Está en la página 1de 11

N U TR IT ION RE S EA RCH 3 2 ( 2 0 12 ) 62 6 –6 36

Available online at www.sciencedirect.com

w w w. n r j o u r n a l . c o m

Aqueous extract of tamarind seeds selectively increases


glucose transporter-2, glucose transporter-4, and islets'
intracellular calcium levels and stimulates β-cell proliferation
resulting in improved glucose homeostasis in rats with
streptozotocin-induced diabetes mellitus☆

Sushant Shivdas Sole⁎, B.P. Srinivasan


Department of Pharmacology, Delhi Institute of Pharmaceutical Sciences and Research, University of Delhi, PushpVihar,
New Delhi 110017, India

ARTI CLE I NFO A BS TRACT

Article history: Tamarindus indica Linn. has been in use for a long time in Asian food and traditional medicine for
Received 15 January 2012 different diseases including diabetes and obesity. However, the molecular mechanisms of these
Revised 23 June 2012 effects have not been fully understood. In view of the multidimensional activity of tamarind
Accepted 26 June 2012 seeds due to their having high levels of polyphenols and flavonoids, we hypothesized that the
insulin mimetic effect of aqueous tamarind seed extract (TSE) might increase glucose uptake
Keywords: through improvement in the expression of genes of the glucose transporter (GLUT) family and
GLUT-2 sterol regulatory element-binding proteins (SREBP) 1c messenger RNA (mRNA) in the liver. Daily
GLUT-4 oral administration of TSE to streptozotocin (STZ)–induced (90 mg/kg intraperitoneally) type 2
Sterol regulatory element-binding diabetic male Wistar rats at different doses (120 and 240 mg/kg body weight) for 4 weeks showed
proteins-1c positive correlation with intracellular calcium and insulin release in isolated islets of
Islet's cytosolic calcium Langerhans. Tamarind seed extract supplementation significantly improved the GLUT-2
Glucagon protein and SREBP-1c mRNA expression in the liver and GLUT-4 protein and mRNA
Somatostatin expression in the skeletal muscles of diabetic rats. The elevated levels of serum nitric oxide
Tamarindus indica seeds (NO), glycosylated hemoglobin level (hemoglobin A1c) and tumor necrosis factor α (TNF-α)
STZ-induced model of type 2 decreased after TSE administration. Immunohistochemical findings revealed that TSE
diabetes mellitus abrogated STZ-induced apoptosis and increased β-cell neogenesis, indicating its effect on
islets and β-cell mass. In conclusion, it was found that the antidiabetic effect of TSE on STZ-
induced diabetes resulted from complex mechanisms of β-cell neogenesis, calcium handling,
GLUT-2, GLUT-4, and SREBP-1c. These findings show the scope for formulating a new herbal
drug for diabetes therapy.
© 2012 Elsevier Inc. All rights reserved.

Abbreviations: ANOVA, analysis of variance; BW, body weight; [Ca2 +]i, intracellular calcium; DM, diabetes mellitus; dUTP, deoxyuridine
5-triphosphate; ELISA, enzyme-linked immunosorbent assay; GLUT, glucose transporter; HbA1c, hemoglobin A1c; mRNA, messenger RNA;
NO, nitric oxide; PBS, phosphate-buffered saline; PCR, polymerase chain reaction; SREBP, sterol regulatory element-binding proteins; STZ,
streptozotocin; TNF-α, tumor necrosis factor α; TSE, tamarind seed extract; TUNEL, terminal deoxynucleotidyl transferase–mediated
deoxyuridine 5-triphosphate nick-end labeling.

Conflict of interest: The authors declare that there is no conflict of interest.
⁎ Corresponding author. Tel.: +91 11 29554327; fax: + 91 11 29554503.
E-mail address: sush.sole@gmail.com (S.S. Sole).

0271-5317/$ – see front matter © 2012 Elsevier Inc. All rights reserved.
doi:10.1016/j.nutres.2012.06.015
N U TR IT ION RE S E ARCH 3 2 ( 2 0 12 ) 62 6 –6 3 6 627

Pakistan, Indochina, Philippines, Java, and Spain. T indica


1. Introduction
pulp fruit is used for seasoning, as a food component, and in
juices. Its fruit is regarded as a digestive, carminative,
Diabetes mellitus (DM) is a disease that results in chronic laxative, expectorant, and blood tonic [8]. Other parts of the
inflammation and apoptosis in pancreatic islets in patients plant present antioxidant [9], antihepatotoxic [10], and anti-
with either type 1 or 2 DM and is characterized by abnormal inflammatory [11] activity. T indica was used as a traditional
insulin secretion or insulin receptor or postreceptor events medicine for the management of DM [12]. Furthermore,
affecting the metabolism in addition to damaging the liver, aqueous extract of tamarind seeds was found to have potent
kidney, and β cells of the pancreas. Chronic damage is antidiabetic and antihyperlipidemic activities in STZ-induced
associated with elevated oxidative or inflammatory activities diabetic rats [13]. Tamarind seeds are regularly used in the
with a continuum of tissue insults leading to more severe preparation of curry and spices for human consumption in
diabetic complications [1]. Insulin-resistant glucose use in Southeast Asian countries including India. Therefore, the
peripheral tissues such as muscle and adipose tissues is a strong rationale of tamarind seed extract (TSE) for antihy-
universal feature of both insulin-dependent DM and non– perglycemic potential obtained from the present study may
insulin-dependent DM. In this process, glucose transporter serve the use of tamarind as an antidiabetic alone or in
(GLUT) and sterol regulatory element-binding proteins (SREBP) combination with other hypoglycemic agents for human
1c along with other components play crucial roles [2]. Glucose subjects of diabetes.
transporter 4, a member of the GLUT family, is mainly The antihyperglycemic potential of tamarind seeds with
expressed in skeletal muscle, heart, and adipose tissues. It respect to GLUT proteins and islets' physiology such as
plays a critical role in insulin-stimulated glucose transport in cytosolic calcium oscillation and endocrine function (insulin,
these tissues, with glucose uptake occurring when insulin glucagon, and somatostatin) has not been explored as yet. In
stimulates the translocation of GLUT-4 from the intracellular view of the multidimensional activity of plant drugs, we
pool to the plasma membrane [3]. Glucose transporter 2, being hypothesized that tamarind seeds would have a complex
the primary GLUT isoform in the liver, plays a pivotal in mechanism of action; therefore, it was imperative to
glucose homeostasis by mediating bidirectional transport of consider different approaches involved in antidiabetic
glucose [4]. Furthermore, it has been reported that SREBP-1c mechanisms. We specifically assumed that insulin secreta-
expression and its nuclear abundance is low in livers of STZ- gogue action might be due to improvement in calcium of
induced diabetic rats and increases markedly with insulin islets in addition to its effect on transport proteins and fatty
treatment. This fact presents a direct relation between insulin acid metabolism.
and fatty acid metabolism [5]. A rise in cytoplasmic-free Therefore, the present study was designed to investigate
calcium concentration (intracellular calcium [Ca2 +]i) owing to the insulin mimetic impact of TSE on the molecular
influx through voltage-gated L-type Ca2 + channels in the mechanisms of glucose uptake on STZ-induced diabetic
plasma membrane is a central component of the stimulus- rats. With these views, we have studied the effect of TSE on
secretion coupling mechanism leading to insulin release by GLUT-2 protein and SREBP-1c messenger RNA (mRNA)
pancreatic β cells [6]. This pulsatility of calcium is lost in expression in the liver and GLUT-4 protein and mRNA
patients with type 2 DM and thereby affects insulin secretion. expression in skeletal muscle in STZ model of type 2 DM. In
Oxidative stress is one of the key mechanisms in the addition, we evaluated the effect of TSE treatment on β-cell
pathogenesis of diabetes-related vascular dysfunction. Strep- proliferation, apoptosis, and neogenesis in pancreatic islets
tozotocin (STZ)–induced β-cell damage involves many com- of STZ rats.
plicated mechanisms, one of which is production of reactive
oxygen species, particularly nitric oxide (NO), either from STZ
or neighboring macrophages. Peroxynitrite and proinflama- 2. Methods and materials
tory cytokines such as TNF-α have been found to cause DNA
damage and apoptosis in human and rat islets [7]. However, 2.1. Drugs and chemicals
the prevention from selective destruction of insulin-produc-
ing β cells of pancreatic islets can be one of the targets for The biochemical kits used in the experiment were obtained as
either the cure or delay of DM. follows: serum NO (Biovision, Milpitas, CA, USA; Cat no. K262-
Besides drugs in modern medicine, several species of 200), apo-BrdU-IHCTM, in situ DNA Fragmentation Assay Kit
plants have been described in the scientific and popular (catalogue no. K403-50; Biovision), TNF-α (catalog no. ELR-
literature as having hypoglycemic activity. Because of their TNF-α-001; Raybiotech, Inc, Norcross, GA, USA), STZ (Sigma
perceived effectiveness, minimal side effects in clinical Chemicals), metformin (Ranbaxy Ltd, Gurgaon, India), Taq
experience and relatively low-cost herbal drugs are pre- DNA polymerase (catalog-A5209, 0200; Bioline Ltd, London,
scribed widely even when their biologically active com- UK), HbA1c assay kit (catalog #COD 11044; Biosystem, Barce-
pounds are unknown. Tamarindus indica Linn. (commonly lona, Spain), glimepiride (manufacturer: Hetero Labs Ltd,
known as tamarind), a tree-type plant belonging to the Hyderabad, India; provided by Panacea Biotech Ltd, Malpur,
Leguminosae (Caesalpiniaceae) family, occurs in the tropical Baddi, Solan, India), benzylpenicillin and streptomycin (Sigma
regions of the world and can be found in more than 50 Aldrich, Munich, Germany), horseradish peroxidase–conju-
countries. It is indigenous to tropical Africa but has become gated immunoglobin G (Jackson Immuno Research Laborato-
naturalized in North and South America from Florida to ries, West Grove, PA, USA), and anti-GLUT-2 and GLUT-4
Brazil and is also cultivated in subtropical China, India, antibodies (Abcam, Cambridge, UK).
628 N U TR IT ION RE S EA RCH 3 2 ( 2 0 12 ) 62 6 –6 36

2.2. Preparation of aqueous extract of seeds of T indica sectioned at 5-μm thickness, and used for immunostaining.
Serial sections of the rat pancreas were immunostained by
Seeds of T indica were purchased from Khari Baoli, New Delhi, streptavidin-biotin peroxidase method using prediluted poly-
in May 2011, and authentication was done by a scientist of the clonal antibodies. All sections were deparaffinized in xylene
National Bureau of Plant Genetic Resources, Pusa, New Delhi bath to remove the excess wax. The slides were placed in 2
(voucher no. NHCP/NBPGR/2010-52). Aqueous extract of seeds changes of absolute alcohol for 3 minutes each. The same
of T indica was prepared by the method of the National procedure was repeated with 90% alcohol. The slides were
Institute of Health and Family Welfare, India [14]. Briefly, after placed in a blocking reagent to block the endogenous
incubation for 2 days at 40°C, the seeds of T indica were peroxidase activity for 5 minutes, which was prediluted with
powdered in a grinder; 100 g powder was suspended in 500 mL 5 volumes of 100% ethanol. The slides were placed in 2
redistilled water, and the extraction was performed in Soxhlet changes of 70% alcohol for 3 minutes each. The excess alcohol
apparatus for 18 hours. A deep-brown aqueous extract was around the sections was removed, and the slides were quickly
obtained, which was filtered using a coarse sieve filter paper. immersed in Tris buffer, pH 7.6 for 5 minutes. Two drops of
The filtrate was then dried under reduced pressure and finally tissue conditioner were added, and the sections were incu-
lyophilized. Phytochemical screening and standardization of bated for 5 minutes and then rinsed in a buffer solution.
the extract were done before commencement of the in vivo Prediluted primary polyclonal antiguinea pig antibody to
study. The aqueous extract yielded 2.8-g lyophilized powder insulin (1:1000; Genetex, Irvin, CA, USA) raised against
(0.28%) from 1 kg of tamarind seeds. human insulin, antirabbit glucagon antibody (1:375; Novus
Biologicals, Littleton, CO, USA) raised against Procine glucagon
2.3. Animals conjugated to bovine serum albumin, and antirabbit somato-
statin antibody (1:500; Novus Biologicals) raised against
Male Wistar rats weighing 150 to 200 g used for the study synthetic somatostatin were added to the sections and
were obtained from the Animal House of the Delhi Institute incubated for 1 hour. The secondary antibodies for insulin,
of Pharmaceutical Sciences and Research. Rats were housed glucagon, and somatostatin were antirabbit polyclonal anti-
in colony cages (4 rats per cage) at an ambient temperature bodies. After incubation for 0.5 hour, the sections were rinsed
of 25°C with 12-hour light to 12-hour dark cycle. Rats had free with Tris buffer, peroxidise solution was added, incubated for
access to standard food and water ad libitum. The Principles 30 minutes, and later rinsed with the buffer. 3-amino, 9-ethyl
of Laboratory Animal Care (NIH, 1985) were followed carbazole chromogen substrate was added to the sections and
throughout the duration of the experiment. All experimental was incubated for 15 minutes and rinsed with distilled water.
procedures were conducted according to the institutional The sections were counterstained with Harris hematoxylin for
animal ethical committee (protocol no. 2/DIPSAR/IAEC/2010) 45 seconds to facilitate nuclear identification [16].
and Committee for the Purpose of Control and Supervision
on Experiments on Animals (CPCSEA) guidelines. 2.6. DNA fragmentation assay

2.4. Experimental design/animal group For detection and localization of apoptosis in the pancreas, we
used the technique of terminal deoxynucleotidyl transferase–
2.4.1. Induction of type 2 DM mediated deoxyuridine 5-triphosphate (dUTP) nick-end label-
Streptozotocin was injected at a dose level of 90 mg/kg ing (TUNEL). Briefly, sections were deparaffinized, hydrated,
intraperitoneally in 0.1 mol/L freshly prepared citrate buffer and digested with proteinase K (20 μg/mL), and then,
pH 4.5 to 2-day-old neonatal rats. Controls were injected with biotinylated dUTP was added to the 3′ end of DNA fragments
an equivalent volume of citrate buffer. After 6 weeks of by incubating sections in 0.05 mol/L Tris-HCl buffer (pH 7.6)
injection, animals were evaluated for fasting blood glucose with 0.03 U/μL TdT and 0.04 nmol/μL biotin-11-dUTP at 37°C
level. A fasting glucose level of 140 mg/dL was the criterion for for 1 hour. The sections were rinsed in phosphate-buffered
selection of diabetic rats [15]. A total of 40 male rats were used, saline (PBS). Endogenous peroxidase was blocked with 0.3%
and they were divided into 5 groups with 8 rats per group: H2O2 in distilled H2O. The sections were rinsed with PBS and
group 1, normal untreated rats; group 2, diabetic control rats; covered with 2% blocking solution in 0.1 mol/L sodium
group 3, diabetic rats treated with TSE (120 mg/kg); group 4, maleate to reduce background staining. The sections were
diabetic rats treated with TSE (240 mg/kg); and group 5, then incubated with avidin-peroxidase complexes in PBS
diabetic rats treated with metformin (100 mg/kg). The doses in (1:50) for 30 minutes and rinsed with PBS (3 × 5 minutes).
the present study were selected based on antidiabetic and Peroxidase activity was visualized with 3,3′-diaminobenzidine
hypolipidemic action of tamarind seeds proposed by Maiti et until the brown product was clearly visible. The sections were
al [13]. After final administration of treatment of 4 weeks, then counterstained with methyl green. The positive apopto-
blood samples were collected under fasting conditions, and tic cells were the cells with brown nucleus [17].
the body weights (BWs) were measured.
2.7. Isolation and culture of islets from normal rats
2.5. Immunocytochemistry
Animals were killed by decapitation and pancreatic islets of
Whole pancreas from the rats was removed under anesthesia Langerhans isolated as previously described [18] using diges-
and fixed in 10% buffered formalin for 24 hours. Tissues were tion with collagenase obtained from Clostridium histolyticum
dehydrated in graded series of alcohol, embedded in paraffin, (Sigma Aldrich). Digestion and sedimentation of islets were
N U TR IT ION RE S E ARCH 3 2 ( 2 0 12 ) 62 6 –6 3 6 629

carried out in Hanks solution containing 5.5 mmol/L glucose. reactions were performed in a Light-Cycler 480 (Roche)
Islets were then handpicked under a stereomicroscope and instrument. Fluorescence was detected at the end of the
transferred to petri dishes containing culture medium RPMI 56°C segment in the PCR step. Polymerase chain reaction
1640 (Sera Laboratories International Ltd, West Sussex, UK) products of β-actin primer gene were used as an internal
supplemented with 100 U/mL benzylpenicillin, 0.1 mg/mL standard. All assays were carried out in triplicate. Real-time
streptomycin, 2 mmol/L L-glutamine, and 10% (vol/vol) heat- PCR analysis and subsequent calculations were performed
inactivated fetal calf serum (Sigma Chemicals Ltd). Islets were on Light-Cycler 480 software (version LCS480 1.2.0.169;
cultured free-floating at 37°C, with an atmosphere of 5% CO2, Roche) [20].
95% O2. The period of culture was 48 hours.
2.10. Glucose transporter 2 expressions in the liver and
2.8. Measurement of cytosolic Ca 2 + concentrations GLUT-4 expressions in the soleus muscle

Pancreatic islet cells were prepared from adult rats and For determination of GLUT-2 protein expressions in the liver
cultured overnight in RPMI 1640 culture medium supplemen- and GLUT-4 protein expressions in the skeletal muscle, each
ted with 10% (vol/vol) fetal calf serum, 100 IU/mL penicillin, sample prepared was mixed with 1% sodium dodecyl sulfate
100μg/mL streptomycin, and 60 μg/mL gentamycin. The cells and 50 mmol/L dithiothreitol, and the mixture was subjected
were loaded with Ca2 + indicator by incubation with 2 μM fura- to electrophoresis with 10% polyacrylamide gel. The separated
2/AM for 45 minutes in the culture medium. The Ca2 +-loaded proteins in the gel were electrotransferred to a polyvinylide-
islets cells were then washed in a medium containing 140 nedifluoride membrane. After blocking with 5% skimmed milk
mmol/L NaCl, 5.9 mmol/L KCl, 1.28 mmol/L CaCl2, 1.2 mmol/L solution including 0.05% Poly (oxyethylene) sorbitan mono-
MgCl2, 25 mmol/L 4-(2-hydroxyethyl)-1-piperazineethanesul- laurate (Tween 20) overnight at 4°C, the membrane was
fonic acid (HEPES), pH 7.4, and 1 mg/mL bovine serum reacted with anti–GLUT-2 antibody and anti–GLUT-4 antibody
albumin. The cells (about 4 × 106) were suspended in 2.5 mL for 2 hours. Subsequently, it was incubated with horseradish
of fresh washing medium, stirred and placed in a spectroflu- peroxidase-conjugated immunoglobin G (diluted 1:2000) for 2
orometer cuvette 50 B; (Perkin Elmer, Waltham, MA, USA) for hours at room temperature. The blots were detected with
monitoring [Ca2 +]i changes by 340 or 340/380 nm excitation chemiluminescence reagents (Western blot) [21].
ratio, and emitted fluorescence of 510 nm at 37°C. Intracellular
Ca2 + levels could not be directly calibrated, and the [Ca2 +]i 2.11. Influence on biochemical metabolic parameters
changes presented are relative changes in fluorescence. After
a stable baseline was reached, islets were supplemented with The fasting blood glucose and BWs were measured on days 0,
0 or 100 μg/mL TSE or 0 or 50 μmol/L glimepiride in the 14, 21, and 28. Plasma insulin was determined by enzyme-
presence or absence of 20 mmol/L D-glucose. The time that the linked immunosorbent assay (ELISA) (Mercodia, Uppsala,
cells spent in the absence of glucose, that is, the time needed Sweden) on day 28. Four weeks after the administration of
for washing, suspending, and establishment of baseline, was drugs (TSE, metformin), blood was collected under ether
10 to 15 minutes [19]. anesthesia from the hearts of animals by cardiac puncture
before killing, placed in EDTA vacutainer tubes and centrifuged
2.9. RNA isolation and real-time polymerase chain at 4000g at 4°C for 15 minutes and stored at −80°C until
reaction analysis analysis. The aliquoted plasma samples were used for the
quantification of TNF-α and insulin levels. Serum was
Real-time polymerase chain reaction (PCR) amplifications for separated by subjecting the coagulated blood to centrifugation
SREBP-1c (gene ID: 78968) and GLUT-4 (gene ID: 25139) were at 4000g for 15 minutes and further used for NO determination.
conducted using Light-Cycler 480 SYBR Green I Master
(Roche, Basel, Switzerland) according to the manufacturer's 2.11.1. TNF-α, serum NO, and HbA1c
instructions. Reaction conditions were 10× PCR buffer (20 Plasma TNF-α level was determined using commercially
mmol/L Tris, pH 8.4, 50 mmol/L KCl, and 2.5 mmol/L MgCl2), available ELISA kits. Briefly, wells of ELISA plate were first
20 pM oligonucleotide, 300 μmol/L deoxyribonucleotides coated with a TNF-α capturing antibody followed by washing
(dNTP), 1:1000 SYBR Green I nucleic acid stain, 0.5 U Taq and drying. Samples were added in wells. Once the samples
DNA polymerase (recombinant), and 50 ng templates com- were loaded, a biotinylated antirat TNF-α antibody was
plementary DNA in a 25-mL reaction volume. Sets of PCR subsequently added in each well. Both the antigen and
primers of GLUT-4 sense 5′-AGC GTAGGTACCAACACTTTCT- antibody were allowed to coincubate for 3 hours followed by a
3′ and antisense 5′-CCG CCC TTA GTT GGT CAG AAG-3′; for series of washes. A solution of horseradish peroxidise-strepta-
SREBP-1C gene sense 5′-GGAGCCATGGATTGCACATT-3′ and vidin complex was added to each well and incubated for 20
antisense 5′- AGGAAGGCTTCCAGAGAGGA-3′; and for β-actin minutes. After this, a ready-to-use chromogen was added, and
sense 5′-TCA CCC ACA CTG TGC CCC ATC TAC GA-3′ and the plates were kept in the dark for 10 to 15 minutes for color to
antisense 5′ CAG CGG AAC CGC TCA TTG CCA ATGG-3′ were develop. The reaction was stopped using a stop solution, and
designed with reference to the National Centre for Biotech- the absorbance was read at 450 nm (Biorad, Hercules, CA, USA)
nology Information (NCBI) database of conserved coding using reference filter at 630 nm. Simultaneously, a series of
regions. Thermal cycling conditions were 95°C for a 3-minute dilutions of standard TNF-α was also run to obtain a standard
denaturation step followed by 40 PCR cycles (94°C for 30 graph. The concentrations of TNF-α in the samples were
seconds, 56°C for 30 seconds, and 72°C for 1 minute), and determined using the standard curve obtained [22].
630 N U TR IT ION RE S EA RCH 3 2 ( 2 0 12 ) 62 6 –6 36

Serum NO concentration was determined by the Griess


reagent method using available reagents and the manufac-
turer's protocol [23]. Briefly, serums were added into wells
(96-well enzymatic assay plate). A sulfanilamide solution
was added to all experimental samples, and after incubation,
N-1-naphtylethylenediamine dihydrochloride solution was
added. Then, absorbance was measured by a microreader at
520 nm wavelength. The concentration of NO samples was
determined by comparison with the nitrite standard refer-
ence curve.
The HbA1c concentration was measured by the principle of
turbidometric assay using available reagents and standards
(Biosystems, Barcelona, Spain). After preparing the hemoly-
sate using tetradecyltrimethylammonium bromide as a
detergent, the HbA1c concentration was determined by
spectrometry [24].

2.12. Statistical analyses

All values are means ± SEM. Data analysis was done with
1-way analysis of variance (ANOVA) followed by Dunnet's
multiple test where a diabetic group was considered a
positive control (Sigma Plot11, San Jose, CA, USA). Group means
were considered to significantly differ at P < .05, as determined
by Dunnet's multiple range analysis. In case of islets Ca2 + P < .05
and ⁎⁎P < .01 for difference from 0 mmol/L glucose, #P < .05, and
###
P < .001 for difference from 20 mmol/L glucose.
Fig. 1 – Effect of TSE on BW (grams) in diabetic rats. The
values are the means ± SEM from 8 animals in each group.
*
P < .05 vs diabetic group. Bar graph with dark shed ( )
3. Results
represents BW before treatment, whereas with light shed ( )
after treatment (TSE, 120 and 240 mg/kg; metformin, 100 mg/
3.1. Effect of TSE on BW, blood glucose, and HbA1c levels
kg; saline, for normal rats, orally for 4 weeks). Data analysis
was done with 1-way ANOVA followed by Dunnet's multiple
Before the supplementation of TSE and metformin, there were
test (Sigma Plot11). The diabetic group without drug treatment
no significant differences of baseline BW of the rats (Fig. 1). The
was compared with the normal, TSE-treated, and metformin
rats treated with TSE (120 and 240 mg/kg) and metformin (100
(standard) groups.
mg/kg) showed significant increase in BW as compared with
diabetic groups after 4 weeks of study. Total HbA1c content is an
indicator of irreversible condensation of glucose with the N-
terminal residue of the β-chain of hemoglobin A. The HbA1c Vehicle-treated diabetic rats led to significant increase in
concentration in blood directly denotes the mean concentration NO and TNF-α concentrations (10.02 ± 0.50 nmol/μL, 24.50 ±
of glucose prevailing in the previous 4 to 8 weeks, equivalent to 0.75 pg/mL) compared with the corresponding vehicle-treated
the lifetime of the erythrocytes. Before treatment, there were normal rats (1.85 ± 0.27 nmol/μL, 11.25 ± 0.59 pg/mL),
significantly higher fasting glucose and HbA1c levels (P < .05) in respectively. Daily administration of TSE at both dose levels
all groups when compared with the normal. The daily (120 and 240 mg/kg) showed dose-dependent decrease in TNF-
administration of TSE at both dose levels (120 and 240) for the α concentrations when compared with corresponding diabetic
period of 4 weeks resulted in dose-dependent reduction of controls (P < .05, Table). In the TSE-supplemented groups, the
fasting glucose (milligrams per deciliter) and HbA1c (percentage) levels of NO were decreased by 22% and 43% at 120 and 240
vs the diabetic group (P < .05, Table). mg/kg dose levels, respectively. This suggests that TSE could
be a potential NO scavenger along with its anti-inflammatory
3.2. Effect of TSE on plasma insulin, serum NO, and activity that was represented by reduction in TNF-α level.
TNF-α concentration
3.3. Effect of TSE on immunohistochemical changes and
Insulin levels were significantly decreased in diabetic rats in cell apoptosis in the pancreas of diabetic rats
comparison with nondiabetic controls. After 28 days of TSE
supplementation to the diabetic rats, there was a significant Immunohistochemical evaluation of the pancreas of diabetic
elevation in insulin levels in respect to diabetic control group rats revealed a high frequency of degenerative changes, such
in a dose-dependent manner (P < .05, Table). Of the 2 doses of as atrophy, pyknosis, and deterioration in the islets. The
TSE tested, the 240 mg/kg dose was found to be more effective numbers of immunoreactive β cells in diabetic rats were
in reducing fasting blood glucose levels. extremely low, and the cells stained weakly, suggesting
N U TR IT ION RE S E ARCH 3 2 ( 2 0 12 ) 62 6 –6 3 6 631

Table – Effect of TSE on TNF-α, serum NO, and HbA1c level


TNF-α NO HbA1c (%) Fasting blood glucose (mg/dL)
(pg/mL) (nmol/μL)
Before treatment After treatment Before treatment After treatment

Normal 11.25 ± 0.59 ⁎ 1.85 ± 0.27 ⁎ 3.45 ± 1.85 ⁎ 3.95 ± 0.17 ⁎ 87.62 ± 2.94 ⁎ 89.25 ± 11.31 ⁎
Diabetic 24.50 ± 0.75 10.02 ± 0.50 7.89 ± 0.87 8.58 ± 0.51 173.50 ± 6.59 174.87 ± 9.74
TSE, 120 mg/kg 17.87 ± 0.61 ⁎ 7.74 ± 0.23 ⁎ 8.57 ± 0.89 5.74 ± 0.13 ⁎ 167.37 ± 4.13 ⁎ 140.12 ± 9.94 ⁎
TSE, 240 mg/kg 14.37 ± 0.73 ⁎ 5.64 ± 0.13 ⁎ 8.14 ± 0.58 5.34 ± 0.12 ⁎ 166.87 ± 4.80 ⁎ 130.00 ± 8.70 ⁎
Metformin 12.75 ± 0.36 ⁎ 5.46 ± 0.18 ⁎ 7.94 ± 1.02 4.85 ± 0.06 ⁎ 171.25 ± 12.03 ⁎ 113.50 ± 7.10 ⁎

The values are the means ± SEM. Data analysis was done with 1-way ANOVA followed by Dunnet's multiple test (Sigma Plot11). The diabetic
group without drug treatment was compared with the normal, TSE-treated (120 and 240 mg/kg), and metformin (standard) groups. The route of
drug (saline in case of the normal group) administration for all the groups was oral. Duration of treatment was 4 weeks after diabetes induction.
⁎ P < .05 vs diabetic group (n = 8).

Fig. 2 – Effect of TSE on immunohistopathologic changes and apoptosis (deoxynucleotidyl-transferase nick-end labeling
[TUNEL] assay) in the pancreata of diabetic rats; antiglucagon, antisomatostatin, and anti-insulin antibody immunostaining
from (A, D, and G) normal rats, (B, E, and H) vehicle-treated diabetic rats, and (C, F, and I) TSE-treated (TSE, 120 and 240 mg/kg;
saline, for normal rats, orally for 4 weeks) diabetic rats, respectively. In the TSE-treated pancreas, antirabbit glucagon antibody
(1:375) and antirabbit somatostatin antibody (1:500) raised against synthetic somatostatin yielded a staining pattern where
glucagon and somatostatin positive cells were decreased compared with diabetic rat. In contrast, prediluted primary polyclonal
antiguinea pig antibody to insulin (1:1,000) raised against human insulin revealed that β cells were abundant and intensely
stained as that in control (G) in TSE-treated rats. I, K, and L denotes effect of TSE on apoptosis (deoxynucleotidyl-transferase
nick-end labeling [TUNEL] assay) changes in the pancreata of normal (I), diabetic (K), and TSE-treated (L) rats. Morphological
features of apoptosis, including pyknotic nuclei, were readily detectable in pancreatic sections from diabetic rats that were
reduced to considerable extent in TSE-treated rats (the arrows denote TUNEL-positive cells). Original magnification ×400.
632 N U TR IT ION RE S EA RCH 3 2 ( 2 0 12 ) 62 6 –6 36

dysfunction of the β cells because of a decrease in their nuclei, were readily detectable in pancreatic sections from
number or failure to synthesize and secrete adequate diabetic rats (Fig. 2).
amounts of immunoreactive insulin or both. In contrast,
degenerative changes occurred at a low frequency in TSE- 3.4. Effects of TSE on muscle GLUT-4 and liver SREBP-1c
treated groups, with only a slight decrease in the number of mRNA concentrations of diabetic rats
insulin-positive granules and no marked islet atrophy,
degeneration, or necrosis. In the TSE-treated pancreas, anti- In view of the fact that acetyl coenzyme A carboxylase and
rabbit glucagon antibody (1:375) and antirabbit somatostatin cytosolic 3-hydroxyl-3-methylglutaryl coenzyme A synthase,
antibody (1:500) yielded a staining pattern where glucagon and the key enzymes of fatty acid synthesis and cholesterol
somatostatin positive cells were decreased compared with synthesis, respectively, are regulated by SREBP-1c during the
diabetic rats. The STZ-induced diabetic rats showed several transcriptional step [25], we investigated the influence of TSE
somatostatin positive cells (Fig. 2), suggesting the role of a on SREBP-1c mRNA. As shown in Fig. 3, the amplification
feedback mechanism of somatostatin on glucagon. efficiency (E) for genes was determined by linear regression
The TUNEL assay revealed that TSE supplementation in analysis of the fluorescent data from the exponential phase of
diabetic rats reduces pancreatic cell apoptosis. In contrast, PCR [26]. Quantitative reverse transcription–PCR results
morphological features of apoptosis, including pyknotic showed increase in muscle GLUT-4 mRNA and liver SREBP-1c

Fig. 3 – Effects of TSE on muscle GLUT-4 mRNA (A), liver SREBP-1c mRNA (B), GLUT-4 (C) expression in the muscles, and GLUT-2
(D) expression in the liver of diabetic rats (TSE, 120 and 240 mg/kg; saline, for normal rats, orally for 4 weeks). The values are the
means ± SEM from 8 animals in each group. Amplification efficiency (E) for gene was determined by linear regression analysis
of the fluorescent data from the exponential phase of PCR [26]. In the liver and muscle membrane fractions of diabetic rats, the
translocation of GLUT4 (C) and GLUT2 (D) was very much reduced when compared with that of band density of healthy controls.
N U TR IT ION RE S E ARCH 3 2 ( 2 0 12 ) 62 6 –6 3 6 633

mRNA concentrations after 4 weeks treatment of TSE as lowest glucose concentration (Fig. 4). It has been shown before
compared with corresponding vehicle-treated diabetic rats. that Ca2 + oscillations of individual β cells within the mouse
and rats islet are synchronized, so that oscillations are
3.5. Effect of TSE on liver GLUT-2 and muscle observed also at the level of the whole islet [27]. In the
GLUT-4 expression absence of glucose, both TSE (100 μg/mL) and glimepiride (50
μmol/L) significantly increased the islet cytosolic Ca2 + (TSE,
Because there was an increase in islets cytosolic Ca2 +as well as 22.22%; ⁎P < .05; glimepiride, 40%; ⁎⁎P < .01; n = 8) contents. The
plasma insulin levels in TSE-treated diabetic rats and because addition of 20 mmol/L D-glucose alone increased the islet
of the physiologic importance of insulin-dependent GLUT-2 cytosolic Ca2 + contents. In the presence of 20 mmol/L
and GLUT-4 translocation to the cell membrane, attempts have D-glucose, both TSE and glimepiride significantly increased
been made to see the effect of TSE treatment on skeletal muscle islet cytosolic Ca2 + contents (TSE, 42%; #P < .05; glimepiride,
tissue membrane GLUT-4 and liver GLUT-2 levels. In the liver 60%; ###P < .001; n = 8) when compared with islets treated with
and muscle membrane fractions of diabetic rats, the translo- D-glucose alone (Fig. 4).
cation of GLUT-2 and GLUT-4 was very much reduced when
compared with the band density of healthy controls. This is
quite rational because the deficiency of insulin in the diabetic 4. Discussion
state would decrease the translocation of GLUT-2 and GLUT-4
from the vesicles to cell membranes. Treatment with TSE The neonatal-STZ Wistar model is a well-characterized model
resulted in the reversal of membrane GLUT-2 and GLUT-4 of type 2 diabetes. The STZ rats develop persistent diabetes
levels. The modulation of GLUT-2 and GLUT-4 protein could rapidly after 6 weeks of age and shows diabetes-like symptoms
thus be one of the mechanisms of antidiabetic properties of such as lack of insulin release in response to glucose, glucose
TSE. The GLUT-2 and GLUT-4 levels of normal control, diabetic, intolerance, and depletion of pancreatic insulin store [15]. In
and TSE-treated diabetic rats are shown in Fig. 3. the current study, TSE was tested after chronic dosing (once a
day) in a preclinical model of STZ-induced type 2 diabetes for
3.6. Cytosolic Ca 2 + concentrations period of 4 weeks. The BW of diabetic rats was found to be less
during the course of the development, which may be because
Stimulation of isolated islets with glucose produced the of accelerated lipolysis, whereas weight gain was significantly
expected increase of intracellular Ca2 +, especially at the observed in rats treated for 4 weeks with TSE and metformin.

Fig. 4 – Effect of TSE on the pattern of cytosolic calcium concentration in islets of pancreas. The values are the means ± SEM from
8 animals in each group. Data analysis was done with 1-way ANOVA followed by Dunnet's multiple test. Islets maintained in
culture for 2 to 4 days were loaded with fura 2. The cells (about 4 × 106) were suspended in 2.5 mL of fresh washing medium and
placed in a spectrofluorometer cuvette (Perkin Elmer 50 B) for monitoring [Ca2 +]i changes by 340 or 340/380 nm excitation ratio
and emitted fluorescence 510 nm, respectively, at 37°C. The [Ca2 +]i was calculated using a standard curve of fura 2 values as a
function of calcium concentration as determined using calibrated solutions of calcium.*P < .05 and ** P < .01 for difference from
0 mmol/L glucose, #P < .05 and ###P < .001 for difference from 20 mmol/L glucose. Glime indicates glimeperide; Glu, glucose.
634 N U TR IT ION RE S EA RCH 3 2 ( 2 0 12 ) 62 6 –6 36

The TSE-treated groups had shown significant antihypergly- Significant changes are observed after TSE treatment in the
cemic effects associated with increase in plasma insulin distribution of insulin, glucagon, and somatostatin-positive
activity. The reduced glucose levels suggested that TSE might cells in the islets of Langerhans. The number of insulin-
either promote glucose uptake by inhibiting hepatic gluconeo- positive cells decreases markedly in both the TSE-treated and
genesis or by absorption of glucose into the muscle and an untreated diabetic rat when compared with control
adipose tissues either through the stimulation of a regenera- animals, but the decrease is much greater in the untreated
tion process and revitalization of the remaining β cells to rats. The effect may be due to prevention of β-cell death by
release insulin or through the insulin mimetic response. decreasing the oxidative stress caused by STZ in diabetic rats.
Because TSE is reported to have antidiabetic action, it Pons and Aoki [33] reported that the number of glucagon-
was of interest to analyze whether TSE affects islet [Ca2+]i producing α and somatostatin-producing δ cells was increased
and insulin release. In the pancreatic β cell, the oscillations in in diabetes, and it appears that somatostatin-producing cells
[Ca2 +]i may be of particular physiologic importance. An rise in number to compensate for the relative reduction in
elevated concentration of glucose within the β cell ultimately insulin-secreting cells. This up-regulation of α cells of the
leads to membrane depolarization and an influx of extracel- pancreas controls the glucagon release to maintain glucose
lular calcium. The resulting increase in [Ca2 +]i is thought to be homeostasis by way of the feedback mechanism of somato-
one of the primary triggers for exocytosis of insulin-contain- statin. This plausible mechanism supports the results of the
ing secretory granules [28]. The present results indicate that present study, where there is a decrease in glucagon-positive
the TSE increased Islet [Ca2 +]i significantly in the presence or cells, whereby somatostatin-positive cells were noted in TSE-
absence of 20 mmol/L D-glucose. The data are consistent with supplemented rats.
the role of adenosine triphosphate and adenosine diphos- The GLUT-4 expression is down-regulated when there is
phate as the connecting linkages between increased glucose relative insulin deficiency, such as in STZ-induced diabetes
metabolism and the ionic events that lead to release of [34]. The GLUT-4 protein level was measured by immunoblot
insulin. Borman and Melzig [29] reported that flavonoids analysis in the homogenate of skeletal muscles and was
might be responsible for inhibition of metallopeptidases that decreased in the diabetic control group. After treatment with
are involved in degradation of neuropeptides. The ability of TSE, the contents of GLUT-4 protein and GLUT-4 mRNA were
TSE to mobilize intracellular Ca2 + in pancreatic islets both in restored to near normal values in skeletal muscles. The
the absence and presence of glucose might be due to the decrease in GLUT-4 levels is essentially one of the main
presence of flavonoids, suggesting inhibition of degradation of reasons of hyperglycemia in the diabetic state, which is due to
neuropeptides in the cells of the islets of Langerhans and in decreased uptake of glucose by the skeletal muscles. Resto-
the basolateral surfaces of pancreatic acinar cells involved in ration of GLUT-4 levels would, therefore, enhance the uptake
increase of [Ca2 +]i and enhanced insulin secretion [30]. of glucose in the skeletal muscles and thus help to combat
The Maillard reaction (excess blood glucose + hemoglobin = hyperglycemic conditions. Glucose unresponsiveness associ-
HbA1c) occurs between lipids and glucose, which results in ated with GLUT-2 impairment is typically demonstrated in
glycation of phospholipids in the cell membrane or the type 2 diabetes [35]. The effects of insulin on SREBP-1c have
organelle under hyperglycemic conditions. These glycated been corroborated by in vivo studies showing that SREBP-1c
lipids are causative agents of oxidative stress (lipid peroxida- expression and nuclear abundance were low in the livers of
tion) and are related to exacerbation of chronic diabetic STZ-induced diabetic rats and markedly increased after
complications [31]. In the present study, administration of insulin treatment [36]. Therefore, the stimulating action of
the extract for 4 weeks tended to significantly bring down the TSE on GLUT-2 protein and SREBP-1c mRNA expression might
level of HbA1c in both TSE-treated diabetic groups. This might be due to the insulin secretogogue effect.
have been due to improved glycemic control produced by Although our data are compatible with the previous
the extract. findings on T indica, some limitations in this study may be
The destruction of β cells in diabetes is mediated by considered. The increased cytosolic calcium in the present
changing expression levels of antiapoptotic or proapoptotic investigation is thought to be one of the mechanisms for
proteins. The STZ could induce NO formation and further lead insulin release; nevertheless, the possibility of other sites of
to potential mitochondrial membrane changes, hence the TSE action on the insulin-secretary mechanisms cannot be
release of cytochrome C, which triggers apoptosis [32]. In the eliminated, for example, interaction with calmodulin and
present study, the diabetic rats showed significant increase in protein kinase C. Furthermore, a glucagon-like peptide 1 and
NO and decrease in insulin secretions and vice versa in gastrointestinal functions, which also play an important role
normal rats. The TSE treatment to diabetic rats shows in postprandial hyperglycemia, are also criteria that need to be
decrease in NO but significant increase in insulin secretions. studied. However, the present study has highlighted the
Our results demonstrate that TSE suppresses STZ-induced relationship between β-cell regeneration and TSE, which
apoptosis significantly. These data suggest that TSE possesses represents a challenge for future studies. Better knowledge
potent antiapoptotic effects when β cells have STZ-induced of the mechanisms underlying the β-cell function in regener-
impairment. Correlations could be found in our study between ating islets should be of more help for the development of
the total phenolic content and NO scavenging ability of the diabetes treatment.
extract. The results are in agreement with those reported by To conclude, the data presented in this study support and
Komutarin et al [8], where the seed coat extract of tamarind thus accept our primary hypothesis that the antidiabetic
seeds suppressed NO in the murine macrophage cell line and action of tamarind seeds comprises an array of actions and
freshly isolated peritoneal macrophages. provides direct evidence for the involvement of GLUTs (GLUT-
N U TR IT ION RE S E ARCH 3 2 ( 2 0 12 ) 62 6 –6 3 6 635

2 and GLUT-4) in controlling glucose homeostasis. In addition, [11] Rimbau V, Cerdan C, Vila R, Iglesia J. Antiinflammatory
the β cell–preserving action of TSE along with the insulin activity of some extracts from plants used in traditional
medicines of North-African Countries (II). Phytother Res
mimectic potential (mediated by increase in cytosolic Ca2 + of
1999;13:128–32.
islets) also helps to control hyperglycemic excursions in
[12] Iyer SR. Tamarindus indica Linn. In: Warrier PK, Nambiar VPK,
diabetes. Furthermore, the study has also provided a plausible Kutty CR, editors. Indian Medicinal Plants, Vol. 5. Orient
mechanistic explanation for the effect of seeds and fruits of T Longman Limited; 1995. p. 235–6.
indica on lipid metabolism [13,37], as the SREBP-1c mRNA [13] Maiti R, Das UK, Ghosh D. Attenuation of hyperglycemia and
concentration in the liver was improved. Therefore, polyphe- hyperlipidemia in streptozotocin induced diabetic rats by
nol-rich seeds of tamarind and its aqueous extract could be aqueous extract of seed of Tamarindus indica. Biol Pharm Bull
2005;28:1172–6.
added as a supplement in human health care food or drugs
[14] Khillare B. Orientation training course on research method-
or combined with other hypoglycemic agents. The present ology in reproductive biomedicine. Department of Repro-
study on the antidiabetic potential of tamarind seeds ductive Biomedicine, National Institute of Health and Family
provides a strong rationale for the study of compounds Welfare, Government of India; 2000.
and functionalities involved in its antihyperglycemic action, [15] Weir GC, Clore EE, Zma-Chinsky CJ, Bonnier-weir S. Islet
thereby offering a foundation for a new herbal drug in secretion in new experimental model of non-insulin depen-
dent diabetes. Diabetes Metab Rev 1981;30:590–4.
diabetes therapy.
[16] Hsu SM, Raine L, Fanger H. Use of avidin-biotin peroxidase
complex (ABC) in immunoperoxidase techniques: a compar-
ison between ABC and unlabelled antibody (PAP) procedures.
Acknowledgment J Histochem Cytochem 1981;29:577–80.
[17] Matsuno T, Sasaki H, Nakagawa K, Nakagawa K, Ishido N,
Matsuda H, et al. Fas antigen expression and apoptosis in
The authors acknowledge the help of Panacea Biotech and
kidney allografts. Transplant Proc 1997;29:177–8.
Ranbaxy, India, for providing glimeperide and metformin,
[18] Lacy PE, Kostianovsky M. Method for the isolation of intact
respectively. Funding for the study was by the Govt of NCT, islets of Langerhans from the rat pancreas. Diabetes 1967;16:
Delhi, India (MH-2203-O.E.1.1[1][1][1][4]). The authors thank 35–9.
Dhiren Bahl of Wordsway Copyediting for editorial assistance. [19] Thomas PJ, Gaspers LD, Pharr C, Thomas JA. Continuous
measurement of mitochondrial pH gradients in isolated
hepatocytes by difference ratio spectroscopy. Arch Biochem
REFERENCES Biophys 1991;288:250–61.
[20] Suh YH, Kim Y, Bang JH, Choi KS, Lee JW, Kim WH, et al.
Analysis of gene expression profiles in insulin-sensitive
[1] Ndsing JF. Role of hemeoxygenase in inflammation insulin tissues from pre-diabetic and diabetic Zucker diabetic fatty
signaling diabetes and obesity. Mediat Inflamm 2010;359: rats. J Mol Endocrinol 2005;34:299–315.
732–8. [21] Yoshihiko H, Takuya I, Kaoru Y, Nobuo K. Effects of Hachimi-
[2] Charron MJ, Brosius FC, Alper SL, Lodish HF. A glucose jio-gan (Ba-Wei-Di-Huang-Wan) on hyperglycemia in strepto-
transport protein expressed predominantly in insulin-re- zotocin induced diabetic rats. Biol Pharm Bull 2007;30:1015–20.
sponsive tissues. Proc Natl Acad Sci 1989;86:2535–9. [22] Nepal S, Malik S, Sharma AK, Bhartia S, Narender Kumar,
[3] Shepherd PR, Kahn BB. Glucose transporters and insulin Siddiqui KM, et al. Abresham ameliorates dyslipidemia,
action: implications for insulin resistance and diabetes hepaticsteatosis and hypertension in high fat diet fed rats by
mellitus. N Engl J Med 1999;341:248–57. repressing oxidative stress, TNF-α and normalizing NO
[4] Bell GI, Kayano T, Buse JB, Burant CF, Takeda J, Lin D. production. Exp Toxicol Pathol 2011, http://dx.doi.org/10.1016/
Molecular biology of mammalian glucose transporters. Dia- j.etp.2011.01.003.
betes Care 1990;13:198–208. [23] Nematbakhsh M, Khazaei M. The effect of estrogen on serum
[5] Shimomura I, Bashmakov Y, Ikemoto S, Horton JD, Brown MS, nitric oxide concentrations in normotensive and DOCA salt
Goldstein JL. Insulin selectively increases SREBP-1c mRNA in hypertensive ovariectomized rats. Clin Chim Acta 2004;344:
the livers of rats with streptozotocininduced, diabetes. Proc 53–7.
Natl Acad Sci 1999;96:13656–61. [24] Karl J, Burns G, Engel WD, Finke A, Kratzer M, Rollinger W.
[6] Prentki M, Matschinsky FM. Ca2 +, cAMP, and phospholipid- Development and standardization of a new immunoturbidi-
derived messengers in coupling mechanisms of insulin metric assay HbA1c Assay. Klin Lab 1993;39:991–6.
secretion. Physiol Rev 1987;67:1185–248. [25] Lopez JM, Bennett MK, Sanchez HB, Rosenfeld JM, Osborne TE.
[7] Hadjivassiliou V, Green MHL, James RFL, Swift, Clayton HL, Sterol regulation of acetyl coenzyme A carboxylase: a
Green IC. Insulin secretion DNA damage and apoptosis in mechanism for coordinate control of cellular lipid. Proc Natl
human and rat islets following exposure to nitric oxide, Acad Sci 1996;93:1049–53.
peroxynitrite and cytokines. Nitric Oxide 1998;2:429–41. [26] Peirson SP, Butler JN, Foster RG. Experimental validation of
[8] Komutarin T, Butterworth AL, Keil D, Chitsomboon B, Suttajit novel and conventional approaches to quantitative real-
M, Meade BJ. Extract of seed coat of Tamarindus indica L. timePCR data analysis. Nucleic Acids Res 2003;31:e73.
Inhibits nitric oxide production by murine macrophages in [27] Valdeolmillos M, Nadal A, Soria B, Garcia-Sancho J. Fluorescence
vitro and in vivo. Food Chem Toxicol 2004;42:649–59. digital image analysis of glucose-induced [Ca2 +]i oscillations in
[9] Tsuda T, Watanable M, Ohshima K, Yamamoto A, Kawakishi mouse pancreatic islets of Langerhans. Diabetes 1993;42:1210–4.
S, Osawa T. Antioxidative components isolated from the seed [28] Weigle DS. Pulsatile secretion of fuel-regulatory hormones.
of Tamarind (Tamarindus indica L). Agric Food Chem 1994;42: Diabetes 1987;36:764–75.
2671–4. [29] Borman H, Melzig MF. Inhibition of metallopeptidases by
[10] Joyeux M, Mortier F, Flurentin J. Screening of antiradical, flavonoids and related compounds. Die Pharmazie 2000;55:
antilipoperoxidant and hepatotoprotective effects of nine 129–32.
plant extracts used in Caribbean folk medicine. Phytother Res [30] Adeghate E, Donath T. Distribution of neuropeptide-Y and
1995;9:228–30. vasoactive intestinal polypeptide immunoreactive nerves in
636 N U TR IT ION RE S EA RCH 3 2 ( 2 0 12 ) 62 6 –6 36

normal and transplanted pancreatic tissue. Peptides 1990;11: [35] Thorens B, Wu YJ, Leahy JL, Weir GC. The loss of GLUT2
1087–92. expression by glucose unresponsive beta cells of db/db mice
[31] Nagai R, Fujiwara Y, Mera K. Usefulness of antibodies for is reversible and is induced by the diabetic environment.
evaluating the biological significance of AGEs. Ann N Y Acad J Clin Invest 1992;90:77–80.
Sci 2008;1126:38–41. [36] Valerio A, Cardile A, Cozzi V, Bracale R, Tedesco L, Pisconti A,
[32] Hirst DG, Robson T. Nitrosative stress as a mediator of apoptosis: et al. TNF-α downregulates eNOS expression and mitochon-
implications for cancer therapy. Curr Pharm Des 2010;16:45–55. drial biogenesis in fat and muscle of obese rodents. J Clin
[33] Pons P, Aoki A. Differential proliferation of somatostatin and Invest 2006;116:2791–8.
glucagons cells in rat pancreatic islets submitted to different [37] Martinello FSM, Soares JJ, Franco AC, Santos A, Sugohara SB,
stimuli. Ann Anat 1995;177:221–7. Garcia C, et al. Hypolipemic and antioxidant activities from
[34] Charron MJ, Katz EB, Olson AL. GLUT 4 gene regulation and Tamarindus indica L. pulp fruit extract in hypercholesterolemic
manipulation. J Biol Chem 1999;274:3253–6. hamsters. Food Chem Toxicol 2006;44:810–8.

También podría gustarte