ArticlePDF AvailableLiterature Review

Alloxan-induced diabetes, a common model for evaluating the glycemic-control potential of therapeutic compounds and plants extracts in experimental studies

Authors:

Abstract and Figures

Glycemic homeostasis refers to glucose balance or control within circulation in living organisms. It is normally and largely compromised in diabetes. The compromise when exacerbated, leads to several complications including retinopathy, nephropathy and neuropathy which are collectively known as diabetic complications and are the principal actors in co-morbidity and eventual mortality often associated with diabetes. The ability of therapeutic compounds including medicinal plants to restore glycemic balance or homeostasis in hyperglycemic condition is an index of their antidiabetic function and relevance. Alloxan and streptozotocin are the most popular diabetogenic agents used for assessing the antidiabetic or hypoglycemic capacity of test compounds. Notably, alloxan is far less expensive and more readily available than streptozotocin. On this ground, one will logically expect a preference for use of alloxan in experimental diabetes studies. Surprisingly, a sub meta-analysis of randomly selected studies conducted within the last one and half decade revealed otherwise. This observation necessitated the review of alloxan as a diabetogenic agent in animal studies.
Content may be subject to copyright.
Review
Alloxan-induced
diabetes,
a
common
model
for
evaluating
the
glycemic-control
potential
of
therapeutic
compounds
and
plants
extracts
in
experimental
studies
Osasenaga
Macdonald
Ighodaro
a,b,
*,
Abiola
Mohammed
Adeosun
a,b
,
Oluseyi
Adeboye
Akinloye
b
a
Department
of
Biochemistry,
Faculty
of
Sciences,
Lead
City
University,
Ibadan,
Nigeria
b
Department
of
Biochemistry,
College
of
Biosciences,
Federal
University
of
Agriculture,
Abeokuta
(FUNAAB),
Abeokuta,
Nigeria
1.
Introduction
Alloxan
which
is
chemically
known
as
5,5-dihydroxyl
pyrimi-
dine-2,4,6-trione
is
an
organic
compound,
a
urea
derivative,
a
carcinogen
and
cytotoxic
glucose
analog
[1].
The
compound
has
the
molecular
formulae,
C
4
H
2
N
2
O
4
and
a
relative
molecu-
lar
mass
of
142.06.
Alloxan
is
one
of
the
common
diabetogenic
agents
often
used
to
assess
the
antidiabetic
potential
of
both
pure
compounds
and
plant
extracts
in
studies
involving
diabetes.
Among
the
known
diabetogenic
agents
which
include
dithizone,
monosodium
glutamate,
gold
thioglucose,
high
fructose
load,
high
glucose
load
and
anti-insulin
serum;
alloxan
and
streptozotocin
(STZ)
are
the
most
widely
used
in
diabetes
studies.
The
current
average
cost
of
one
gram
of
m
e
d
i
c
i
n
a
5
3
(
2
0
1
7
)
3
6
5
3
7
4
*
Corresponding
author
at:
Department
of
Biochemistry,
Lead
City
University,
Ibadan,
Nigeria.
E-mail
addresses:
Ighodaro.macdonald@lcu.edu.ng,
macigho@gmail.com
(O.M.
Ighodaro).
a
r
t
i
c
l
e
i
n
f
o
Article
history:
Received
14
September
2017
Accepted
8
February
2018
Available
online
27
February
2018
Keywords:
Alloxan
Diabetes
mellitus
Diabetogenic
agent
Streptozotocin
Animals
a
b
s
t
r
a
c
t
Glycemic
homeostasis
refers
to
glucose
balance
or
control
within
circulation
in
living
organisms.
It
is
normally
and
largely
compromised
in
diabetes.
The
compromise
when
exacerbated,
leads
to
several
complications
including
retinopathy,
nephropathy
and
neu-
ropathy
which
are
collectively
known
as
diabetic
complications
and
are
the
principal
actors
in
co-morbidity
and
eventual
mortality
often
associated
with
diabetes.
The
ability
of
therapeutic
compounds
including
medicinal
plants
to
restore
glycemic
balance
or
homeo-
stasis
in
hyperglycemic
condition
is
an
index
of
their
antidiabetic
function
and
relevance.
Alloxan
and
streptozotocin
are
the
most
popular
diabetogenic
agents
used
for
assessing
the
antidiabetic
or
hypoglycemic
capacity
of
test
compounds.
Notably,
alloxan
is
far
less
expensive
and
more
readily
available
than
streptozotocin.
On
this
ground,
one
will
logically
expect
a
preference
for
use
of
alloxan
in
experimental
diabetes
studies.
Surprisingly,
a
sub
meta-analysis
of
randomly
selected
studies
conducted
within
the
last
one
and
half
decade
revealed
otherwise.
This
observation
necessitated
the
review
of
alloxan
as
a
diabetogenic
agent
in
animal
studies.
©
2018
The
Lithuanian
University
of
Health
Sciences.
Production
and
hosting
by
Elsevier
Sp.
z
o.o.
This
is
an
open
access
article
under
the
CC
BY-NC-ND
license
(http://creative-
commons.org/licenses/by-nc-nd/4.0/).
Available
online
at
www.sciencedirect.com
ScienceDirect
journal
homepage:
http://www.elsevier.com/locate/medici
https://doi.org/10.1016/j.medici.2018.02.001
1010-660X/©
2018
The
Lithuanian
University
of
Health
Sciences.
Production
and
hosting
by
Elsevier
Sp.
z
o.o.
This
is
an
open
access
article
under
the
CC
BY-NC-ND
license
(http://creativecommons.org/licenses/by-nc-nd/4.0/).
alloxan
and
STZ
are
respectively
1.5
and
200
US
dollars
respectively.
Due
to
relative
affordability
and
availability,
one
will
logically
expect
that
alloxan
will
be
more
used
compared
to
STZ
[2].
However,
a
literature
survey
and
sub
meta-analysis
that
we
carried
out
on
the
use
of
both
compounds
in
experimental
diabetes
studies
conducted
within
the
last
one
and
half
decade
(20002016)
suggested
otherwise
(Table
1).
Analysis
of
the
data
obtained
showed
that
30.3%
of
the
studies
used
alloxan
while
57.9%
made
used
of
STZ
as
a
diabetogenic
agent,
and
others
which
used
glucose,
fructose
and
genetic
diabetic
mice
constituted
the
remaining
11.8%
(Table
1).
2.
Alloxan-induced
diabetes
Alloxan-induced
diabetes
is
a
form
of
insulin-dependent
diabetes
mellitus
that
occurs
as
a
result
of
alloxan
adminis-
tration
or
injection
to
animals
[78,79].
It
has
been
successfully
induced
in
a
variety
of
animal
species;
rabbits,
mice,
rats,
monkeys,
cats
and
dogs
[80,81].
Alloxan
has
been
administered
in
single
or
multiple
doses,
through
different
routes
(intraper-
itoneal,
intravenous
and
subcutaneous);
with
single
intraperi-
toneal
administration
apparently
the
most
employed
mode.
The
dosage
of
the
drug
also
varies
across
studies,
ranging
between
90
and
200
mg/kg
of
body
weight
(BW),
with
150
mg/
kg
BW
being
the
most
frequently
used
dosage.
Animal
species,
route
of
administration
and
nutritional
status
have
been
considered
to
play
a
role
in
determining
the
dose
of
alloxan
appropriate
for
induction
of
diabetes
[2].
However,
single
intraperitoneal
administration
of
the
drug
at
170200
mg/kg
BW
appears
to
be
most
effective
[2].
Alloxan
was
rst
isolated
by
Brugnatelli
in
1818
and
initially
described
by
Frederick
Wohler
and
Justin
Liebig
in
1838
[83].
Alloxan
causes
diabetes
by
a
mechanism
which
basically
involves
partial
degradation
of
the
beta
(b)
cells
of
pancreatic
islets
and
subsequent
compromise
in
the
quality
and
quantity
of
insulin
produced
by
these
cells.
Its
use
as
a
diabetogenic
drug
in
experimental
animals
was
rst
reported
by
Dunn
and
McLetchie
in
their
study
in
which
they
successfully
induced
diabetes
in
experimental
rabbits
[78].
Thereafter,
several
authors
have
used
alloxan-induced
diabetes
model
as
a
‘‘study
tool’’
to
elucidate
the
pathophysiology
of
the
disease
and
much
more
as
a
‘‘search
engine’’
for
antidiabetic
compounds
with
better
therapeutic
characteristics.
The
model
employs
two
distinct
pathological
effects
which
include
selective
inhibition
of
glucose-stimulated
insulin
secretion,
and
induced
formation
of
reactive
oxygen
species
(ROS)
which
promotes
selective
necrosis
of
beta
cells
of
the
pancreas.
Both
effects
collectively
result
in
a
pathophysiologi-
cal
state
of
insulin-dependent
diabetes
or
type
1-like
diabetes
mellitus
in
cells
[78,84].
The
former
is
associated
with
specic
inhibition
of
a
pancreatic
glucose
sensor
enzyme,
glucokinase
by
alloxan
whereas
the
latter
is
rather
connected
with
the
redox
cycling
ability
of
alloxan
which
results
in
ROS
genera-
tion.
More
importantly,
both
effects
have
been
linked
to
the
chemical
properties
of
alloxan
as
well
as
its
structure.
2.1.
Chemical
features
of
alloxan
and
their
contribution
to
its
diabetogenicity
The
diabetogenicity
of
alloxan
is
underlined
by
its
selective
cellular
uptake
by
beta
cells
of
the
pancreas
and
consequent
Table
1
Randomly
selected
experimental
diabetic
studies
conducted
within
the
last
two
decades
(19952016).
Authors
Diabetogenic
agent
used
Authors
Diabetogenic
agent
used
Authors
Diabetogenic
agent
used
Kameswararao
et
al.
[3]
Alloxan
Yadav
et
al.
[28]
Fructose
Ighodaro
et
al.
[53]
Alloxan
Pari
and
Saravanan
[4]
Alloxan
Al-Azzawie
and
Alhamdani
[29]
Alloxan
Nyomaan
et
al.
[54]
STZ
Eidi
et
al.
[5]
STZ
Verspoh
et
al.
[30]
Glucose
Petchi
et
al.
[55]
STZ
Maiti
et
al.
[6]
STZ
Jaiswal
et
al.
[31]
STZ
Akaladi
et
al.
[56]
STZ
Gupta
et
al.
[7]
STZ
Sunil
et
al.
[32]
STZ
Olatunji
et
al.
[57]
Fructose
Bagri
et
al.
[8]
STZ
Jelodar
et
al.
[33]
Alloxan
Daud
et
al.
[58]
STZ
Tabuchi
et
al.
[9]
STZ
Ighodaro
et
al.
[34]
STZ
Cao
et
al.
[59]
STZ
Ragavan
and
Krishnakumari
[10]
Alloxan
Asgary
et
al.
[35]
Alloxan
Saravanan
et
al.
[60]
STZ
Dewanjee
et
al.
[11]
Alloxan
Kumar
et
al.
[36]
Alloxan
Hakkim
et
al.
[61]
Alloxan
Yang
et
al.
[12]
Alloxan
Venkatesh
et
al.
[37]
Alloxan
Lee
et
al.
[62]
STZ
Jala
et
al.
[13]
Fructose
Paril
et
al.
[38]
Alloxan
Poudyal
et
al.
[63]
CHO/High
fat
Jemai
et
al.
[14]
Alloxan
Shanmugasundaram
et
al.
[39]
Alloxan
Dzeuet
et
al.
[64]
STZ
Sridhar
et
al.
[15]
STZ
Nugroho
et
al.
[40]
Fructose
Anathan
et
al.
[65]
Alloxan
Pandit
et
al.
[16]
STZ
Jelastin
et
al.
[41]
Alloxan
Miura
et
al.
[66]
KK
mice
Papato
et
al.
[17]
STZ
Satheesh
and
Paril
[42]
Alloxan
Oliveira
et
al.
[67]
STZ
Zhang
and
Tan
[18]
STZ
Wainstein
et
al.
[43]
STZ
Singh
et
al.
[68]
STZ
Sarkhail
et
al.
[19]
STZ
Moon
[44]
STZ
Bnouham
et
al.
[69]
STZ
Habibuddin
et
al.
[20]
STZ
Chung
et
al.
[45]
STZ
Surana
et
al.
[70]
Alloxan
Liu
et
al.
[21]
STZ
Ahangarpour
et
al.
[46]
Fructose
Anwar
et
al.
[71]
STZ
Abdelmoaty
et
al.
[22]
STZ
Prince
et
al.
[47]
Alloxan
Ju
et
al.
[72]
STZ
Oku
et
al.
[23]
STZ
Abedinzade
et
al.
[48]
STZ
Gandhi
et
al.
[73]
STZ
Orhan
et
al.
[24]
STZ
Kook
et
al.
[49]
STZ
Shirwaikar
et
al.
[74]
STZ
Ezquer
et
al.
[25]
STZ
Kumar
et
al.
[50]
STZ
Chen
et
al.
[75]
STZ
Zhao
et
al.
[26]
STZ
Shajeela
et
al.
[51]
Alloxan
Djomeni
et
al.
[76]
STZ
Singh
et
al.
[27]
STZ
Sowmia
and
Kokilavani
[52]
Alloxan
Veerapur
et
al.
[77]
STZ
STZ,
streptozotocin.
m
e
d
i
c
i
n
a
5
3
(
2
0
1
7
)
3
6
5
3
7
4366
accumulation
in
these
cells
[85].
The
chemical
properties
of
alloxan
and
how
they
contribute
to
its
toxicity
or
diabeto-
genicity
are
shown
below.
2.1.1.
Alloxan
shares
semblance
with
glucose
in
molecular
shape
and
hydrophilicity
Alloxan
shares
huge
structural
(molecular
shape)
similarity
with
glucose
[86].
It
is
a
b-cell
toxic
glucose
analog
with
hydrophilic
characteristic
(with
a
partition
coefcient
of
1.8)
and
exists
as
alloxan
monohydrate
in
aqueous
solutions
[1].
Glucose
is
a
hydrophilic
molecule
and
hence,
incapable
of
crossing
the
lipid
bilayer
of
the
plasma
membrane
on
its
own
into
the
cytosol.
Rather
it
is
transported
via
a
facilitated
diffusion
transport
mechanism
involving
a
transport
protein
known
as
glucose
transporter
2
(GLUT2)
which
is
located
in
plasma
membranes
of
cells.
In
the
same
manner,
due
to
structural
similarity
of
alloxan
to
glucose,
its
movement
across
the
plasma
membrane
into
the
cytosol
of
the
beta
cells
is
also
carried
out
by
GLUT2
[1,87].
Interestingly,
alloxan
does
not
in
any
way
inhibit
the
activity
of
GLUT2
and
this
attribute
signicantly
enhances
its
uptake
by
beta
cells,
resulting
in
its
selective
bio-accumulation
and
toxicity
in
these
cells
[88,89].
This
view
is
substantiated
by
the
fact
that
alloxan
has
been
reported
to
be
non-toxic
to
insulin-producing
cells
which
lack
or
are
decient
in
the
GLUT
2.
Secondly,
N-substitution
with
the
alkyl
group
in
alloxan
produces
alloxan
derivatives
with
lipophilic
characteristic
and
these
compounds
have
been
noted
to
easily
transit
the
lipid
bilayer
of
the
plasma
membrane
without
the
assistance
of
glucose
transporter
2,
GLUT2
[90].
Consequently,
they
can
permeate
all
cell
types
including
those
which
do
not
express
GLUT2
and
cause
systemic
toxicity
rather
than
diabetogenicity
[91].
It
is
important
to
note
that
the
mechanism
of
glucose
uptake
in
humans
is
contrastingly
different
from
that
of
animals
(rodents)
and
this
probably
accounts
for
why
alloxan,
even
at
high
concentrations
are
non-toxic
to
humans.
2.1.2.
Alloxan
is
a
weak
acid
Alloxan
is
a
weak
acid,
a
barbituric
acid
derivative
(5-
ketobarbituric
acid)
and
hence,
readily
attacks
thiol
reagents
or
the
sulfhydryl
group
(SH)
present
in
proteins.
The
selective
inhibition
of
glucose-stimulated
insulin
secretion
has
been
described
as
the
major
pathological
effect
of
alloxan
[1,85]
and
is
directly
linked
with
the
ability
of
alloxan
to
oxidized
or
attack
the
thiol
group
present
in
glucokinase,
a
glucose
phosphorylating
enzyme
which
plays
a
key
role
as
glucose
sensor
in
the
pancreas
and
liver.
2.1.3.
The
chemical
structure
of
alloxan
has
a
5-carbonyl
group
The
chemical
structure
of
alloxan
(Fig.
1)
has
a
5-carbonyl
group
which
is
hyper
reactive
with
thiol
groups,
and
this
is
indicative
of
a
structure-function
relationship
in
alloxan
toxicity
or
diabetogenicity.
Glucokinase
has
two
thiol
groups
(SH)
in
its
binding
site
which
makes
it
exceptionally
susceptible
to
oxidation
by
alloxan
[92].
The
binding
of
alloxan
to
glucokinase
results
in
the
formation
of
a
disulphide
bond
and
consequent
inactivation
of
the
enzyme.
This
phenome-
non
occurs
as
fast
as
within
the
rst
minute
of
exposure
of
the
enzyme
to
alloxan
and
accounts
for
the
selective
inhibition
of
glucose-stimulated
insulin
secretion
usually
observed
within
minutes
of
alloxan
injection
[86].
Although,
alloxan
can
inhibit
the
activities
of
several
other
functionally
important
thiol-
enzymes
such
as
phosphofructokinase
[93],
aconitase
[92],
hexokinase
[94]
and
Ca
2+
/calmodulin-dependent
protein
kinase
[95]
but
glucokinase
is
the
most
susceptible
thiol
enzyme
to
alloxan
attack
in
the
beta
cells
[96,97].
The
inhibitory
action
of
alloxan
on
glucokinase
hinders
glucose
oxidation,
and
by
extension
the
formation
of
adenosine
triphosphate
(ATP)
In
turn,
lack
of
ATP
suppresses
the
signal
generating
metabolic
ux
necessary
for
glucose-stimulated
insulin
secretion
[98].
The
same
mechanism
may
likely
be
responsible
for
the
inhibitory
action
of
alloxan
on
insulin
biosynthesis
[99].
2.1.4.
Alloxan
is
a
very
unstable
compound
In
addition,
alloxan
is
a
very
unstable
compound,
a
property
that
enables
it
to
readily
undergo
redox
cycling.
In
the
presence
of
intracellular
thiols
especially
glutathione
(GSH),
alloxan
undergoes
a
persisting
continuous
cyclic
reaction
to
generate
reactive
oxygen
species
(ROS)
such
as
superoxide
radical
anion
(O
2

)
and
hydroxyl
radical
(
OH)
via
the
auto-
oxidation
of
its
reduction
product,
dialuric
acid.
The
process
involves
the
reduction
of
alloxan
to
dialuric
acid
and
re-
oxidization
of
dialuric
acid
to
alloxan
[100].
Re-oxidation
of
alloxan
to
dialuric
acid
causes
a
release
of
alloxan
radical
that
in
the
presence
of
oxygen
generates
O
2

(Fig.
2).
O
2

is
usually
dismutated
to
a
relatively
harmless
hydrogen
peroxide
(H
2
O
2
)
by
superoxide
dismutase
(SOD),
an
antioxidant
enzyme
present
in
virtually
in
all
tissues
(Fig.
2).
Catalase,
another
antioxidant
enzyme
is
required
to
prevent
the
accumulation
of
H
2
O
2
and
its
consequent
conversion
to
hydroxyl
radical,
by
quick
degradation
of
the
compound
to
water
and
molecular
oxygen.
However,
catalase
activity
is
very
low
in
the
pancreas
[101]
and
as
a
result
H
2
O
2
accumulates,
leading
to
its
conversion
to
highly
reactive
hydroxyl
radical
through
Fenton
reaction
(Fig.
2).
Hydroxyl
radical
is
apparently
the
most
dangerous
radical
in
the
cell
and
considered
to
be
the
principally
culprit
in
beta
cell
toxicity
and
alloxan
diabeto-
genicity.
Damage
of
pancreatic
beta
cells
by
ROS
has
been
linked
to
fragmentation
of
DNA
of
these
cells,
leading
to
the
stimulation
of
poly
ADP-ribose
polymerase
1,
an
enzyme
that
plays
an
important
role
in
DNA
repair
process
[102].
Naturally,
compounds
with
sulfhydryl
group
(GSH,
cysteine
and
dithiothreitol)
should
protect
glucokinase
against
alloxan
inhibition
by
a
reductive
process,
but
they
have
to
continu-
ously
maintain
the
reduction
product,
dialuric
acid
in
its
reduced
form
in
order
to
effectively
protect
the
enzyme
Fig.
1
Chemical
structure
of
alloxan.
m
e
d
i
c
i
n
a
5
3
(
2
0
1
7
)
3
6
5
3
7
4
367
[92,103].
Unfortunately,
this
is
not
the
case,
as
the
thiols
are
often
exhausted
by
the
persisting
continuous
cyclic
reaction
of
alloxan.
Winterbourn
and
Munday
[104]
informed
that
the
amount
of
reduced
GSH
available
in
a
cell
for
redox
cycling
diminishes
gradually
and
thus
fosters
a
lower
pro-oxidative
ratio
between
alloxan
and
GSH,
rather
than
a
higher
antioxidative
ratio.
This
explains
why
co
administration
of
thiols
such
as
GSH
or
cysteine
with
alloxan
tends
to
ameliorate
the
toxic
and
diabetogenic
effects
of
alloxan
as
reported
by
a
number
of
other
studies
[105107].
2.2.
Alloxan-induced
diabetes
is
characterized
by
multiphasic
blood
glucose
response
Alloxan
induces
a
multiphasic
blood
glucose
response
when
injected
into
to
experimental
animals
as
noted
in
our
recent
study
in
which
we
examined
the
time
course
effects
of
alloxan
in
Wistar
rats.
Within
36
h
post
alloxan
administration,
several
phases
of
glucose
response
were
observed
in
the
animals
administered
170
and
200
mg/kg
BW
alloxan
(Fig.
3).
The
observation
is
similar
to
previous
reports
on
blood
glucose
behavior
or
response
to
alloxan
[1,2,85,108].
Some
of
these
authors
also
noted
that
changes
in
blood
glucose
concentra-
tion
are
accompanied
by
corresponding
inverse
changes
in
the
plasma
insulin
concentration.
Lenzen
[1]
postulated
that
blood
glucose
multiphasic
response
to
alloxan
injection
begins
in
the
rst
few
minutes
with
a
transient
hypoglycemic
phase
that
lasts
maximally
for
30
min.
This
event
has
been
adduced
to
a
transient
hyper-
insulinemia
that
is
probably
due
to
a
momentary
increase
in
ATP
level
resulting
from
the
temporary
effects
of
alloxan
inhibition
of
glucokinase.
The
second
phase
that
usually
takes
place
1
h
post
alloxan
administration
is
characterized
by
upsurge
in
blood
glucose
concentration
and
concomitant
decrease
in
plasma
insulin
concentration.
This
is
the
rst
hyperglycemic
effect
of
alloxan
and
it
lasts
for
a
period
of
24
h.
In
our
study,
alloxan
induced
diabetic
hyperglycemia
(blood
glucose
200
mg/dL
or
11.1
mmol/
L)
in
rats
1
h
post
its
administration.
Previous
authors
have
also
noted
the
immediate
diabetogenicity
of
alloxan
following
its
administration
to
experimental
animals.
Lenzen
[1]
in
his
study
titled
‘‘Alloxan
and
streptozotocin
diabetes’’
informed
that
notable
hyperglycemia
commenced
in
experimental
rats
1
h
after
alloxan
injection.
Similar
reports
that
alloxan
administra-
tion
to
rats
causes
immediate
hyperglycemia
which
reaches
its
peak
within
two
or
three
hours'
had
been
earlier
communicated
by
Goldner
and
Gomori
[80].
Inhibition
of
insulin
secretion
from
the
pancreatic
beta
cells
due
to
ROS
attack
accounts
for
this
phase
of
alloxan
diabetogenicity
[109].
According
to
Szkudelski
[85],
alloxan
is
a
hydrophilic
and
unstable
substance
with
a
half-
life
of
1.5
min
at
neutral
pH
and
37
8C.
This
implies
that
the
time
for
alloxan
degradation
(metabolism)
is
sufciently
short
enough
to
allow
it
to
reach
the
pancreas
very
fast
and
in
deleterious
amount.
This
also
explains
and
buttresses
the
1
h
post
hyperglycemic
effect
of
alloxan.
As
alloxan
uptake
by
the
insulin-secreting
beta
cells
of
the
pancreas
reaches
its
maximum,
its
toxicity
via
ROS
increases,
leading
to
induced
rupture
of
the
secretory
granules
and
cell
membrane
of
the
beta
cells
[1,85,97,110,111].
The
attendant
effect
of
this
burst
up
is
ooding
of
the
circulation
with
insulin,
a
pathophysiological
condition
that
results
in
a
severe
transitional
hypoglycemic
phase
which
is
observable
a
couple
of
hours
after
alloxan
injection.
The
hypoglycemic
phase
in
alloxan
diabetogenicity
has
also
been
associated
with
the
ability
of
alloxan
to
cause
signicant
inux
of
free
Ca
2+
into
the
cytosol
of
pancreatic
islet
beta
cells,
thereby
compromising
the
intracellular
calcium
homeostasis
[112].
The
process
involves
the
depolarization
of
the
pancreatic
beta
cells,
which
facil-
itates
further
calcium
entry
into
pancreatic
cells
via
voltage
dependent
calcium
channels.
High
intracellular
level
of
Ca
2+
has
been
noted
to
contribute
signicantly
to
super
high
level
of
insulin
release
[85].
Fig.
2
Formation
of
ROS
through
redox
cycling
of
alloxan.
m
e
d
i
c
i
n
a
5
3
(
2
0
1
7
)
3
6
5
3
7
4368
Hypoglycemia
is
characteristic
of
experimental
diabetes
and
the
phase
has
been
noted
to
last
for
at
least
3
h
or
more
[1,113,114]
and
is
largely
responsible
for
the
mortality
associated
with
alloxan-induced
diabetes.
This
view
is
consistent
with
the
opinion
of
Goldner
and
Gomori
[80],
who
hinted
that
alloxan-induced
hyperglycemia
in
rats
is
followed
by
a
severe
and
fatal
hypoglycemia,
which
after
a
duration
of
several
hours
yields
to
a
nal
hyperglycemia,
last
phase
of
alloxan
induced
diabetes.
The
last
phase
of
the
blood
glucose
response
to
alloxan
administration
is
touted
to
be
a
permanent
diabetic
hypergly-
cemic
phase
that
takes
place
between
24
and
48
h
after
alloxan
administration.
Supposedly,
there
is
complete
degranulation
and
loss
of
structural
integrity
of
the
beta
cells
during
this
phase
[1,113].
The
incidence
in
this
phase
indicates
that
other
cell
types
of
the
pancreas
are
spared
of
alloxan
toxicity,
substantiating
the
theory
of
selective
uptake
of
alloxan
by
the
insulin-producing
cells
(pancreatic
beta
cells)
[115,116].
2.3.
Protective
effects
of
glucose
against
alloxan
toxicity
and
diabetogenicity
It
is
a
popular
postulation
that
blood
glucose
protects
the
pancreatic
islet
beta
cells
against
the
toxicity
and
diabeto-
genicity
of
alloxan
[98].
There
are
a
couple
of
reasons
to
believe
that
this
is
possible.
Principally,
the
mechanism
by
which
alloxan
elicits
its
toxicity
and
diabetogenicity
provides
that
possibility.
As
already
stated,
the
toxic
and
diabetogenic
effects
of
alloxan
are
underlined
by
its
selective
inhibition
of
glucose-stimulated
insulin
secretion
via
inactivation
of
gluco-
kinase
and
selective
necrosis
of
the
beta
cells
via
induced
ROS.
These
two
processes
are
sequel
to
alloxan
uptake
by
beta
cells
and
subsequent
accumulation
in
these
cells.
Alloxan
uptake
by
beta
cells
is
facilitated
by
GLUT2.
Since
both
glucose
and
alloxan
due
to
similarity
in
molecular
shape
competes
for
the
same
transport
protein,
it
therefore
implies
that
high
level
of
glucose
in
circulation
will
automatically
lower
the
chance
of
alloxan
binding
to
GLUT2.
Even
at
a
relatively
equal
concentration
of
both
molecules,
GLUT2
has
a
stronger
afnity
for
glucose
than
alloxan
and
thus
favors
the
binding
of
glucose
compared
with
alloxan
[90,116118].
This
effect
will
consequently
minimize
alloxan
uptake
and
invariably
affects
its
ability
to
induce
diabetes.
It
has
also
been
suggested
that
glucose
through
the
pentose
phosphate
pathway
has
the
ability
to
supply
reduced
nicotinamide
adenine
dinucleotide
phosphate
(NADPH)
and
reduced
nico-
tinamide
adenine
dinucleotide
(NADH)
which
are
capable
of
recycling
GSH,
i.e.
keeps
it
in
its
reduced
form
[1].
This
in
itself
will
effectively
attenuate
the
ability
of
alloxan
to
generate
reactive
radicals
through
redox
cycling.
More
so,
high
level
of
blood
glucose
will
protect
glucokinase
and
prevent
its
exposure
to
alloxan
attack.
This
happens
in
such
a
way
that
once
glucokinase
is
bound
by
glucose,
the
sulfhydryl
groups
in
its
glucose-binding
site
is
no
more
available
for
alloxan
attack
[98].
The
protection
offered
by
glucose
explains
why
fed
animals
are
less
sensitive
to
alloxan
induced
diabetes
compared
with
fasted
animals
with
relatively
lower
blood
glucose
level
[119].
In
the
same
manner,
animals
fed
on
fat
diet
have
been
observed
to
be
more
susceptible
to
alloxan
diabetogenicity
relative
to
those
fed
on
high
carbohydrate
and
protein
prior
to
alloxan
injection.
2.4.
Limitations
of
alloxan-induced
diabetes
The
effectiveness
of
alloxan
for
induction
of
experimental
diabetes
has
been
queried
by
a
number
of
investigators.
This
is
rightly
so
as
noticeable
limitations
have
been
associated
with
the
use
of
alloxan
as
a
diabetogenic
agent.
Jain
and
Arya
[120]
highlighted
several
anomalies
and
inconsistencies
in
alloxan-
induced
diabetes
model,
and
we
are
of
the
opinion
that
the
concerns
raised
by
these
authors
should
be
given
some
level
of
consideration
and
attention.
Instability
and
auto-reversibility
of
alloxan-induced
hyper-
glycemia
is
particularly
of
utmost
concern.
Alloxan
when
administered
causes
multiphasic
glucose
response
character-
ized
by
inconsistent
increase
and
decrease
in
blood
glucose
Fig.
3
Time
course
effect
of
alloxan
on
blood
glucose
level
of
rat.
m
e
d
i
c
i
n
a
5
3
(
2
0
1
7
)
3
6
5
3
7
4
369
concentration
[1,2,108].
In
other
words,
the
hyperglycemia
induced
by
alloxan
is
not
sufciently
stable
for
proper
evaluation
of
the
antidiabetic
or
hypoglycemic
potential
of
test
compounds.
Even
in
few
cases
where
apparent
stability
is
achieved,
the
duration
of
such
stable
hyperglycemia
is
on
the
average
less
than
a
month
and
this
period
is
not
adequate
for
proper
evaluation
of
a
test
drug.
This
often
leads
to
illusive
conclusion
on
the
antidiabetic
relevance
of
the
test
compound.
According
to
Misra
and
Aiman
[108],
a
wide
range
of
uctuations
in
the
blood
glucose
level
and
auto
reversal
from
conrmed
diabetic
hyperglycemia
to
the
non-diabetic
range
is
a
major
setback
as
regards
alloxan-induced
diabetes
model.
Another
problem
with
alloxan
is
that
its
diabetogenic
and
toxic
effects
on
animals
vary
widely,
even
among
those
belonging
to
the
same
species.
Such
inconsistent
effect
makes
the
drug
an
unreliable
model
for
afrming
the
antidiabetic
potency
of
test
compounds,
a
view
shared
by
previous
authors
[108,120].
Moreover,
alloxan
does
not
exactly
induce
the
human
type
2
diabetes
mellitus
[121]
which
accounts
for
about
9095%
of
all
diabetic
cases.
In
support
of
this,
Jain
and
Arya
[120]
drew
our
attention
to
a
couple
of
test
compounds
reported
to
have
exhibited
notable
antidiabetic
activities
against
alloxan-
induced
diabetes
but
were
found
to
be
ineffective
against
human
diabetes.
Alloxan
has
been
noted
to
stimulate
a
type
1
form
of
diabetes
when
used
in
animals.
This
form
of
diabetes
is
often
associated
with
high
level
of
ketoacidosis
that
arguably
is
partly
responsible
for
the
high
animal
mortality
rate
(3060%)
[120]
usually
observed
with
use
of
alloxan
as
a
diabetogenic
agent.
Besides,
the
mechanism
of
alloxan
diabetogenicity
encloses
a
chronic
measure
of
toxicity
involving
free
radical
generation,
particularly
(
OH).
No
doubt,
this
play
a
bigger
role
in
the
mortality
of
experimental
animals
exposed
to
alloxan.
Mortality
from
diabetes
has
been
adduced
to
either
initial
hypoglycemic
shock
or
emergence
of
diabetic
complications
or
direct
kidney
tubular
cell
toxicity
[85].
The
practice
of
placing
alloxan-treated
animals
on
510%
glucose
solution
in
a
bid
to
prevent
hypoglycemic
shock
is
often
observed
but
this
intervention
appears
not
to
be
signicantly
helpful,
and
thus
the
problem
of
mortality
persists.
High
mortality
rate
is
a
major
drawback
in
the
use
of
alloxan
diabetic
model.
First,
it
increases
the
nancial
burden
of
the
study
as
several
animals
more
than
required
have
to
be
used
in
attempt
to
carry
the
study
to
a
meaningful
end.
Secondly,
it
does
not
allow
for
proper
evaluation
of
the
antidiabetic
potential
of
the
investigated
compound
or
test
drug.
2.5.
Comparing
alloxan
with
streptozotocin
as
diabetogenic
agents
STZ
has
notable
advantages
over
alloxan
as
chemical
agents
for
induction
of
experimental
diabetes,
thus,
is
often
preferred
to
the
latter
(alloxan).
For
instance,
STZ
has
longer
half-life
(15
min
against
1.5
min
of
alloxan)
[122].
This
makes
it
more
stable
in
solution
before
and
after
injection
into
animals.
STZ-
induced
hyperglycemia
is
relatively
more
stable
and
for
a
longer
duration
(as
much
as
three
months
compared
to
alloxan-induced
hyperglycemia
that
can
only
be
sustained
for
less
than
a
month).
Moreover,
the
mechanism
of
STZ
diabetogenicity
is
less
associated
with
cellular
toxicity,
hence,
lesser
animal
mortality.
Alloxan
on
the
contrary,
induces
diabetes
by
a
mechanism
characterized
by
incidences
of
ketosis,
ROS
toxicity,
and
high
mortality
rate
which
is
particularly
a
major
setback
in
experimental
diabetes
studies
[85].
One
reason
for
this
is
that
STZ
is
more
selective
to
islet
beta
cells
than
alloxan
which
causes
severe
damage
to
other
cell
types
which
express
GLUT2
(systemic
toxicity).
More
so,
STZ-induce
diabetes
is
associated
with
well
characterized
diabetic
complications
unlike
alloxan-induced
diabetes
[1].
In
addition,
compared
to
alloxan,
STZ
diabeto-
genicity
is
not
severely
interfered
with
by
blood
glucose
level.
Overall,
STZ
diabetogenicity
is
more
effective
and
with
lesser
variation
with
animal
species.
2.6.
Suggestions
to
improve
the
use
of
alloxan
as
a
diabetogenic
drug
1.
Alloxan
is
very
unstable,
and
with
a
half-life
of
about
1.5
min,
it
could
easily
disintegrate
when
left
to
stand
in
aqueous
solutions.
Therefore,
when
used
as
a
diabetogenic
agent,
it
should
be
freshly
prepared.
In
a
case
where
the
animals
to
be
injected
are
quite
many,
it
is
advisable
that
the
appropriate
amount
of
alloxan
for
a
specic
number
of
animals
(i.e.
5)
is
measured
in
replicates
for
different
batches
of
animals.
This
means
that
alloxan
for
a
batch
of
animals
(n
=
5)
is
dissolved
in
freshly
prepared
0.9%
saline
just
before
the
commencement
of
administration.
This
practice
improves
alloxan
diabetogenicity.
On
the
contrary,
when
all
the
animals
in
a
large
group
is
injected
from
the
same
alloxan
preparation,
there
is
a
possibility
that
the
last
set
of
animals
administered
the
drug
may
not
receive
sufcient
amount
of
the
active
drug
due
to
disintegration.
According
to
Lenzen
and
Munday
[123],
alloxan
when
left
to
stand
in
aqueous
solutions
is
readily
converted
to
non-
diabetogenic
alloxanic
acid
due
to
spontaneous
decompo-
sition.
2.
Poor
diabetogenicity
and
easy
auto-reversal
of
alloxan-
induced
hyperglycemia
is
very
common
with
intraperito-
neal
doses
of
150
mg/kg
and
below
[85,124].
In
the
use
of
alloxan,
higher
dose
between
170
and
200
mg/kg
BW
have
been
noted
to
be
more
effective.
3.
Very
young
animals
have
been
observed
to
be
highly
resistant
or
less
susceptible
to
the
diabetogenic
effect
of
alloxan
[120,125].
Older
animals
should
be
preferably
used
in
diabetic
studies
involving
the
use
of
alloxan.
Antioxidant
defense
system
has
been
reported
to
decrease
with
age;
this
may
be
responsible
for
this
difference
in
agedependent
response
to
alloxan.
4.
Fed
animals
due
to
the
effect
of
blood
glucose
are
less
susceptible
to
alloxan
toxicity
and
diabetogenicity
[84,85].
Animals
should
therefore
be
fasted
for
at
least
12
h
prior
to
alloxan
injection.
Fasted
animals
have
relatively
low
blood
glucose
level.
This
physiological
condition
enhances
allox-
an
uptake
by
the
islet
beta
cells
and
consequently
improve
alloxan
diabetogenicity.
5.
Exogenous
GSH
has
been
reported
to
protect
well
against
alloxan
toxicity
which
is
often
connected
with
animal
mortality
[1].
Probably,
co-administration
of
very
low
m
e
d
i
c
i
n
a
5
3
(
2
0
1
7
)
3
6
5
3
7
4370
concentration
of
GSH
and
higher
dose
of
alloxan
(170
200
mg/kg)
should
be
considered
for
improved
diabetogeni-
city
of
alloxan.
6.
The
route
and
speed
of
administration
have
been
reported
to
affect
the
diabetogenicity
of
alloxan,
with
fast
or
rapid
intravenous
administration
preferred
to
slow
intravenous
and
intraperitoneal
(I.P.)
administration.
But
higher
rate
of
mortality
have
also
been
associated
with
rapid
intravenous
injection
[108].
In
alloxan
diabetes
studies,
intraperitoneal
injection
is
commonly
used.
Perhaps,
increasing
the
speed
of
intraperitoneal
administration
may
improve
alloxan
diabetogenicity.
Conict
of
interest
The
authors
state
no
conict
of
interest.
r
e
f
e
r
e
n
c
e
s
[1]
Lenzen
S.
The
mechanisms
of
alloxan-
and
streptozotocin-
induced
diabetes.
Diabetologia
2008;51(2):21626.
[2]
Federiuk
IF,
Casey
HM,
Quinn
MJ,
Wood
MD,
Ward
KW.
Induction
of
type-1
diabetes
mellitus
in
laboratory
rats
by
use
of
alloxan:
route
of
administration,
pitfalls,
and
insulin
treatment.
Compar
Med
2004;54(June
(3)):2527.
[3]
Kameswararao
B,
Kesavulu
MM,
Apparao
C.
Evaluation
of
antidiabetic
effect
of
Momordica
cymbalaria
fruit
in
alloxan-diabetic
rats.
Fitoterapia
2003;74(1):713.
[4]
Pari
L,
Saravanan
G.
Antidiabetic
effect
of
Cogent
db,
a
herbal
drug
in
alloxan-induced
diabetes
mellitus.
Compar
Biochem
Physiol
C:
Toxicol
Pharmacol
2002;131(January
(1)):1925.
[5]
Eidi
A,
Eidi
M,
Esmaeili
E.
Antidiabetic
effect
of
garlic
(Allium
sativum
L.)
in
normal
and
streptozotocin-induced
diabetic
rats.
Phytomedicine
2006;13(November
(9)):6249.
[6]
Maiti
R,
Jana
D,
Das
UK,
Ghosh
D.
Antidiabetic
effect
of
aqueous
extract
of
seed
of
Tamarindus
indica
in
streptozotocin-induced
diabetic
rats.
J
Ethnopharmacol
2004;92(May
(1)):8591.
[7]
Gupta
RK,
Kesari
AN,
Murthy
PS,
Chandra
R,
Tandon
V,
Watal
G.
Hypoglycemic
and
antidiabetic
effect
of
ethanolic
extract
of
leaves
of
Annona
squamosa
L.
in
experimental
animals.
J
Ethnopharmacol
2005;99(May
(1)):7581.
[8]
Bagri
P,
Ali
M,
Aeri
V,
Bhowmik
M,
Sultana
S.
Antidiabetic
effect
of
Punica
granatum
owers:
effect
on
hyperlipidemia,
pancreatic
cells
lipid
peroxidation
and
antioxidant
enzymes
in
experimental
diabetes.
Food
Chem
Toxicol
2009;47(January
(1)):504.
[9]
Tabuchi
M,
Ozaki
M,
Tamura
A,
Yamada
N,
Ishida
T,
Hosoda
M,
et
al.
Antidiabetic
effect
of
Lactobacillus
GG
in
streptozotocin-induced
diabetic
rats.
Biosci
Biotechnol
Biochem
2003;67(6):14214.
[10]
Ragavan
B,
Krishnakumari
S.
Antidiabetic
effect
of
T.
arjuna
bark
extract
in
alloxan
induced
diabetic
rats.
Indian
J
Clin
Biochem
2006;21(September
(2)):123.
[11]
Dewanjee
S,
Bose
S,
Sahu
R,
Mandal
S.
Antidiabetic
effect
of
matured
fruits
of
Diospyros
peregrina
in
alloxan-induced
diabetic
rats.
Int
J
Green
Pharm
2008;2(April
(2)):95.
[12]
Yang
XB,
Huang
ZM,
Cao
WB,
Zheng
M,
Chen
HY,
Zhang
JZ.
Antidiabetic
effect
of
Oenanthe
javanica
avone.
Acta
Pharmacol
Sin
2000;21(March
(3)):23942.
[13]
Jalal
R,
Bagheri
SM,
Moghimi
A,
Rasuli
MB.
Hypoglycemic
effect
of
aqueous
shallot
and
garlic
extracts
in
rats
with
fructose-induced
insulin
resistance.
J
Clin
Biochem
Nutr
2007;41(3):21823.
[14]
Jemai
H,
El
Feki
A,
Sayadi
S.
Antidiabetic
and
antioxidant
effects
of
hydroxytyrosol
and
oleuropein
from
olive
leaves
in
alloxan-diabetic
rats.
J
Agric
Food
Chem
2009;57
(September
(19)):8798804.
[15]
Sridhar
SB,
Sheetal
UD,
Pai
MR,
Shastri
MS.
Preclinical
evaluation
of
the
antidiabetic
effect
of
Eugenia
jambolana
seed
powder
in
streptozotocin-diabetic
rats.
Braz
J
Med
Biol
Res
2005;38(March
(3)):4638.
[16]
Pandit
R,
Phadke
A,
Jagtap
A.
Antidiabetic
effect
of
Ficus
religiosa
extract
in
streptozotocin-induced
diabetic
rats.
J
Ethnopharmacol
2010;128(March
(2)):4626.
[17]
Pepato
MT,
Folgado
VB,
Kettelhut
IC,
Brunetti
IL.
Lack
of
antidiabetic
effect
of
a
Eugenia
jambolana
leaf
decoction
on
rat
streptozotocin
diabetes.
Braz
J
Med
Biol
Res
2001;34
(March
(3)):38995.
[18]
Zhang
XF,
Tan
BH.
Anti-diabetic
property
of
ethanolic
extract
of
Andrographis
paniculata
in
streptozotocin-
diabetic
rats.
Acta
Pharm
Sin
2000;2(December
(12)):1157
64.
[19]
Sarkhail
P,
Rahmanipour
S,
Fadyevatan
S,
Mohammadirad
A,
Dehghan
G,
Amin
G,
et
al.
Antidiabetic
effect
of
Phlomis
anisodonta:
effects
on
hepatic
cells
lipid
peroxidation
and
antioxidant
enzymes
in
experimental
diabetes.
Pharmacol
Res
2007;56(September
(3)):2616.
[20]
Habibuddin
M,
Daghriri
HA,
Humaira
T,
Al
Qahtani
MS,
Hefzi
AA.
Antidiabetic
effect
of
alcoholic
extract
of
Caralluma
sinaica
L.
on
streptozotocin-induced
diabetic
rabbits.
J
Ethnopharmacol
2008;117(May
(2)):21520.
[21]
Liu
CT,
Wong
PL,
Lii
CK,
Hse
H,
Sheen
LY.
Antidiabetic
effect
of
garlic
oil
but
not
diallyl
disulde
in
rats
with
streptozotocin-induced
diabetes.
Food
Chem
Toxicol
2006;44(August
(8)):137784.
[22]
Abdelmoaty
MA,
Ibrahim
MA,
Ahmed
NS,
Abdelaziz
MA.
Conrmatory
studies
on
the
antioxidant
and
antidiabetic
effect
of
quercetin
in
rats.
Indian
J
Clin
Biochem
2010;25
(April
(2)):18892.
[23]
Oku
A,
Ueta
K,
Nawano
M,
Arakawa
K,
Kano-Ishihara
T,
Matsumoto
M,
et
al.
Antidiabetic
effect
of
T-1095,
an
inhibitor
of
Na
+
-glucose
cotransporter,
in
neonatally
streptozotocin-treated
rats.
Eur
J
Pharmacol
2000;391
(March
(1)):18392.
[24]
Orhan
N,
Aslan
M,
Hosbas
S,
Deliorman
OD.
Antidiabetic
effect
and
antioxidant
potential
of
Rosa
canina
fruits.
Pharmacogn
Mag
2009;5(October
(20)):309.
[25]
Ezquer
F,
Ezquer
M,
Contador
D,
Ricca
M,
Simon
V,
Conget
P.
The
antidiabetic
effect
of
mesenchymal
stem
cells
is
unrelated
to
their
transdifferentiation
potential
but
to
their
capability
to
restore
Th1/Th2
balance
and
to
modify
the
pancreatic
microenvironment.
Stem
Cells
2012;30
(August
(8)):166474.
[26]
Zhao
LY,
Lan
QJ,
Huang
ZC,
Ouyang
LJ,
Zeng
FH.
Antidiabetic
effect
of
a
newly
identied
component
of
Opuntia
dillenii
polysaccharides.
Phytomedicine
2011;18
(June
(8)):6618.
[27]
Singh
RK,
Mehta
S,
Jaiswal
D,
Rai
PK,
Watal
G.
Antidiabetic
effect
of
Ficus
bengalensis
aerial
roots
in
experimental
animals.
J
Ethnopharmacol
2009;123(May
(1)):1104.
[28]
Yadav
H,
Jain
S,
Sinha
PR.
Antidiabetic
effect
of
probiotic
dahi
containing
Lactobacillus
acidophilus
and
Lactobacillus
casei
in
high
fructose
fed
rats.
Nutrition
2007;23(January
(1)):628.
[29]
Al-Azzawie
HF,
Alhamdani
MS.
Hypoglycemic
and
antioxidant
effect
of
oleuropein
in
alloxan-diabetic
rabbits.
Life
Sci
2006;78(February
(12)):13717.
m
e
d
i
c
i
n
a
5
3
(
2
0
1
7
)
3
6
5
3
7
4
371
[30]
Verspohl
EJ,
Bauer
K,
Neddermann
E.
Antidiabetic
effect
of
Cinnamomum
cassia
and
Cinnamomum
zeylanicum
in
vivo
and
in
vitro.
Phytotherapy
Res
2005;19(March
(3)):2036.
[31]
Jaiswal
D,
Rai
PK,
Watal
G.
Antidiabetic
effect
of
Withania
coagulans
in
experimental
rats.
Indian
J
Clin
Biochem
2009;24(January
(1)):8893.
[32]
Sunil
C,
Duraipandiyan
V,
Agastian
P,
Ignacimuthu
S.
Antidiabetic
effect
of
plumbagin
isolated
from
Plumbago
zeylanica
L.
root
and
its
effect
on
GLUT4
translocation
in
streptozotocin-induced
diabetic
rats.
Food
Chem
Toxicol
2012;50(December
(12)):435663.
[33]
Jelodar
G,
Maleki
M,
Motadayen
M,
Sirus
S.
Effect
of
fenugreek,
onion
and
garlic
on
blood
glucose
and
histopathology
of
pancreas
of
alloxan-induced
diabetic
rats.
Indian
J
Med
Sci
2005;59(February
(2)):64.
[34]
Ighodaro
OM,
Akinloye
OA,
Ugbaja
RN,
Omotainse
SO.
Sapium
ellipticum
(Hochst)
Pax
Ethanol
Leaf
Extract
modulates
glucokinse
and
glucose-6-phosphatase
activities
in
streptozotocin
induced
diabetic
rats.
J
Coastal
Life
Med
2017;4(5):1626.
[35]
Asgary
S,
Parkhideh
S,
Solhpour
A,
Madani
H,
Mahzouni
P,
Rahimi
P.
Effect
of
ethanolic
extract
of
Juglans
regia
L.
on
blood
sugar
in
diabetes-induced
rats.
J
Med
Food
2008;11
(September
(3)):5338.
[36]
Kumar
V,
Khanna
AK,
Khan
MM,
Singh
R,
Singh
S,
Chander
R,
et
al.
Hypoglycemic,
lipid
lowering
and
antioxidant
activities
in
root
extract
of
Anthocephalus
indicus
in
alloxan
induced
diabetic
rats.
Indian
J
Clin
Biochem
2009;24
(January
(1)):659.
[37]
Venkatesh
S,
Reddy
BM,
Reddy
GD,
Mullangi
R,
Lakshman
M.
Antihyperglycemic
and
hypolipidemic
effects
of
Helicteres
isora
roots
in
alloxan-induced
diabetic
rats:
a
possible
mechanism
of
action.
J
Nat
Med
2010;64(July
(3)):295304.
[38]
Pari
L,
Amarnath
Satheesh
M.
Antidiabetic
effect
of
Boerhavia
diffusa:
effect
on
serum
and
tissue
lipids
in
experimental
diabetes.
J
Med
Food
2004;7(December
(4)):4726.
[39]
Shanmugasundaram
R,
Devi
KV,
Soris
TP,
Maruthupandian
A,
Mohan
VR.
Antidiabetic,
antihyperlipidaemic
and
antioxidant
activity
of
Senna
auriculata
(L.)
Roxb.
leaves
in
alloxan
induced
diabetic
rats.
Int
J
Pharm
Tech
Res
2011;3(2):74756.
[40]
Nugroho
AE,
Andrie
M,
Warditiani
NK,
Siswanto
E,
Pramono
S,
Lukitaningsih
E.
Antidiabetic
and
antihiperlipidemic
effect
of
Andrographis
paniculata
(Burm.
f.)
Nees
and
andrographolide
in
high-fructose-fat-fed
rats.
Indian
J
Pharmacol
2012;44(May
(3)):377.
[41]
Jelastin
KS,
Tresina
PS,
Mohan
VR.
Antioxidant,
antihyperlipidaemic
and
antidiabetic
activity
of
Eugenia
occosa
Bedd
leaves
in
alloxan
induced
diabetic
rats.
J
Basic
Clin
Pharm
2011;3(December
(1)):235.
[42]
Satheesh
MA,
Pari
L.
Effect
of
red
hogweed
(Boerhavia
diffusa
L.)
on
plasma
antioxidants
in
alloxan-induced
diabetes.
J
Herbs
Spices
Med
Plants
2004;10(March
(4)):1139.
[43]
Wainstein
J,
Ganz
T,
Boaz
M,
Bar
Dayan
Y,
Dolev
E,
Kerem
Z,
et
al.
Olive
leaf
extract
as
a
hypoglycemic
agent
in
both
human
diabetic
subjects
and
in
rats.
J
Med
Food
2012;15
(July
(7)):60510.
[44]
Moon
HI.
Antidiabetic
effects
of
dendropanoxide
from
leaves
of
Dendropanax
morbifera
Leveille
in
normal
and
streptozotocin-induced
diabetic
rats.
Hum
Exp
Toxicol
2011;30(August
(8)):8705.
[45]
Chung
IM,
Kim
EH,
Yeo
MA,
Kim
SJ,
Seo
MC,
Moon
HI.
Antidiabetic
effects
of
three
Korean
sorghum
phenolic
extracts
in
normal
and
streptozotocin-induced
diabetic
rats.
Food
Res
Int
2011;44(January
(1)):12732.
[46]
Ahangarpour
A,
Mohammadian
M,
Dianat
M.
Antidiabetic
effect
of
hydroalcholic
Urtica
dioica
leaf
extract
in
male
rats
with
fructose-induced
insulin
resistance.
Iran
J
Med
Sci
2012;37(September
(3)):181.
[47]
Prince
PS,
Kamalakkannan
N,
Menon
VP.
Antidiabetic
and
antihyperlipidaemic
effect
of
alcoholic
Syzigium
cumini
seeds
in
alloxan
induced
diabetic
albino
rats.
J
Ethnopharmacol
2004;91(April
(2)):20913.
[48]
Abedinzade
M,
Nikokar
I,
Nasri
S,
Nursabaghi
F.
Effect
of
hexanic
and
alcoholic
extracts
of
fenugreek
seed
in
male
diabetic
rats.
Zahedan
J
Res
Med
Sci
2013;15(June
(6)):503.
[49]
Kook
SJ,
Kim
GH,
Choi
KH.
The
effects
of
Panax
Ginseng
on
streptozotocin-induced
diabetic
rats:
meta
analysis.
Korean
J
Appl
Stat
2009;22(1):10714.
[50]
Kumar
GP,
Arulselvan
P,
Kumar
DS,
Subramanian
SP.
Anti-
diabetic
activity
of
fruits
of
Terminalia
chebula
on
streptozotocin
induced
diabetic
rats.
J
Health
Sci
2006;52
(3):28391.
[51]
Shajeela
PS,
Kalpanadevi
V,
Mohan
VR.
Potential
antidiabetic,
hypolipidaemic
and
antioxidant
effects
of
Nymphaea
pubescens
extract
in
alloxan
induced
diabetic
rats.
J
Appl
Pharm
Sci
2012;838.
[52]
Sowmia
C,
Kokilavani
R.
Antidiabetic
and
antihypercholesterolemic
effect
of
Hemidesmus
indicus
Linn.
R.
root
in
Alloxan
induced
diabetic
rats.
Ancient
Sci
Life
2007;26(April
(4)):4.
[53]
Ighodaro
OM,
Omole
JO,
Adejuwon
AO,
Odunaiya
AA.
Effects
of
Parinari
polyandra
seed
extract
on
blood
glucose
level
and
biochemical
indices
in
Wistar
Rats.
Int
J
Diabetes
Res
2012;1(4):6872.
[54]
Suarsana
N,
Utama
IH,
Made
Kardena
I.
Immunohistochemical
expression
of
insulin
and
glucagon.
Superoxide
dismutase
and
catalase
activity
in
pancreas
in
hyperglycaemia
condition.
Asian
J
Biochem
2016;11:17785.
[55]
Petchi
RR,
Vijaya
C,
Parasuraman
S.
Antidiabetic
activity
of
polyherbal
formulation
in
streptozotocinnicotinamide
induced
diabetic
Wistar
rats.
J
Trad
Complement
Med
2014;4(June
(2)):10817.
[56]
Alkaladi
A,
Abdelazim
AM,
Afifi
M.
Antidiabetic
activity
of
zinc
oxide
and
silver
nanoparticles
on
streptozotocin-
induced
diabetic
rats.
Int
J
Molec
Sci
2014;15(January
(2)):201523.
[57]
Olatunji
LA,
Okwusidi
JI,
Soladoye
AO.
Antidiabetic
effect
of
Anacardium
occidentale.
Stem-bark
in
fructose-diabetic
rats.
Pharm
Biol
2005;43(January
(7)):58993.
[58]
Daud
N,
Rosidah
M,
Nasution
P.
Antidiabetic
activity
of
Ipomoea
batatas
L.
leaves
extract
in
streptozotocin-induced
diabetic
mice.
Int
J
PharmTech
Res
2016;9(3):16770.
[59]
Cao
J,
Li
C,
Zhang
P,
Cao
X,
Huang
T,
Bai
Y,
et
al.
Antidiabetic
effect
of
burdock
(Arctium
lappa
L.)
root
ethanolic
extract
on
streptozotocin-induced
diabetic
rats.
Afr
J
Biotechnol
2012;11(May
(37)):9079.
[60]
Saravanan
G,
Ponmurugan
P,
Kumar
GS,
Rajarajan
T.
Antidiabetic
effect
of
S-allylcysteine:
effect
on
plasma
and
tissue
glycoproteins
in
experimental
diabetes.
Phytomedicine
2010;17(December
(14)):10869.
[61]
Hakkim
FL,
Girija
S,
Kumar
RS,
Jalaludeen
MD.
Effect
of
aqueous
and
ethanol
extracts
of
Cassia
auriculata
L.
owers
on
diabetes
using
alloxan
induced
diabetic
rats.
Int
J
Diabetes
Metab
2007;January
(15):1006.
[62]
Lee
SO,
Chung
SK,
Lee
IS.
The
antidiabetic
effect
of
dietary
persimmon
(Diospyros
kaki
L.
cv.
Sangjudu
ngsi)
peel
in
streptozotocin-induced
diabetic
rats.
J
Food
Sci
2006;71
(April
(3)).
[63]
Poudyal
H,
Campbell
F,
Brown
L.
Olive
leaf
extract
attenuates
cardiac,
hepatic,
and
metabolic
changes
in
high
carbohydrate-,
high
fat-fed
rats.
J
Nutr
2010;140(May
(5)):94653.
[64]
Dzeuet
PD,
Ohandja
DY,
Tédong
L,
Asongalem
EA,
Dimo
T,
Sokeng
SD,
et
al.
Antidiabetic
effect
of
ceiba
pentandra
m
e
d
i
c
i
n
a
5
3
(
2
0
1
7
)
3
6
5
3
7
4372
extract
on
streptozo-tocin-induced
non-insulin-
dependent
diabetic
(NIDDM)
rats.
Afr
J
Trad
Complement
Altern
Med
2007;4(1):4754.
[65]
Ananthan
R,
Baskar
C,
NarmathaBai
V,
Pari
L,
Latha
M,
Ramkumar
KM.
Antidiabetic
effect
of
Gymnema
montanum
leaves:
effect
on
lipid
peroxidation
induced
oxidative
stress
in
experimental
diabetes.
Pharmacol
Res
2003;48(December
(6)):5516.
[66]
Miura
T,
Koike
T,
Ishida
T.
Antidiabetic
activity
of
green
tea
(Thea
sinensis
L.)
in
genetically
Type
2
diabetic
mice.
J
Health
Sci
2005;51(6):70810.
[67]
Oliveira
AC,
Endringer
DC,
Amorim
LA,
Maria
das
Graças
LB,
Coelho
MM.
Effect
of
the
extracts
and
fractions
of
Baccharis
trimera
and
Syzygium
cumini
on
glycaemia
of
diabetic
and
non-diabetic
mice.
J
Ethnopharmacol
2005;102(3):4659.
[68]
Singh
SK,
Kesari
AN,
Gupta
RK,
Jaiswal
D,
Watal
G.
Assessment
of
antidiabetic
potential
of
Cynodon
dactylon
extract
in
streptozotocin
diabetic
rats.
J
Ethnopharmacol
2007;114(November
(2)):1749.
[69]
Bnouham
M,
Merhfour
FZ,
Ziyyat
A,
Aziz
M,
Legssyer
A,
Mekh
H.
Antidiabetic
effect
of
some
medicinal
plants
of
Oriental
Morocco
in
neonatal
non-insulin-dependent
diabetes
mellitus
rats.
Hum
Exp
Toxicol
2010;29(October
(10)):86571.
[70]
Surana
SJ,
Gokhale
SB,
Jadhav
RB,
Sawant
RL,
Wadekar
JB.
Antihyperglycemic
activity
of
various
fractions
of
Cassia
auriculata
Linn.
in
alloxan
diabetic
rats.
Indian
J
Pharm
Sci
2008;70(March
(2)):227.
[71]
Anwar
MM,
Meki
AR.
Oxidative
stress
in
streptozotocin-
induced
diabetic
rats:
effects
of
garlic
oil
and
melatonin.
Compar
Biochem
Physiol
A:
Molec
Integr
Physiol
2003;135
(August
(4)):53947.
[72]
Ju
C,
Yue
W,
Yang
Z,
Zhang
Q,
Yang
X,
Liu
Z,
et
al.
Antidiabetic
effect
and
mechanism
of
chitooligosaccharides.
Biol
Pharm
Bull
2010;33(September
(9)):15116.
[73]
hi
GR,
Ignacimuthu
S,
Paulraj
MG,
Sasikumar
P.
Antihyperglycemic
activity
and
antidiabetic
effect
of
methyl
caffeate
isolated
from
Solanum
torvum
Swartz.
fruit
in
streptozotocin
induced
diabetic
rats.
Eur
J
Pharmacol
2011;670(November
(2)):62331.
[74]
Shirwaikar
A,
Rajendran
K,
Barik
R.
Effect
of
aqueous
bark
extract
of
Garuga
pinnata
Roxb.
in
streptozotocin-
nicotinamide
induced
type-II
diabetes
mellitus.
J
Ethnopharmacol
2006;107(September
(2)):28590.
[75]
Chen
F,
Xiong
H,
Wang
J,
Ding
X,
Shu
G,
Mei
Z.
Antidiabetic
effect
of
total
avonoids
from
Sanguis
draxonis
in
type
2
diabetic
rats.
J
Ethnopharmacol
2013;149(October
(3)):
72936.
[76]
Djomeni
PD,
Tédong
L,
Asongalem
EA,
Dimo
T,
Sokeng
SD,
Kamtchouing
P.
Hypoglycaemic
and
antidiabetic
effect
of
root
extracts
of
Ceiba
pentandra
in
normal
and
diabetic
rats.
Afr
J
Trad
Complement
Altern
Med
(AJTCAM)
2006;3
(January
(1)):12936.
[77]
Veerapur
VP,
Prabhakar
KR,
Thippeswamy
BS,
Bansal
P,
Srinivasan
KK,
Unnikrishnan
MK.
Antidiabetic
effect
of
Ficus
racemosa
Linn.
stem
bark
in
high-fat
diet
and
low-
dose
streptozotocin-induced
type
2
diabetic
rats:
A
mechanistic
study.
Food
Chem
2012;132(May
(1)):18693.
[78]
Dunn
JS,
McLetchie
NG.
Experimental
alloxan
diabetes
in
the
rat.
Lancet
1943;242(September
(6265)):3847.
[79]
Gomori
G,
Goldner
MG.
Acute
nature
of
alloxan
damage.
Proc
Soc
Exp
Biol
Med
1945;58(March
(3)):2323.
[80]
Goldner
MG,
Gomori
G.
Studies
on
the
mechanism
of
alloxan
diabetes.
Endocrinology
1944;35(October
(4)):2418.
[81]
Cruz
AB,
Amatuzio
DS,
Grande
F,
Hay
LJ.
Effect
of
intra-
arterial
insulin
on
tissue
cholesterol
and
fatty
acids
in
alloxan-diabetic
dogs.
Circ
Res
1961;9(January
(1)):3943.
[83]
Wöhler
F,
Liebig
J.
Untersuchungen
über
die
Natur
der
Harnsäure.
Eur
J
Organ
Chem
1838;26(January
(3)):241336.
[84]
Jorns
A,
Munday
R,
Tiedge
M,
Lenzen
S.
Comparative
toxicity
of
alloxan,
N-alkylalloxans
and
ninhydrin
to
isolated
pancreatic
islets
in
vitro.
J
Endocrinol
1997;155
(November
(2)):28393.
[85]
Szkudelski
T.
The
mechanism
of
alloxan
and
streptozotocin
action
in
B
cells
of
the
rat
pancreas.
Physiol
Res
2001;50(6):53746.
[86]
Weaver
DC,
Barry
CD,
Mcdaniel
ML,
Marshall
GR,
Lacy
PE.
Molecular
requirements
for
recognition
at
a
glucoreceptor
for
insulin
release.
Molec
Pharmacol
1979;16(2):3618.
[87]
Gorus
FK,
Malaisse
WJ,
Pipeleers
DG.
Selective
uptake
of
alloxan
by
pancreatic
B-cells.
Biochem
J
1982;208(2):5135.
[88]
Malaisse
WJ,
Doherty
M,
Ladriere
LA,
Malaisse-Lagae
FR.
Pancreatic
uptake
of
[2-14C]
alloxan.
Int
J
Molec
Med
2001;7
(3):3115.
[89]
Elsner
M,
Hashimoto
H,
Nilsson
T.
Cisternal
maturation
and
vesicle
transport:
join
the
band
wagon!
Molec
Membr
Biol
2003;20(3):2219.
[90]
Elsner
M,
Tiedge
M,
Guldbakke
B,
Munday
R,
Lenzen
S.
Importance
of
the
GLUT2
glucose
transporter
for
pancreatic
beta
cell
toxicity
of
alloxan.
Diabetologia
2002;45(11):15429.
[91]
Brückmann
G,
Wertheimer
E.
Alloxan
studies:
the
action
of
alloxan
homologues
and
related
compounds.
J
Biol
Chem
1947;168(April
(1)):24156.
[92]
Lenzen
S,
Mirzaie-Petri
M.
Inhibition
of
aconitase
by
alloxan
and
the
differential
modes
of
protection
of
glucose,
3-O-methylglucose,
and
mannoheptulose.
Naunyn-Schmiedeberg's
Arch
Pharmacol
1992;346
(November
(5)):5326.
[93]
Garland
PB,
Randle
PJ,
Newsholme
EA.
Citrate
as
an
intermediary
in
the
inhibition
of
phosphofructokinase
in
rat
heart
muscle
by
fatty
acids,
ketone
bodies,
pyruvate,
diabetes
and
starvation.
Nature
1963;200(October
(4902)):16970.
[94]
Lenzen
S,
Freytag
S,
Panten
U,
Flatt
PR,
Bailey
CJ.
Alloxan
and
ninhydrin
inhibition
of
hexokinase
from
pancreatic
islets
and
tumoural
insulin-secreting
cells.
Basic
Clin
Pharmacol
Toxicol
1990;66(March
(3)):15762.
[95]
Colca
JR,
Brooks
CL,
Landt
M,
McDANIEL
ML.
Correlation
of
Ca
2+
-and
calmodulin-dependent
protein
kinase
activity
with
secretion
of
insulin
from
islets
of
Langerhans.
Biochem
J
1983;212(June
(3)):81927.
[96]
Tiedge
M,
Elsner
M,
McClenaghan
NH,
Hedrich
HJ,
Grube
D,
Klempnauer
J,
et
al.
Engineering
of
a
glucose-responsive
surrogate
cell
for
insulin
replacement
therapy
of
experimental
insulin-dependent
diabetes.
Hum
Gene
Therapy
2000;11(3):40314.
[97]
Borg
LH,
Eide
SJ,
Andersson
A,
Hellerström
C.
Effects
in
vitro
of
alloxan
on
the
glucose
metabolism
of
mouse
pancreatic
B-cells.
Biochem
J
1979;182(3):797802.
[98]
Lenzen
S,
Panten
U.
Signal
recognition
by
pancreatic
B-
cells.
Biochem
Pharmacol
1988;37(3):3718.
[99]
Niki
A,
Niki
H,
Miwa
I,
Lin
BJ.
Interaction
of
alloxan
and
anomers
of
D-glucose
on
glucose-induced
insulin
secretion
and
biosynthesis
in
vitro.
Diabetes
1976;25(7):5749.
[100]
Munday
R.
Dialuric
acid
autoxidation:
effects
of
transition
metals
on
the
reaction
rate
and
on
the
generation
of
‘‘active
oxygen’’
species.
Biochem
Pharmacol
1988;37(3):40913.
[101]
Burkart
V,
Koike
T,
Brenner
HH,
Imai
Y,
Kolb
H.
Dihydrolipoic
acid
protects
pancreatic
islet
cells
from
inammatory
attack.
Inamm
Res
1993;38:605.
[102]
Brownlee
M.
The
pathobiology
of
diabetic
complications:
a
unifying
mechanism.
Diabetes
2005;54(6):161525.
[103]
Elsner
M,
Gurgul-Convey
E,
Lenzen
S.
Relative
importance
of
cellular
uptake
and
reactive
oxygen
species
for
the
m
e
d
i
c
i
n
a
5
3
(
2
0
1
7
)
3
6
5
3
7
4
373
toxicity
of
alloxan
and
dialuric
acid
to
insulin-producing
cells.
Free
Radic
Biol
Med
2006;41(September
(5)):82534.
[104]
Winterbourn
CC,
Munday
R.
Glutathione-mediated
redox
cycling
of
alloxan:
mechanisms
of
superoxide
dismutase
inhibition
and
of
metal-catalyzed
OH
formation.
Biochem
Pharmacol
1989;38(2):2717.
[105]
Lazarow
A.
Protective
effect
of
glutathione
and
cysteine
against
alloxan
diabetes
in
the
rat.
Proc
Soc
Exp
Biol
Med
1946;61(4):4417.
[106]
Lazarow
A,
Patterson
JW,
Levey
S.
The
mechanism
of
cysteine
and
glutathione
protection
against
alloxan
diabetes.
Science
1948;108(2803):3089.
[107]
Sen
PB,
Bhattacharya
G.
Protection
against
alloxan
diabetes
by
glucose.
Indian
J
Physiol
1952;6:1124.
[108]
Misra
M,
Aiman
U,
Alloxan:.
An
unpredictable
drug
for
diabetes
induction?
Indian
J
Pharmacol
2012;44(4):538.
[109]
Kliber
J,
Schindler
I.
Recrystallization/precipitation
behaviour
in
microalloyed
steels.
J
Mater
Process
Technol
1996;60(14):597602.
[110]
Bailey
CC.
Alloxan
diabetes.
Vitam
Horm
1949;7:36582.
[111]
Mythili
M,
Vyas
R,
Akila
G,
Gunasekaran
S.
Effect
of
streptozotocin
on
the
ultrastructure
of
rat
pancreatic
islets.
Microsc
Res
Tech
2004;63(5):27481.
[112]
Park
BH,
Rho
HW,
Park
JW,
Cho
CG,
Kim
JS,
Chung
HT,
et
al.
Protective
mechanism
of
glucose
against
alloxan-
induced
pancreatic
b-cell
damage.
Biochem
Biophys
Res
Commun
1995;210(1):16.
[113]
Tasaka
Y,
Inoue
Y,
Matsumoto
H,
Hirata
Y.
Changes
in
plasma
glucagon,
pancreatic
polypeptide
and
insulin
during
development
of
alloxan
diabetes
mellitus
in
dog.
Endocrinol
Japon
1988;35(3):399404.
[114]
Tripathi
V,
Verma
J.
Different
models
used
to
induce
diabetes:
a
comprehensive
review.
Int
J
Pharm
Sci
2014;6
(6):2932.
[115]
Elsner
M,
Gurgul-Convey
E,
Lenzen
S.
Relation
between
triketone
structure,
generation
of
reactive
oxygen
species,
and
selective
toxicity
of
the
diabetogenic
agent
alloxan.
Antioxid
Redox
Signal
2008;10(4):691700.
[116]
Bansal
R,
Ahmad
N,
Kidwai
JR.
Alloxan-glucose
interaction:
effect
on
incorporation
of
14
C-leucine
into
pancreatic
islets
of
rat.
Acta
Diabetol
1980;17(2):
13543.
[117]
Bhattacharya
G.
On
the
protection
against
alloxan
diabetes
by
hexoses.
Science
1954;120(3125):8413.
[118]
Scheynius
A,
Täljedal
IB.
On
the
mechanism
of
glucose
protection
against
alloxan
toxicity.
Diabetologia
1971;7
(4):2525.
[119]
Boquist
L.
A
new
hypothesis
for
alloxan
diabetes.
APMIS
1980;88(16):2019.
[120]
Jain
D,
Arya
R.
Anomalies
in
alloxan-induced
diabetic
model:
it
is
better
to
standardize
it
rst.
Indian
J
Pharmacol
2011;43(1):91.
[121]
Lukens
FD.
Alloxan
diabetes.
Physiol
Rev
1948;28(3):
30430.
[122]
Rossini
AA,
Like
AA,
Chick
WL,
Appel
MC,
Cahill
GF.
Studies
of
streptozotocin-induced
insulitis
and
diabetes.
Proc
Natl
Acad
Sci
U
S
A
1977;74(6):24859.
[123]
Lenzen
S,
Munday
R.
Thiol-group
reactivity,
hydrophilicity
and
stability
of
alloxan,
its
reduction
products
and
its
N-
methyl
derivatives
and
a
comparison
with
ninhydrin.
Biochem
Pharmacol
1991;42(7):138591.
[124]
Katsumata
K,
Katsumata
Y,
Ozawa
T.
Potentiating
effects
of
combined
usage
of
three
sulfonylurea
drugs
on
the
occurrence
of
alloxan
diabetes
in
rats.
Hormone
Metab
Res
1993;25(02):1256.
[125]
Rerup
CC.
Drugs
producing
diabetes
through
damage
of
the
insulin
secreting
cells.
Pharmacol
Rev
1970;22(4):
485518.
m
e
d
i
c
i
n
a
5
3
(
2
0
1
7
)
3
6
5
3
7
4374

Supplementary resource (1)

... Alloxan can be administrated through different routes (intraperitoneal, intravenous, and subcutaneous), but intraperitoneal administration is the most employed. Alloxan dosage ranges from 90 to 200 mg/kg of body weight, but the most effective is between 170 to 200 mg/kg of body weight [93]. ...
... The most common is the instability and auto-reversibility of hyperglycemia, which lasts less than a month. This period is inadequate for proper evaluation to test antidiabetic compounds, leading to a false conclusion on the antidiabetic relevance of the test compound [93]. Another limitation is that alloxan might not induce human T2DM; it has been noted to stimulate type 1. ...
... In addition, alloxan is very unstable, with a half-life of about 1.5 min. Therefore, it is recommended that the solution be prepared freshly [93]. ...
Article
Background: Plants are used in medicine because they are low-cost, widely available, and have few side effects (compared to pharmacological treatment). Plants have phytocompounds with antidiabetic properties that can be delivered using nanoparticles (NPs). Objective: To describe the antidiabetic properties of green synthesized NPs (GSNPs) and their characterization methods. Methods: Three databases were searched using the terms “type 2 diabetes mellitus,” “antidiabetic effects,” “phytochemicals,” “plants,” and “nanoparticles.” Studies describing the antidiabetic effects (in vitro or animal models) of NPs synthesized by plant extracts and characterizing them through UV-Vis spectroscopy, FTIR, XRD, SEM, TEM, and DLS were included. Results: 16 studies were included. In vitro studies reported enzyme inhibition values between 11% (H. polyrhizus) and 100% (A. concinna) for alfa-amylase and between 41.1% (M. zapota) and 100% (A. concinna) for alfa-glucosidase. Animal studies with Wistar Albino rats having diabetes (induced by alloxan or streptozotocin) reported improved blood glucose, triglycerides, total cholesterol, LDL, and HDL after treatment with GSNPs. Regarding characterization, NP sizes were measured with DLS (25-181.5 nm), SEM (52.1-91 nm), and TEM (8.7-40.6 nm). The surface charge was analyzed with zeta potential (-30.7 to -2.9 mV). UV-Vis spectroscopy was employed to confirm the formations of AgNPs (360-460 nm), AuNPs (524-540 nm), and ZnONPs (300-400 nm), and FTIR was used to identify plant extract functional groups. Conclusions: GSNP characterization (shape, size, zeta potential, and others) is essential to know the viability and stability, which are important to achieve health benefits for biomedical applications. Studies reported good enzyme inhibition percentages in in vitro studies, decreasing blood glucose levels and improving lipid profiles in animal models with diabetes. However, these studies had limitations in the methodology and potential risk of bias, so results need careful interpretation.
... Among the various methods for inducing diabetes in animal models, the administration of alloxan, a cytotoxic agent, is widely used because of its ability to selectively destroy pancreatic beta cells and induce hyperglycemia (Ighodaro et al., 2017). Alloxan-induced diabetic models have been instrumental in studying the metabolic and vascular complications associated with diabetes, but their application in shoulder periarthritis research has been limited due to the lack of a combined model that addresses both conditions (Longkumer et al., 2021). ...
... Over time, the demand for insulin exceeds the supply, leading to persistent hyperglycemia. Experimental studies have shown that after the injection of alloxan, there are three stages: the rst stage is the hyperglycemic period, which usually lasts 2-4 hours; the second stage is the hypoglycemic period, during which blood sugar begins to decrease and lasts approximately 12-16 hours, a dangerous period during which severe cases may die from hypoglycemia; and the third stage begins the morning after the second day of medication, during which blood sugar levels gradually increase and maintain a state of sustained hyperglycemia (Ighodaro et al., 2017). Therefore, appropriate prevention is needed during the hypoglycemic period, with attention given to the mental changes and reactions of the rabbits, as well as random blood sugar measurements for observation and timely intervention during the hyperglycemic period to avoid excessively high blood sugar levels, which could lead to diabetic ketoacidosis. ...
Preprint
Full-text available
Background Diabetic shoulder periarthritis is a prevalent form of shoulder periarthritis that causes significant discomfort to patients. However, the pathogenesis and treatment of this condition remain unresolved, highlighting the need for a stable and effective experimental animal model. The use of animal models is the primary experimental method for studying the pathogenesis and treatment of human shoulder periarthritis. The absence of an animal model for diabetic shoulder periarthritis is a major obstacle to the advancement of related research. Methods This study aimed to develop a rapid, simple, and naturally pathologically consistent model of diabetic shoulder periarthritis using a combination of alloxan and continuous strain and ice compression methods. Postinduction, pathological specimens were collected from the long head of the biceps tendon, shoulder joint synovium, and pancreas for macroscopic, histological, immunohistochemical, and biochemical assessments, as well as radiological evaluation through MRI of the shoulder joint. Results MRI revealed that the diabetic shoulder periarthritis model group exhibited more pronounced joint effusion and tendon structural disorders at various time points than did the control group. An increase in signal intensity within the joint cavity was observed at 14 days postinduction compared to 7 days, indicating an increase in effusion. The tendon fibers in the model group were disorganized, the synovial tissue structure was dense, with significant vascular proliferation and synovial cell hyperplasia, and the degree of fibrosis increased over time. Pancreatic islet observation revealed a significant reduction in islet number and sparse islet cells in the diabetic shoulder periarthritis model group compared with those in the control group. These results indicate that the diabetic shoulder periarthritis model group exhibited more severe pathological changes in structure and function. Conclusion The combination of alloxan and continuous strain and ice compression can be used to successfully and rapidly and easily induce a rabbit model of diabetic shoulder periarthritis. This study provides further options for the establishment of an animal model for diabetic shoulder periarthritis.
... This study demonstrates that lemon extract can help lower blood sugar levels in mice due to its content, such as ascorbic acid (vitamin C) and flavonoid. Vitamin C, structurally similar to glucose, acts as a glucose competitor for cellular entry, thereby reducing protein glycosylation within the body [16]. This action can decrease glycated hemoglobin levels, lower high blood sugar, and reduce oxidative stress produced by this process. ...
Article
Full-text available
Background: High blood sugar levels that exceed normal limits or commonly refered to as hyperglycemia, is an early symptom of diabetes mellitus. Objectives: This study explored the effect of lemon (Citrus limon) extract on blood sugar levels and pancreatic b cell regeneration in alloxan-induced hyperglycemic mice (Mus musculus). Method: This research is an experimental study using a post-test group. The sampling was carried out using a randomization method. By administering 125 mg/kg BW of alloxan, the sample was conditioned for hyperglycemia. The samples were divided into five groups: normal control, alloxan-induced control, and three treatment groups that received lemon extract with dosages of 100 mg/kg BW, 300 mg/kg BW and 500 mg/kg BW, respectively. Semiquantitative analysis was used to evaluate pancreatic damage. Results: The results showed that lemon extract can decrease blood sugar levels. Histopathological imaging revealed a significant improvement in b cell distribution and decreased vacuolization in the Langerhans islets of mice administered lemon extract. No significant differences were observed among different dosages of lemon extract (p>0.05). Conclusion: Our findings underscore the potential therapeutic benefits of lemon extract in managing blood sugar levels and promoting pancreatic b cell regeneration in alloxan-induced mice.
... [26,27] Alloxan monohydrate is one of the chemical agents used to induce diabetes by partial destruction of the -cells of islets of langerhan of the pancrease resulting in decreased insulin secretion and hyperglycaemia. [17,28] In the present study, the effect of Abelmoschus esculentus on blood glucose was investigated. The result showed that the extract of Abelmoschus esculentus showed time dependent reduction of alloxan monohydrate-induced elevation in blood glucose levels. ...
... Part 1). The animals in groups 2-5 remained without food for 16 h, upon which we induced diabetes mellitus with a single intraperitoneal injection of alloxan solution at a dose of 150 mg/kg per 1 mL in line with experimentally selected conditions [26]. ...
Article
Diseases associated with metabolic disorders seem to affect more and more people worldwide. Biologically active supplements may prevent or relieve metabolic disorders. Quercetin is known for its potential to inhibit metabolic syndrome. This paper introduces an in vivo experiment on rodents. It featured hypoglycemic, hypocholesterolemic, and hepatotoxic properties of quercetin. Quercetin was obtained from the hairy root extract of Hedysarum neglectum Ledeb. Two doses (50 and 100 mg/kg) were used to evaluate its hypoglycemic potential. Rats with induced diabetes were tested for body weight, glucose, and cholesterol while mice with induced hypercholesterolemia were checked for blood cholesterol changes. Potential biochemical and pathological changes in the liver were also studied on rats. Quercetin treatment caused neither significant health problems nor death in the model animals. It had no effect on body weight, even in the animals with induced diabetes. In addition, quercetin did not increase glucose and cholesterol in the blood and triggered no pathological changes in the liver. Quercetin isolated from H. neglectum hairy root extract demonstrated no hepatotoxicity. Unfortunately, it showed no beneficial effect on cholesterol and glucose levels and had no efficacy against metabolic syndrome. Further research is needed to assess the effect of quercetin on other metabolic markers, e.g., genes associated with the metabolism of lipids, carbohydrates, etc.
... The study presented findings indicating the potential antihyperglycemic, hypo-lipidemic and antiatherogenic effects of DTSE in alloxan-induced diabetic rats. Alloxan-induced diabetes involves the alteration of pancreatic beta cells responsible for insulin production 12 . In vitro studies have highlighted alloxan's toxicity to these cells, causing necrosis due to reactive oxygen species and elevated calcium levels 13,14 . ...
Article
Full-text available
Copyright©2024; The Author(s): This is an open-access article distributed under the terms of the CC BY-NC 4.0 which permits unrestricted use, distribution, and reproduction in any medium for non-commercial use provided the original author and source are credited _______________________________________________ Cite this article: Alaebo PO, Egbuonu ACC, Achi NK, Obike CA, Ukpabi-Ugo JC, Ezeigwe OC, Chukwu UD, Wisdom CE. In-vivo antidiabetic potentials and toxicological effect of Dennettia tripetala seeds on alloxan-induced diabetic male albino rats. Abstract ____________________________________________________________________________________________________ Background: Hyperglycemia and dyslipidemia are hallmarks of diabetes mellitus. Diabetes mellitus is a complex metabolic disorder characterized by disturbances in carbohydrates, protein, and lipid metabolism due to insufficient insulin production. Aim: Present study was aimed to estimate the effects of Dennettia tripetala seed methanol extract (DTSE) on blood sugar and toxicological effect in alloxan-induced diabetes models using male wistar rats. Methods: A total of thirty (30) male albino rats were divided into six distinct groups. Group A served as normal control group, where rats were neither induced nor treated. Group B acted as the negative control group, where rats were induced but not treated. Group C served as the positive control group, where rats were induced and treated with glibenclamide. Group D consisted of rats that were induced and treated with 100 mg of DTSE. Group E included rats that were induced and treated with 200 mg of DTSE. Lastly, group F comprised rats that were induced and treated with 400 mg of DTSE. Liver and kidney functions were determined using established analytical procedures. Blood samples were collected through ocular puncture for the evaluation of biochemical parameters. Results: Blood glucose level in all the alloxan-induced diabetic rats treated with DTSE showed a relative significant (p<0.05) reduction when compared with the controls. Histological investigation of diabetic rat's liver and kidney indicated degradation of normal tissue architecture, however after the treatment with DTSE minor reparative alteration were seen. Conclusions: The study suggests that DTSE possesses a hypoglycemic effect, hepatoprotective and anti-atherogenic effect but has toxicological effect on the kidney of the rats treated with high dose of DTSE as pathological changes were elicited in the organ of the rats.
... 86 A freshly prepared aqueous alloxan was used owing to the fact that alloxan is unstable with a half-life of about 1.5minutes and thus could easily disintegrate when left to stand in aqueous solutions into nondiabetogenic alloxanic acid due to spontaneous decomposition. [87][88][89] During the course of the experiment, 5% dextrose solution was provided in a drinking bottle so as to counter the initial hypoglycemic effect which accounts for high mortality rate. 90 The result from the studies indicated that intraperitoneal administration of alloxan produced consistent hyperglycemia in experimental animal. ...
Article
Full-text available
Diabetes remains a major public health concern affecting about 2.8% of the global population. Chronic hyperglycaemia of diabetes is frequently associated with long-term damage, dysfunction, and failure of different organs. With multiple risk factors, delayed diagnosis, life-threatening complications, failure of the current therapies, and financial costs, there is a need to look for alternative treatment. Methods: Acute toxicity and anti-diabetic property of the methanolic extract of P.santaliniodes were evaluated on mice and alloxan induced diabetic rats respectively. Twenty alloxan induced diabetic rats were used for both chronic and sub chronic test. For both test methods, two dose levels (250mg/kg, 500mg/kg) were chosen. Water (5ml) and glybenclamide (5mg/kg) were used for negative and positive control respectively. Data obtained were analysed using analysis of varaiance (ANOVA) and T-test. The plant material was subjected to pharmacognostic studies, and it includes physicochemical analysis, phytochemical evaluation, determination of extractive value, histo-chemical analysis and microscopic analysis of the powdered crude drug. Results: The 250mg/kg dose and 500mg/kg dose were statistically significant at p<0.05 at day 1,6,9,12,24 and 6, 9, 12, 24 relative to the placebo for chronic respectively. For sub chronic study, statistical significance was seen only for 500mg/kg on day 10 relative to placebo at p<0.05. Phytochemical analysis of the plant revealed alkaloids, resins, steroids,terpenoids,flavonoids, proteins, carbohydrates, reducing sugars, oils, acidic compounds,cardiac glycosides, tannins and saponins. Physicochemical analysis (total ash {4.7%}, water soluble ash {1%}, acid insoluble ash {12%}, sulphated ash 1.25%}); extractive value (ethanol extractive value {2.2%}, chloroform extractive value {10%}); histochemical analysis (lignified tissue, calcium oxalate, protein, starch, fat and oil and cellulose cell wall) and microscopic analysis of the powdered (branched multicellular non glandular trichomes, elongated unicellular non-glandular trichomes, epidermal cell of the testa,starch globules, annular xylem vessel, peristerm of raphae, large irregularly shaped calcium oxalate and layer of peristerm containing pigment). Conclusions: Finally, P. santaliniodes possess anti-diabetic property which may be linked to the phytoconstituent and thus could serve as lead drug.
... The more sugar transporters in the cells, the more reactive oxygen stress (ROS) increases and pancreatic beta cells become lesions so that the body experiences insulin deficiency and increased hyperglycemia. Researchers have chosen alloxan because of its availability and it is cheaper than streptomyzin (Ighodaro et al., 2018). ...
Chapter
Chemical, Material Sciences & Nano technology book series aims to bring together leading academic scientists, researchers and research scholars to exchange and share their experiences and research results on all aspects of Chemical, Material Sciences & Nano technology. The field of advanced and applied Chemical, Material Sciences & Nano technology has not only helped the development in various fields in Science and Technology but also contributes the improvement of the quality of human life to a great extent. The focus of the book would be on state-of-the-art technologies and advances in Chemical, Material Sciences & Nano technology and to provides a remarkable opportunity for the academic, research and industrial communities to address new challenges and share solutions.
Article
Full-text available
In light of trad itional anti-diabetic claim, therapeutic effect of coconut water extract of Parinari polyandra seeds on fasting blood sugar level and seru m biochemical indices in allo xan-induced diabetic rats was investigated. Ten normoglycemic and forty diabetic rats (randomly assigned to four groups, n=10) (mean weight of 222±13.01g) were used for the study. The normoglycemic rats (group1) served as normal control and a group of diabetic rats (group II) served as diabetic control. Other groups of diabetic rats (Group III, IV and V) were t reated differently with (1) coconut water ext ract of Parinari polyandra seeds (2ml), coconut water alone (2ml) and (3) Glibenclamide, a standard antidiabetic drug (5mg/Kg of body weight). The treat ments were done orally for seven days. Blood glucose level, aspartate aminotransferase (AST) and alanine aminotransferase (ALT) activit ies, total cholesterol, total triglyceride(TG), low density lipoprotein (LDL) and high density lipoprotein (HDL) were examined. The results obtained revealed that rats treated with Parinari polyandra seeds extract expressed significant (p≤0.05) decrease in the levels of glucose, ALT, AST, TA G, total cholesterol, LDL with slight increase in HDL as compared with diabetic control rats. Collectively, the data of the current study indicates that coconut water extract of Parinari polyandra has antidiabetic, anti-hyperlipidemia and anticholesterolemia potentials. Hence, its local use in the management of diabetes should be encouraged.
Article
Full-text available
Objective: To examine the effects of Sapium ellipticum (SE) leaf extract on the hepatic activities of glucokinase and glucose-6-phosphatase in streptozotocin-induced diabetic Wistar rats. Methods: STZ-induced diabetic Wistar rats (four groups, n = 8) were used in this study. SE was assessed at two different doses, 400 and 800 mg/kg BW, in comparison with metformin (METF) (12 mg/kg BW) as a reference antidiabetic drug. All treatments were done orally (p.o), twice daily at 8 h interval for a period of 21 days. Glucokinase and glucose-6-phosphatase activities were respectively determined using standard protocols. Hepatic and muscle glycogen contents were estimated as well. Results: STZ caused significant decrease in glucose-6-phosphatase activity and concomitant increase in glucokinase activity. SE extract especially at 400 mg dosage significantly reversed the alterations by increasing glucokinase activity by 40.31% and inhibiting glucose-6-phosphatase activity by 37.29% compared to diabetic control animals. However, the effects were significantly lower than that of METF which enhanced glucokinase activity by 94.76% and simultaneously inhibited glucose-6-phosphatase activity by 49.15%. The extract also improved hepatic glycogen level by 32.37 and 27.06% at 400 and 800 mg dosage respectively. HPLC-MS analysis of some SE fractions in dynamic MRM mode (using the optimized compound-specific parameters) revealed among other active compounds, the presence of amentoflavone, which has been associated with antidiabetic function. Conclusions: The ability of SE extract to concurrently inhibit glucose-6-phosphatase and activate glucokinase in this study suggests that it may be a treatment option for type 2 diabetes patients, and the presence of amentoflavone in the plant extract may account for its anti-diabetic potential.
Article
Full-text available
Objective: To examine the effects of Sapium ellipticum (S. ellipticum) extract on hepatic activities of glucokinase and glucose-6-phosphatase in streptozotocin (STZ)-induced diabetic Wistar rats. Methods: STZ-induced diabetic Wistar rats (four groups, n = 8) were used in this study. S. ellipticum was assessed at two different doses, 400 and 800 mg/kg BW, in comparison with metformin (12 mg/kg BW) as a standard antidiabetic drug. All treatments were done orally (p.o), twice daily at 8 h interval for a period of 21 days. Glucose-6-phosphatase and glucokinase activities were respectively determined. Hepatic and muscle glycogen contents were estimated as well. Results: STZ caused significant decrease in glucose-6-phosphatase activity and concomitant increase in glucokinase activity. S. ellipticum especially at 400 mg/kg significantly restored glucokinase activity by 40.31% and glucose-6-phosphatase activity by 37.29%. These effects were though significantly lower than that of metformin which enhanced glucokinase activity by 94.76% and simultaneously inhibited glucose-6-phosphatase activity by 49.15%. S. ellipticum also improved hepatic glycogen level by 32.37% and 27.06% at 400 and 800 mg/kg, respectively. HPLC-MS analysis of active S. ellipticum fractions in dynamic MRM mode (using the optimized compound-specific parameters) revealed among other active compounds, the presence of amentoflavone, which has been associated with anti-diabetic function. Conclusions: The ability of S. ellipticum extract to concurrently inhibit glucose-6-phosphatase and activate glucokinase in this study suggests that it may be a treatment option for type 2 diabetes patients and the presence of amentoflavone in the plant extract may account for its anti-diabetic potential.
Article
Background and Objective: Hyperglycemia leads to changes on a cellular level that potentially accelerates to induce cell damage. This study investigated the effect of chronic hyperglycemia to superoxida dismutase and catalase activity and expression of insulin and glucagon by immunohistochemistry in rat pancreas. Methodology: Sprague dawley albino male rats (200-225 g) were divided into 2 groups. Group I was normal control, group II was hyperglycemia control. Observations were carried on the levels of blood glucose, superoxida dismutase (SOD) and catalase activities, malondialdehyde (MDA) levels, β and αcells pancreatic immunohistochemically and histology analysis. Results: The results showed that rats experienced hyperglycemia from the 4th week and suffered hyperglycemia for 1 month with final glucose level was 139.5±5.2 mg dL⁻¹, whereas, the glucose level of the control group was 97.8±4.3 mg dL⁻¹. Hyperglycemia caused pathological changes in pancreatic tissue, namely increased malondialdehyde level at 31.85±5.69 pmol g⁻¹, while MDA levels in control group only at 22.94±3.82 pmol g⁻¹. The SOD antioxidant enzymes and catalase activities on control groups were at 31.37±3.60 and 0.85±0.08 U g⁻¹, respectively, then they decreased to 21.18±2.34 and 0.67±0.03 U g⁻¹ in the hyperglycemia group. Total percentages of β and "-cells in control group were 87.30±6.70 and 48.66±2.64, respectively, then they decreased to 74.54±4.35 and 41.96±2.56 in the hyperglycemia group. In hyperglycemia group, degeneration with mild degree was observed in the pancreatic cells islet of langerhans. Conclusion: Hyperglycemia lead to pathological conditions in rat’s pancreatic tissue with increased levels of malonaldehyde (MDA), decreased SOD and catalase activity, reduced expression of insulin in β-cells and nucleus of β-cell showed picnotics.
Article
Rosa canina L. fruits (Rosaceae) are used to treat diabetes in Anatolia traditionally. In this study, the ethanol extract of R. canina fruits and its fractions were screened for their antioxidant, hypoglycaemic and antidiabetic activities. The ethanol extract that was administered for 7 days possessed a remarkable hypoglycemic effect at 250 mg/kg dose in streptozotocin (STZ) induced diabetic rats. Then it was fractionated through successive solvent extractions to yield CHCl 3 Fr., EtOAc Fr., n -BuOH Fr. and R-H 2 O Fr. respectively. These fractions were administrated to normal plus glucose hyperglycemic rats. Additionally the subacute antidiabetic activities of the fractions were studied in diabetic rats for 7 days. The experimental data indicated that R-H 2 O Fr. Possessed significant antidiabetic activity (50-62%) in diabetic rats. Also, a minor hypoglycemic effect was observed in normoglycemic plus glucose-hyperglycemic animals treated with R-H 2 O Fr. (15%). In vitro antioxidant experiments revealed that EtOAc Fr. Showed the highest radical scavenging activity on DPPH (79.5±0.4%), whereas CHCl 3 Fr. exhibited the maximum reducing power. The highest total phenolic content was observed in CHCl 3 Fr. (18.5±0.6% gallic acid equivalent g/g fraction) but no correlation was observed between the antidiabetic activity of fractions and their phenolic contents. Our findings support the traditional usage of R. canina fruits as a folk remedy in the treatment of diabetes in Turkey.
Article
Diabetes mellitus (DM) is caused by the deficiency of insulin production that functions in the utilization of glucose as the source of energy and fat synthesis so that the lack of insulin hormone will increases the blood glucose level. Traditionally, Ipomoea batatas L. leaves have been used for the treatment of diabetes, cancer, as antioxidant, hyperlipidemic, by natives in different regions and also to cure dengue fever. The objectives of this study were to analyze antidiabetic activity of ethylacetate extract in streptozotocin-induced mice This study consisted of plant material procurement and extract preparation, phytochemical screening, mice blood glucose level examination, and data analysis. Analysis of their antidiabetic activity was started by measuring glucose tolerance to identify the extract of the highest activity at varied dosages (100, 200, and 300 mg/kg bw) of this extract was examined on the streptozotocininduced mice. At the fifteenth day of treatment, all extracts at dosages of 100, 200, and 300 mg/kg bw exerted similar effects to those of metformin, except 0.5% CMC. Antidiabetict effect exerted by EEA of Ipomoea batatas L. 300 mg/kg bw was significantly different from that produced by EAE 100 mg/kg bw (α = 0.05). © 2016, International Journal of PharmTech Research. All right reserved.
Article
Diabetes mellitus is a group of heterogeneous metabolic disorders. A large number of pharmacological agents and animal models are used in study of diabetes for understanding the pathogenesis, complications, genetic and environmental influences. Animal models for type 1 diabetes range from animals with spontaneously developing autoimmune diabetic to chemical ablation of the pancreatic beta cells and Type 2diabetes is studied in both obese and non-obese animal models with varying degrees of insulin resistance and beta cell failure. In recent years, a large number of new genetically modified animals, chemical agents, surgical manipulations, viruses and diabetogenic hormones have been engineered for the study of diabetes.
Article
The present study was designed to investigate the possible antidiabetic, hypolipidaemic and antioxidant effects of ethanol extract of Nymphaea pubescens tuber. Diabetes was induced in Albino rats by administration of alloxan monohydrate (150 mg/kg, body weight i.p). The ethanol extract of Nymphaea pubescens tuber at a dose of 200mg/kg and 500mg/kg body weight were administered at single dose per day to diabetes induced rats for a period of 14 days. The effect of ethanol extract of Nymphaea pubescens tuber extract on blood glucose, plasma insulin, urea creatinine, glycosylated haemoglobin, serum lipid profile (total cholesterol (TC), triglyceride (TG), low density lipoprotein- cholesterol (LDL-C), very low density lipoprotein- cholesterol (VLDL-C), high density lipoprotein- cholesterol (HDLC) and phospholipids (PL)), serum protein, albumin, globulin, serum enzymes (Serum glutamate pyruvate transaminases (SGPT), serum glutamate oxaloacetate transaminases (SGOT) and alkaline phosphate (ALP)), lipoprotein peroxidation (LPO), blood reduced glutathione (GSH), oxidative glutathione (GSSG), GSH/GSSG ratio, erythrocytes glutathione reductase (GR), glutathione peroxidase (GPX) and glutathione S-transferase (GST) were measured in the diabetic rats. The ethanol extracts of Nymphaea pubescens tuber elicited significant (p<0.05) reductions of blood glucose, lipid parameters except HDL-C, serum enzymes and significantly increased HDL-C and antioxidant. The extract also caused significant increase in plasma insulin (p<0.05) in the diabetic rats. In conclusion, ethanol extract of Nymphaea pubescens tuber offers promising antidiabetic and hypolipidaemic effects that may be mainly attributed to its potent antioxidant potential. Further studies will be needed in future in order to determine which one or more of its active constituents have the main antidiabetic and hypolidaemic effects.