ArticlePDF Available

Discovery of adrenomedullin in rat ischemic cortex and evidence for its role in exacerbating focal brain ischemic damage.

Authors:

Abstract and Figures

Focal brain ischemia is the most common event leading to stroke in humans. To understand the molecular mechanisms associated with brain ischemia, we applied the technique of mRNA differential display and isolated a gene that encodes a recently discovered peptide, adrenomedullin (AM), which is a member of the calcitonin gene-related peptide (CGRP) family. Using the rat focal stroke model of middle cerebral artery occlusion (MCAO), we determined that AM mRNA expression was significantly increased in the ischemic cortex up to 17.4-fold at 3 h post-MCAO (P < 0.05) and 21.7-fold at 6 h post-MCAO (P < 0.05) and remained elevated for up to 15 days (9.6-fold increase; P < 0.05). Immunohistochemical studies localized AM to ischemic neuronal processes, and radioligand (125I-labeled CGRP) displacement revealed high-affinity (IC50 = 80.3 nmol) binding of AM to CGRP receptors in brain cortex. The cerebrovascular function of AM was studied using synthetic AM microinjected onto rat pial vessels using a cranial window or applied to canine basilar arteries in vitro. AM, applied abluminally, produced dose-dependent relaxation of preconstricted pial vessels (P < 0.05). Intracerebroventricular (but not systemic) AM administration at a high dose (8 nmol), prior to and after MCAO, increased the degree of focal ischemic injury (P < 0.05). The ischemia-induced expression of both AM mRNA and peptide in ischemic cortical neurons, the demonstration of the direct vasodilating effects of the peptide on cerebral vessels, and the ability of AM to exacerbate ischemic brain damage suggests that AM plays a significant role in focal ischemic brain injury.
Content may be subject to copyright.
Proc.
Natl.
Acad.
Sci.
USA
Vol.
92,
pp.
11480-11484,
December
1995
Neurobiology
Discovery
of
adrenomedullin
in
rat
ischemic
cortex
and
evidence
for
its
role
in
exacerbating
focal
brain
ischemic
damage
XINKANG
WANG,
TIAN-LI
YUE,
FRANK
C.
BARONE,
RAYMOND
F.
WHITE,
ROBERT
K.
CLARK,
ROBERT
N.
WILLET-FE,
ANTONY
C.
SULPIZIO,
NAMBI
V.
AIYAR,
ROBERT
R.
RUFFOLO,
JR.,
AND
GIoRA
Z.
FEUERSTEIN
Department
of
Cardiovascular
Pharmacology,
SmithKline
Beecham
Pharmaceuticals,
King
of
Prussia,
PA
19406
Communicated
by
Stephen
J.
Benkovic,
The
Pennsylvania
State
University,
University
Park
PA,
August
7,
1995
(received
for
review
March
3,
1995)
ABSTRACT
Focal
brain
ischemia
is
the
most
common
event
leading
to
stroke
in
humans.
To
understand
the
molec-
ular
mechanisms
associated
with
brain
ischemia,
we
applied
the
technique
of
mRNA
differential
display
and
isolated
a
gene
that
encodes
a
recently
discovered
peptide,
adrenomedullin
(AM),
which
is
a
member
of
the
calcitonin
gene-related
peptide
(CGRP)
family.
Using
the
rat
focal
stroke
model
of
middle
cerebral
artery
occlusion
(MCAO),
we
determined
that
AM
mRNA
expression
was
significantly
increased
in
the
ischemic
cortex
up
to
17.4-fold
at
3
h
post-MCAO
(P
<
0.05)
and
21.7-fold
at
6
h
post-MCAO
(P
<
0.05)
and
remained
elevated
for
up
to
15
days
(9.6-fold
increase;
P
<
0.05).
Immuno-
histochemical
studies
localized
AM
to
ischemic
neuronal
processes,
and
radioligand
(12II-labeled
CGRP)
displacement
revealed
high-affinity
(IC50
=
80.3
nmol)
binding
of
AM
to
CGRP
receptors
in
brain
cortex.
The
cerebrovascular
function
of
AM
was
studied
using
synthetic
AM
microinjected
onto
rat
pial
vessels
using
a
cranial
window
or
applied
to
canine
basilar
arteries
in
vitro.
AM,
applied
abluminally,
produced
dose-
dependent
relaxation
of
preconstricted
pial
vessels
(P
<
0.05).
Intracerebroventricular
(but
not
systemic)
AM
administra-
tion
at
a
high
dose
(8
nmol),
prior
to
and
after
MCAO,
increased
the
degree
of
focal
ischemic
injury
(P
<
0.05).
The
ischemia-induced
expression
of
both
AM
mRNA
and
peptide
in
ischemic
cortical
neurons,
the
demonstration
of
the
direct
vasodilating
effects
of
the
peptide
on
cerebral
vessels,
and
the
ability
of
AM
to
exacerbate
ischemic
brain
damage
suggests
that
AM
plays
a
significant
role
in
focal
ischemic
brain
injury.
Adrenomedullin
(AM)
is
a
recently
discovered
peptide
that
was
initially
identified
from
human
pheochromocytoma
(1).
Biologically
active
AM
consists
of
52
amino
acids
in
humans
and
50
amino
acids
in
rats
(1,
2),
and
both
AMs
exhibit
potent
vasodilator
activity
in
vitro
and
in
vivo
(3-6).
AM
bears
homology
to
a
family
of
peptides
that
includes
calcitonin
gene-related
peptide
(CGRP)
(7-10)
and
amylin
(11,
12).
CGRP
is
a
widely
distributed
neuropeptide
best
known
for
its
potent
vasodilator
actions
(13-15)
and
its
effect
on
insulin
functions
(16).
Amylin
is
the
major
protein
found
in
islet
amyloid
(11,
16)
in
humans
with
non-insulin-dependent
dia-
betes
mellitus.
CGRP
and
amylin
have
been
found
to
have
a
wide
range
of
biological
activities,
including
energy
metabo-
lism,
central
nervous
system
and
cardiovascular
functions,
and
calcium
metabolism
(for
review
see
refs.
16
and
17).
In
contrast,
little
is
known
of
the
biological
function
of
AM
beyond
vasodilation.
In
contrast
to
CGRP,
AM
mRNA
and
peptide
have
not
been
detected
in
normal
brain
(1, 2).
Very
recently,
AM
has
been
associated
with
congestive
heart
failure,
since
elevated
levels
of
AM
were
found
in
cardiac
tissue
of
the
failing
hearts
(18).
In
the
present
report,
we
used
a
recently
developed
mRNA
differential
display
technique
(19)
to
identify
genes
expressed
The
publication
costs
of
this article
were
defrayed
in
part
by
page
charge
payment.
This
article
must
therefore
be
hereby
marked
"advertisement"
in
accordance
with
18
U.S.C.
§1734
solely
to
indicate
this
fact.
in
response
to
brain
ischemia
induced
by
permanent
occlusion
of
the
middle
cerebral
artery
(MCAO)
in
the
rat.
We
herein
report
the
upregulation
of
AM
mRNA
expression
and
peptide
production
after
focal
cerebral
ischemia.
Furthermore,
we
have
demonstrated
that
AM
is
a
potent
cerebral
vasodilator
and
that
intracerebroventricular
(i.c.v.),
but
not
i.v.,
adminis-
tration
of
AM
exacerbated
focal
ischemic
injury.
Taken
to-
gether,
our
data
suggest
a
significant
role
for
AM
in
the
evolution
of
stroke.
MATERIALS
AND
METHODS
Focal
Brain
Ischemia.
Focal
cerebral
ischemia
was
carried
out
in
spontaneously
hypertensive
rats
(Taconic
Farms)
by
permanent
MCAO
as
described
in
detail
previously
(20,
21).
mRNA
Differential
Display.
Total
cellular
RNA
was
iso-
lated
from
ipsilateral
(ischemic)
cortex
and
contralateral
(nonischemic)
cortex
samples
as
described
(22,
23).
RNA
samples
from
2
h
and
12
h
after
MCAO
were
used
for
differential
display
essentially
as
described
(19,
24)
using
an
RNAmap
kit
(GenHunter).
The
differentially
expressed
bands
of
interest
were
isolated,
reamplified,
and
subcloned
into
a
pCRII
vector
(Invitrogen).
Northern
Blot
Analysis.
Northern
hybridization
was
carried
out
as
described
earlier
(23,
25).
cDNA
Library
Construction,
Screening,
and
DNA
Sequence
Analysis.
A
rat
cerebral
ischemia
cDNA
library
in
A
ZAP
II
vector
(Stratagene)
was
constructed
according
to
manufactur-
er's
specifications
using
poly(A)
RNA
isolated
from
ischemic
cortex
2
and
12
h
after
MCAO.
This
library
was
screened
using
the
PMCAO-9
cDNA
probe
isolated
from
differential
display
as
described
in
detail
previously
(25).
Four
individual
positive
clones
were
isolated,
and
the
corresponding
phagemids
were
produced
by
in
vivo
excision.
The
complete
cDNA
sequence*
of
AM
was
determined
from
both
strands.
DNA
sequence
analysis
and
computer
data
base
searches
were
performed
using
the
Genetics
Computer
Group
program.
Primer
Extension
Analysis.
Primer
extension
was
carried
out
as
described
(24)
in
the
presence
of
a
32P-labeled
primer
complementary
to
bases
46-64
(5'-GATGAGAAGCCGAG-
AAACC-3'),
and
5
,tg
of
poly(A)
RNA
isolated
from
rat
ischemic
cortex
at
12
h
after
MCAO.
Immunohistochemical
Study.
Six-micron-thick
frozen
sec-
tions
were
incubated
with
rabbit
anti-AM-(1-52)
(human)
IgG
(Peninsula
Laboratories)
and
then
with
fluorescein-conjugated
goat
anti-rabbit
IgG
antiserum
(Organon
Teknika-Cappel).
Antisera
were
diluted
in
phosphate-buffered
saline
(PBS)
containing
3%
(wt/vol)
bovine
serum
albumin.
Parallel
sec-
tions
were
incubated
with
either
preabsorbed
antiserum
[i.e.,
preincubating
anti-AM
antiserum
with
human
AM-(1-52)
Abbreviations:
AM,
adrenomedullin;
CGRP,
calcitonin
gene-related
peptide;
MCAO,
middle
cerebral
artery
occlusion;
ET-1,
endothelin
1;
i.c.v.,
intracerebroventricular.
*The
sequence
reported
in
this
paper
has
been
deposited
in
the
GenBank
data
base
(accession
no.
U15419).
11480
Proc.
Natl.
Acad.
Sci.
USA
92
(1995)
11481
(Peninsula
Laboratories)
at
10
,g/ml]
or
secondary
antiserum
only.
Double
labeling
was
performed
on
sections
by
first
treating
for
AM
immunoreactivity
as
above
and
then
applying
monoclonal
antibodies
for
glial
fibrillary
acidic
protein
(Boehringer
Mannheim)
or
neurofilaments
(ICN)
followed
by
rhodamine-conjugated
goat
anti-mouse
IgG
secondary
anti-
serum.
Sections
were
analyzed
using
OPTIMAS
image
analysis
(BioScan,
Edmonds,
WA).
In
Vitro
Canine
Cerebral
Vessels.
Four
adult
mongrel
dogs
were
euthanized
via
an
intravenous
overdose
of
pentobarbital.
The
basilar
artery
with
intact
endothelium
was
removed,
and
segments
were
prepared
for
isometric
tension
recording
(for
details,
see
ref.
26).
An
optimum
resting
tension
of
750
mg
was
applied
to
each
segment. During
the
equilibration
period
(-2
h),
the
segments
included
in
this
study
generated
substantial
spontaneous
tone.
The
concentration-related
effects
of
AM
on
spontaneous tone
were
then
determined.
In
control
experiments,
pretreatment
of
the
tissues
with
the
appropriate
volume
of
the
AM
vehicle
(saline)
had
no
significant
effect
on
the
spontaneous
tone.
The
relaxation
observed
with
AM
was
expressed
as
the
mean
±
SEM
of
the
percentage
of
spontaneous
tone.
Rat
Pial
Arteries.
The
effects
of
AM
on
pial
arteries
in
the
rat
were
investigated
in
situ
with
video
microscopic
techniques
in
a
cranial
window
preparation
with
the
dura
intact
(27).
Three
male
Sprague-Dawley
rats
were
prepared
and
main-
tained
as
described
(28).
Since
in
preliminary
experiments
AM
produced
inconsistent
relaxation,
endothelin
1
(ET-1;
0.2
pmol)
in
saline
(0.5
,ul)
was
microinjected
to
elicit
local
vasospasm;
this
was
followed
by
a
second
dose
of
ET-1
(0.2
pmol)
in
the
control
group
or
ET-1
(0.2
pmol)
plus
AM
(50
pmol)
in
the
AM
group.
All
responses
were
recorded
on
video
tape
and,.analyzed
using
National
Institutes
of
Health
image
analysis
software.
AM
Administration
in
MCAO.
Focal
ischemia
was
produced
in
spontaneously
hypertensive
rats
as
described
earlier.
AM
(i.v.
at
1
,ug/kg
per
min;
n
=
6)
or
vehicle
(PBS
with
1%
bovine
serum
albumin;
n
=
6)
was
administrated
continuously
for
1
h
pre-MCAO
and
4
h
post-MCAO.
i.c.v.
AM
at
doses
of
0
(n
=
13),
0.2
(n
=
9),
2
(n
=
10),
or
8
(n
=
13)
nmol
in
5
,ul
was
administered
into
the
ipsilateral
cerebral
ventricle
at
1
h
pre-MCAO
and
6
h
post-MCAO.
After
24
h
of
MCAO,
forebrain
sections
were
removed,
stained,
and
analyzed
as
described
(20,
21)
for
percent
hemispheric
swelling
and
percent
hemispheric
infarct
(normalized
to
the
contralateral
control
hemisphere)
and
the
actual
infarct
volume
in
cubic
millimeters.
RESULTS
AND
DISCUSSION
Identification
of
Upregulated
PMCAO-9
Gene
Expression
in
Rat
Ischemic
Cortex
Using
mRNA
Differential
Display.
Fig.
1A
illustrates
a
representative
autoradiograph
showing
the
ischemia-induced
expression
of
a
band
designated
as
PM-
CAO-9
at
2
and
12
h
post-MCAO
using
mRNA
differential
display.
The
upregulation
of
this
gene
expressed
in
ischemic
cortex
was
confirmed
using
Northern
blot
analysis
(Fig.
1B).
Thereafter,
PMCAO-9
was
subcloned
into
a
pCRII
vector
and
subjected
to
DNA
sequencing
analysis.
Computer
data
base
searches
of
the
PMCAO-9
sequence
(Fig.
2)
demonstrated
that
the
sequence
represented
an
unreported
cDNA.
Isolation
and
Analysis
of
AM
cDNA.
A
rat
brain
ischemia
cDNA
library
was
constructed
and
screened
using
the
PM-
CAO-9
DNA
as
a
probe.
Four
individual
positive
clones
were
isolated,
which
differed
in
length
at
the
5'
ends
(Fig.
2).
Upon
completion
of
the
full-length
cDNA
sequencing,
we
found
that
the
clone
matched
a
rat
precursor
AM
cDNA
in
the
GenBank
data
base
(accession
no.
D15069).
However,
several
sequence
differences
were
noted:
18
bases
have
been
extended
at
the
5'
end
of
our
clone
compared
to
the
sequence
reported
previ-
ously
(2).
In
addition,
a
single
base
(T)
deletion
after
base
1085
and
a
base
replacement
of
C
for
G
at
base
982
were
observed
A
1
2
3
4
_
PMCAO-9
B
PMCAO-9
1
2 3
4
kb
-
1.5
j~~~~A
WkL_
A-
j
rpL32
-
0.6
FIG.
1.
Identification
of
altered
gene
expression
in
rat
ischemic
cortex
after
MCAO
by
mRNA
different
display.
The
differential
display
PCR
was
carried
out
using
a
5'
arbitrary
primer
(5'-
GACCGCTTGT-3')
and
a
3'
T12NA
primer
and
resolved
by
electro-
phoresis
in
the
following
order:
lane
1,
2
h
ischemic;
lane
2,
2
h
nonischemic;
lane
3,
12
h
ischemic;
lane
4,
12
h
nonischemic
(A).
The
candidate
gene
indicated
with
an
arrowhead
(PMCAO-9)
was
con-
firmed
by
Northern
analysis
(B).
Two
micrograms
of
poly(A)
RNA
per
lane
was
used
with
the
same
loading
order
as
shown
in
A.
The
ribosomal
protein
L32
(rpL32)
probe
was
used
as
a
loading
control
(23).
in
all
of
our
four
clones
compared
to
the
previously
reported
sequence
(2).
Also,
a
three-base
deletion
(CCT)
after
base
1227
was
observed
in
three
of
our
four
clones,
whereas
the
other
clone
has
five
bases
(CCTGT)
deleted
compared
to
the
published
rat
AM
sequence
(2).
The
position
of
this
deleted
sequence
is
tandemly
followed
by
23
GT
repeats
in
all
four
clones.
This
variation
is
likely
to
be
caused
by
in
vivo
recom-
bination
events
as
pointed
out
by
a
recent
study
(29).
Further-
more,
primer
extension
experiments
revealed
that
the
AM
mRNA
contains
a
single
transcription
initiation
site
located
about
47
bases
upstream
of
our
longest
cDNA
clone
(data
not
shown).
Temporal
Expression
of
AM
mRNA
in
Rat
Ischemic
Cortex
After
MCAO.
Fig.
3A
shows
a
representative
Northern
blot
for
the
AM
mRNA
expression
in
the
ipsilateral
(ischemic)
and
contralateral
(nonischemic)
cortical
samples
at
different
time
points
after
MCAO.
Quantitative
Northern
blot
data
(n
=
4),
after
normalizing
to
an
rpL32
probe,
are
illustrated
graphically
in
Fig.
3B.
Very
low
levels
of
AM
mRNA
were
detected
in
normal
(data
not
shown)
or
sham-operated
cortical
samples.
The
AM
mRNA
expression
was
induced
significantly
in
the
ischemic
cortex
3
h
after
MCAO
(17.4-fold
increase
compared
to
sham;
P
<
0.05),
reached
its
peak
expression
at
6
h
(21.7-fold
increase;
P
<
0.05),
and
remained
elevated
for
at
least
15
days
(9.6-fold
increase;
P
<
0.05)
after
MCAO
(Fig.
3).
The
temporal
expression
profile
of
AM
mRNA
is
distinctly
differ-
ent
from
that
of
the
immediate
early
genes,
such
as
c-fos
and
zif268
(30),
which
exhibit
a
more
acute
response
profile
(significant
increase
at
1
h
and
peak
at
3
h),
or
the
delayed
response
genes,
including
inflammatory
cytokines,
such
as
tumor
necrosis
factor
a
and
interleukin
1(3
(31,
32),
which
are
significantly
elevated
at
6
h
and
peak
at
12
h
after
MCAO,
in
the
same
focal
ischemia
model.
The
significantly
prolonged
increase
in
AM
mRNA
after
MCAO
(Fig.
3)
suggests
both
an
early
and
late
involvement
in
ischemic
injury.
As
is
the
case
for
c-fos
and
other
acute
response
genes,
AM
mRNA
contains
two
AUUUA
motifs
in
the
3'-untranslated
region,
which
are
believed
to
be
associated
with
rapid
degradation
of
mRNA
(33,
34),
suggesting
that
prolonged
elevation
of
AM
mRNA
expression
after
MCAO
may
represent
a
de
novo
transcriptional
event.
Neurobiology:
Wang
et
al.
11482
Neurobiology:
Wang
et
al.
Proc.
Natl.
Acad.
Sci.
USA
92
(1995)
1
GACACTAGGC
AGAACAACTC
CAGCCTTTAC
CGCTCCTGGT
TTCTCGGCTT
CTCATCGCAG
TCAGTCTTGG
ACTTTGCGGG
TTTTGCCGCT
91
GTCAGAAGGA
CGTCTCGGAC
TTTCTGCTTC
AAGTGCTTGA
CAACTCACCC
TTTCAGCAGG
GTATCGGAGC
ATCGCTACAG
A
172
ATG
AAG
CTG
GTT
TCC
ATC
GCC
CTG
ATG
TTA
TTG
GGT
TCG
CTC
GCC
GTT
CTC
GGC
GCG
GAC ACC
GCA
CGG
CTC
GAC
M
K
L
V
S
I
A
L
M
L
L
G
S
L
A
V
L
G
A
D
TA
R
T.
D
25
247
ACT
TCC
TCG
CAG
TTC
CGA AAG
AAG
TGG
AAT
AAG
TGG
GCG CTA
AGT
CGT
GGG
AAG
AGG
GAA
CTA
CAA
GCG
TCC
AGC
T
S
S
0
F
R
K
K
W
N
K
W A
L
S
R
G
K R
E L
Q
A
S
S
50
322
AGC
TAC
CCT
ACG
GGG
CTC
GTT
GAT
GAG
AAG
ACA
GTC
CCG
ACC
CAG ACT
CTT
GGG
CTC
CAG GAC
AAG
CAG
AGC
ACG
S
Y
P
T
G
L
V
D
E
K
T
V
P
T
Q
T
L
G
L
Q
D
K
Q
S
T
75
397
TCT
AGC
ACC
CCA CAA
GCC
AGC
ACT
CAG
AGC
ACA
GCC
CAC
ATT
CGA
GTC
AAA
CGC
TAC
CGC
CAG
AGC
ATG
AAC
CAG
S
S
T
P
Q
A
S
T
Q
S
T A
H
I
R
V
K R
Y R
0
S
M
N
0
100
472
GGG
TCC CGC
AGC
ACT
GGA
TGC
CGC
TTT
GGG
ACC
TGC
ACA
ATM
CAG
AAA
CTG
GCT
CAC
CAG
ATC
TAC
CAG
TTT
ACA
G
S
R
S
T
G
C
R
F
G
T
C
T
M
0
K
L
A
H
0
I
Y
0
F
T
125
547
GAC
AAA
GAC AAG
GAC GGC
ATG GCC
CCC
AGA
AAC
AAG
ATC
AGC
CCT
CAA
GGC
TAT
GGC
CGC
CGG
CGC
CGG
CGT
TCC
D
K
D
K
D
G
M
A
P
R
N
K
I
S
P
0
a
Y
G
R
R
R
R
R
S
150
622
CTG
CCA
GAG
GTC
CTC
CGA
GCC
CGG
ACT
GTG
GAG
TCC
TCC
CAG
GAG
CAG
ACA
CAC
TCA
GCT
CCA
GCC
TCC
CCG
GCG
L
P
E
V
L
R
A
R
T
V
E
S
S
Q
E
Q
T
H
S
A
P
A
S
P
A
175
697
CAC
CAA
GAC
ATC
TCC
AGA
GTC
TCT
AGG
TTA
TAG
GTGCGGGTGG
CAGCATTGAA
CAGTCGGGCG
AGTATCCCAT
TGGCGCCTGC
H
Q
D
I
S
R
V
S
R
L
*
186
780
GGAATCAGAG
AGCTTCGCAC
CCTGAGCGGA
CTGAGACAAT
CTTGCAGAGA
TCTGCCTGGC
TGCCCCTAGG
GGAGGCAGAG
GAACCCAAGA
870
TCAAGCCAGG
CTCACGTCAG
AAACCGAGAA
TTACAGGCTG
ATACTCTCTC
CGGGCAGGGG
TCTGAGCCAC
TGCCTTGCCC
GCTCATAAAC
960
TGGTTTTCTC
ACGGGGCATA
CGCCTCATTA
CTTACTTGAA
CTTTCCAAAA
CCTAGCGAGG
AAAAGTGCAA
TGCTTGTTAT
ACAGCCAAAG
1050
GTAACTATCA
T&A&AGTT
TGTTGATGTC
AAGAGGTTTT
TIMMTGTAA
CTTCAAATAT
ATAGAAATAT
T7TTGTACGT
TATATATTGT
1140
ATTAAGGGCA
ITTTTAAAGCG
ATTATATTGT
CACCTTCCCC
TATTTTAAGA
AGTGAATGTC
TCAGCAAGGT
GTAAGGTTGT
TTGGTTCCCC
1230
GTGTGTGTG
GTTTGT
GTGTGTGTGT
GTGTGTGTGT
GITIGTAAGG
TGGAGAGCGC
CTGATTACCG
CCTGTGGATG
AAGAAAAAAC
1320
ATTGTGTCTT
CTATAATCT&_.nCATAAA
ATATGTGATC
TGGGAAAAAG
CAAACCAAIA
AACTGTCTCA
ATGCTG
(A)
n
FIG.
2.
Nucleotide
sequence
of
rat
AM
cDNA.
The
numbers
to
the
left
refer
to
the
nucleotide
positions
and
those
to
the
right
refer
to
the
amino
acid
positions.
The
vertical
arrows
indicate
the
5'
end
of
the
individual
cDNA
clones,
and
the
horizontal
arrows
are the
primers
for
the
differential
display,
where
the
unmatched
bases
are
marked
with
a
dot.
The
position
of
the
two
AUUUA
motifs
in
the
3'-untranslated
region
are
bold-faced
and
underlined,
and
the
position
of
the
polyadenylylation
signal,
AATAAA,
is
bold-faced.
The
TG
repeats
are
underlined.
The
amino
acid
sequences
predicted
to
be
the
proteolytic
cleavage
products
are
underlined.
The
AM
peptide
(amino
acids
93-143)
has
been
shown
to
have
vasodilator
properties
and
was
used
for
functional
analysis
in
Fig.
5
and
Table
1.
Immunostaining
of
AM
in
the
Ischemic
Cortex
After
12
h,
24
h,
and
5
days
after
MCAO.
AM
immunoreactivity
MCAO.
Immunohistochemical
analysis
was
carried
out
using
revealed
the
presence
of
short
fiber
processes
(highly
fluores-
brain
tissues
after
sham
surgery
and
from
animals
(n
=
3)
6
h,
cent
in
a
granular
or
vacuolar
pattern
within
the
ischemic
A
lpsilateral:+
-
+
-
+
-
+ +
-
+
-
+
-
+
-
+
-
Contralateral:
-
+
-
+
-
+
-
+
-
+
-
+
-
+
-
+
-
+
-
+
Time:
S
S
lh
lh
3h
3h
6h
6h
12h
12h
24h 24h
2d
2d
Sd
5d
10d
10d
lSd
15d
kb
AM
.
-1.5
rpL32
*
B
S
20-
*
Ischemic
*
*
0
~~~~~~~Nonischemic
Z
10
*
*
E
Sham
1
h
3h
6h
1
2h
24h
2d
5d
1Od
15d
Time
post-MCAO
FIG.
3.
Time
course
study
of
AM
mRNA
induction
in
rat
ischemic
cortex
after
MCAO.
(A)
Representive
Northern
blot
for
AM
and
rpL32
probes
to
the
samples
isolated
at
various
time
points
and
conditions
from
rats
subjected
to
MCAO.
Total
cellular
RNA
(40
,ug
per
lane)
was
used
for
this
analysis.
Ipsilateral
and
contralateral
cortex
samples
(denoted
by
+)
from
individual
rats
after
sham
surgery
(S;
12
h
sacrifice)
or
1,
3,
6,
12,
and
24
h,
and
2,
5,
10,
and
15
days
(d)
of
MCAO
are
depicted.
(B)
Quantitative
Northern
blot
data
for
AM
mRNA
expression
after
focal
brain
ischemic
injury.
The
data
were
analyzed
using
a
Phosphorlmager
and
are
presented
as
the
mean
values
±
SE
of
four
animals
for
each
time
point.
The
data
were
analyzed
using
one-way
ANOVA
followed
by
Bonferroni-adjusted
post
hoc
t
test.
*,
P
<
0.05
vs
sham
samples.
Proc.
Natl.
Acad.
Sci.
USA
92
(1995)
11483
cortex).
These
fibers
were
rare
and
only
weakly
fluorescent
6
and
12
h
after
MCAO
but
were
intensely
immunoreactive
and
abundant
in
tissues
examined
24
h
and
5
days
after
MCAO
(Fig.
4A
and
C).
No
immunoreactivity
was
observed
in
sham-operated
rats
or
outside
of
the
ischemic
zone
after
MCAO.
When
AM
antiserum
was
preincubated
with
AM
prior
to
incubation
of
tissue
sections,
immunofluorescence
was
completely
eliminated
(Fig.
4,
E
compared
with
C).
Also,
no
immunoreactivity
was
observed
in
sections
incubated
with
the
second
antibody
only
(data
not
shown).
When
double
labeled
with
intermediate
filament
markers
for
neurons
(neurofila-
ments)
or
astroglia
(glial
fibrillary
acidic
protein),
the
immuno-
fluorescent
data
indicated
colocalization
of
AM
with
neuro-
filaments
(Fig.
4
B,
D,
and
F),
which
is
indicative
of
nerve
fiber
processes.
In
contrast,
no
colocalization
was
seen
between
AM
and
glial
fibrillary
acidic
protein
(data
not
shown).
AM
Effects
on
Rat
Pial
Arteries
in
Situ.
The
effects
of
rat
AM
on
rat
pial
microvessels
(43
±
7
,um)
were
determined
using
videomicroscopic
techniques
(Fig.
5A).
The
local
appli-
cation
of
ET-1
(0.2
pmol)
via
subarachnoid
microinjection
rapidly
elicited
a
prolonged
submaximal
constriction
of
adjacent
pial
arteries
(Fig.
5
A
and
B).
There
was
little
or
no
change
in
the
pial
artery
diameter
when
the
microinjection
of
ET-1
was
re-
peated
within
10
min
of
the
first
microinjection.
In
contrast,
when
AM
(50
pmol)
was
added
to
the
second
ET-1
microinjection,
a
significant
vasodilation
of
the
adjacent
pial
artery
was
observed.
AM
(up
to
10
,gmol)
did
not
interfere
with
ETA
receptor
binding
(data
not
shown).
The
percent
of
the
control
diameter
was
44%
±
8%
after
ET-1
alone
compared
to
98%
±
18%
after
ET-1
plus
AM
(Fig.
SB).
There
were
no
changes
in
systemic
arterial
pressure
after
ET-1
or
AM
microinjection.
In
addition,
AM
had
no
effect
on
cortical
perfusion
when
administered
systemically
at
the
same
doses
(data
not
shown).
AM
Effects
on
Cerebral
Vessels
in
Vitro.
The
effects
of
rat
AM
on
segments
of
the
canine
basilar
artery
were
determined
in
vitro.
The
canine
basilar
arteries
used
in
this
study
slowly
generated
spontaneous
tone,
which
reached
a
steady-state
A
CONTROL
ET-1
ET-1
+
AM
SOPm
B
120
100]
CD
-
E
80-
.U
L
a
5
60-
o
40-
20-
0-
I
(n=3)
ET-1
(0.2
pmoles)
ET-1
(0.2
pmoles)
AM
(50
pmoles)
C
CANINE
BASILAR
ARTERY
c
0
x
to
Ox
FIG.
4.
Immunohistochemical
detection
of
AM
expression
in
ischemic
cortex
after
MCAO
in
the
rats.
Matched
computer-captured
images
of
ischemic
rat
cerebral
cortex
immunolabeled
with
antiserum
against
AM
(A,
C,
and
E)
and
neurofilaments
(B,
D,
and
F).
A
and
B
are
from
the
same
matched
field,
as
are
C-F.
Ischemic
cortex
at
24
h
or
5
d
after
MCAO
demonstrated
intense
AM
immunoreactivity
in
a
granular
or
vacuolar
pattern
(A
and
C,
24
h
after
MCAO).
This
immunoflourescence
was
restricted
to
the
ischemic
cortex.
When
the
anti-AM
antiserum
was
preincubated
with
AM,
the
immunoreactivity
was
eliminated
(E
compared
with
C).
Combining
computer
images
of
brain
tissue
immunofluorescence
for
AM
(green
in
A
and
C)
and
neurofilaments
(red
in
F)
allow
demonstration
of
double-labeling
in
neuronal
processes
(yellow
in
double
labeled
B
and
D).
Arrows
indicate
location
of
neuronal
processes.
100-
90-/
80-
70
60
50
40
/
30
20
10-
/
n=4
3-
,
-
10
100
10
00
3
10
100
1000
AM
(nM)
FIG.
5.
The
cerebrovascular
effects
of
AM
were
assessed
in
rat
pial
arteries
in
situ
(A
and
B)
and
canine
basilar
artery
segments
in
vitro
(C).
Microinjection
of
ET-1
(0.2
pmol)
produced
significant
vasoconstric-
tion
of
rat
pial
arteries
(A
Middle
and
B).
AM
(50
pmol)
administration
reversed
ET-1-induced
vasoconstriction
(A
Right
and
B).
(C)
AM
also
produced
a
concentration-dependent
relaxation
of
spontaneous
tone
in
the
canine
basilar
artery.
*P
<
0.02,
compared
to
ET-1
treatment
alone
as
determined
by
Student's
t
test.
Neurobiology:
Wang
et
al.
Proc.
Natl.
Acad.
Sci.
USA
92
(1995)
Table
1.
Effects
of
AM
in
focal
ischemic
injury
i.v.,
ng/kg
per
min
i.c.v.,
nmol
Measure
0
1
0
0.2
2
8
Swelling,
%
1
±
1
2
±
2
2
±
1
0
±
1
0
±
1
5
±
1
Infarct,
%
18
±
2
17
±
2
16
±
1
13
±
1
13
±
1
20
±
2*
Infarct
volume,
mm3
110
±
13
105
±
14
92
±
8
67
±
8
72
±
9
120
±
9*
*P
<
0.05
compared
to
vehicle,
analyzed
by
a
one-way
ANOVA
followed
by
Fisher's
protected
least-significant
difference
post
hoc
t
test.
maximum
(841
±
115
mg)
prior
to
the
addition
of
AM
to
the
tissue
bath.
AM
produced
a
prolonged,
concentration-
dependent
relaxation
of
spontaneous
tone.
The
concentration
required
to
produce
half-maximal
relaxation
(EC5o)
was
56
nmol,
and
relaxation
was
complete
at
300
nmol
(Fig.
SC).
AM
produced
similar
concentration-dependent
vasodilation
of
the
canine
basilar
artery
segments
when
precontracted
with
0.1
nmol
ET-1
(data
not
shown).
Furthermore,
additional
studies
indicate
that
the
vasodilation
induced
by
AM
can
be
blocked
by
CGRP
antagonists
(CGRP8-37),
indicating
AM
action
at
CGRP
or
CGRP-like
receptors.
AM
Effects
on
MCAO
Injury.
Continuous
i.v.
infusion
of
AM
beginning
1
h
before
and
4
h
after
MCAO
did
not
produce
any
significant
changes
in
ischemic
injury
produced
by
focal
ischemia
(Table
1).
Low
doses
(0.2
and
2
nmol)
of
i.c.v.
AM
administration
at
1
h
prior
to
and
6
h
after
MCAO
tended
to
decrease
hemispheric
swelling,
hemispheric
infarct,
and
infarct
volume
(P
>
0.05;
not
significant),
whereas
the
high
i.c.v.
dose
(8
nmol)
of
AM
tended
to
increase
percent
hemispheric
swelling
(P
>
0.05;
not
significant)
and
significantly
increased
(P
<
0.05;
Table
1)
percent
hemispheric
infarct
(25.0%)
and
infarct
volume
(30.4%).
Furthermore,
we
have
established
that
AM
may
act
on
CGRP
or
CGRP-like
receptors
by
performing
radioligand
(125I-labeled
CGRP)
displacement
studies
(rat
cor-
tical
membranes)
with
AM;
these
binding
studies
(n
=
3)
revealed
an
IC50
=
80.3
nmol
for
AM
displacement
of
125I-labeled
CGRP.
Although
the
expression
of
AM
has
not
been
observed
in
the
normal
brain
(1,
2),
the
effects
of
AM
on
rat
pial
arteries
in
situ
and
on
canine
basilar
arteries
in
vitro
demonstrated
the
similar
vasodilation
function
of
AM
in
the
brain
as
in
the
non-central
nervous
system
vasculature
(1,
3-6).
i.v.
administration
of
CGRP,
a
related
vasodilator,
can
produce
a
significant
im-
provement
in
ischemic
blood
flow
to
cerebral
ischemic
tissue
and
reduce
brain
injury
after
focal
stroke
(35).
However,
dilation
of
the
microcirculation
at
the
site
of
injury
may
also
facilitate
inflammatory
cell
infiltration
that
occurs
in
the
ischemic
cortex
shortly
after
MCAO
(20,
36,
37).
Although
the
role
of
AM
in
ischemic
brain
injury
requires
further
explora-
tion,
unlike the
intraluminal
vasodilatory
and
protective
ef-
fects
of
CGRP
in
focal
ischemia
(35),
AM
vasodilatory
effects
are
abluminal.
AM
administration,
i.c.v.,
but
not
i.v.,
exacer-
bates
focal
ischemic
injury
at
a
relatively
high
dose
range
(8
nmol;
Table
1),
suggesting
that
ischemic
tissue
AM
may
contribute
to
permeability
and
disruption
of
the
blood
brain
barrier
and
thereby
to
increase
ischemic
brain
damage.
In
conclusion,
the
upregulation
of
AM
in
the
ischemic
cortex
suggests
its
involvement
in
the
acute
response
to
ischemia.
The
potential
action
of
AM
to
dilate
cerebral
vessels
and
to
increase
permeability
and
infarct
size
call
for
a
pathogenic
role
of
locally
expressed
AM
in
focal
stroke,
although
that
requires
further
exploration.
We
thank
G.
Sathe,
J.
Mao,
and
R.
Morris
for
oligonucleotide
synthesis
and
DNA
sequencing.
1.
Kitamura,
K.,
Kangawa,
K.,
Kawamoto,
M.,
Ichiki,
Y.,
Sakakibara,
S.,
Matsuo,
H.
&
Eto,
T.
(1993)
Biochem.
Biophys.
Res.
Commun.
192,
553-560.
2.
Sakata,
J.,
Shimokubo,
T.,
Kitamura,
K.,
Nakamura,
S.,
Kangawa,
K.,
Matsuo,
H.
&
Eto,
T.
(1993)
Biochem.
Biophys.
Res.
Commun.
195,
921-927.
3.
Ishiyama,
Y.,
Kitamura,
K.,
Ichiki,
Y.,
Nakamura,
S.,
Kida,
O.,
Kanagawa,
K.
&
Eto,
T.
(1993)
Eur.
J.
Pharmacol.
241,
271-273.
4.
Nuki,
C.,
Kawasaki,
H.,
Kitamura,
K,
Takenaga,
M.,
Kangawa,
K.,
Eto,
T.
&
Wada,
A.
(1993)
Biochem.
Biophys.
Res.
Commun.
196,
245-251.
5.
Perret,
M.,
Broussard,
H.,
LeGros
T.,
Bums,
A.,
Chang,
J.
K.,
Summer,
W.,
Hyman,
A.
&
Lippton,
H.
(1993)
Life
Sci.
53,
377-379.
6.
DeWitt,
C.,
Cheng,
D.
Y.,
Caminiti,
G.
N.,
Nossaman,
B.
D.,
Coy,
D.
H.,
Murphy,
W.
A.
&
Kadowitz,
P.
J.
(1994)
Eur.
J.
Pharmacol.
257,303-306.
7.
Amara,
S.
G.,
Jonas,
V.,
Rosenfeld,
M.
G.,
Ong,
E.
S.
&
Evans,
R.
M.
(1982)
Nature
(London)
298,
240-244.
8.
Morris,
H.
R.,
Panico,
M.,
Etienne,
T.,
Tippins,
J.,
Girgis,
S.
I.
&
McIntyre,
I.
(1984)
Nature
(London)
308,
746-748.
9.
Steenbergh,
P.
H.
Hoppener,
J.
W.
M.,
Zandberg,
J.,
Lips,
C.
J.
M.
&
Jansz,
H.
S.
(1985)
FEBS
Lett.
183,
403-407.
10.
Amara,
S.
G.,
Arriza,
J.
L.,
Leff,
S.
E.,
Swanson,
L.
W.,
Evans,
R.
M.
&
Rosenfeld,
M.
G.
(1985)
Science
229,
1094-1097.
11.
Cooper,
G.
J.
S.,
Willis,
A.
C.,
Clark,
A.,
Turner,
R.
C.,
Sim,
R.
B.
&
Reid,
K.
B.
M.
(1987)
Proc.
Natl.
Acad.
Sci.
USA
84,
8628-8632.
12.
Leffert,
J.
D.,
Newgard,
C.
B.,
Okamoto,
H.,
Milburn,
J.
L.
&
Luskey,
K.
L.
(1989)
Proc.
Natl.
Acad.
Sci.
USA
86,
3127-3130.
13.
Brain,
S.
D.,
Williams,
T.
J.,
Tippins,
J.
R.,
Morris,
H.
R.
&
MacIn-
tyre,
I.
(1985)
Nature
(London)
313,
54-56.
14.
Girgis,
S.
I.,
MacDolnald,
D.
W.
R.,
Stevenson,
J.
C.,
Bevis,
P.
J.
R.,
Lynch,
C.,
Wimalawansa,
S.
J.,
Self,
C.
H.,
Morris,
H.
R.
&
MacIntyre,
I.
(1985)
Lancet
2,
14-16.
15.
O'Halloran,
D.
J.
&
Bloom,
S.
R.
(1991)
Br.
Med.
J.
302,
739-740.
16.
Cooper,
G.
J.
S.
(1994)
Endocr.
Rev.
15,
163-201.
17.
Zaidi,
M.,
Moonga,
B.
S.,
Bevis,
P.
J.
R.,
Towhidul,
A.,
Legon,
S.,
Wimalawansa,
S.,
Maclntyre,
I.
&
Breimer,
L.
H.
(1991)
Vitam.
and
Horm.
(San
Diego)
46,
87-163.
18.
Jougasaki,
M.,
Wei,
C.
M.,
McKinley,
L.
J.
&
Burnett,
J.
C.
(1995)
Circulation
92,
286-289.
19.
Liang,
P.
&
Pardee,
A.
B.
(1992)
Science
257,
967-971.
20.
Barone,
F.
C.,
Hillegass,
L.
M.,
Price,
W.
J.,
White,
R.
F.,
Lee,
E.
V.,
Feuerstein,
G.
Z.,
Sarau,
H.
M.,
Clark,
R.
K.
&
Griswold,
D.
E.
(1991)
J.
Neurosci.
Res.
29,
336-345.
21.
Barone,
F.
C.,
Price,
W.
J.,
White,
R.
F.,
Willette,
R.
N.
&
Feuerstein,
G.
Z.
(1992)
Neurosci.
Biobehav.
Rev.
16,
219-233.
22.
Chomczynski,
P.
&
Sacchi,
N.
(1987)
Anal.
Biochem.
162,
156-159.
23.
Wang,
X.
K.,
Siren,
A.
L.,
Liu,
Y.,
Yue,
T.
L.,
Barone,
F.
C.
&
Feuerstein,
G.
Z.
(1994)
Mol.
Brain
Res.
26,
61-68.
24.
Wang,
X.
K.
&
Feuerstein,
G.
Z.
(1995)
BioTechniques
18,
448-453.
25.
Wang,
X.
K.,
Lee,
G.,
Liebhaber,
S.
A.
&
Cooke,
N.
E.
(1992)
J.
Biol.
Chem.
267,
9176-9184.
26.
Willette,
R.
N.,
Zhang,
H.,
Mitchell,
M.,
Sauermelch,
C.
F.,
Ohlstein,
E.
H.
&
Sulpizio,
A.
C.
(1994)
Stroke
25,
2450-2456.
27.
Willette,
R.
N.
&
Ohlstein,
E.
H.
(1994)
Drug
News
Perspect.
7,
75-81.
28.
Willette,
R.
N.
&
Sauermelch,
C.
F.
(1990)
Am.
J.
Physiol.
259,
H1688-H1693.
29.
Gaillard,
C.
&
Strauss,
F.
(1994)
Science
264,
433-436.
30.
Wang,
X.
K.,
Yue,
T.
L.,
Young,
P.
R.,
Barone,
F.
C.
&
Feuerstein,
G.
Z.
(1995)
J.
Cereb.
Blood
Flow
Metab.
15,
166-171.
31.
Liu,
T.,
McDonnell,
P.
C.,
Young,
P.
R.,
White,
R.
F.,
Siren,
A.
L.,
Hallenbeck,
J.
M.,
Barone,
F.
C.
&
Feuerstein,
G.
Z.
(1993)
Stroke
24,
1746-1751.
32.
Liu,
T.,
Clark,
R.
K.,
McDonnell,
P.
C.,
Young,
P.
R.,
White,
R.
F.,
Barone,
F.
C.
&
Feuerstein,
G.
Z.
(1994)
Stroke
25,
1481-1488.
33.
Shaw,
G.
&
Kamen,
R.
(1986)
Cell
46,
659-667.
34.
Wilson,
T.
&
Treisman,
R.
(1988)
Nature
(London)
336,
396-399.
35.
Holland,
J.
P.,
Sydserff,
S.
G.
C.,
Taylor,
W.
A.
S.
&
Bell,
B.
A.
(1994)
Stroke
25,
2055-2059.
36.
Hallenbeck,
J.
M.,
Dutka,
A.
J.,
Tanishima,
T.,
Kochaniek,
P.
M.,
Kumaroo,
K.
K.,
Thompson,
C.
B.,
Obrenovitch,
T.
P.
&
Contreras,
T.
J.
(1986)
Stroke
17,
246-253.
37.
Clark,
R.
K.,
Lee,
E.
V.,
Fish,
C.
J.,
White,
R.
F.,
Price,
W.
J.,
Jonak,
Z.
L.,
Feuerstein,
G.
Z.
&
Barone,
F.
C.
(1993)
Brain
Res.
Bull.
31,
565-572.
11484
Neurobiology:
Wang
et
al.

Supplementary resource (1)

... The neuroprotective properties of ADM have been demonstrated in several rodent models [15][16][17]. ADM mRNA is induced as early as 1 h after occlusion of the middle cerebral artery (MCA) in the rat cortex, which precedes obvious progression of infarction [18,19], further supporting the notion that ADM could act as a biomarker for the penumbra. Although reliable measurement of circulating ADM is difficult because of its short half-life [20], mid-regional pro-ADM (MR-proADM), a stable peptide fragment of ADM precursor, can be accurately measured as a surrogate marker for ADM [21]. ...
... Moreover, MR-proADM levels increased in HAIS without ischemic core, which indicated a highly sensitive tissue-based reaction to brain ischemia. These results were consistent with previous studies using the MCA occlusion rodent models, which showed early ADM mRNA upregulation in the cortex before obvious infarct development [18,19]. A prospective study showed that circulating ADM levels were significantly increased in acute cerebral hemorrhage, compared to healthy volunteers [37], but the increase was milder than ischemic stroke with a similar severity [32], supporting MR-proADM as a reliable candidate biomarker accurately reflecting ischemia. ...
... In the ischemic core, as the vascular endothelial cells are severely compromised [40], ADM secretion is presumed to be almost absent, corresponding to no correlation between the infarct size and the MR-proADM levels in our study. Moreover, a rodent MCA occlusion model study showed elevated ADM mRNA levels in the ischemic cortex even before the development of brain infarction [18,19]. Therefore, it is reasonable to assume that ADM is mainly secreted from ischemic, but not yet infarcted, region of brain tissue, namely the ischemic penumbra. ...
Article
Full-text available
Reperfusion therapy has improved the outcomes of ischemic stroke but also emphasized the importance of ischemic penumbra. However, blood biomarkers are currently unavailable for this region. Adrenomedullin (ADM) is a neuroprotective peptide, secreted in a compensatory response to brain ischemia. We thus investigated whether an increase in mid‐regional pro‐ADM (MR‐proADM), a stable peptide fragment of the ADM precursor, could act as a biomarker by predicting the ischemic penumbra in hyperacute ischemic stroke (HAIS). We prospectively enrolled consecutive HAIS patients (n = 119; median age, 77 years; male, 59.7%) admitted to our institutes from July 2017 to March 2019 and evaluated plasma MR‐proADM levels within 4.5 h of onset. MR‐proADM levels in HAIS were compared to healthy controls (n = 1298; median age, 58 years; male, 33.2%) in the Japan Multi‐Institutional Collaborative Cohort Study from 2013 to 2017. Furthermore, we evaluated whether MR‐proADM levels were associated with the penumbra estimated by clinical‐diffusion mismatch (CDM) (National Institute of Health Stroke Scale [NIHSS] ≥8, diffusion ischemic core volume ≤25 ml), or magnetic resonance angiography‐diffusion‐weighted imaging mismatch (MDM) (NIHSS ≥5, a proximal vessel occlusion with core volume ≤25 ml, or a proximal vessel stenosis/distal vessel occlusion with core volume ≤15 ml). In a case–control study, multivariate logistic analysis showed a significant association between HAIS and MR‐proADM ≥0.54 nmol/L (adjusted odds ratio, 7.92 [95% CI, 4.17–15.02], p < 0.001). Though MR‐proADM levels in HAIS did not correlate with the ischemic core volume (rs = 0.09, p = 0.348), they were higher in HAIS with CDM (n = 34; 0.81 vs. 0.61 nmol/L, p < 0.001) or MDM (n = 26; 0.83 vs. 0.62 nmol/L, p = 0.002). These differences remained significant after adjusting baseline factors (adjusted odds ratio, 4.06 [95% CI, 1.31–12.55], p = 0.015 and 4.65 [1.35–16.11], p = 0.015, respectively). Plasma MR‐proADM is elevated in HAIS, especially in those with a substantial penumbra, suggesting potential as a blood biomarker in this region.
... Blood AM levels on admission can predict outcomes after acute intracerebral hemorrhage (Wang et al., 2014a) and traumatic brain injury ). Another study found that AM was increased in cerebrospinal fluid after traumatic brain injury (Robertson et al., 2001), and its mRNA expression was significantly increased in the ischemic cortex of an animal model of focal stroke produced by middle cerebral artery occlusion (Wang et al., 1995). Moreover, intracerebroventricular administration of AM exacerbated focal ischemic brain injury, which seems to show the pathogenic role of locally expressed AM in focal stroke (Wang et al., 1995). ...
... Another study found that AM was increased in cerebrospinal fluid after traumatic brain injury (Robertson et al., 2001), and its mRNA expression was significantly increased in the ischemic cortex of an animal model of focal stroke produced by middle cerebral artery occlusion (Wang et al., 1995). Moreover, intracerebroventricular administration of AM exacerbated focal ischemic brain injury, which seems to show the pathogenic role of locally expressed AM in focal stroke (Wang et al., 1995). ...
Article
Full-text available
Adrenomedullin, a peptide with multiple physiological functions in nervous system injury and disease, has aroused the interest of researchers. This review summarizes the role of adrenomedullin in neuropathological disorders, including pathological pain, brain injury and nerve regeneration, and their treatment. As a newly characterized pronociceptive mediator, adrenomedullin has been shown to act as an upstream factor in the transmission of noxious information for various types of pathological pain including acute and chronic inflammatory pain, cancer pain, neuropathic pain induced by spinal nerve injury and diabetic neuropathy. Initiation of glia-neuron signaling networks in the peripheral and central nervous system by adrenomedullin is involved in the formation and maintenance of morphine tolerance. Adrenomedullin has been shown to exert a facilitated or neuroprotective effect against brain injury including hemorrhagic or ischemic stroke and traumatic brain injury. Additionally, adrenomedullin can serve as a regulator to promote nerve regeneration in pathological conditions. Therefore, adrenomedullin is an important participant in nervous system diseases.
... ANP is a wellknown physiologically important cardiovascular peptide that exerts natriuretic, diuretic, and vasorelaxant properties, and it is expressed in cardiac and cerebral tissues[56]. In the search for stroke-related genes, another experimental model was investigated, the SHR with MCAO-induced ischemic stroke[57]. ...
Article
Cardiovascular disease is actually a major cause of mortality, illness and hospitalization worldwide. Several risk factors have been identified that are strongly associated with the development of cardiovascular disease. Public prevention strategies have relied predominately on managing environmental factors that contribute to cardiovascular disease, such as obesity, smoking and lack of exercise. The understanding of the role of genetics in cardiovascular disease development has become much more important to link genetics with the onset of disease and response to therapy. This seeks to examine how genes can predispose individuals to cardiovascular disease and how this knowledge might be applied to more comprehensive preventive strategies in the future. In addition, the review explores possibilities for genetics in cardiovascular disease treatment, particularly through the use of identified driver genes and gene therapy. To fully understand the biological implications of these associations, there is a need to relate them to the exquisite, multilayered regulation of protein expression and regulatory elements, mutation, microRNAs and epigenetics. Understanding how the information contained in the DNA relates to the operation of these regulatory layers will allow us not only to better predict the development of cardiovascular disease but also to develop more effective therapies.
... These results suggest that the ADM, ANXA3, SLC22A4 and VIM genes play a key role in the pathological process of ischaemic stroke. Previous research has proven that the expression levels of adrenomedullin (ADM) are significantly increased in ischaemic cortical neurons induced by ischaemic injury in patients with ischaemic cerebrovascular disease [29]. Ischaemic cerebrovascular disease involves not only ischaemic brain cell injury but also arterial injury. ...
Article
Full-text available
Background The immune system plays a vital role in the pathological process of ischaemic stroke. However, the exact immune-related mechanism remains unclear. The current research aimed to identify immune-related key genes associated with ischaemic stroke. Methods CIBERSORT was utilized to reveal the immune cell infiltration pattern in ischaemic stroke patients. Meanwhile, a weighted gene coexpression network analysis (WGCNA) was utilized to identify meaningful modules significantly correlated with ischaemic stroke. The characteristic genes correlated with ischaemic stroke were identified by the following two machine learning methods: the support vector machine-recursive feature elimination (SVM-RFE) algorithm and least absolute shrinkage and selection operator (LASSO) logistic regression. Results The CIBERSORT results suggested that there was a decreased infiltration of naive CD4 T cells, CD8 T cells, resting mast cells and eosinophils and an increased infiltration of neutrophils, M0 macrophages and activated memory CD4 T cells in ischaemic stroke patients. Then, three significant modules (pink, brown and cyan) were identified to be significantly associated with ischaemic stroke. The gene enrichment analysis indicated that 519 genes in the above three modules were mainly involved in several inflammatory or immune-related signalling pathways and biological processes. Eight hub genes (ADM, ANXA3, CARD6, CPQ, SLC22A4, UBE2S, VIM and ZFP36) were revealed to be significantly correlated with ischaemic stroke by the LASSO logistic regression and SVM-RFE algorithm. The external validation combined with a RT‒qPCR analysis revealed that the expression levels of ADM, ANXA3, SLC22A4 and VIM were significantly increased in ischaemic stroke patients and that these key genes were positively associated with neutrophils and M0 macrophages and negatively correlated with CD8 T cells. The mean AUC value of ADM, ANXA3, SLC22A4 and VIM was 0.80, 0.87, 0.91 and 0.88 in the training set, 0.85, 0.77, 0.86 and 0.72 in the testing set and 0.87, 0.83, 0.88 and 0.91 in the validation samples, respectively. Conclusions These results suggest that the ADM, ANXA3, SLC22A4 and VIM genes are reliable serum markers for the diagnosis of ischaemic stroke and that immune cell infiltration plays a crucial role in the occurrence and development of ischaemic stroke.
... The role of AM in angiogenesis has been extensively researched due to its expression in ECs and VSMCs. AM is one of several proangiogenic targets up-regulated in hypoxia to maintain tumor growth and progression [42]. Transcription factor HIF-1α synthesizes AM through hypoxic response elements [43,44]. ...
Article
Full-text available
Introduction: Adrenomedullin (AM) is a peptide responsible for many physiological processes including vascular health and hormone regulation. Dysregulation of AM signaling can stimulate cancers by promoting proliferation, angiogenesis and metastasis. Two AM receptors contribute to tumor progression in different ways. Adrenomedullin-1 receptor (AM1R) regulates blood pressure and blocking AM signaling via AM1R would be clinically unacceptable. Therefore, antagonizing adrenomedullin-2 receptor (AM2R) presents as an avenue for anti-cancer drug development. Areas covered: We review the literature to highlight AM's role in cancer as well as delineating the specific roles AM1R and AM2R mediate in the development of a pro-tumoral microenvironment. We highlight the importance of exploring the residue differences between the receptors that led to the development of first-in-class selective AM2R small molecule antagonists. We also summarize the current approaches targeting AM and its receptors, their anti-tumor effects and their limitations. Expert opinion: As tool compounds, AM2R antagonists will allow the dissection of the functions of CGRPR (calcitonin gene-related peptide receptor), AM1R and AM2R, and has considerable potential as a first-in-class oncology therapy. Furthermore, the lack of detectable side effects and good drug-like pharmacokinetic properties of these AM2R antagonists support the promise of this class of compounds as potential anti-cancer therapeutics.
... Thus, they have variable efficacy and substantial tolerability issues that often lead to discontinuation. 2 Erenumab is a first-in-class fully human monoclonal antibody targeting the calcitonin gene-related peptide receptor. [3][4][5][6] Randomized controlled trials (RCTs) demonstrated a clinically meaningful reduction in monthly migraine days (MMD), monthly headache days (MHD), and monthly migraine-specific medication treatment days (MSMD) in erenumab-treated patients with migraine. [7][8][9][10][11] While erenumab RCTs included many patients with no prior ineffective prophylactic migraine treatments and included many others with fewer than two prior ineffective migraine prophylactic treatments, a recent randomized double-blind placebo-controlled study has shown efficacy for erenumab in patients with EM who had experienced inadequate efficacy of two to four migraine prophylactic drugs. ...
Article
Full-text available
Objectives: To assess real-world effectiveness, safety, and usage of erenumab in Canadian patients with episodic and chronic migraine with prior ineffective prophylactic treatments. Background: In randomized controlled trials, erenumab demonstrated efficacy for migraine prevention in patients with ≤4 prior ineffective prophylactic migraine therapies. The "Migraine prevention with AimoviG: Informative Canadian real-world study" (MAGIC) assessed real-world effectiveness of erenumab in Canadian patients with migraine. Methods: MAGIC was a prospective open-label, observational study conducted in Canadian patients with chronic migraine (CM) and episodic migraine (EM) with two to six categories of prior ineffective prophylactic therapies. Participants were administered 70 mg or 140 mg erenumab monthly based on physician's assessment. Migraine attacks were self-assessed using an electronic diary and patient-reported outcome questionnaires. The primary outcome was the proportion of subjects achieving ≥50% reduction in monthly migraine days (MMD) after the 3-month treatment period. Results: Among the 95 participants who mostly experienced two (54.7%) or three (32.6%) prior categories of ineffective prophylactic therapies and who initiated erenumab, treatment was generally safe and well tolerated; 89/95 (93.7%) participants initiated treatment with 140 mg erenumab. At week 12, 32/95 (33.7%) participants including 17/64 (26.6%) CM and 15/32 (48.4%) EM achieved ≥50% reduction in MMD while 30/86 (34.9%) participants including 19/55 (34.5%) CM and 11/31 (35.5%) EM achieved ≥50% reduction in MMD at week 24. Through patient-reported outcome questionnaires, 62/95 (65.3%) and 45/86 (52.3%) participants reported improvement of their condition at weeks 12 and 24, respectively. Physicians observed improvement in the condition of 78/95 (82.1%) and 67/86 (77.9%) participants at weeks 12 and 24, respectively. Conclusion: One-third of patients with EM and CM achieved ≥50% MMD reduction after 3 months of erenumab treatment. This study provides real-world evidence of erenumab effectiveness, safety, and usage for migraine prevention in adult Canadian patients with multiple prior ineffective prophylactic treatments.
... Despite the clear connection between AM and the cardiovascular system [18,37], understanding the exact contribution of AM to stroke resolution is not an easy task and the literature is full of apparent controversies, which may ultimately reflect the high complexity of the system [42]. For instance, therapeutic treatment of animal models of ischemic stroke with AM peptide may result in either damaging [43] or protective [44][45][46] effects. In another example, from our own laboratory, we described that genetically eliminating the adm gene from neurons increased stroke volume in a mouse model of middle cerebral artery occlusion [23]; but when we eliminated the same gene from endothelial cells, we obtained the reverse result [47]. ...
Article
Full-text available
Hemorrhagic stroke remains an important health challenge. Adrenomedullin (AM) is a vasoactive peptide with an important role in cardiovascular diseases, including stroke. Serum AM and nitrate–nitrite and S-nitroso compounds (NOx) levels were measured and compared between healthy volunteers (n = 50) and acute hemorrhagic stroke patients (n = 64). Blood samples were taken at admission (d0), 24 h later (d1), and after 7 days or at the time of hospital discharge (d7). Neurological severity (NIHSS) and functional prognosis (mRankin) were measured as clinical outcomes. AM levels were higher in stroke patients at all times when compared with healthy controls (p < 0.0001). A receiving operating characteristic curve analysis identified that AM levels at admission > 69.0 pg/mL had a great value as a diagnostic biomarker (area under the curve = 0.89, sensitivity = 80.0%, specificity = 100%). Furthermore, patients with a favorable outcome (NIHSS ≤ 3; mRankin ≤ 2) experienced an increase in AM levels from d0 to d1, and a decrease from d1 to d7, whereas patients with unfavorable outcome had no significant changes over time. NOx levels were lower in patients at d0 (p = 0.04) and d1 (p < 0.001) than in healthy controls. In conclusion, AM levels may constitute a new diagnostic and prognostic biomarker for this disease, and identify AM as a positive mediator for hemorrhagic stroke resolution.
... In this context, AM may therefore have a harmful effect in the endothelium but a neuroprotective effect when administered systemically [29]. In addition, intracerebroventricular injection of AM results in an increase in ischemic injury [30]. The effects of AM may thus be context-and/or cell type-dependent. ...
Article
Full-text available
Adrenomedullin (AM) is an endogenous peptide mainly secreted from endothelial cells, which has multiple physiological actions such as anti-inflammation, vasodilation, vascular permeability regulation and angiogenesis. Blood AM levels are upregulated in a variety of pathological states including sepsis, severe COVID-19, acute ischemic stroke and vascular cognitive impairment with white matter changes, likely serving as a compensatory biological defense response against infection and ischemia. AM is currently being tested in clinical trials for ulcerative colitis, Crohn’s disease, severe COVID-19 for its anti-inflammatory properties and in ischemic stroke for its additional angiogenic action. AM has been proposed as a therapeutic option for vascular cognitive impairment as its arteriogenic and angiogenic properties are thought to contribute to a slowing of cognitive decline in mice after chronic cerebral hypoperfusion. As AM promotes differentiation of oligodendrocyte precursor cells into mature oligodendrocytes under hypoxic conditions, AM could also be used in the treatment of CADASIL, where reduced oxygen delivery is thought to lead to the death of hypoxia-prone oligodendrocytes. AM therefore holds potential as an innovative therapeutic drug, which may regenerate blood vessels, while controlling inflammation in cerebrovascular diseases.
Article
Full-text available
Adrenomedullin (ADM) is a potent hypotensive peptide, which is produced during sepsis and ischemia. We demonstrate here that hypoxia induced a time-dependent increase of both ADM mRNA and protein expressions in cultured astrocytes and endothelial cells from rat brain microvessels. Gene reporter analyses showed a 2-fold increase in ADM gene transcription which was suppressed when the ADM promoter was deleted of its hypoxia responsive element. Hypoxia increased 7-fold the stability of pre-formed ADM mRNAs. Rat brain microvessels expressed mRNAs coding for the different putative ADM receptors but they did not respond to exogenous ADM and calcitonin gene-related peptide by the formation of cAMP. In contrast, ADM and calcitonin gene-related peptide increased the formation of cAMP in astrocytes and their actions were potentiated about 2-fold after hypoxia. Messenger RNA species coding for three putative ADM receptors (the L1 orphan receptor, RDC-1, and calcitonin receptor-like receptor) and accessory proteins (receptor-activity modifying proteins) were present in astrocytes. Hypoxia selectively up-regulated expression of RDC-1 receptor mRNAs. The results indicate that ADM and RDC-1 are hypoxia-sensitive genes and that RDC-1 receptors may mediate some actions of ADM in hypoxic astrocytes.
Article
Full-text available
The expression of interleukin-6 (IL-6) mRNA in the focal ischemic rat cortex was studied by means of Northern hybridization. IL-6 mRNA was induced after permanent occlusion of the middle cerebral artery, reached a significant level at 3 h, and peaked at 12 h, i.e., ~ 10-fold increase in the ischemic zone compared with the nonischemic cortex or sham-operated controls. The increased IL-6 mRNA was elevated for at least 24 h. Low levels of IL-6 mRNA were detected in sham-operated rats or in the contralateral nonischemic cortex. The expression of c-fos and zif268 mRNAs, two early response genes, was rapid (increased by 1 h postischemia) and transient (returned to basal levels by 24 and 12 h, respectively), clearly having different kinetic patterns from that of IL-6 mRNA. The early response kinetic pattern of c-fos and zif268 mRNAs in focal ischemia suggests their transcriptional regulatory roles in response to ischemic insult, while the delayed induction pattern of IL-6 mRNA suggests a role for this pleiotropic cytokine in the inflammatory response to the focal ischemic damage of the brain.Keywords: c-fos; Focal brain ischemia; Inflammation; Interleukin-6; Stroke; zif268
Article
Full-text available
We previously reported the structure of the placentally derived human cysteine-rich (h crp) cDNA and demonstrated that it encodes a highly conserved and widely distributed zinc finger-like protein. We now report that the expression of both the mouse and human crp genes is induced as a primary response to serum in quiescent Balb/c 3T3 cells and in human fibroblasts. The profile of this primary response is remarkably parallel to that of c-myc in the Balb/c 3T3 cell line. The structure of the 23.2-kilobase h crp gene demonstrates that it is a member of a gene superfamily encoding proteins sharing a highly characteristic 52-amino acid "LIM/double-finger" motif. The evolutionarily conserved structure of cysteine-rich protein, its structural similarity to a number of developmentally critical proteins, its distinctive tissue distribution, and its primary response to early events in the cell cycle suggest that crp plays an important role in cell function.
Article
In addition to calcitonin and katacalcin, the human calcitonin gene encodes a novel peptide--calcitonin gene-related peptide (CGRP)--a potent vasodilator. A sensitive and specific radioimmunoassay was developed to study plasma levels of CGRP in normal subjects. CGRP circulates at five times the concentration of calcitonin, suggesting that it may be an important physiological regulator of vascular tone and blood flow.
Article
The expression of intercellular adhesion molecule 1 (ICAM-1) was studied in rat focal ischemic cortex. A significant increase in ICAM-1 mRNA expression in the ischemic cortex over levels in contralateral (nonischemic) site was observed by means of Northern blot analysis following either permanent or temporary occlusion with reperfusion of the middle cerebral artery (PMCAO or MCAO with reperfusion) in spontaneously hypertensive rats. In the ischemic cortex, levels of ICAM-1 mRNA increased significantly at 3 h (2.6-fold, n = 3, P < 0.05), peaked at 6 to 12 h (6.0-fold, P < 0.01) and remained elevated up to 5 days (2.5-fold, P < 0.05) after PMCAO. The profile of ICAM-1 mRNA expression in the ischemic cortex following MCAO with reperfusion was similar to that following PMCAO, except that ICAM-1 mRNA was significantly increased as early as 1 h (6.3-fold, n = 3, P < 0.05) and then gradually reached a peak at 12 h (12-fold, P < 0.01) after reperfusion. ICAM-1 mRNA expression in ischemic cortex following PMCAO was significantly greater in hypertensive rats than in two normotensive rat strains. Immunostaining using anti-ICAM-1 antibodies indicated that upregulated ICAM-1 expression was localized to endothelial cells of intraparenchymal blood vessels in the ischemic but not contralateral cortex. The data suggest that an upregulation of ICAM-1 mRNA and protein on brain capillary endothelium may play an important role in leukocyte migration into ischemic brain tissue.
Article
Effective methods are needed to identify and isolate those genes that are differentially expressed in various cells or under altered conditions. This report describes a method to separate and clone individual messenger RNAs (mRNAs) by means of the polymerase chain reaction. The key element is to use a set of oligonucleotide primers, one being anchored to the polyadenylate tail of a subset of mRNAs, the other being short and arbitrary in sequence so that it anneals at different positions relative to the first primer. The mRNA subpopulations defined by these primer pairs were amplified after reverse transcription and resolved on a DNA sequencing gel. When multiple primer sets were used, reproducible patterns of amplified complementary DNA fragments were obtained that showed strong dependence on sequence specificity of either primer.
Article
A review of the sensitivity of genetically hypertensive rats to cerebral ischemia was presented together with original data describing the systematic comparison of the effects of focal ischemia (permanent and temporary with reperfusion) performed in hypertensive and normotensive rats (i.e., blood pressures verified in conscious instrumented rats). Microsurgical techniques were used to isolate and occlude the middle cerebral artery (MCAO) of spontaneously hypertensive (SHR), Sprague-Dawley (SD) and Wistar Kyoto (WKY) rats at the level of the inferior cerebral vein. Following permanent (24 h) MCAO, persistent and similar decreases in local microvascular perfusion (i.e., to 15.6 +/- 1.7% of pre-MCAO levels) were verified in the primary ischemic zone of the cortex for all strains using Laser-Doppler flowmetry. A contralateral hemiplegia that occurred following MCAO, evidenced by forelimb flexion and muscle weakness, was greater in SHR (neurological grade = 2.0 +/- 0.1) than SD (1.0 +/- 0.4) or WKY (0.7 +/- 0.4) rats (N = 7-9, p less than 0.05). SHR also exhibited sensory motor deficits following MCAO compared to sham-operation, with decreased normal placement response of the hindlimb (% normal = 20 vs. 83, N = 23-30, p decreased rota-rod (41 +/- 7 vs. 126 +/- 19 on rod, N = 10-15, p less than 0.05) and balance beam (25 +/- 5 vs. 116 +/- 29 s on beam, N = 5-7, p less than 0.05) performance. However, an index of general motor activity was not affected by permanent MCAO. Triphenyltetrazolium-stained forebrain tissue analyzed by planimetry revealed a significantly larger and more consistent cortical infarction in SHR (hemispheric infarction = 27.9 +/- 1.5%) compared to SD (15.4 +/- 4.1%) and WKY (4.0 +/- 2.4%) rats (N = 7-9, p less than 0.05), occupying predominantly the frontal and parietal areas. Also, a significant degree of ipsilateral hemispheric swelling (4.6 +/- 0.9%, N = 7-9, p less than 0.05) and increased brain water content (78.4 +/- 0.3% to 80.4 +/- 0.2%, N = 8-9, p less than 0.05) was identified in SHR that was not observed in SD or WKY rats. A novel model of temporary MCAO also was evaluated in the hypertensive and normotensive rat strains. Initially, the effect of increasing MCAO-time followed by 24 h reperfusion in SHR was studied. During temporary MCAO (20 to 300 min), persistent and stable decreases in local microvascular perfusion (i.e., to 15-20% of pre-MCAO levels) were verified in the primary ischemic zones of the cortex.(ABSTRACT TRUNCATED AT 400 WORDS)
Article
Two different techniques were utilized to identify the infiltration of polymorphonuclear leukocytes (PMN) into cerebral tissue following focal ischemia: histologic analysis and a modified myeloperoxidase (MPO) activity assay. Twenty-four hours after producing permanent cortical ischemia by occluding and severing the middle cerebral artery of male spontaneously hypertensive rats, contralateral hemiparalysis and sensory-motor deficits were observed due to cerebral infarction of the frontal and parietal cortex. In hematoxylin-and-eosin-stained histologic sections, PMN, predominantly neutrophils, were identified at various stages of diapedesis from deep cerebral and meningeal vessels at the periphery of the infarct, into brain parenchyma. When MPO activity in normal brain tissue was studied initially, it could not be demonstrated in normal tissues extracted from non-washed homogenates. However, if tissue was homogenized in phosphate buffer (i.e., washed), MPO activity was expressed upon extraction. Utilizing this modified assay, MPO activity was significantly increased only in the infarcted cortex compared to other normal areas of the brain. This was observed in non-perfused animals and after perfusion with isotonic saline to remove blood constituents from the vasculature prior to brain removal. The increased PMN infiltration and MPO activity were not observed in forebrain tissue of sham-operated control rats. Also, MPO activity was not increased in the ischemic cortex of MCAO rats perfused immediately after middle cerebral artery occlusion, indicating that blood was not trapped in the ischemic area. By using a leukocyte histochemical staining assay, activity of peroxidases was identified within vascular-adhering/infiltrating PMN in the infarcted cortex 24 hr after focal ischemia. An evaluation of several blood components indicated that increased MPO activity was selective for PMN. The observed increase of approximately 0.3 U MPO/g wet weight ischemic tissue vs. nonischemic cerebral tissues probably reflects the increased vascular adherance/infiltration of approximately 600,000 PMN/g wet weight infarcted cortex 24 hr after focal ischemia. This combined biochemical and histological study strongly suggests that PMN adhere within blood vessels and infiltrate into brain tissue injured by focal ischemia and that the associated inflammatory response might contribute to delayed progressive tissue damage in focal stroke. This modified MPO assay is a useful, quantitative index of PMN that can be utilized to elucidate the potential deleterious consequences of neutrophils infiltrating into the central nervous system after cerebral ischemia, trauma, or other pro-inflammatory stimuli.
Article
This chapter discusses the expression and function of the calcitonin gene products. The starting point for the study of the calcitonin genes is the isolation and characterization of clones representing calcitonin mRNA. Calcitonin is not the first regulatory peptide sequence to be cloned; however, its cloning proved to be the first step in the discovery of several further regulatory peptides. Although the cloning of the calcitonin genes has led to the discovery of more new and interesting information than almost any other similar gene, there are good reasons to believe that there is more to come. There have been a number of reports that there is a second type of calcitonin in humans that is detectable with antisera raised against salmon calcitonin. A study of the evolution of a gene can provide useful information about which parts of the gene are most important for its biological activity. Studies of single genes in several species generally show that active sites tend to be conserved while the other regions are less stable.