with inorganic phosphate formed by acid hydrolysis not contain any compound. One unit of enzyme activity is
of the substrate. The amount of phosphate liberated per time defined as the amount of enzyme required to release one
unit, determined as the blue phosphor–molybdous complex at micromole of maltose from starch per min under the assay
700 nm, was a measure of the glucose-6-phosphatase activ- conditions. For the determination of the inhibitor concentra-
ity. The protein content of the liver extract was quantified by tion at which 50% inhibition of enzyme activity occurs (IC50),
Bradford reaction (Bio-Rad protein assay kit).26) The G6Pase the assay was performed as above except that the inhibitor/
activity (mU) was expressed as mmol of phosphate released/ compound concentrations were varied from 10–100 µg/mL.
min/mg of protein. Acarbose was used as a positive control at a concentration
GK activity was measured using a spectrophotometric range of 6.5–32.8 µg/mL. The IC50 values were determined
method as previously described by Panserat et al.27) Briefly, from plots of percent inhibition versus log inhibitor concentra-
liver tissues were homogenized and the supernatant obtained tion and calculated by logarithmic regression analysis from
by centrifugation was supplemented with 1 m M NADP, 5 m M the mea n inhibitory values. The IC50 values were defined as
ATP, and l00 or 0.5 m M glucose at pH 7.5. The enzymatic the concentration of the 1, containing the α-amylase inhibitor
reaction was started by the addition of 0.2 unit of glucose- that inhibited 50% of the PPA activity.
6-phosphate dehydrogenase (EC 1.1.1.49) and incubated for Inhibition of α-Glucosidase α-Glucosidase activity
5 min at 37°C. NADPH generated by GK was measured using was determined by incubating a solution (0.1 mL) of enzyme
a spectrophotometer at 340 nm. GK activity was estimated by preparation with Tris buffer, pH 8 (0.1 mL) containing increas-
the standard method, i.e. subtracting the rate of NADPH for- ing concentration (5, 10, 20, 40, 80, 100 µg/mL) of the (0.1 mL)
mation in the presence of 0.5 m M glucose from that obtained inhibitor at 37°C for 60 min. The reaction mixture was heated
in the presence of 100 m M glucose.28) Protein concentration for 2 min in a boiling water bath to stop the reaction. The
was quantified by Bradford and one unit of enzyme activity amount of liberated glucose was measured by glucose oxidase
September 2012 1519

method. The absorbance was measured at 540 nm.31) To fur- Compound 1 has a molecular formula on the basis of the
ther study the inhibitory property of 1 as an α-glucosidase positive HR-EI-MS showing a ion peak at m/z 510.2993 (Calcd
inhibitor, the concentration-dependent effect of 1 on the inhi- for C31H42O6, 510.2981). The IR spectrum revealed absorp-
bition of α-glucosidase was evaluated. Varying concentrations tion bands of hydroxyl (3423 cm−1), ester (1708 cm−1), ether
of 1 were added to the α-glucosidase-catalysing reaction. (1139, 1082 cm−1), and double bond (1462 cm−1) groups. The
13
Statistical Analysis Values obtained were expressed as C-NMR (distortionless enhancement by polarization transfer
mean±standard error mean (S.E.M.) and analyzed using the (DEPT)) exhibited 31 carbon atoms assigned to six methyl
statistical package for social sciences (SPSS) version 10 using group, five methylenes, twelve methines and eight quaternary
analysis of variance (ANOVA) followed by Dunnett’s test and carbon atoms one of which was carbonyl group. The tetran-
p<0.05 was considered to be significant. ortriterpenoid character of 1 was indicated by the presence of
four quaternary methyl singlets at δ H 0.93 (H-18), 0.95 (H-19),
RESULTS 1.12 (H-29), and 1.20 (H-30). The 1H-NMR spectrum further
showed two one-proton signals at δ H 5.32 (t, J=1.5 Hz) and δ H
Structural Elucidation of Tetranortriterpenoid 2.46 (m) responding to H-15 and H-17, respectively. These sig-
24,25,26,27-Tetranor-apotirucalla-(apoeupha)-1α-senecioyl- nals and their corresponding carbons at δC 119.6 and δC 51.9,
oxy-3α,7α-dihydroxy-14,20,22-trien-21,23-epoxy (1). IR ν max respectively, along with the carbons at δC 147.3 (C-14) and
(CHCl3) cm−1: 3423 (OH), 1708 (ester), 1462 (C=C), 1140 δC 34.5 (C-16) were suggestive of ring-D of 14,15 deoxy-ge-
and 1080 (ether linkage); HR-EI-MS (positive) m/z 510.2993 dunin.32) H-15 of 1 appeared downfield as compared to those
[M+H]+ Calcd for C31H42O6, 510.2981; electrospray ionization of deoxy-gedunin and nimocinol which supported the hydroxy
(ESI)-MS-MS m/z 510 [M], (2), 477 [M−CH3−H2O] (5), 443 substituent at C-7.33) Similarly, the double bond at C-14 re-
[C27H39O5] (2), 403 [C24H36O5] (2), 380 [C23H24O5] (100), 335 sulted in a downfield shift of H-7. Further, oxygen-bearing
[C19H27O5] (3), 304 [C18H24O4] (3), 284 [C14H19O3 or C15H24O5], methane signals at δC 79.6 and δ H 4.28 (d, J=3.0 Hz) displaced
(6), 236 [C17H16O] (6), 192 (17), 156 [C8H12O3] (1), 83 [C5H7O] an upfield methylene resonance indicating that a methylene
(3); 1H- and 13C-NMR (CDCl3) data, Table 1. carbon was substituted by a hydroxyl group. The positioning

Table 1. 300 MHz 1H- and 13C-NMR Spectral Data of Compound 1

C/H No. δH J (Hz) δC HMBC correlations protons

1 4.12 t (J=3.0) 74.1 H-3, H-19


2 2.00 m 29.6 H-2
3 4.20 t (J=3.0) 75.2 H-2, H-28, H-29
4 — — 44.2 H-3, H-28, H-29
5 2.35 d (J=12.0) 39.1 H-28, H-29
6 3.98 dd (J=12.0, 3.0) 76.3 H-5, H-7
7 4.28 d (J=3.0) 79.6 H-5, H-6
8 — — 44.2 H-7, H-9, H-15
9 2.28 dd (J=9.5, 3.0) 38.5 H-5, H-7, H-11
10 — — 41.5 H-2, H-5, H-6, H-11
11 1.61 m 17.5 H-9
12 1.65 m 30.6 H-9, H-11, H-18
13 — — 36.4 H-15, H-18
14 — — 147.3 H-7, H-15, H-17, H-18
15 5.32 t (J=1.5) 119.6 H-17
16 2.30 m 34.5 H-15, H-17
17 2.46 m 36.9 H-15, H-18, H-21
18 0.93 s 19.8 H-17
19 0.95 s 14.3 H-5, H-9
20 — — 122.5 H-17, H-21, H-22
21 7.12 dd, (J=1.6, 0.9) 146.0 H-17, H-23
22 6.80 dd, (J=1.6, 0.9) 112.6 H-17, H-21
23 7.36 t (J=1.5) 128.7 H-17, H-21
24a 4.12 d (J=7.5) 77.6 H-3, H-5, H-29
24b 3.72 d (J=7.5) 77.6 H-3, H-5, H-29
25 1.12 s 22.1 H-3, H-5, H-28
26 1.20 s 30.7 H-7, H-9
Osen
1′ — — 171.7 H-2′, H-3
2′ 5.67 (br s) 128.3 H-4′, H-5′
3′ — — 123.7 H-2′, H-4′, H-5′
4′ 1.46 (br s) 27.5 H-2′
5′ 2.12 (br s) 21.0 H-2′
Senecioiloxi substituent (Osen).
1520 Vol. 35, No. 9

74.1, δC-1′ 171.7, δC-2′ 128.3, δC-3′ 123.7δC-4′ 27.5, δC-5′ 21.0. The
interaction of C-1′ with H-1 in the HMBC plot displayed the
senecioyl moiety at C-1, which was also evident from the
mass fragment at m/z 83. Two one-proton double doublets at
δ H 3.98 (J=12.0, 3.0 Hz) and δ H 2.28 (J=9.5, 3.0 Hz) were at-
tributable to H-6 and H-9, respectively. The chemical shift and
multiplicity of H-6 suggested an oxygen function at C-6. The
oxygen at C-6 form an ether linkage between C-6 and C-28
(δ H-6 3.97, dd, J = 12.0, 3.0; δ H-28a 4.12 d, J=7.5 Hz; δ H-28b
3.72, d, J=7.5 Hz; δC-6 76.3, δC-28 77.6. HMBC data Table 1
allowed the connection of the moieties, in particular, cross-
peaks of Me-19, Me-25, and Me-18 indicated the structure
of ring A, including the placement of an senecioyloxy at C-1
and the hydroxyl at C-3, cross-peaks of H-23 (δ H 7.36) with
C-21 (δC 146.0) suggested the presence of an oxygen bridge
between C-23 and C-21, given rise to a furan ring. Interac-
tions of C-1′ with H-3 displayed the osen group at C-3, C-15
had a connectivity with H-17 while C-22 showed connectiv-
ity with H-17. Interactions in the NOESY spectrum showed
connectivity H-3 with H-6, H-19 and H-26. An interaction
Fig. 1. 1
H–1H COSY — Correlations Detected for Meliacinolin H-17 with H-23; and H-17 with H-26. The α orientation of
the side chain could be deduced from the spatial interac-
of the hydroxyl group at C-7 was based on observation of tion of H-18 with H-22 and that of H-17 with H-26 in the
the heteronuclear multiple bond connectivity (HMBC) cor- NOESY spectrum. 1H–1H correlation spectroscopy (COSY)
relation of the proton at δ H 4.28 (1 H, d, J=3.0 Hz, H-7) and correlations are showed in the Fig. 1. Thus, the structure of
the carbons at δC 39.1 (C-5), 44.2 (C-8), 38.5 (C-9), and 147.3 1 was determined as 24,25,26,27-tetranor-apotirucalla-(apo-
(C-14). One oxygenated methine signals appeared at δ H 4.20 eupha)-1α-senecioyloxy-3α,7α-dihydroxy-14,20,22-trien-21,23-
(t, J=3.0 Hz) and δC 75.2, this implied that a hydroxyl group is epoxy named by us as meliacinolin.
located at C-3. The assumption was confirmed by the correla- Normal, STZ-Induced Changes of Glucose Levels, Body
tions of Me-29 to C-3 in de HMBC experiment. According to Weight and Food and Water Intake Table 2 shows effect
the nuclear Overhauser effect spectroscopy (NOESY) correla- of a single dose treatment on blood glucose determined at
tions of H3-19 to H3-29 and H3-29 to H-3. This supported that various time intervals for 12 h. Treatment of 1 the mice with
the hydroxyl groups at C-3 and C-7 are axial. The 1H-NMR doses of 10, 20 and 30 mg/kg, control drug: glibenclamide
spectrum (Table 1) exhibited three downfield signals typi- 4 mg/kg, significantly decreased blood glucose in diabetic
cal of a furan side chain at δ H 7.12 (dd, J=1.6, 0.9 Hz, H-21), mice. The percentage of glucose reduction at 6 h was 22, 29
6.80 (dd, J=1.6, 0.9 Hz, H-22), 7.36 (t, J=1.5 Hz, H-23). This and 40%, respectively. Gibenclamide 4 mg/kg dose produced
was supported by the presence of a pair of doublet bonds at 41.0% of blood glucose reduction at 6 h. Maximum percent-
δC 122.5 (C-20), 146.0 (C-21), and 112.6 (C-22), 128.7 (C-23), age of blood glucose reduction to normoglycemic rats was
in the 13C-NMR spectrum (Table 1). These values for the achieved at 6 h with 30 mg/kg of 1 (57.33%). Gibenclamide
furan ring are comparable with those of deoxygedunin.32) In 4 mg/kg dose produced 30.5% blood glucose reduction at 8 h
ring A, a senecioyl substituent is present at C-1 (δ H-1 4.12, t, in no diabetic rats (Table 1).
J=3.0 Hz; δ H-2′ 5.67, br s; δ H-4′ 41.46, br s; δ H-5′ 2.12 br s; δC-1 In the chronic study, daily administration of 1 the body

Table 2. Effect of 1 on Fasting Blood Glucose Level in Diabetic Mice

At the time of Blood glucose levels (mg/dL) at different time intervals (h)
Group Dose (mg/kg)
grouping 2h 4h 6h 8h 12 h

Non-diabetic — 103.48±1.34 101.61±0.95 102.25±1.38 100.25±0.98 99.54±1.75 98.67±0.85


control
10 103.18±0.92 93.25±4.21a 86.31±1.95a 80.46±0.87a 85.22±3.51a 90.47±0.63a
Non-diabetic
20 102.35±3.45 85.43±5.38a 56.65±2.48a 51.06±1.54a 55.13±2.37a 66.47±1.39a
control+1
30 104.29±1.29 81.31±4.95a 49.61±2.38a 44.50±1.76a 52.56±2.85a 63.25±1.83a
Non-diabetic 4.0 99.56±5.19 89.49±5.22 82.67±4.39 74.13±4.73 69.21±3.89a 54.37±2.76a
control+GB
Diabetic control — 383.34±3.67 378.87±4.17 368.39±4.10 367.84±5.16 363.48±4.47 360.62±5.33
10 346.23±3.29 324.18±4.38b 311.68±4.80b 270.23±3.55b 274.19±2.76b 292.43±2.56b
Diabetic+1 20 369.68±2.81 328.43±3.43b 279.89±2.68b 262.14±5.89b 268.36±3.49b 276.52±4.24b
30 359.34±4.20 305.71±5.17b 258.48±3.36b 215.24±4.19b 219.87±1.87b 227.48±3.98b
Diabetic+GB 4.0 351.46±5.79 276.78±6.87b 205.19±2.59b 206.30±2.94b 212.57±1.98b 229.17±2.83b
Each values represent mean±S.D. (n=6). a p<0.05 compared to normal group (ANOVA) followed by Dunnett’s test. b p<0.05 compared to diabetic group (ANOVA) fol-
lowed by Dunnett’s test. Gibenclamide (GB).
September 2012 1521

weight and food, water intakes in control and experimental Mice Liver and Kidney after Treatment with Meliacinolin
animals are shown in Table 3; the induction of STZ-diabetes There was a significant elevation in serum triglycerides, total
resulted in elevated intake of both, significant decrease in cholesterol and TBARS levels in the liver and kidney of dia-
body weight during the 28 d was observed in the diabetic betic groups in comparison to control. Daily administration of
control mice compared with control mice, showing no differ- 1 at a dose of 20 mg/kg to diabetic mice for 28 d significantly
ence between initial and final values; however, administration reduced total cholesterol and triglycerides by 46.0% and 47%
of 1 to diabetic mice increased body weight gain significantly respectively. TBARS level in diabetic mice also decreased
(from 22.4 to 29.9 g). Diabetic mice showed increase in food after treatment with the compound, a 39.0% in liver and 31%
and water intakes as compared to normal control mice but the in kidney. Administration of this tetranortriterpenoid results
administration of meliacinolin led to a decrease in water and a significant diminution of these parameters and the levels of
food intake in experimental groups. these parameters were resettled toward the control level (Table
Oral Glucose Tolerance Test The effect of 1 on the 5). While HDLc, a friendly lipoprotein, was decreased in
blood glucose tolerance was determined after treatment by diabetic groups in respect to the control (Table 5). After 28 d
28 d. Postprandial blood glucose levels in mice show a sig- of administration of 1 there was a significant elevation of this
nificant change after glucose loading, increasing rapidly in lipoprotein level in serum and was resettled toward the control
all groups of diabetic mice within 30 min and remaining high level. The results also demonstrate a significant control of se-
over the next 120 min in diabetic control mice. Table 4 shows rum lipid profiles in the 1 treated diabetic mice.
the changes in the levels of blood glucose in diabetic control Effect of 1 on G6Pase, GK, Glycogen and Hexokinase
and experimental group after oral administration of glucose (HK) Activity Table 6 shows the effect of 1 on G6Pase, GK,
(2 g/kg). Oral treatment (20 mg/kg) in diabetic control mice HK activity and glycogen content of liver and skeletal muscle.
with 1 showed significant (p<0.05) reduction in blood glucose The administration of 1 increased the content of hepatic gly-
levels over 120 min compared to STZ-diabetic mice showed cogen, GK and HK in diabetic mice while G6Pase decreased.
47% decreased in glucose levels compared with STZ-diabetic The result showed that the hexokinase activity tended to be
mice. reversed to normal values, while normal mice did not exhibit
Determination of Serum Total Cholesterol, Triglyc- any significant alteration. Furthermore, the attenuating effect
erides, LDL-Cholesterol and TBARS Levels in Diabetic of this compound on experimental physiological symptoms of

Table 3. Effect on Physico-Metabolic Symptoms of Meliacinolin (1)

Body weight (g) Intake (g/d)


Group (mg/kg)
Initial Final Gain Food Water
a a a a
No-diabetic 23.7±2.7 39.5±6.3 15.8±5.2 21.6±3.3 40.8±5.6a
Diabetic 21.4±4.6c 23.2±5.8c 1.8±0.3c 30.8±2.9c 140.8±6.5c
1 (20 mg/kg) 22.4±5.5c 29.9±7.4b 7.5±2.7a 24.3±4.1c 100.2±7.0a
Each value represents mean±S.E.M. (n=10), ANOVA followed by multiple two-tail “t” test. In each vertical column, mean with different superscripts (a, b, c) differ from t′
each other significantly, p<0.05.

Table 4. Meliacinolin (1) on Oral Glucose Tolerance Test (OGTT) in Diabetic Rats

Blood glucose levels (mg/dL)


Groups
0 min 30 min 60 min 90 min 120 min
a a a
Normal 92.76±1.82 181.37±3.17 162.49±2.83 129.13±2.49 96.21±1.53a
Diabetic control 270.23±3.42 346.71±1.36b 412.86±5.21c 376.29±5.12c 351.63±3.71a
1 (20 mg/kg) 266.35±4.71 327.80±5.12b 247.36±4.71c 200.62±3.14c 141.29±1.45a
GB (4 mg/kg) 239.73±2.75 312.34±2.67b 221.43±3.28c 172.74±4.96c 138.56±2.53c
Values are mean±S.D. (n=6). ANOVA followed by multiple two tail “t” test. Line with (a, b, c) fixed within duration of measurement differ from each other significantly,
p>0.05.

Table 5. Effect of Meliacinolin (1) on Lipid Profile in STZ-Induced Diabetic Rats

Mean concentration (mg/g)±S.E.M.

Group (mg/kg) TBARS (µM/g)


Triglycerides (mg/dL) Total cholesterol (mg/dL)HDL-cholesterol (mg/dL)
Liver Kidney

Normal 92.26±4.86 130.10±6.71 68.24±5.21 0.98±0.006 1.7±0.03


Diabetic control 186.32±5.19a 246.32±2.74a 34.48±1.26a 1.62±0.05a 2.6±0.08a
1 (20 mg/kg) 90.87±4.19ab 130.68±5.82ab 58.53±2.85ab 0.99±0.08ab 1.8±0.05ab
GB 4 mg/kg 89.94±5.28ab 126.32±2.65ab 54.39±2.75ab 0.93±0.09ab 1.5±0.04ab
All values are expressed as mean±S.E.M., n=6 values. a p<0.05 when compared to normal control group, b p<0.01 when compared to diabetic control group, where the
significance was performed by one-way ANOVA followed by post hoc Dunnett’s test.
1522 Vol. 35, No. 9

Table 6. Effect of Meliacinolin (1) on Hepatic Glucose Regulation Enzyme Activities (G6Pase and GK), Hepatic Glycogen Content and Hexokinase
(HK) in Diabetic Mice

Glycogen (mg/g) HK activity (U/mg


Group (mg/kg) G6Pase activity (mU) GK activity (mU)
Liver Skeletal muscle protein)

Normal control 0.38±0.004 3.32±0.07 19.60±2.43 11.58±2.83 1.62±0.04


Diabetic control 0.69±0.006a 1.22±0.03a 9.25±1.65a 4.40±1.10a 1.30±0.02a
1 (20 mg/kg) 0.40±0.008ab 2.91±0.09ab d
17.86±1.98b d
10.84±2.38b d
1.59±0.05ab
GB 4 0.36±0.009b 3.11±0.07b 17.78±2.31b 11.01±1.98b 1.60±0.03ab
Each values represent mean±S.D. (n=6); ANOVA followed by multiple two tail “t” test. In each vertical column, mean with different superscripts (a, b) differ from each
other. All values are expressed as mean±S.D., n=6 values. Significant difference of diabetic control from normal control a p<0.001. Significant difference of treated groups
from diabetic control b p<0.01, c p<0.05. d p<0.01 when compared with glibenclamide 4 mg/kg treated group.

Table 7. Effect of Meliacinolin (1) on Antioxidant Enzyme Activities in Liver and Kidney in Diabetic Mice

Parameters Normal control Diabetic control Diabetic+1 (20 mg/kg) Diabetic+GB (4 mg/kg)
a b
SOD-Liver 7.51±1.67 3.82±0.76 6.75±0.50 6.80±0.41c
SOD-Kidney 13.79±2.43 7.16±1.81a 12.80±2.23b 13.025±1.57b
CAT-Liver 82.15±4.54 44.64±2.53a 69.75±4.21b 70.26±2.16b
CAT-Kidney 35.07±2.76 20.76±1.65a 33.41±2.47c 34.17±1.79b
GSH-Liver 47.21±2.80 21.80±1.97a 42.23±2.19b 42.38±2.28b
GSH-Kidney 24.18±5.36 5.79±2.06a 19.13±2.12b 19.27±3.58b
GPx-Liver 7.26±1.59 4.31±1.30a 5.72±4.37b 5.92±1.26b
GPx-Kidney 5.92±1.42 3.53±0.92a 4.76±2.48b 4.76±0.94b
All values are expressed as mean±S.E.M., n=6 values. a p<0.01 when compared to normal control group; b p<0.01 c p<0.05 compared to diabetic control group; where the
significance was performed by one-way ANOVA followed by post hoc Dunnett’s test. Glibenclamide (GB). The values are given in U/mg of protein.

Table 8. Effect of Meliacinolin (1) on Plasma Insulin Level and Pancre- Table 9. Inhibitory Effects of Meliacinolin (1) on α-Amylase and
atic Insulin Content in Diabetic Mice α-Glucosidase

Pancreatic insulin Concentration % Inhibition


Plasma insulin (µU/mL)
(µU/mL) (µg/mL)
Groups α-Amylase α-Glucosidase Acarbose
Before 1 adminis- After 1 adminis- After 1 adminis-
tration (0 h) tration (28 d) tration (28 d) 10 34.15±1.98 49.23±1.56 18.6±3.63
20 41.65±1.93 46.21±1.98 23.6±3.75
No-diabetic 3.51±0.78 3.60±0.21 25.78±4.12 40 54.78±2.65 57.30±3.67 28.7±3.21
control
60 68.53±2.87 69.48±2.93 39.16±2.54
Diabetic control 0.75±0.59 1.42±0.18a 14.87±3.76a
d
80 74.68±3.54 76.23±2.73 57.68±2.76
1 (20 mg/kg) 0.73±0.43 3.34±0.42b 22.41±1.84bc
100 82.49±3.17 85.48±3.90 68.39±1.96
Glibenclamide 0.74±0.61 3.46±0.27b 19.35±3.23bc
4 mg/kg
All values are expressed as mean±S.D., n=6 values. Plasma insulin values at 0 h Insulin Content Serum insulin level and pancreatic insulin
before drug administration are significantly different compared to respective days 28
after drug treatment. Significant difference of diabetic control from normal control content were significantly decreased in streptozotozin induced
a
c
p<0.001. Significant difference of treated groups from diabetic control b p<0.01, diabetic mice, as lower as (1.3 µIU/mL) in the STZ untreated
p<0.05. d p<0.01 when compared with glibenclamide 4 mg/kg treated group.
mice compared with the non-diabetic control group (3.1 µIU/
mL). Effects of 1 and glibenclamide for 28 d on elevation of
streptozotocin induced diabetes has been confirmed here by serum insulin and pancreatic insulin levels which were par-
the study of glucose-6-phosphatase activity in liver, as well allel with those anti-hyperglycemic effects in STZ-diabetic
as the quantification of glycogen in liver and skeletal muscle, mice. The effect of the tetranortriterpenoid on insulin is
which are the important indicators of diabetes mellitus. In the shown in Table 8.
present study, 1 had a beneficial effect in terms of peripheral Inhibition of α-Amylase and α-Glucosidase Incuba-
glucose utilization. tion of the α-amylase with the substrate led to generation
Effect of 1 on SOD, CAT, GSH and GPx in Hepatic and of maltose with the addition of 1 significantly inhibited in a
Renal Tissues The antioxidant effect of the 1 over tissue concentration-related manner (IC50: 46.74 µg/mL) as compared
oxidative markers was studied here (Table 7). Diabetic mice with acarbose with a IC50: 12.23 µg/mL. While α-glucosidase
showed a significant reduction in SOD, CAT, GSH and GPx in inhibitory effect was IC50: 32.18 µg/mL as compared with
hepatic and renal tissues indicating persistent oxidative stress. acarbose with a IC50: 78.54 µg/mL (Table 9).
In addition, administration of 1 have shown increased SOD,
GPx, and CAT activities after 28 d of STZ treatment in mice DISCUSSION
indicated that extract can reduce reactive oxygen free radicals
and improve the activities of the antioxidant enzymes. In view of enormous therapeutic and economic
Effect of 1 on Serum Insulin Level and Pancreatic importance34) attributed to Azadirachta indica (neem), studies
September 2012 1523

undertaken by several groups of workers on its various parts oxidative stress as an intensified free radical production, has
have led to the isolation of a host of new constituents.35) In been shown under diabetic conditions.38) The administration
continuation of investigations on the terpenoidal constituents of 1 reduced the renal and hepatic TBA-reactive substance
of the leaves of neem one new tetracyclic triterpenoids meliac- level significantly. These results suggest that 1 may alleviate
inolin (1) has been isolated from the chloroform leaves extract. oxidative stress associated with diabetic pathological condi-
The results of the present study indicated that 1 was an ef- tions through the inhibition of lipid peroxidation.
fective drug against chronic STZ-induced diabetes and al so Digestion of starch by key gastrointestinal enzymes can
in preventing the gain of food and drink intake induced as be retarded and this can benefit the diabetic patient. Release
a consequence of diabetes. A deficiency or insufficiency of of glucose from food sources is the main factor affecting
insulin secretion or insulin resistance in the diabetic state usu- postprandial hyperglycemia.39) Our finding revealed that me-
ally causes a decrease in body weight gain and increases in liacinolin efficiently inhibited α-amylase suggesting that 1 is
food intake, water intake, In our experimental condition, the a starch blocker. α-Glucosidase inhibitors retard the digestion
STZ-induced diabetic rats also showed the same symptoms. of carbohydrates to simpler carbohydrates and slows down the
However, administration of 1 slightly improved these physico- absorption of the later in the small intestine thus preventing
metabolic abnormalities. high glucose concentration in the blood after a meal.40) Meli-
Meliacinolin could reduce blood glucose via increased in- acinolin showed appreciable α-amylase and α-glucosidase in-
sulin, as evidenced by STZ-treated mice (Table 8), which is hibitory activities so this compound plays a role in the carbo-
capable of modulating the pancreatic secretion. The results of hydrate metabolism, thereby causing a delay in carbohydrate
the OGTT study strongly suggest that 1 reduces postprandial digestion and absorption.
blood glucose levels. This postprandial hypoglycaemic effect The mechanism of action for the antidiabetic activity of 1
of 1 could be due to its ability to facilitate or enhance clear- in streptozotocin-induced mildly diabetic type 2 (MD) mice.
ance of postprandial blood glucose in mice. The improving Meliacinolin was orally administered to MD mice at a dose
actions of 1 on serum levels of glucose and insulin were simi- of 20 mg/kg once a day for 28 d. Meliacinolin-treated MD
lar to those in STZ-diabetic mice. These results indicated that mice showed significant reduction in fasting blood glucose.
our STZ-diabetic mice can be released insulin from pancreatic Assessment of plasma insulin and pancreatic insulin follow-
β cells by both glibenclamide and 1. ing treatment with 1 revealed significant elevation in their
Interestingly, there was decrease in the concentration of levels. Administration of 1 enhanced the activity of hepatic
serum triglyceride, total cholesterol and HDL-cholesterol. The hexokinase and suppressed glucose-6-phosphatase in diabetic
mechanism by which 1 induce reduction in lipid concentration mice. A significant rise in glycogen content was also observed
could be explained by stimulation of lipolysis and fatty acid in liver and muscles of MD mice. The findings of the study
utilization and/o suppression of hepatic fatty acid synthesis suggest that the antidiabetic effect of meliacinolin could be
in mice.36) It is well known that insulin activates lipoprotein due to its insulinogenic action. In addition, impaired glucose
lipase, that hydrolyzes triglycerides under normal conditions. homeostasis was improved by 1 through amelioration in the
Destruction of β-cells leads to depletion of plasma insulin, carbohydrate metabolic pathways. Thus, the extract may exert
which results in hyperlipidemia and hypercholesterolemia an antidiabetic effect through pancreatic as well as extrapan-
caused by derangement of metabolic abnormalities.37) We creatic action such as the intestinal inhibition of glucose.
proved that repeated administration of 1 has a beneficial effect In conclusion, the results obtained demonstrated that meli-
lowering hyperlipidemia associated with hyperglycemia. acinolin acts as an antihyperglycemic agent in streptozotocin-
In this study, 1 caused a marked increase of hepatic gly- nicotinamide induced diabetes mice by reducing the severity
cogen content in STZ-induced diabetic mice, which indicates of oxidative stress and acuity of hyperglycemia induced by
that mice cloud decrease hepatic glucose production (HGP) by STZ, through the improvement of hyperlipidemia, insulin
increasing glycogen content. In addition, 1 decreased G6Pase resistance, and antioxidant defense system. In part the anti-
activity and increased GK activity in liver, which indicates diabetic action of meliacinolin, is attributed to the intestinal
that there, could be an increase in hepatic glucose uptake inhibition of glucose absorption. Is for all this reasons that,
and decrease in hepatic glucose release. Therefore, this study meliacinolin can be considered as a compound with a very in-
strongly suggests that meliacinolin enhances hypoglycemic teresting and promising potential as an anti-diabetic agent, for
activity probably by reducing HGP via decreasing G6Pase its hypoglycemic and insulin augmenting effect and inhibition
activity and increasing GK activity. The increased activity of of intestinal α-gluciosidase and pancreatic α-amylase.
hexokinase can promote glycolysis and increase utilization of
glucose for energy production. The administration of meliac- REFERENCES
inolin to the diabetic mice increased the activity of hepatic
hexokinase causing an increase in glycolysis. 1) Gupta RK. Multipurpose trees for agroforestry and wasteland uti-
SOD, CAT, GSH and GPx are defensive agents against lization. International Science Publisher, New York, pp. 236–239
reactive species (ROS). Decrease in the activity of these (1993).
2) Luo XD, Wu SH, Ma YB, Wu DG. A new triterpenoid from Azadi-
enzymes in liver may indicate increased production of free
rachta indica. Fitoterapia, 71, 668–672 (2000).
radicals. Activity of these enzymes particularly catalase and
3) Yanpallewar S. U., Sen S., Tapas S., Kumar M., Raju S. S., Acharya
glutathione peroxidase reached the non-diabetic level with 1. S. B., Effect of Azadirachta indica on paracetamol-induced hepatic
Hepatic lipid peroxidation was also effectively inhibited by 1 damage in albino rats. Phytomedicine, 10, 391–396 (2003).
indicating an anti-diabetic effects. Decreased activities of an- 4) Kausik BI, Ranajet KB. Biological activities and medicinal proper-
tioxidant enzyrnes in kidney were reversed with meliacinolin. ties of neem plant. Curr. Sci., 82, 1336–1345 (2002).
TBARS are an index of endogenous lipid peroxidation and 5) Dixit VP, Sinha R, Tank R. Effect of Neem seed oil on the blood
1524 Vol. 35, No. 9

glucose concentration of normal and alloxan diabetic rats. J. Ethno- terization and comparison with homogenates and microsomes. Free
pharmacol., 17, 95–98 (1986). Radic. Biol. Med., 4, 155–161 (1988).
6) Khosla P, Bhanwra S, Singh J, Seth S, Srivastava RK. A study of 23) Kinney LaPier TL, Rodnick KJ. Effects of aerobic exercise on
hypoglycaemic effects of Azadirachta indica (neem) in normal and energy metabolism in the hypertensive rat heart. Phys. Ther., 81,
alloxan diabetic rabbits. Indian J. Physiol. Pharmacol., 44, 69–74 1006–1017 (2001).
(2008). 24) Davis SN, Granner DK. Insulin oral hypoglycaemic agents in the
7) Halim EM. Lowering of blood sugar by water extract of Azadirach- pharmacology of the endocrine pancreas. The pharmacological
ta indica and Abroma augusta in diabetes rats. Indian J. Exp. Biol., basis of therapeutica (Hardman JG, Limbird LG eds.) 10th ed., Mc-
41, 636–640 (2001). Millan, New York, pp. 1701–1704 (1996).
8) Gupta S, Kataria M, Gupta PK, Murganandan S, Yashroy RC. Pro- 25) Baginiski ES, Foa PP, Zak B. Methods of enzymatic analysis. Berg-
tective role of extracts of neem seeds in diabetes caused by strepto- meyer HU, 22, 158–161 (1969).
zotocin in rats. J. Ethnopharmacol., 90, 185–189 (2004). 26) Bradford MM. A rapid and sensitive method for the quantitation of
9) Ebong PE, Atangwho IJ, Eyong EU, Egbung GE. The Antidiabetic microgram quantities of protein utilizing the principle of protein–
efficacy of combined extracts from two continental plants: Azadi- dye binding. Anal. Biochem., 72, 248–254 (1976).
rachta indica (A. JUSS) (Neem) and Vernonia77 amygdalina (Del.) 27) Panserat S, Capilla E, Gutierrez J, Frappart PO, Vachot C, Plagnes-
(African bitter leaf). Amer. J. Biochem. Biotech., 4, 239–244 (2008). Juan E, Aguirre P, Brèque J, Kaushik S. Glucokinase is highly
10) Ekaidem IS, Akpan HD, Usoh IF, Etim OE, Ebong PE. Effects of induced and glucose-6-phosphatase poorly repressed in liver of
ethanolic extract of Azadirachta indica leaves on lipid peroxidation rainbow trout (Oncorhynchus mykiss) by a single meal with glucose.
and serum lipids of diabetic Wistar rats. Acta Biologica Szegeden- Comp. Biochem. Physiol. B Biochem. Mol. Biol., 128, 275–283
sis, 51, 17–20 (2007). (2001).
11) Perez GRM, Gomez YG, Guzman MD. Attenuation of antiglycation 28) Tranulis MA, Christophersen B, Blom AK, Borrebaek B. Glucose
non-enzimatic, hypoglycemia and hypolipidemia in streptozotocin- dehydrogenase, glucose-6-phosphate dehydrogenase and hexokinase
induced diabetic rats by chloroform extract of leaves of Azadirachta in liver of rainbow trout (Salmo gairdneri). Effects of starvation and
indica. Phcog. Mag., 7, 254–259 (2011). temperature variations. Comp. Biochem. Physiol. B, 99, 687–691
12) Mostafa M, Choudhury ME, Hossain MA, Islam MZ, Sunon MH. (1991).
Antidiabetic effects of Catharanthus roseus, Azadirachta indica, Al- 29) Hissin PJ, Foley JE, Wardzala LJ, Karnieli E, Simpson IA, Salans
lium sativum and glimepride in experimentally diabetic induced rat. LB, Cushman SW. Mechanism of insulin-resistant glucose transport
Bangl. J. Vet. Med., 5, 99–102 (2007). activity in the enlarged adipose cell of the aged, obese rat. J. Clin.
13) Kochhar A, Sharma N, Sachdeva R. Effect of supplementation of Invest., 70, 780–790 (1982).
tulsi (Ocímum sanctum) and neem (Azadirachta indica) leaf powder 30) Hansawasdi C, Kawabata J, Kasai T. α-Amylase inhibitors from
on diabetic symptoms, anthropometric parameters and blood pres- roselle (Hibiscus sabdariffa LINN.) tea. Biosci. Biotechnol. Bio-
sure of non-insulin dependent male diabetics. Ethno-Med., 3, 5–9 chem., 64, 1041–1043 (2000).
(2009). 31) Matsui T, Yoshimoto C, Osajima K, Oki T, Osajima Y. In vitro
14) Pillai NR, Santhakumari G. Hypoglycemic activity of Melia azadi- survey of α-glucosidase inhibitory food components. Biosci. Bio-
rachta. Indian J. Med. Res., 74, 931–933 (1981). technol. Biochem., 60, 2019–2022 (1996).
15) Kumar DB, Miltra A, Manjunatha M. Azadirachtolide: Anti-diabet- 32) Lavie D, Levy EC, Jain MK. Limonoids of biogenetic interest from
ic ant hypolidemic effects from Azadirachta indica leaves. Pharma- Melia Azdiracta L. Tetrahdedron, 27, 3927–3929 (1971).
cog. Comm., 1, 78–84 (2011). 33) Siddiqui S, Siddiqui BS, Faiza S. Tetracyclic triterpenoids of the
16) Chattopadhyay R. R., Possible mechanism of antihyperglycemic ef- fruit coats of Azadirachta indica. J. Nat. Prod., 54, 408–415 (1991).
fect of Azadirachta indica leaf extract: Part V. J. Ethnopharmacol. 34) Kraus W, Cramer R, Sawitzki G. Tetranotriterpenoids from the
67, 373–376 (1999). seeds of Azadirachta indica. Phytochemistry, 20, 117–120 (1981).
17) Tahara A, Matsuyama-Yokono A, Nakano R, Someya Y, Shibasaki 35) Goh SY, Cooper ME. Clinical review: The role of advanced glyca-
M. Hypoglycaemic effects of antidiabetic drugs in streptozotocin- tion end products in progression and complications of diabetes. J.
nicotinamide-induced mildly diabetic and streptozotocin-induced Clin. Endocrinol. Metab., 93, 1143–1152 (2008).
severely diabetic rats. Basic Clin. Pharmacol. Toxicol., 103, 560– 36) Pushparaj PN, Low HK, Manikandan J, Tan BK, Tan CH. Anti-
568 (2008). diabetic effects of Cichorium intybus in streptozotocin-induced
18) Scott LJ, Spencer CM. Miglitol: a review of its therapeutic potential diabetic rats. J. Ethnopharmacol., 111, 430–434 (2001).
in type 2 diabetes mellitus. Drugs, 59, 521–549 (2000). 37) Gohil T, Pathak N, Jivani N, Devmurari Y, Atel J. Treatment
19) Eichler HG, Korn A, Gasic S, Pirson W, Businger J. The effect of with extract of Eugenia jambolana seed and Aegle marmelos leaf
a new specific α-amylase inhibitor on post-prandial glucose and extracts prevents hyperglycemia and hyperlipidemia in alloxan-
insulin excursions in normal subjects and Type 2 (non-insulin- induced diabetic. African J. Pharm. Pharmacol., 4, 270–275 (2010).
dependent) diabetic patients. Diabetologia, 26, 278–281 (1984). 38) Quine SD, Raghu PS. Effects of (−)-epicatechin, a flavonoid on
20) Punitha ISR, Shirwaikar A, Shirwaikar A. Antidiabetic activity lipid peroxidation and antioxidants in streptozotocin-induced dia-
of benzyl tetra isoquinoline alkaloid berberine in streptozotocin- betic liver, kidney and heart. Pharmacol. Rep., 57, 610–615 (2005).
nicotinamide induced type 2 diabetic rats. Diabetologia Croatica, 39) Matkovies B, Kotorman M, Varga IS, Quyhai DQ, Varga C. Oxida-
34, 117–128 (2005). tive stress in experimental diabetes induced by streptozotocin. Acta
21) Park SE, Cho MH, Lim JK, Kim JS, Kim JH, Kwon DY, Park CS. Physiol. Hung., 85, 29–38 (1998).
A new colorimetric method for determining the isomerization activ- 40) Franco OL, Rigden DJ, Melo FR, Bloch C Jr, Silva CP, Grossi de
ity of sucrose isomerase. Biosci. Biotechnol. Biochem., 71, 583–586 Sá MF. Activity of wheat α-amylase inhibitors towards bruchid
(2007). α-amylases and structural explanation of observed specificities. Eur.
22) Fraga CG, Leibovitz BE, Tappel AL. Lipid peroxidation measured J. Biochem., 267, 2166–2173 (2000).
as thiobarbituric acid-reactive substances in tissue slices: charac-

You might also like