ArticlePDF Available

Requirement for matrix metalloproteinase-9 (Gelatinase B) expression in metastasis by murine prostate carcinoma

Authors:

Abstract

Although a number of effective therapies are available for localized prostate cancer, metastatic prostate cancer is difficult to treat and impossible to cure. Identification of the gene products that enable a prostatic carcinoma cell to metastasize should facilitate an understanding of the processes leading to metastasis. To characterize the contribution of matrix metalloproteinase-9 (MMP-9, gelatinase B or the 92-kd type IV gelatinase/collagenase) to the development of metastasis in prostate cancer, we reduced MMP-9 expression in metastatic murine prostatic carcinoma cells using a ribozyme. The ribozyme transfected cells had lower basal levels of MMP-9 as well as decreased levels after stimulation by transforming growth factor-beta or phorbol 12-myristate 13-acetate when compared with the parental cells or with control transfectants. The cells with down-regulated MMP-9 were unable to form lung colonies in the experimental metastasis assay, whereas the controls and parental cells readily formed metastases. All cell types readily formed tumors after injection and down-regulation of MMP-9 did not adversely affect the rate of tumor growth. Thus, MMP-9 expression is required for hematogenous metastasis in a murine prostate model system raising the possibility that it may play an equivalent role in human prostate cancer.
American
Journal
of
Pathology,
Vol.
152,
No.
2,
February
1998
Copyright
©)
American
Society
for
Investigative
Pathology
Requirement
for
Matrix
Metalloproteinase-9
(Gelatinase
B)
Expression
in
Metastasis
by
Murine
Prostate
Carcinoma
Geeta
Sehgal,*
Jin
Hua,*
Eric
J.
Bernhard,t
Inder
Sehgal,*
Timothy
C.
Thompson,f
and
Ruth
J.
Muschel*
From
the
Departments
of
Pathology
and
Laboratory
Medicine,*
and
Radiation
Oncology,t
University
of
Pennsylvania,
Philadelphia,
Pennsylvania
and
the
Department
of
Urology,*
Baylor
College
of
Medicine,
Houston,
Texas
Although
a
number
of
effective
therapies
are
available
for
localized
prostate
cancer,
metastatic
prostate
can-
cer
is
difficult
to
treat
and
impossible
to
cure.
Identi-
fication
of
the
gene
products
that
enable
a
prostatic
carcinoma
cell
to
metastasize
should
facilitate
an
un-
derstanding
of
the
processes
leading
to
metastasis.
To
characterize
the
contribution
of
matrix
metallopro-
teinase-9
(MMP-9,
gelatinase
B
or
the
92-kd
type
IV
gelatinase/collagenase)
to
the
development
of
metas-
tasis
in
prostate
cancer,
we
reduced
MMP-9
expres-
sion
in
metastatic
murine
prostatic
carcinoma
cells
using
a
ribozyme.
The
ribozyme
transfected
cells
had
lower
basal
levels
of
MMP-9
as
well
as
decreased
levels
after
stimulation
by
transforming
growth
factor-8
or
phorbol
12-myristate
13-acetate
when
compared
with
the
parental
cells
or
with
control
transfectants.
The
cells
with
down-regulated
MMP-9
were
unable
to
form
lung
colonies
in
the
experimental
metastasis
assay,
whereas
the
controls
and
parental
cells
readily
formed
metastases.
All
cell
types
readily
formed
tu-
mors
after
injection
and
down-regulation
of
MMP-9
did
not
adversely
affect
the
rate
of
tumor
growth.
Thus,
MMP-9
expression
is
required
for
hematoge-
nous
metastasis
in
a
murine
prostate
model
system
raising
the
possibility
that
it
may
play
an
equivalent
role
in
human
prostate
cancer.
(Am
J
Patbol
1998,
152:591-596)
In
individual
patients
it
is
currently
not
possible
to
predict
whether
metastasis
has
occurred
from
the
size
or
other
clinical
parameters
of
the
primary
tumor.
For
patients
with
prostatic
carcinoma,
the
determination
of
whether
metas-
tases
are
present
has
considerable
clinical
significance
as
prostate
cancer
can
often
be
effectively
treated
if
the
tumor
has
remained
confined
to
the
gland,
but
for
pa-
tients
with
metastatic
disease,
long
term
survival
is
rare.
Thus,
an
understanding
of
the
gene
products
that
are
required
for
metastasis
in
prostate
cancer
could
be
valu-
able
both
for
delineation
of
the
alterations
that
must
occur
in
the
cell
for
metastasis
to
occur
and
to
lead
to
the
identification
of
markers
that
might
predict
metastasis.
To
determine
some
of
the
factors
that
are
required
for
metastasis
by
prostate
carcinoma
cells,
we
turned
to
the
mouse
prostate
reconstitution
system
developed
by
Thompson
et
al1
in
which
neonatal
urogenital
sinus
tis-
sue,
the
fetal
precursor
of
the
adult
prostate,
was
ex-
planted,
infected
with
a
recombinant
retrovirus
that
ex-
presses
the
oncogenes
rasH
and
myc,
and
reimplanted
into
the
renal
capsule.
This
procedure
resulted
in
the
formation
of
tumors
that
histologically
were
virtually
iden-
tical
to
human
prostatic
carcinomas
and
that
displayed
characteristic
murine
prostate
markers.
These
tumors
were
generally
nonmetastatic,
but
when
male
p53
-/-
or
p53
+/-
mice
were
used
as
the
source
of
urogenital
sinus
tissue,
the
resultant
tumors
frequently
gave
rise
to
metastases
in
the
lung,
liver,
bone,
and
mesentery.2
Sehgal
et
al,3
using
cells
isolated
from
both
primary
tumors
and
metastases
generated
in
this
way,
found
that
the
105-kd
gelatinase
activity
characteristic
of
murine
matrix
metalloproteinase-9
(MMP-9)
or
murine
gelatinase
B
could
be
induced
by
transforming
growth
factor-,8
(TGF-3)
in
five
of
the
six
lines
isolated
from
metastatic
tumors
but
in
only
one
of
six
cell
cultures
isolated
from
the
primary
tumors.
These
data
correlating
MMP-9
produc-
tion
to
prostate
cancer
metastasis
suggested
that
MMP-9
expression
might
be
important
for
metastasis
in
this
sys-
tem
and
were
consistent
with
observations
linking
MMP-9
to
metastasis
in
other
systems.
MMP-9,
gelatinase
B,
or
the
92-kd
type
IV
gelatinase/
collagenase
is
one
member
of
a
family
of
matrix
metallo-
proteinases
(MMPs),
an
enzyme
family
defined
by
a
con-
served
catalytic
domain
that
requires
Zn2+
for
activity.4`6
MMP-9
is
capable
of
degrading
a
variety
of
substrates
including
molecules
found
in
the
extracellular
matrix
such
as
collagens
IV,
V,
elastin,
entactin,
as
well
as
casein,
the
precursor
of
tumor
necrosis
factor-a,
and
a
cell
surface
Supported
by
grants
from
the
National
Cancer
Institute
National
Institutes
of
Health,
CA-46830
(R.
J.
Muschel),
CA
50588,
and
CA
68814
(T.
C.
Thompson).
Accepted
for
publication
November
15,
1997.
Address
reprint
requests
to
Dr.
Ruth
J.
Muschel,
Room
269
John
Morgan
Building,
36
and
Hamilton
Walk,
University
of
Pennsylvania,
Phil-
adelphia,
PA
19104.
E-mail:
muschel@mail.med.upenn.edu.
591
592
Sehgal
et
al
AJP
February
1998,
Vol.
152,
No.
2
molecule,
galectin.7-9
MMP-9
is
secreted
in
an
inactive
form
of
molecular
mass
of
92
kd
in
humans
and
rats
but
of
105
kd
in
mice
because
of
a
16-amino
acid
insert.10'11
In
addition
to
the
widely
recognized
secreted
form
of
MMP-9,
Toth
et
al12
have
also
identified
a
cell
surface-
associated
form.
Successive
cleavages
of
the
N-terminal
region,
which
has
inhibitory
activity
against
the
enzyme
itself,
results
in
the
active
82-kd
form
and
sometimes
an
active
65-kd
form
of
the
human
enzyme.13
In
vitro
treat-
ment
of
pro-MMP-9
with
either
stromelysin,
MMP-2,
tissue
kallikerien,
matrilysin,
type
collagenase,
14-19
or
plas-
minogen
activator
generates
the
active
form,
but
the
physiological
activators
are
unknown.
The
tissue
inhibi-
tors
of
matrix
metalloproteinases,
TIMPs-1,
-2,
-3,
or
-4
can
inhibit
MMP-9
activity,
but
TIMP-1
has
a
higher
affin-
ity
for
MMP-9
than
does
TIMP-2,
and
MMP-9
is
frequently
isolated
in
a
complex
with
TIMP-1
.20-31
The
importance
of
MMP-9
in
metastasis
was
under-
scored
by
results
in
a
rat
sarcoma
model
system.
Bern-
hard
et
a132
noted
that
rat
embryo
fibroblasts
transformed
by
rasH
plus
myc
were
metastatic
and
secreted
MMP-9,
but
that
the
same
cells
transformed
by
ras
plus
adenovi-
rus
ElA
were
nonmetastatic
and
did
not
secrete
MMP-9.
Both
cell
types
were
equally
tumorigenic.
Introduction
of
ElA
expression
vectors
into
metastatic
transformed
rat
embryo
fibroblasts
inhibited
both
metastasis
and
MMP-9
expression.
ElA
will
also
block
metastasis
by
murine
mammary
cells.33
To
establish
the
role
of
MMP-9
in
this
system,
Bernhard
et
a134
introduced
an
expression
vector
for
MMP-9
into
the
nonmetastatic
rasH
plus
ElA
transfor-
mants.
This
led
to
metastasis
in
the
experimental
metas-
tasis
assay.
In
complementary
work,
Hua
and
Muschel35
showed
that
inhibition
of
MMP-9
expression
using
an
expression
vector
for
a
ribozyme
against
MMP-9
in
the
fibroblasts
that
were
transformed
by
rasH
plus
myc
blocked
experimental
metastasis
but
did
not
measurably
alter
tumorigenicity.
These
data
established
the
impor-
tance
of
MMP-9
in
metastasis
in
a
rat
sarcoma
model
system
as
well
as
its
independence
from
tumorigenicity
and
led
us
to
ask
whether
inhibition
of
MMP-9
expression
would
affect
metastasis
by
the
cell
lines
isolated
from
the
metastatic
tumors
generated
in
the
mouse
prostate
re-
constitution
system.
Materials
and
Methods
Plasmids
MMP-9
ribozyme
and
the
control
hammerhead
expres-
sion
vectors
were
constructed
by
subcloning
DNA
frag-
ments
coding
for
the
ribozyme
directed
against
MMP-9
or
for
a
hammerhead
structure
into
the
expression
vector
pRC/CMV
(Invitrogen,
San
Diego,
CA)
as
described
in
Hua
and
Muschel.35
Cell
Culture
Murine
prostate
carcinoma
cells,
the
metastatic
(148-
1,LMD)
and
primary
cells
(148-1,PA),
were
generated
as
described
in
Thompson
et
al.2
Cell
cultures
were
routinely
maintained
in
Dulbecco's
modified
Eagle's
me-
dium
supplemented
with
10%
heat-inactivated
fetal
bovine
serum,
penicillin,
and
streptomycin.
Metastatic
(148-1,LMD)
cells
were
transfected
with
30
,ug
of
pRC/
CMV,
pRC/CMV
with
the
MMP-9
ribozyme,
or
the
control
hammerhead
construct
by
the
calcium
phosphate
pre-
cipitation
method
as
described
previously
(Muschel
et
al,
1985).36
The
parental
cells
already
contain
one
copy
of
the
neomycin
resistance
gene
because
of
the
homolo-
gous
recombination
to
eliminate
p53.
The
parental
cells
retained
neomycin
resistance
at
600
to
700
,ug/ml.
Intro-
duction
of
the
ribozyme-expressing
plasmids
was
ac-
complished
by
selection
after
transfection
for
greatly
en-
hanced
resistance
to
geneticin.
Selection
was
with
geneticin
at
1.4
mg/ml.
Individual
colonies
were
isolated
using
cloning
cylinders.
Clones
AR
1.51
and
AR
1.52
were
isolated
after
transfection
of
148-1,LMD
with
the
expression
vector
for
the
MMP-9
ribozyme,
and
sub-
clones
AAS
1.22
and
AAS
1.74
were
isolated
after
trans-
fection
of
the
same
cells
with
the
expression
vector
for
the
hammerhead
control
ribozyme.
Zymography
Gelatin
substrate
gel
electrophoresis
was
accomplished
as
previously
described.32
To
prepare
conditioned
me-
dium,
cells
were
washed
with
serum-free
medium
and
resupplied
with
fresh
serum-free
media.
They
were
treated
with
TGF-f3
(80
pmol/L)
or
with
TPA
(50
ng/ml).
After
24
hours,
the
serum-free
conditioned
media
was
harvested,
adjusted
for
cell
number,
centrifuged
at
1500
x
g
to
remove
cellular
debris,
and
concentrated
to
650
,ul
(Centriprep-10
concentrator,
Millipore,
Burlington,
MA).
Concentrated
media
was
electrophoresed
under
nonreducing
conditions
and
without
heating
through
a
7%
sodium
dodecyl
sulfate-polyacrylamide
gel
electro-
phoresis
gel
containing
0.1%
porcine
gelatin
(Sigma,
St.
Louis,
MO).
Gels
were
washed
in
0.05
mol/L
Tris
(pH
7.4),
2%
Triton
X-100,
0.2
m
of
NaCI,
and
0.02%
NaN3.
Positive
controls
were
human
recombinant
MMP-9
generously
provided
by
R.
Fridman
(Wayne
State
University,
Detroit,
Ml)
or
conditioned
medium
from
2.10.10
cells
from
trans-
formed
rat
embryo
fibroblasts.37
Gels
were
stained
in
0.2%
Coomassie
blue
for
1
hour
and
destained
in
20%
(v/v)
methanol
and
10%
(v/v)
acetic
acid.
The
clear
bands
represent
gelatinase
activity.
Northern
Blotting
Total
cellular
RNA
(60
,ug)
or
polyadenylated
RNA
(5
,ug)
(direct
mRNA
mini
kit,
Qiagen,
Chatsworth,
CA)
was
elec-
trophoresed
in
a
formaldehyde/agarose
gel
and
trans-
ferred
to
a
Hybond-N+
membrane
(Amersham,
Arlington
Heights,
IL).
RNA
blot
hybridization
was
performed
with
a
32P-labeled
probe
derived
from
the
1.3-kb
EcoRI
insert
fragment
released
from
cDNA
clone
p8P2a37
or
from
the
ribosomal
protein
rPL32
as
an
internal
standard.3839
A
probe
that
hybridizes
to
the
ribozyme
was
derived
from
the
146-bp
insert
fragment
released
from
pRC/CMV
MMP-9
in
Prostate
Cancer
593
AJP
February
1998,
Vol.
152,
No.
2
0
Oo
C-
2
92
_
kDa
Primary
Metastatic
_.O
105
kDa
Figure
1.
Gelatinolytic
activity
is
present
in
conditioned
media
from
meta-
static
murine
prostate
carcinoma
cells.
Concentrated,
conditioned
medium
from
cells
derived
from
a
primary
prostate
carcinoma
148-1,PA
or
from
a
metastatic
tumor
148-1,LMD
were
subjected
to
gelatin
substrate
gel
electro-
phoresis.
The
cells
were
treated
for
24
hours
with
TGF-,3
or
with
TPA
as
indicated.
MMP-9
ribozyme
construct
by
digestion
with
Sacd
and
Apal.
Lung
Colonization,
Metastasis,
and
Tumorigenicity
Assays
Four-
to
six-week-old
NCr-nu/nu
mice
were
obtained
from
Taconic
Farms
(Germantown,
NY)
and
housed
asepti-
cally.
On
the
day
of
injection,
cells
were
washed
in
phos-
phate-buffered
saline,
trypsinized,
resuspended
in
se-
rum-free
Dulbecco's
modified
Eagle's
medium,
and
microscopically
examined
to
ensure
single
cell
suspen-
sions.
For
metastasis
studies,
mice
were
injected
in
the
lateral
tail
vein
with
5
x
104
single
cells/0.1
ml.
Animals
were
sacrificed
when
exhibiting
labored
breathing
or
at
4
weeks.
Lungs
were
fixed
in
10%
(w/v)
formalin.
A
dissect-
ing
microscope
was
used
to
count
the
lung
tumors
for
evidence
of
metastasis.
Tumorigenicity
was
assessed
after
injection
of
5
x
105
subcutaneously
in
the
flanks
of
4-
to
6-week-old
male
nu/nu
mice.
Six
tumors
were
mea-
sured
for
each
point.
The
tumor
volume
was
determined
by
measurement
of
the
tumor
in
three
dimensions
with
calipers
at
the
indicated
times.
Results
Cells
derived
either
from
a
primary
tumor
or
from
a
met-
astatic
tumor
generated
in
the
mouse
prostate
reconsti-
tution
system
were
evaluated
for
expression
of
MMP-9.3
Conditioned
medium
from
the
cells
isolated
from
a
pri-
mary
tumor
(148.1,PA)
showed
little
basal
expression
of
the
105-kd
gelatinase
activity
characteristic
of
murine
MMP-9,
and
there
was
no
increase
after
exposure
of
the
cells
to
either
TGF-,3
or
TPA
(Figure
1).
In
contrast,
the
conditioned
medium
from
the
cells
from
the
metastasis
(148.1,LMD)
contained
detectable
gelatinolytic
activity
at
105
kd
after
24
hours,
and
the
amount
of
gelatinase
released
increased
after
exposure
of
the
cells
to
either
TGF-,B
or
to
TPA.
The
conditioned
medium
from
the
pri-
mary
cells
had
a
high
level
of
gelatinolytic
activity
in
the
range
of
72
kd
likely
to
be
MMP-2,
but
this
activity
was
not
seen
in
the
medium
from
the
metastatic
cells.
Thus,
we
chose
to
use
the
148.1,LMD
prostatic
carcinoma
cells
isolated
from
a
metastasis
as
a
vehicle
to
test
whether
down-regulation
of
MMP-9
would
have
an
effect
on
met-
astatic
potential.
Hua
and
Muschel35
had
previously
shown
that
the
introduction
of
an
expression
vector
for
a
ribozyme
di-
rected
against
the
rat
MMP-9
could
down-regulate
MMP-9
expression
in
rat
cells.
This
ribozyme
consists
of
a
hammerhead-type
catalytic
site
flanked
by
15
bases
(8
on
the
5'
and
7
on
the
3'
side)
complementary
to
the
rat
MMP-9
mRNA.
Thirteen
of
these
15
bases
are
identical
between
rats
and
mice
suggesting
that
this
ribozyme
might
also
be
effective
against
murine
MMP-9.
After
transfection
of
this
vector
into
parental
148.1,LMD
cells,
11
transfected
clones
were
isolated;
six
showed
de-
creased
levels
of
released
MMP-9
gelatinase
activity.
Transfection
with
an
expression
vector
that
codes
for
a
ribozyme
hammerhead
structure
flanked
by
the
sense
MMP-9
sequence,
which
would
not
target
MMP-9,
was
used
as
a
control.
Of
the
six
clones
isolated
after
trans-
fection
with
the
control
ribozyme
expression
vector,
none
showed
decreased
levels
of
105-kd-released
gelatinase
activity
compared
with
the
parental
cells.
Two
clones
transfected
with
the
MMP-9
ribozyme
and
two
with
the
control
ribozyme,
were
selected
for
additional
analysis.
These
clones
were
shown
to
contain
the
plasmid
DNA
using
Southern
blotting
(data
not
shown).
Figure
2A
shows
that
mRNA
corresponding
to
the
ribozyme
could
be
detected
in
the
transfectants
but
not
in
the
parental
cells.
MMP-9
mRNA
could
not
be
detected
in
the
cells
transfected
with
the
MMP-9
ribozyme
under
the
same
conditions
in
which
MMP-9
mRNA
could
be
found
in
the
clones
transfected
with
the
control
ribozyme
(Figure
2B).
Increased
levels
of
MMP-9
mRNA
were
seen
in
the
con-
trol
cells
after
treatment
with
either
TGF-f
or
TPA,
but
MMP-9
mRNA
was
still
not
detected
in
either
of
the
two
clones
transfected
with
the
expression
vector
for
the
MMP-9
ribozyme.
Loading
in
each
lane
was
shown
to
be
equivalent
by
reprobing
the
blot
for
rPL32
mRNA,
a
gene
coding
for
a
ribosomal
protein.38'39
Thus,
introduction
of
the
ribozyme
directed
against
rat
MMP-9
resulted
in
de-
ceased
levels
of
MMP-9
mRNA.
The
decreased
levels
of
MMP-9
mRNA
in
cells
trans-
fected
with
the
ribozyme
against
MMP-9
were
reflected
in
decreased
levels
of
MMP-9
gelatinase
released
into
the
medium.
(Figure
3).
Gelatinase
activity
at
105
kd
as
de-
tected
by
substrate
gel
electrophoresis
indicated
that
the
parental
148.1,LMD
cells
and
the
control
transfectants
secreted
equivalent
amounts
of
105-kd
gelatinase
activ-
ity,
but
conditioned
medium
from
the
two
ribozyme
trans-
fected
clones
had
5-
to
10-fold
less.
After
stimulation
with
either
TGF-,B
or
TPA,
the
parental
and
control
cells
re-
leased
greater
amounts
of
MMP-9
activity
into
the
me-
dium.
The
ribozyme
transfected
cells
also
released
greater
amounts
of
gelatinase
activity
after
stimulation,
but
the
absolute
level
was
still
considerably
reduced
in
comparison
with
the
controls.
No
lower
molecular
mass
bands
indicative
of
activation
were
seen
in
any
case.
To
determine
the
effect
of
lowered
MMP-9
expression
on
the
behavior
of
these
prostatic
carcinoma
cells,
we
594
Sehgal
et
al
AJP
February
1998,
Vol.
152,
No.
2
UMMp-9
ribozyme
A
Ib
2
q
Control
ribozyme
r
Ribozyme
Probe
rpL
32
MMP-9
Control
B
nbozyme
nbozyme
1.51
1.52
1.74
1.22
O
t
.
L
.
.
L-
.
<-
MMP-9
L
F
F
EL
<
u:(1:
UL
c<
a.
uLc-
-S
Parent
1.51
1.22
(Ribozyme)
(Control)
Ca
(DO<
OI-H
lLCC
O
a.
-
F- F-
ec
-
F- F-
Parent
1.52
1.74
(Ribozyme)
(Control)
Figure
3.
Ninety-two-kd
gelatinase
activity
is
reduced
in
conditioned
me-
dium
from
ribozyme-transfected
cells.
Conditioned
medium
was
collected
from
cells
treated
with
the
indicated
agent
and
subjected
to
gelatin
substrate
gel
electrophoresis.
r
p
L
3
2
Figure
2.
The
MMP-9
ribozyme
results
in
decreased
MMP-9
mRNA
expres-
sion.
A:
Expression
of
the
ribozymes
in
transfected
cells.
Poly(A)
plus
RNA
was
isolated
from
the
parental
cells
148-1,
from
cells
transfected
with
the
control
ribozyme
1.22
and
1.74,
and
from
the
MMP-9
ribozyme
1.51
and
1.52.
The
upper
panel
shows
the
autoradiograph
after
hybridization
with
a
probe
derived
from
the
ribozyme
expression
vector,
whereas
the
lower
panel
shows
a
loading
control
with
the
same
blot
reprobed
using
rPL32.
B:
Ex-
pression
of
MMP-9
mRNA
in
the
ribozyme-transfected
cells.
Total
RNA
(10
,xg)
from
each
of
the
indicated
cells
treated
with
TGF-(3
or
with
TPA
were
subjected
to
Northern
blotting
with
an
MMP-9
probe.
The
upper
panel
shows
this
autoradiograph,
and
the
lower
panel
shows
the
autoradiograph
after
reprobing
with
a
loading
control
rPL32.
tested
their
metastatic
potential
using
the
lung
coloniza-
tion
assay
(Table
1).
After
injection
of
5
x
105
cells
into
the
tail
veins
of
female
nude
mice,
both
the
parental
cells
and
the
controls
gave
rise
to
lung
colonies
(22
to
31
per
mouse),
but
injection
of
the
same
number
of
MMP-9
ribozyme
transfected
cells
gave
rise
to
at
most
1
to
3
per
mouse.
Although
the
rasH
plus
myc-transformed
prostate
cells
derived
from
the
murine
prostate
reconstitution
sys-
tem
have
been
reported
to
be
androgen
independent,
we
also
wished
to
determine
whether
the
cells
would
re-
spond
differently
after
injection
in
a
male
mouse.40
The
experimental
metastasis
assay
was
performed
using
male
nude
mice.
In
this
case
the
controls
and
parental
cells
again
readily
gave
rise
to
lung
colonization,
from
28
to
35
nodules/mouse,
whereas
only
3
to
4
nodules/mouse
were
seen
after
injection
of
the
clones
with
down-regu-
lated
MMP-9.
Thus,
metastasis
was
inhibited
by
down-
regulation
of
MMP-9.
Tumor
growth
rate
was
evaluated
after
subcutaneous
injection
in
male
nude
mice
with
no
significant
differences
seen
regardless
of
their
MMP-9
expression
(Figure
4).
Discussion
The
metastatic
potential
of
the
prostate
cells
with
re-
duced
MMP-9
was
drastically
reduced
compared
with
either
control
transfected
clones
or
the
parental,
meta-
static
cells.
In
contrast,
tumor
growth
was
unaffected.
The
finding
that
MMP-9
contributes
to
metastasis
in
a
murine
prostate
model
system
leads
to
the
question
of
whether
MMP-9
might
be
involved
in
tumor
progression
in
human
Table
1.
Effect
of
MMP-9
Ribozyme
on
Experimental
Metastasis
Male
(lung
nodules/
Female
(lung
nodules/
Cell
type
Cell
lines
mouse
±
SD)
mouse
±
SD)
Parental
148-1,LMD
29
±
8.5
26.2
±
5.3
Control
ribozyme
AAS
1.22
37
±
11
22.2
±
6.7
AAS
1.74
38
±
8
31.8
±
9.9
MMP-9
Ribozyme
AR
1.51
3.25
±
2.5
0.63
±
0.3
AR
1.52
3.0
±
2.2
1.5
±
0.5
Cells
(5
x
104)
from
the
parental
population,
cells
from
two
MMP-9
ribozyme
transfected
clones
AR1.51
and
AR1.52,
and
from
two
control
transfectants
AAS
1.22
and
AAS
1.74
were
each
injected
intravenously
into
male
or
female
nude
mice.
After
4
weeks,
the
animals
were
sacrificed
and
the
number
of
lung
nodules
scored.
The
results
shown
are
from
one
experiment
with
five
mice
in
each
group.
Similar
results
were
obtained
in
two
repeat
experiments.
The
range
of
nodule
number
in
all
experiments
for
the
cells
in
the
parental
group
was
from
12
to
55,
for
the
control
ribozyme
cells
from
22
to
82,
and
for
the
MMP-9
ribozyme
cells
from
0
to
3.
The
difference
between
the
results
from
the
control
ribozyme
and
the
MMP-9
ribozyme
groups
is
significant
to
P
<
0.001
by
the
Mann-Whitney
test.
MMP-9
in
Prostate
Cancer
595
AJP
February
1998,
Vol.
152,
No.
2
I>---
1.74
Control
Ribozyme
0.8-
---0O----
1.22
---
1.51
MMP-9
-YD-w
---
1.52
Ribozyme
T
.2.
0.6
~~~~
Parental
E
cE
4
0
Tj
0
E
0.2-
2.5
5
7.5
10
12.5
15
Time
(days)
Figure
4.
Tumor
growth
rate
is
not
affected
by
MMP-9
expression.
The
diameter
of
the
tumors
formed
after
injection
of
the
indicated
cells
was
measured.
The
standard
deviation
is
shown
with
error
bars.
When
the
error
bars
cannot
be
seen,
they
were
smaller
than
the
diameter
of
the
data
point.
prostate
cancer.
Wood
et
al41
found
low
levels
of
MMP-9
mRNA
in
normal
prostate
tissue,
tissues
from
patients
with
benign
prostatic
hypertrophy,
or
carcinoma
with
a
low
Gleason
score
(5
or
less).
In
contrast,
MMP-9
mRNA
levels
were
highly
expressed
in
specimens
from
carcino-
mas
with
high
Gleason
scores,8-10
and
there
was
a
sig-
nificant
correlation
with
high
MMP-9
expression
and
local
invasion
as
well
as
survival.
Elevated
MMP-9
levels
were
found
in
cell
cultures
from
prostate
carcinomas
with
high
Gleason
scores,42
and
gelatinase
profiles
from
tissue
samples
revealed
a
correlation
with
MMP-9
gelatinase
activity
and
metastasis.43
These
data
are
consistent
with
MMP-9
filling
a
role
in
human
prostate
cancer
similar
to
its
role
in
the
mouse
reconstitution
model
of
prostate
cancer.
Other
MMPs
have
also
been
implicated
as
playing
a
role
in
the
behavior
of
prostate
carcinoma.
MMP-7
(ma-
trilysin
or
PUMP-1)
has
been
found
to
be
overexpressed
in
the
epithelial
cells
of
a
high
frequency
of
human
pros-
tate
carcinoma
specimens
and
prostatic
carcinoma
cell
lines.544
Transfection
of
an
expression
vector
for
matri-
lysin
into
the
prostatic
carcinoma
line
DU
145
resulted
in
increased
invasion
into
the
diaphragm
by
these
cells,
and
this
correlated
with
invasion
in
the
prostate
itself.
MMP-2
(gelatinase
A
or
72-kd
type
IV
collagenase)
is
also
overexpressed
in
prostatic
carcinoma
cells
indicat-
ing
that
it
too
may
play
an
important
role
in
the
biological
behavior
of
this
tumor.
Whether
TGF-,3
acts
in
vivo
to
stimulate
MMP-9
expres-
sion
and
thus
lead
to
metastatic
behavior
is
an
unre-
solved
issue
at
this
point.
TGF-,3
is
expressed
at
higher
levels
in
prostatic
carcinoma
specimens
from
patients
with
more
aggressive
disease.4548
It
has
also
been
re-
ported
that
latent
TGF-,j
binding
protein
is
absent
from
prostatic
carcinoma
specimens
but
present
in
normal
tissues
and
those
from
patients
with
benign
diseases.49
It
has
also
been
suggested
that
response
of
TGF-,3
recep-
tor
1
may
be
altered
in
some
prostatic
carcinomas
po-
tentially
leading
to
differential
responses
to
TGF-f3;
al-
though
this
does
not
appear
to
be
the
case
in
the
mouse
prostate
reconstitution
system
as
both
receptors
were
present
in
cells
derived
from
these
tumors,
and
TGF-f
led
to
equivalent
plasminogen
activator
inhibitor
1
stimulation
in
metastatic
and
nonmetastatic
cells.3,50
The
effects
of
TGF-p,
however,
must
be
pleiotrophic
as
its
effects
on
cell
growth
are
likely
to
be
independent
of
its
effects
on
MMP-9
production.
These
results
complement
our
previous
results
in
a
murine
sarcoma
model
system
in
which
down-regulation
of
MMP-9
also
led
to
a
decrease
in
metastatic
potential
without
a
decrease
in
tumorigenicity
or
tumor
growth
rate.
Whereas
it
has
often
been
assumed
that
MMPs
were
involved
in
metastasis,
these
studies
now
begin
to
pro-
vide
some
evidence
that
MMP-9
in
fact
does
play
a
role
in
metastasis.
As
MMP-9
is
overexpressed
in
many
hu-
man
tumor
types,
these
results
raise
the
possibility
that
MMP-9
expression
may
be
linked
to
tumor
progression
and
to
the
development
of
metastatic
potential
in
a
variety
of
human
cancers,
including
prostate
cancer,
and
that
MMP-9
expression could
be
a
potential
target
for
the
modulation
of
metastatic
potential.
References
1.
Thompson
T,
Southgate
J,
Kitchener,
Land
H:
Multistage
carcinogen-
esis
induced
by
ras
and
myc
oncogenes
in
a
reconstituted
organ.
Cell
1989,
56:917-930
2.
Thompson
TC,
Park
SH,
Timme
TL,
Ren
C,
Eastham
JA,
Donehower
LA,
Bradley
A,
Kadmon
D,
Yang
G:
Loss
of
p53
function
leads
to
metastasis
in
ras+myc-initiated
mouse
prostate
cancer.
Oncogene
1995,
10:869-879
3.
Sehgal
I,
Baley
PA,
Thompson
TC:
Transforming
growth
factor
p1
stimulates
contrasting
responses
in
metastatic
versus
primary
mouse
prostate
cancer
derived
cell
lines
in
vitro.
Cancer
Res
1996,
56:
3359-3365
4.
Stetler-Stevenson
WG,
Hewitt
R,
Corcoran
M:
Matrix
metalloprotein-
ases
and
tumor
invasion:
from
correlation
and
causality
to
the
clinic.
Semin
Cancer
Biol
1996,
7:147-154
5.
Powell
WC,
Knox
JD,
Navre
M,
Grogan
TM,
Kittelson
J,
Nagle
RB,
Bowden
GT:
Expression
of
the
metalloproteinase
matrilysin
in
DU-1
45
cells
increases
their
invasive
potential
in
severe
combined
immuno-
deficient
mice.
Cancer Res
1993,
53:417-422
6.
Birkedal-Hansen
H:
Proteolytic
remodeling
of
extracellular
matrix.
Curr
Opin
Cell
Biol
1995,
7:728-735
7.
Ochieng
J,
Fridman
R,
Nangia-Makker
P,
Kleiner
DE,
Liotta
LA,
Stetler-Stevenson
WG,
Raz
A:
Galectin-3
is
a
novel
substrate
for
human
matrix
metalloproteinases-2
and
-9.
Biochemistry
1994,
33:
14109-14114
8.
Gearing
AJ,
Beckett
P,
Christodoulou
M,
Churchill
M,
Clements
JM,
Crimmin
M,
Davidson
AH,
Drummond
AH,
Galloway
WA,
Gilbert
R,
et
al:
Matrix
metalloproteinases
and
processing
of
pro-TNF-a.
J
Leuko-
cyte
Biol
1995,
57:774-777
9.
Sires,
U.,
Griffin,
G.,
Broekelmann,
T.,
Mecham,
R.,
Murphy,
G.,
Chung,
A.,
Welgus,
H.,
and
RM,
S.:
Degradation
of
entactin
by
matrix
metalloproteinases.
J
Biol
Chem
1993,
268:2069-2074
10.
Tanaka
H,
Hojo
K,
Yoshida
H,
Yoshioka
T,
Sugita
K:
Molecular
cloning
and
expression
of
the
mouse
105-kd
gelatinase
cDNA.
Biochem
Biophys
Res
Commun
1993,
190:732-740
11.
Reponen
P,
Sahlberg
C,
Munaut
C,
Thesleff
I,
Tryggvason
K:
High
expression
of
92
kd
type
IV
collagenase
(gelatinase
B)
in
the
oste-
oclast
lineage
during
mouse
development.
J
Cell
Biol
1994,
124:
1091-1102
12.
Toth
M,
Gervasi
DC,
Fridman
R:
Phorbol
ester-induced
cell
surface
association
of
matrix
metalloproteinase-9
in
human
MCF10A
breast
epithelial
cells.
Cancer
Res
1997,
57:3159-3167
13.
Ogata
Y,
Itoh
Y,
Nagase
H:
Steps
involved
in
activation
of
the
pro-
matrix
metalloproteinase
9
(progelatinase
B)-tissue
inhibitor
of
met-
596
Sehgal
et
al
AJP
February
1998,
Vol.
152,
No.
2
alloproteinases-1
complex
by
4-aminophenylmercuric
acetate
and
proteinases.
J
Biol
Chem
1995,
270:18506-18511
14.
Ogata
Y,
Enghild
J,
Nagase
H:
Matrix
metalloproteinase
3
(stromely-
sin)
activates
the
precursor
for
the
human
matrix
metalloproteinase
9.
J
Biol
Chem
1992,
267:3581-3584
15.
Shapiro
SD,
Fliszar
CJ,
Broekelmann
TJ,
Mecham
RP,
Senior
RM,
Welgus
HG:
Activation
of
the
92-kd
gelatinase
by
stromelysin
and
4-aminophenylmercuric
acetate:
differential
processing
and
stabili-
zation
of
the
carboxyl-terminal
domain
by
tissue
inhibitor
of
metallo-
proteinases
(TIMP).
J
Biol
Chem
1995,
270:6351-6356
16.
Desrivieres
S,
Lu
H,
Peyri
N,
Soria
C,
Legrand
Y,
Menashi
S:
Activa-
tion
of
the
92
kd
type
IV
collagenase
by
tissue
kallikrein.
J
Cell
Physiol
1993,
157:587-593
17.
Fridman
R,
Toth
M,
Pena
D,
Mobashery
S:
Activation
of
progelatinase
B
(MMP-9)
by
gelatinase
A
(MMP-2).
Cancer
Res
1995,
55:
2548-2555
18.
Sang
QX,
Birkedal-Hansen
H,
Van
Wart
HE:
Proteolytic
and
non-
proteolytic
activation
of
human
neutrophil
progelatinase
B.
Biochim
Biophys
Acta
1995,
1251
:99-108
19.
Atkinson
SJ,
Ward
RV,
Reynolds
JJ,
Murphy
G:
Cell-mediated
deg-
radation
of
type
IV
collagen
and
gelatin
films
is
dependent
on
the
activation
of
matrix
metalloproteinases.
Biochem
J
1992,
288:
605-611
20.
Willenbrock
F,
Crabbe
T,
Slocombe
P,
Sutton
C,
Docherty
A,
Cockett
M,
O'Shea
M,
Brocklehurst
K,
Phillips
I,
Murphy
G:
The
activity
of
the
tissue
inhibitors
of
metalloproteinases
is
regulated
by
C-terminal
do-
main
interactions:
a
kinetic
analysis
of
the
inhibition
of
gelatinase
A.
Biochemistry
1993,
32:4330-4337
21.
O'Connell
J,
Willenbrock
F,
Docherty
A,
Eaton
D,
Murphy
G:
Analysis
of
the
role
of
the
COOH-terminal
domain
in
the
activation,
proteolytic
activity,
and
tissue
inhibitor
of
metalloproteinase
interactions
of
gela-
tinase
B.
J
Biol
Chem
1994,
269:14967-14973
22.
Strongin
A,
Collier
I,
Krasnov
P,
Genrich
T,
Marmer
B,
Goldberg
G:
Human
92
kd
type
IV
collagenase:
functional
analysis
of
fibronectin
and
carboxyl-end
domains.
Kidney
Int
1993,
43:158-162
23.
Goldberg
GI,
Strongin
A,
Collier
IE,
Genrich
LT,
Marmer
BL:
Interac-
tion
of
92-kd
type
IV
collagenase
with
the
tissue
inhibitor
of
metallo-
proteinases
prevents
dimerization,
complex
formation
with
interstitial
collagenase,
and
activation
of
the
proenzyme
with
stromelysin.
J
Biol
Chem
1992,
267:4583-4591
24.
DeClerck
YA,
Imren
S:
Protease
inhibitors:
role
and
potential
thera-
peutic
use
in
human
cancer.
Eur
J
Cancer
1994,
30A:2170-2180
25.
Yang
TT,
Hawkes
SP:
Role
of
the
21-kd
protein
TIMP-3
in
oncogenic
transformation
of
cultured
chicken
embryo
fibroblasts.
Proc
Natl
Acad
Sci
USA
1992,
89:10676-10680
26.
Pavloff
N,
Staskus
PW,
Kishnani
NS,
Hawkes
SP:
A
new
inhibitor
of
metalloproteinases
from
chicken:
ChIMP-3.
A
third
member
of
the
TIMP
family.
J
Biol
Chem
1992,
267:17321-17326
27.
Howard
EW,
Bullen
EC,
Banda
MJ:
Preferential
inhibition
of
72-
and
92-kd
gelatinases
by
tissue
inhibitor
of
metalloproteinases-2.
J
Biol
Chem
1991,
266:13070-13075
28.
Howard
EW,
Banda
MJ:
Binding
of
tissue
inhibitor
of
metalloprotein-
ases
2
to
two
distinct
sites
on
human
72-kd
gelatinase:
identification
of
a
stabilization
site.
J
Biol
Chem
1991,
266:17972-17977
29.
Greene
J,
Wang
M,
Liu
YE,
Raymond
LA,
Rosen
C,
Shi
YE:
Molecular
cloning
and
characterization
of
human
tissue
inhibitor
of
metallopro-
teinase
4.
J
Biol
Chem
1996,
271:30375-30380
30.
Leco
KJ,
Apte
SS,
Taniguchi
GT,
Hawkes
SP,
Khokha
R,
Schultz
GA,
Edwards
DR:
Murine
tissue
inhibitor
of
metalloproteinases-4
(Timp-4):
cDNA
isolation
and
expression
in
adult
mouse
tissues.
FEBS
Lett
1997,
401:213-217
31.
Murphy
G,
Willenbrock
F:
Tissue
inhibitors
of
matrix
metalloendopep-
tidases.
Methods
Enzymol
1995,
248:496-510
32.
Bernhard
E,
Muschel
R,
Hughes
E:
92
kd
gelatinase
release
corre-
lates
with
the
metastatic
phenotype
in
transformed
rat
embryo
cells.
Cancer
Res
1990,
50:3872-3877
33.
Yu
D,
Wang
SS,
Dulski
KM,
Tsai
CM,
Nicolson
GL,
Hung
MC:
c-erbB-
2/neu
overexpression
enhances
metastatic
potential
of
human
lung
cancer
cells
by
induction
of
metastasis-associated
properties.
Can-
cer
Res
1994,
54:3260-3266
34.
Bernhard
E,
Hagner
B,
Lubensky
I,
Wong
C,
Muschel
R:
The
effect
of
ElA
transfection
on
MMP-9
expression
and
metastatic
potential.
Int
J
Cancer
1995,
60:718-724
35.
Hua
J,
Muschel
RJ:
Inhibition
of
matrix
metalloproteinase
9
expres-
sion
by
a
ribozyme
blocks
metastasis
in
a
rat
sarcoma
model
system.
Cancer
Res
1996,
56:5279-5284
36.
Muschel
R,
Williams
J,
Lowy
D,
Liotta
L:
Harvey
ras
induction
of
metastatic
potential
depends
upon
oncogene
activation
and
the
type
of
recipient
cell.
Am
J
Pathol
1985,
121:1-8
37.
Bernhard
E,
Gruber
S,
Muschel
R:
Direct
evidence
linking
MMP-9
(92
kd
gelatinase/collagenase)
expression
to
the
metastatic
phenotype
in
transformed
rat
embryo
cells.
Proc
Natl
Acad
Sci
USA
1994,
91:
4293-4297
38.
Meyuhas
0,
Perry
RP:
Construction
and
identification
of
cDNA
clones
for
mouse
ribosomal
proteins:
applications
for
the
study
of
r-protein
gene
expression.
Gene
1980,
10:113-129
39.
Maity
A,
McKenna
WG,
Muschel
RJ:
Posttranscriptional
regulation
of
cyclin
Bi
mRNA
through
the
cell
cycle
and
after
irradiation.
EMBO
J
1994,
14:603-609
40.
Thompson
TC,
Egawa
S,
Kadmon
D,
Miller
GJ,
Timme
TL,
Scardino
PT,
Park
SH:
Androgen
sensitivity
and
gene
expression
in
ras
+
myc-induced
mouse
prostate
carcinomas.
J
Steroid
Biochem
Mol
Biol
1992,
43:79-85
41.
Wood
M,
Fudge
K,
Mohler
JL,
Frost
AR,
Garcia
F,
Wang
M,
Stearns
ME:
In
situ
hybridization
studies
of
metalloproteinases
2,
and
9,
and
TIMP-1,
and
TIMP-2
expression
in
human
prostate
cancer.
Clin
Exp
Metastasis
1997,
15:246-58
42.
Festuccia
C,
Bologna
M,
Vicentini
C,
Tacconelli
A,
Miano
R,
Violini
S,
Mackay
AR:
Increased
matrix
metalloproteinase-9
secretion
in
short-
term
tissue
cultures
of
prostatic
tumor
cells.
Int
J
Cancer
1996,
69:386-393
43.
Hamdy
FC,
Fadlon
EJ,
Cottam
D,
Lawry
J,
Thurrell
W,
Silcocks
PB,
Anderson
JB,
Williams
JL,
Rees
RC:
Matrix
metalloproteinase
9
ex-
pression
in
primary
human
prostatic
adenocarcinoma
and
benign
prostatic
hyperplasia.
Br
J
Cancer
1994,
69:177-182
44.
Pajouh
MS,
Nagle
RB,
Breathnach
R,
Finch
JS,
Brawer
MK,
Bowden
GT:
Expression
of
metalloproteinase
genes
in
human
prostate
can-
cer.
J
Cancer
Res
Clin
Oncol
1991,
117:144-150
45.
Merz
VW,
Miller
GJ,
Krebs
T,
Timme
TL,
Kadmon
D,
Park
SH,
Egawa
S,
Scardino
PT,
Thompson
TC:
Elevated
transforming
growth
fac-
tor-f1
and
83
mRNA
levels
are
associated
with
ras
+
myc-induced
carcinomas
in
reconstituted
mouse
prostate:
evidence
for
a
paracrine
role
during
progression.
Mol
Endocrinol
1991,
5:503-513
46.
Steiner
MS,
Zhou
ZZ,
Tonb
DC,
Barrack
ER:
Expression
of
transform-
ing
growth
factor-,B1
in
prostate
cancer.
Endocrinology
1994,
135:
2240-2247
47.
Glynne-Jones
E,
Harper
ME,
Goddard
L,
Eaton
CL,
Matthews
PN,
Griffiths
K:
Transforming
growth
factor
,B1
expression
in
benign
and
malignant
prostatic
tumors.
Prostate
1994,
25:210-218
48.
Merz
VW,
Arnold
AM,
Studer
UE:
Differential
expression
of
transform-
ing
growth
factor-p1
and
,B3
as
well
as
c-fos
mRNA
in
normal
human
prostate,
benign
prostatic
hyperplasia
and
prostatic
cancer.
World
J
Urol
1994,
12:96-98
49.
Eklov
S,
Funa
K,
Nordgren
H,
Olofsson
A,
Kanzaki
T,
Miyazono
K,
Nilsson
S:
Lack
of
the
latent
transforming
growth
factor
,3
binding
protein
in
malignant,
but
not
benign
prostatic
tissue.
Cancer
Res
1993,
53:3193-3197
50.
Kim
IY,
Ahn
HJ,
Zelner
DJ,
Shaw
JW,
Sensibar
JA,
Kim
JH,
Kato
M,
Lee
C:
Genetic
change
in
transforming
growth
factor
/3
(TGF-,3)
receptor
type
gene
correlates
with
insensitivity
to
TGF-31
in
human
prostate
cancer
cells.
Cancer
Res
1996,
56:44-48
... A number of MDEs such as urokinase plasminogen activator (uPA) and matrix metalloproteinases (MMPs) have been described [24]. Regulation of matrix-degrading activity is highly complex; no MDE is completely specific for one element of the ECM, and both uPA and MMPs have played a role in the necessary steps of tumour metastasis [24,[28][29][30][31][32][33][34][35][36][37][38]. ...
Article
Full-text available
We present two different methods to estimate parameters within a partial differential equation model of cancer invasion. The model describes the spatio-temporal evolution of three variables—tumour cell density, extracellular matrix density and matrix degrading enzyme concentration—in a one-dimensional tissue domain. The first method is a likelihood-free approach associated with approximate Bayesian computation; the second is a two-stage gradient matching method based on smoothing the data with a generalized additive model (GAM) and matching gradients from the GAM to those from the model. Both methods performed well on simulated data. To increase realism, additionally we tested the gradient matching scheme with simulated measurement error and found that the ability to estimate some model parameters deteriorated rapidly as measurement error increased.
... The invasive properties of tumor cells in the bone niche are closely linked to the expression of MMP-2 and MMP-9 metalloproteases stimulated by CXCL12/CXCR4 signaling, allowing their implantation in their new environment [32,132,133]. Therefore, inhibition of these metalloproteases reduces invasion and metastasis formation by prostate cancer cells in vivo [164]. ...
Article
Full-text available
Breast and prostate cancers have a great propensity to metastasize to long bones. The development of bone metastases is life-threatening, incurable, and drastically reduces patients’ quality of life. The chemokines CCL2 and CXCL12 and their respective receptors, CCR2 and CXCR4, are central instigators involved in all stages leading to cancer cell dissemination and secondary tumor formation in distant target organs. They orchestrate tumor cell survival, growth and migration, tumor invasion and angiogenesis, and the formation of micrometastases in the bone marrow. The bone niche is of particular importance in metastasis formation, as it expresses high levels of CCL2 and CXCL12, which attract tumor cells and contribute to malignancy. The limited number of available effective treatment strategies highlights the need to better understand the pathophysiology of bone metastases and reduce the skeletal tumor burden in patients diagnosed with metastatic bone disease. This review focuses on the involvement of the CCL2/CCR2 and CXCL12/CXCR4 chemokine axes in the formation and development of bone metastases, as well as on therapeutic perspectives aimed at targeting these chemokine-receptor pairs.
... These genes were upregulated in PCa of African-American patients and in EO-PCa 25 . CCR7 and MMP9 are genes associated with PCa progression and metastases [56][57][58] . MMP-9 is involved in several hallmarks of PCa progression, such as proliferation, angiogenesis, epithelial to mesenchymal transition, apoptosis, and metastasis 59 . ...
Article
Full-text available
The incidence of patients under 55 years old diagnosed with Prostate Cancer (EO-PCa) has increased during recent years. The molecular biology of PCa cancer in this group of patients remains unclear. Here, we applied weighted gene coexpression network analysis of the expression of miRNAs from 24 EO-PCa patients (38–45 years) and 25 late-onset PCa patients (LO-PCa, 71–74 years) to identify key miRNAs in EO-PCa patients. In total, 69 differentially expressed miRNAs were identified. Specifically, 26 and 14 miRNAs were exclusively deregulated in young and elderly patients, respectively, and 29 miRNAs were shared. We identified 20 hub miRNAs for the network built for EO-PCa. Six of these hub miRNAs exhibited prognostic significance in relapse‐free or overall survival. Additionally, two of the hub miRNAs were coexpressed with mRNAs of genes previously identified as deregulated in EO-PCa and in the most aggressive forms of PCa in African-American patients compared with Caucasian patients. These genes are involved in activation of immune response pathways, increased rates of metastasis and poor prognosis in PCa patients. In conclusion, our analysis identified miRNAs that are potentially important in the molecular pathology of EO-PCa. These genes may serve as biomarkers in EO-PCa and as possible therapeutic targets.
... Increased expression of MMP-2 in PCa cells is an independent predictor of shorter disease-free survival (125). Moreover, expression of MMP-9 in prostatic carcinoma cells results in reduced lung metastases but does not affect the tumor growth rate (121,126). The most common site of metastasis for PCa is the bone where MMP expression promotes metastatic seeding. ...
Chapter
Full-text available
Localized prostate cancer (confined to the gland) generally is considered curable, with nearly a 100% 5-year-survival rate. When the tumor escapes the prostate capsule, leading to metastasis, there is a poorer prognosis and higher mortality rate, with 5-year survival dropping to less than 30%. A major research question has been to understand the transition from indolent (low risk) disease to aggressive (high risk) disease. In this chapter, we provide details of the changing tumor microenvironments during prostate cancer invasion and their role in the progression and metastasis of lethal prostate cancer. Four microenvironments covered here include the muscle stroma, perineural invasion, hypoxia, and the role of microvesicles in altering the extracellular matrix environment. The adaptability of prostate cancer to these varied microenvironments and the cues for phenotypic changes are currently understudied areas. Model systems for understanding smooth muscle invasion both in vitro and in vivo are highlighted. Invasive human needle biopsy tissue and mouse xenograft tumors both contain smooth muscle invasion. In combination, the models can be used in an iterative process to validate molecular events for smooth muscle invasion in human tissue. Understanding the complex and interacting microenvironments in the prostate holds the key to early detection of high-risk disease and preventing tumor invasion through escape from the prostate capsule.
... The results clearly showed that there was a significant inhibition of MMP-9 than MMP-2 in both tests. In murine prostate cancer cells the hematogenous metastasis has been affected by the expression of these MMP-9 (Sehgal et al., 1998)). Therefore, cucurmin by decreasing these MMPs particularly by reducing the activity of MMP-9 helps to prevent the pulmonary metastasis . ...
... The results clearly showed that there was a significant inhibition of MMP-9 than MMP-2 in both tests. In murine prostate cancer cells the hematogenous metastasis has been affected by the expression of these MMP-9 (Sehgal et al., 1998)). Therefore, cucurmin by decreasing these MMPs particularly by reducing the activity of MMP-9 helps to prevent the pulmonary metastasis (Guo et al., 2015). ...
Article
Cucurmin, a naturally yellow component isolated from turmeric, ability to prevent various life-style related disorders. The current review article mainly emphasizes on different anticancer perspectives of cucurmin i.e. colon, cervical, uterine, ovarian, prostate head and neck, breast, pulmonary, stomach and gastric, pancreatic, bladder oral, oesophageal and bone cancer. It holds a mixture of strong bioactive molecule known as cucurminoids that has ability to reduce cancer/tumor at initial, promotion and progression stages of tumor development. In particular, these compounds block several enzymes required for the growth of tumors and may therefore involve in tumor treatments. Moreover, it modulates an array of cellular progressions i.e. nitric oxide synthetase activity, protein kinase C activity, epidermal growth factor (EGF) receptor intrinsic kinase activity, nuclear factor kappa (NF-kB) activity, inhibiting lipid peroxidation and production of reactive oxygen species. However, current manuscript summarizes most of the recent investigations of cucurmin but still further research should be conducted to explore the role of curcumin to mitigate various cancers.
Article
Full-text available
Prostate cancer remains a leading cause of cancer-related morbidity in men. Potentially important regulators of prostate cancer progression are members of the metzincin superfamily of proteases, principally through their regulation of the extracellular matrix. It is therefore timely to review the role of the metzincin superfamily in prostate cancer and its progression to better understand their involvement in this disease. A systematic-like search strategy was conducted. Articles that investigated the roles of members of the metzincin superfamily and their key regulators in prostate cancer were included. The extracted articles were synthesized and data presented in tabular and narrative forms. Two hundred and five studies met the inclusion criteria. Of these, 138 investigated the role of the Matrix Metalloproteinase (MMP) subgroup, 34 the Membrane-Tethered Matrix Metalloproteinase (MT-MMP) subgroup, 22 the A Disintegrin and Metalloproteinase (ADAM) subgroup, 8 the A Disintegrin and Metalloproteinase with Thrombospondin Motifs (ADAMTS) subgroup and 53 the Tissue Inhibitor of Metalloproteinases (TIMP) family of regulators, noting that several studies investigated multiple family members. There was clear evidence that specific members of the metzincin superfamily are involved in prostate cancer progression, which can be either in a positive or negative manner. However, further understanding of their mechanisms of action and how they may be used as prognostic indicators or molecular targets is required.
Article
Serum amyloid A (SAA) concentration and plasma matrix metalloproteinase-9 (MMP-9) levels are increased in cats with lymphoma. In the present study, the association between SAA and MMP-9 production was evaluated using recombinant feline SAA (rfSAA) and three feline lymphoma-derived cell lines: 3201, MS4, and MCC. MMP-9 mRNA expression was significantly increased by rfSAA stimulation only in MCC cells. Secreted MMP-9 protein in culture media was confirmed by gelatin zymography, with clear bands of MMP-9 detected in MCC cells following rfSAA stimulation. A significant increase in semi-quantified MMP-9 levels was observed with 5 and 25 μg/ml of rfSAA stimulation. The infiltrative activities of feline lymphoma cells, assessed by the matrigel transwell assay, showed that rfSAA stimulated cell infiltration in MCC cells, in addition to MMP-9 expression. Although the response to rfSAA stimulation varied between cell lines, the results showed that rfSAA can stimulate MMP-9 production and infiltration of feline lymphoma-derived cells. The findings of this study have identified a novel role for SAA in the progression of some forms of feline lymphoma.
Article
Full-text available
Secreted metalloproteases initiating proteolytic degradation of collagens and proteoglycans play a critical role in remodeling of the connective tissue. Activation of the secreted proenzymes and interaction with their specific inhibitors TIMP and TIMP-2 are responsible for regulation of enzyme activity in extracellular space. We have previously demonstrated that 92- and 72-kDa Type IV procollagenases, in contrast to interstitial collagenase (ClI), form specific complexes with TIMP and the related inhibitor TIMP-2, respectively. The physiologic significance of the proenzyme-inhibitor complex and the mechanism of activation of Type IV collagenases remained unclear. Here, we demonstrate that in the absence of TIMP, 92-kDa Type IV procollagenase (92T4Cl) can form a covalent homodimer and a novel complex with ClI. In the presence of TIMP, the formation of a 92T4Cl proenzyme complex with TIMP prevents dimerization, formation of the complex with ClI, and activation of the 92T4Cl proenzyme by stromelysin, a related metalloprotease. The proenzyme homodimer is unable to form a complex with TIMP. All TIMP-free forms of the proenzyme can be activated by stromelysin. The 92T4Cl-ClI complex can be activated to yield a complex active against both gelatin and fibrillar Type I collagen, suggesting a mechanism for cooperative action of two enzymes in reducing collagen fibrils to small peptides under physiologic conditions.
Article
Full-text available
Matrix metalloproteinase 9 (MMP-9), also known as 92-kDa gelatinase/type IV collagenase, is secreted from neutrophils, macrophages, and a number of transformed cells in zymogen form. Here we report that matrix metalloproteinase 3 (MMP-3/stromelysin) is an activator of the precursor of matrix metalloproteinase 9 (proMMP-9). MMP-3 initially cleaves proMMP-9 at the Glu40-Met41 bond located in the middle of the propeptide to generate an 86-kDa intermediate. Cleavage of this bond triggers a change in proMMP-9 that renders the Arg87-Phe88 bond susceptible to the second cleavage by MMP-3, resulting in conversion to an 82-kDa form. alpha 2-Macroglobulin binding studies of partially activated MMP-9 demonstrate that the 82-kDa species is proteolytically active, but not the initial intermediate of 86 kDa. This stepwise activation mechanism of proMMP-9 is analogous to those of other members of the MMP family, but the action of MMP-3 on proMMP-9 is the first example of zymogen activation that can be triggered by another member of the MMP family. The results imply that MMP-3 may be an effective activator of proMMP-9 in vivo.
Article
Full-text available
We report cDNA cloning and primary structure of a new metalloproteinase inhibitor (ChIMP-3) produced by chicken embryo fibroblasts. ChIMP-3, formerly called the 21-kDa protein, is one of five ChIMPs (Chicken Inhibitor of MetalloProteinases). In this paper, we report that of the three most abundant ChIMPs, ChIMP-3 and ChIMP-a are extracellular matrix components, whereas ChIMP-2 is found in the media conditioned by the cells. Treatment of ChIMP-3 and ChIMP-a with N-glycosidase-F indicates that ChIMP-a is N-glycosylated whereas ChIMP-3 is not. The deduced amino acid sequence of ChIMP-3 predicts a protein whose properties are consistent with experimental measurements. Analysis of sequence alignments with the two previously described members of the TIMP (tissue inhibitor of metalloproteinases) family, TIMP-1 and TIMP-2, from various species indicates that ChIMP-3 is a related but distinct protein. This conclusion is supported by lack of significant binding with anti-TIMP-1 and anti-TIMP-2 antibodies. Based on these data, its unusual solubility properties, and its exclusive location in the matrix, we propose that ChIMP-3 is a new member of this family of metalloproteinase inhibitors, a TIMP-3.
Article
Full-text available
Transformed human fibroblasts secrete two structurally and functionally related inhibitors of matrix metalloproteinases, tissue inhibitor of metalloproteinases (TIMP) 1 and 2. In assays measuring the relative inhibitory capability of TIMP-1 and TIMP-2 against autoactivated 72-kDa gelatinase, which consists of two major active peptides and several inactive fragments, TIMP-2 was more effective than TIMP-1. The isolated 42.5-kDa active fragment that formed as a result of the autoactivation of 72-kDa gelatinase showed the greatest preference for TIMP-2; at half-maximal inhibition, TIMP-2 was greater than 10-fold more effective than TIMP-1. TIMP-2 was also greater than 2-fold more effective than TIMP-1 at inhibiting 72-kDa gelatinase-TIMP-2 complexes activated with 4-aminophenylmercuric acetate, and greater than 7-fold more effective than TIMP-1 at inhibiting 92-kDa gelatinase activated with 4-aminophenylmercuric acetate. Furthermore, these active gelatinases preferentially bound 125I-TIMP-2 when incubated with equal amounts of radiolabeled TIMP-1 and TIMP-2. The ratios of 125I-TIMP-2/125I-TIMP-1 binding to 92-kDa gelatinase, autoactivated 72-kDa gelatinase, and 42.5-kDa fragment were 4.4, 10, and 33, respectively. On the other hand, interstitial collagenase was inhibited by TIMP-1 greater than 2-fold more effectively than TIMP-2 in assays measuring cleavage of loose collagen fibrils.
Article
Full-text available
We have identified a binding site for tissue inhibitors of metalloproteinases 2 (TIMP-2) on human 72-kDa gelatinase that is distinct from the active site. 72-kDa progelatinase is found in a complex with TIMP-2 in the medium of cultured cells and can be activated with organomercurial compounds to yield a gelatinolytic proteinase that remains bound to TIMP-2. Removal of TIMP-2 from 72-kDa progelatinase by reverse-phase high performance liquid chromatography, followed by reconstitution of the progelatinase in neutral pH buffer, results in autocatalytic activation. When samples of autoactivated gelatinase were blotted onto nitrocellulose, then probed with 125I-TIMP-2, we found a 29-kDa peptide that was capable of binding TIMP-2. We isolated this fragment and identified it as the region of gelatinase from amino acid 414 to the carboxyl terminus in the primary amino acid sequence of progelatinase. This portion of the molecule does not contain the putative zinc- or gelatin-binding sites and is proteolytically inactive. Incubation of 125I-TIMP-2 with 72-kDa progelatinase-TIMP-2 complexes resulted in a concentration-dependent exchange of labeled TIMP-2 with unlabeled TIMP-2, in both the presence and absence of the metalloproteinase inhibitor 1,10-phenanthroline. Saturation binding kinetics for the active site of 72-kDa gelatinase were measured in pools of the 43-kDa active fragment that results from the autoactivation of 72-kDa progelatinase; this fragment has no carboxyl-terminal TIMP-2 binding capability. Binding of 125I-TIMP-2 to the active site was completely inhibited by 1,10-phenanthroline. Binding kinetics for the putative stabilization site were determined with isolated 72-kDa progelatinase. In the presence of 1,10-phenanthroline, 72-kDa progelatinase bound 125I-TIMP-2 but not 125I-TIMP-1. Scatchard analysis yielded an approximate dissociation constant (Kd) of 0.72 nM for the active site and 0.42 nM for the stabilization site.
Article
Twenty-five surgical specimens of malignant human prostate, 3 lymph nodes with metastatic prostate carcinoma, 11 normal human prostates, as well as 3 human prostate cell lines (DU-145, PC3 and LNCaP) were examined for the expression of the human matrix metalloproteinase-7 gene (MMP-7) from the human collagenase family (originally called PUMP-1 for putative metalloproteinase-1) [Quantin et al. (1989) Biochemistry 28:5327–5334; Muller et al. (1988) Biochem J 253:187–192; Matrisian and Bowden (1990) Semin Cancer Biol 1:107–115]. Northern blots were prepared using total RNA extracted from 18 prostate adenocarcinomas, 2 lymph nodes with metastatic prostate carcinoma and 11 normal human prostates. When the northern blots were hybridized with a32P-labeledMMP-7 cDNA probe, a 1.2-kb mRNA was detected in 14 out of 18 prostate adenocarcinomas, 1 out of 2 metastatic lymph nodes, and 3 out of 11 normal prostates. The 3 human prostate cell lines did not show any evidence of theMMP-7 transcript. In situ hybridization was conducted to localize theMMP-7 mRNA to individual cells using a35S-labeledMMP-7 cRNA. In situ hybridization was carried out on snap-frozen tissue sections of 7 prostate adenocarcinomas and 3 lymph nodes containing metastatic prostate adenocarcinoma using the same tissues previously probed by northern analysis as well as new samples. In situ hybridization revealed that theMMP-7 gene was expressed in the epithelial cells of primary prostate adenocarcinoma as well as in invasive and metastatic cells.MMP-7 expression was also seen focally in some dysplastic glands but not in stroma. Additional northern blot analysis was performed using probes to human type-IV collagenase, type-I collagenase and stromelysin I in human prostate adenocarcinoma as well as normal prostate tissue. Our results indicated that 6 out of 10 adenocarcinoma samples and none of the 4 normal samples were positive for type-IV collagenase transcripts. Tissue samples were also examined for the expression of type-I collagenase (9 adenocarcinomas and 4 normal) and stromelysin I (13 adenocarcinomas) by northern analysis. None of the tissues was found to express the transcripts of interest at detectable levels. These data suggest that certain metalloproteinases are present in prostatic adenocarcinoma and may play a role in invasion and metastasis.
Article
A set of recombinant DNAs with inserts of mRNA sequences coding for various mouse ribosomal proteins (r- proteins) were isolated. The recombinants were selected from a library of cDNA clones representing the small (⩽12S) poly(A)+ mRNA of mouse L cells, which is relatively enriched for the r-protein mRNAs. Selection consisted of testing the various recombinant plasmids for their ability to hybridize selectively with an mRNA that directs the synthesis of a recognizable r-protein in a cell-free translational system. Translation products were subjected to a preliminary screen by electrophoresis on one-dimensional Triton X-100/urea slab gels and then more definitively identified by the two-dimensional gel analyses conventionally used for ribosomal proteins. The set of recombinants represents the mRNAs specifying ribosomal proteins S16, L7, L13, L18, L19, L30, L32/33 and probably L10.
Article
The 21-kDa protein is an extracellular matrix (ECM) component whose synthesis is stimulated transiently during oncogenic transformation of chicken embryo fibroblasts (CEF) or after treatment of normal cells with the tumor promoter phorbol 12-myristate 13-acetate. Biochemical characterization indicates that the protein is related, but not identical, to two members of the family of tissue inhibitors of metalloproteinases, TIMP-1 and TIMP-2. The cDNA of the 21-kDa protein was recently cloned, and based upon its deduced amino acid sequence and other supporting data we propose that it is another member of this family, a TIMP-3. We now report electrophoretic purification of sufficient quantities of this protein to determine its function. The protein promotes the detachment of transforming cells from the ECM. Although its presence in the matrix may be necessary for cell release it is not the only factor involved because it does not influence the adhesive properties of nontransformed cells. It also appears to accelerate the morphological changes associated with cell transformation and stimulates the proliferation of growth-retarded, nontransformed cells maintained under low serum conditions. Based on these data we hypothesize that the 21-kDa protein promotes the development of the transformed phenotype in cultured cells.
Article
The ability of normal rabbit dermal fibroblasts to degrade films of type IV collagen and gelatin when stimulated by phorbol ester was shown to be dependent on the induction, secretion and activation of 95 kDa gelatinase B and the secretion and activation of 72 kDa gelatinase A and stromelysin. Degradation was inhibited by exogenous human recombinant tissue inhibitor of metalloproteinases-1, specific antibodies to gelatinase and stromelysin and by the reactive-oxygen-metabolite inhibitor catalase. We discuss the various pathways for activation of matrix metalloproteinases in this model system and conclude that, although plasmin may play a key role in the activation of gelatinase B and stromelysin, gelatinase A is activated by a mechanism which has yet to be elucidated. The involvement of oxygen radicals in the direct activation of matrix metalloproteinases in this model is thought to be unlikely.
Article
We established an androgen-sensitive cell line (BR31-5) from a ras + myc-induced mouse prostate carcinoma and used this cell line together with a previously reported transplantable androgen-independent mouse prostate carcinoma to investigate patterns of expression for apoptosis-related genes in an androgen-deprived environment. Single cell suspensions derived from the BR31-5 cell line were inoculated into the flank of intact or castrated adult male C57BL/6 mice and tumors were harvested 12 days post-inoculation for Northern blotting. A transplantable androgen-independent prostate cancer was also inoculated into intact or castrated mice and tumors harvested 21 days later. Tumor volume analyses showed that BR31-5 carcinomas were androgen-sensitive. Northern blotting showed that mRNA levels for two apoptosis-related genes, transforming growth factor-beta 1 and c-myc, were significantly elevated to a similar extent in carcinomas grown in castrated hosts compared to intact hosts for both the androgen-sensitive BR31-5 and androgen-independent carcinomas. Levels of mRNA for tissue type plasminogen activator, shown previously to be elevated in androgen-independent carcinomas following growth in castrates, were also increased in BR31-5 carcinomas under similar androgen-deprived conditions but to a lesser extent. Interestingly, testosterone repressed prostate mRNA No. 2 levels shown previously to be similar in both the intact and castrated groups for androgen-independent carcinomas were significantly increased in the castrated group compared to the intact group for BR31-5 carcinomas. Therefore, specific patterns of expression for apoptosis-related genes may be able to discriminate androgen-sensitive and androgen-independent prostate cancer under androgen-deprived conditions.