ArticlePDF Available

Maspin is angiogenesis inhibitor

Authors:

Abstract and Figures

Maspin, a unique member of the serpin family, is a secreted protein encoded by a class II tumor suppressor gene whose downregulation is associated with the development of breast and prostate cancers. Overexpression of maspin in breast tumor cells limits their growth and metastases in vivo. In this report we demonstrate that maspin is an effective inhibitor of angiogenesis. In vitro, it acted directly on cultured endothelial cells to stop their migration towards basic fibroblast growth factor and vascular endothelial growth factor and to limit mitogenesis and tube formation. In vivo, it blocked neovascularization in the rat cornea pocket model. Maspin derivatives mutated in the serpin reactive site lost their ability to inhibit the migration of fibroblasts, keratinocytes, and breast cancer cells but were still able to block angiogenesis in vitro and in vivo. When maspin was delivered locally to human prostate tumor cells in a xenograft mouse model, it blocked tumor growth and dramatically reduced the density of tumor-associated microvessels. These data suggest that the tumor suppressor activity of maspin may depend in large part on its ability to inhibit angiogenesis and raise the possibility that maspin and similar serpins may be excellent leads for the development of drugs that modulate angiogenesis.
Content may be subject to copyright.
196 NATURE MEDICINE VOLUME 6 NUMBER 2 FEBRUARY 2000
ARTICLES
Maspin, a unique member of the serpin family, is a secreted
protein encoded by a class II tumor suppressor gene whose
downregulation is associated with the development of breast
and prostate cancers1,2. Overexpression of maspin in breast
tumor cells limits their growth and metastases in vivo. In this
report we demonstrate that maspin is an effective inhibitor of
angiogenesis. In vitro, it acted directly on cultured endothelial
cells to stop their migration towards basic fibroblast growth
factor and vascular endothelial growth factor and to limit mi-
togenesis and tube formation. In vivo, it blocked neovascular-
ization in the rat cornea pocket model. Maspin derivatives
mutated in the serpin reactive site lost their ability to inhibit
the migration of fibroblasts, keratinocytes, and breast cancer
cells but were still able to block angiogenesis in vitro and in
vivo. When maspin was delivered locally to human prostate
tumor cells in a xenograft mouse model, it blocked tumor
growth and dramatically reduced the density of tumor-associ-
ated microvessels. These data suggest that the tumor suppres-
sor activity of maspin may depend in large part on its ability to
inhibit angiogenesis and raise the possibility that maspin and
similar serpins may be excellent leads for the development of
drugs that modulate angiogenesis.
To study the potential anti-angiogenic properties of maspin,
the mouse protein was produced in Escherichia coli as a recom-
binant glutathione S-transferase (GST) fusion protein and
tested in a variety of angiogenesis assays. Recombinant maspin
blocked endothelial cell migration induced by vascular en-
dothelial growth factor (VEGF) in a dose dependent manner
with a median effective dose (ED50) of 0.2 µM–0.3 µM (Fig. 1a).
Similar results were obtained using basic fibroblast growth fac-
tor (bFGF) as an inducer (Fig. 1b; data not shown). At 1 µM,
maspin completely blocked the response of the endothelial
cells to both angiogenic inducers, whereas the GST control was
inactive. Maspin also inhibited the growth of endothelial cells
(Fig. 1c) and prevented them from forming tubes in a matrigel
assay (data not shown).
Purified maspin effectively inhibited neovascularization
in vivo. Rat corneas were surgically implanted with non-
inflammatory, slow release pellets containing maspin with
bFGF and examined 6 or 7 days later for ingrowth of vessels.
Maspin completely blocked bFGF-induced neovascularization
(Fig. 2; compare i and v).
Maspin, a member of the large family of serine protease in-
hibitors (serpins), has been shown to serve as a substrate rather
than an inhibitor for trypsin-like serine proteinases3, suggest-
ing that it may fall into the growing category of noninhibitory
serpins that lack antiprotease activity. One recent study, using
purified reagents in vitro, suggested that maspin might have
some antiprotease activity as it inhibited tissue plasminogen
activator in vitro4. We were unable to confirm these results in
our laboratory5, but to determine if the anti-angiogenic activity
of maspin depended on the inhibition of some undefined pro-
tease, we constructed, expressed, and tested several mutants.
The RSL (reactive serpin loop) near the C-terminus of serpin
family members is essential for their antiprotease activity.
Mutations at the RSL region of other serpins, especially at the
P1 site, abolish serpin activity6. To disrupt this loop in maspin
two different mutations were introduced in the RSL region: a
C-terminal deletion downstream of P7residue7(maspin RSL)
and a conversion of the critical P1 arginine of the RSL loop to a
glutamine (maspin*). A third mutant was constructed in which
the first 139 amino acids were removed but the serpin region
was left intact (maspinN). We tested recombinant maspin and
two RSL mutants in a quantitative tissue-type plasminogen
activator (tPA) assay. As previously demonstrated5, neither
wild-type maspin nor its derivatives displayed any tPA inhibi-
tion (data not shown).
We used migration assays, with nonendothelial cells, to
demonstrate that the constructed mutants were indeed defec-
tive. An intact RSL region is essential for maspin to block the
migration of breast tumor cells in vitro8. When we repeated this
assay with mutant proteins both maspin* and maspinN had
no inhibitory effect (Fig. 1d). All three mutants were also un-
able to inhibit the migration towards bFGF of normal human
fibroblasts (Fig. 1e) or normal human keratinocytes (Fig. 1f)
although wild-type maspin protein produced in the same way
was an effective inhibitor.
When we tested these defective mutants on endothelial cells
(see Fig. 1b and c), they behaved somewhat differently. Those
with RSL defects that were unable to block the migration of
other cells, retained the ability to inhibit endothelial cell mi-
gration and mitogenesis. Protein with mutations in the RSL re-
gion also retained the ability to inhibit neovascularization in
vivo (Fig. 2, compare i and vii). The N-terminal deletion,
maspinN, was defective in all assays so it was not possible to
determine if a crucial active region had been deleted or if it was
just a dead protein.
To determine if the ability of maspin to inhibit angiogenesis
is involved in its antitumor activity, we used an athymic mouse
xenograft model. We implanted LNCaP prostate tumor cells
subcutaneously on the bidorsal back of nude mice and moni-
tored tumor growth and neovascularization after systemic
treatment with exogenous maspin. Maspin-treated tumors con-
tained considerably fewer vessels as determined by CD31 im-
munostaining than GST-treated controls (Fig. 3) To determine
whether maspin effects on the tumor-induced vasculature were
maintained during a more prolonged treatment, the above ex-
Maspin is an angiogenesis inhibitor
MING ZHANG1, OLGA VOLPERT2, YIHUI H. SHI1, AND NOËL BOUCK2
1Department of Molecular & Cellular Biology, Baylor College of Medicine, Houston, Texas 77030, USA
2Department of Microbiology-Immunology and Robert H. Lurie Comprehensive Cancer Center, Northwestern
University Medical School, Chicago, Illinois 60611, USA
Correspondence should be addressed to M.Z.; email: mzhang@bcm.tmc.edu
© 2000 Nature America Inc. • http://medicine.nature.com
© 2000 Nature America Inc. • http://medicine.nature.com
NATURE MEDICINE VOLUME 6 NUMBER 2 FEBRUARY 2000 197
ARTICLES
periment was replicated with tumors harvested after 7–8 weeks.
We treated 32 tumor sites with maspin and 37 with GST. At
week 8, the growth of 53% of the maspin-treated tumors had
been completely inhibited. The remaining 15 maspin-treated
tumors were reduced in size to 29.2% of GST control-treated tu-
mors. The effect of maspin was reversible. Among those sites
that exhibited no detectable tumors at week 8, most developed
palpable tumors within 1–3 weeks after cessation of maspin
treatment, indicating that some viable tumor cells remained.
To determine if the reduced size of maspin-treated tumors
coincided with reduced neovascularization, we used 20 repre-
sentative tumors from either maspin-treated (10 sites) or GST-
treated tumors (10 sites) to quantify the density of microvessels
after immunostaining with CD31 antibody (Fig. 3 and Table1).
The density of vessels in maspin-treated tumors was
reduced to 37.8% of control tumors and this differ-
ence was highly significant. We also compared the
treated and control tumors of similar size. There was
also reduction of vessel density (ranging from 33.3%
to 45.5%, n = 4) in the maspin treated samples.
There is little evidence to indicate exactly how
maspin blocks angiogenesis. It could act through a re-
ceptor-mediated event, as does thrombospondin-19.
The discrepancy in the effect of the RSL domain on en-
dothelial and breast tumor cells may be due to the
difference of receptors located on both cells.
Alternatively, it could mimic the protease-indepen-
dent effects of plasminogen activator inhibitor-1 (PAI-
1). PAI-1 has a unique domain located at the
N-terminal that regulates cell motility10. Experiments
are underway to investigate this domain of maspin.
The observed ED50 of 0.2–0.3 µM in the capillary en-
dothelial cell migration assay indicates that maspin is
less potent than inhibitors like angiostatin (10 nM)11,
but more effective than small molecules such as cap-
topril (10 µM)12. However, in epithelial tumor cells,
exogenously added maspin localizes at the cell sur-
face13. If this also happens on endothelial cells, then
the concentration of soluble protein may not be par-
ticularly meaningful. Maspin was effective against
several inducers in vitro, and in vivo it blocked angio-
Fig. 1 Effect of maspin and its derivatives
on cultured cells. a,GST–maspin was
tested at a range of concentrations for its
ability to inhibit endothelial cell migration
induced by 100 pg/ml VEGF. VEGF, migra-
tion towards VEGF alone; BSA, background
migration in the absence of a gradient.
b, GST–maspin and its mutants (1 µM)
were tested for their ability to inhibit en-
dothelial cell migration towards 10 ng/ml
bFGF. Maspin, GST–maspin fusion protein;
maspin RSL, maspin with a deletion at the
C-terminus; maspin*, maspin containing an
R to Q mutation in the P1 residue of the RSL
loop; maspin N, maspin with a deletion at
the N-terminus. Glutathione-S-transferase
tested alone was neutral in this assay. *, P <
0.01 compared with migration towards
bFGF (bFGF-----). c, Maspin and its mutants
(1 µM) were tested for their ability to in-
hibit the growth of endothelial cells after
5 d. *, P < 0.05 compared with GST.
d, Maspin and its mutants were tested at a
concentration of 0.3 µM for their ability to
inhibit the invasion and migration of breast
tumor cells. *, P < 0.03. e, Maspin and its
mutants were tested at a concentration of 1
µM for their ability to inhibit the migration of normal human fibroblasts
towards bFGF (20 ng/ml). *, P < 0.05. f, Maspin and its mutants were
Fig. 2 Maspin inhibition of corneal neovascularization. Pellets containing 10 µM test
substances at with or without 100 ng/ml bFGF were incorporated into Hydron slow-
release pellets and implanted in rat corneas. After 6 or 7 d, rats were perfused with col-
loidal carbon to visualize vessels, and excised corneas were photographed with a ×20
objective (b) and scored for neovascularization (a). +/–, corneas in which only one or two
vessels were induced that did not reach the pellet.
tested for their ability to block the migration of normal human
keratinocytes towards 20 ng/ml bFGF. *, P < 0.03.
a b
c
d
a b
e f
v
i
vii
ix
i
ii
iii
iv
v
vi
vii
viii
ix
© 2000 Nature America Inc. • http://medicine.nature.com
© 2000 Nature America Inc. • http://medicine.nature.com
198 NATURE MEDICINE VOLUME 6 NUMBER 2 FEBRUARY 2000
ARTICLES
genesis induced by bFGF and reduced tumor
angiogenesis in response to the LNCaP, a cell
line that produces VEGF as its major angio-
genic factor (J. D. and N. B., unpublished data).
Complete inhibition of endothelial cell mi-
gration in vitro was achieved between 0.5 and 1
µM, in the same concentration range where
maspin also inhibits tumor cell motility and in-
vasion13, but the mechanisms underlying these
two maspin activities seem to be different. The
former requires that the protein have an intact
RSL (refs. 4, 8), whereas this feature was not es-
sential for the inhibition of angiogenesis. Thus,
even if maspin does inhibit some unidentified
protease, this serpin activity is probably not in-
volved in the inhibition of angiogenesis.
It is probable that maspin produced by
tumor-associated normal tissue as well as that
produced by developing tumor cells them-
selves can influence tumor growth. Maspin is produced selec-
tively and at high levels by myoepithelial cells, which surround
the normal mammary ducts. The myoepithelial cells them-
selves form only low-grade tumors and that they also may
delay the progression of adjacent ductal carcinomas in situ to
invasive carcinomas14. The anti-angiogenic nature of the
maspin they secrete offers a possible explanation for both phe-
nomena.
The ability to inhibit angiogenesis is only one of several activ-
ities associated with the intact maspin protein. Other serpins
also have multiple functions and several of them are linked to
angiogenesis and tumor growth. Plasminogen activator in-
hibitor-1 is involved in modulating both proteolysis and angio-
genesis. Pigment epithelium-derived factor (PEDF), a known
regulator of cell differentiation, is also a very potent anti-angio-
genic factor15. Such results indicate that a variety of molecules,
whose structure places them in the serpin family can be impor-
tant regulators of natural tumor growth through their influence
on neovascularization.
Methods
Cell culture. The human prostate carcinoma cell line LNCaP from
American Type Culture Collection (ATCC; Manassas, Virginia) was grown
in RPMI 1640 with 10% fetal bovine serum. MDA-MB-435 cells were
maintained in DMEM (Life Technologies) supplemented with 10% fetal
calf serum. Normal human foreskin fibroblasts (HFF-S1) were established
in the laboratory by S. Tolsma (Northwester College, Orange City, Iowa)
and maintained in DME (Life Technologies) supplemented with 10% fetal
calf serum. Normal human keratinocytes (NHOK) were a gift from
M.Lingen (Loyola University Medical School, Maywood, Illinois) and
maintained in keratinocyte growth medium (Clonetics Cell Systems, San
Diego, California) with supplements recommended by the manufacturer.
Bovine adrenal capillary endothelial cells, a gift from J. Folkman
(Children’s Hospital, Harvard Medical School, Boston, Massachusetts
02115) were maintained in DMEM supplemented with 10% calf serum
and 100 µg/ml endothelial cell mitogen, and were used at passage 15.
Human dermal microvascular endothelial cells (HMVEC, passage 9) were
from Clonetics Cell Systems and maintained in endothelial cell basal
medium with 5% fetal bovine serum and an EC (endothelial cell) ‘bullet’
kit as recommended by supplier.
Protein production and purification. The pGST-maspin and maspinRSL
vectors were constructed as described7. MaspinN was generated by di-
gestion of the pGST-maspin vector with BamH1, which was blunt ended
and digested again with Sst1. The adhesive ends were filled by T4 poly-
merase and ligated to remove the N-terminal 139 amino acids. To con-
struct pmaspin, oligonucleotides encoding a mutation in the P1 residue
were generated (sense primer 5–GGGTCCCAGATCTTA–3and antisense
primer 5– TAAGATCTGGGACCC–3). Site-directed mutagenesis accom-
plished using the pGST-maspin vector and the above oligos (Stratagene,
La Jolla, California). All mutant constructs were sequenced to ensure the
in-frame fusion. The GST fusion proteins were produced as described7.
The size and purity of recombinant maspin and mutants were confirmed
by SDS-PAGE gel electrophoresis and western blot analysis using a poly-
clonal AbS4A antibody7.
Endothelial cell assays. Migrations were done using bovine adrenal cap-
illary endothelial cells as described16. Cells were starved overnight in DME
containing 0.1% bovine serum albumin (BSA), collected, re-suspended in
DME with 0.1% BSA, and plated at a concentration of 3 ×104cells/well
on the lower surface of a gelatinized 5.0 µm filter (Nucleopore,
Pleasanton, California) in an inverted, modified Boyden chamber. Cells
were allowed to adhere for 2 h at 37 °C, the chambers were re-inverted,
test samples were added to the top wells and the chambers incubated 4 h
at 37 °C to allow migration. Chambers were then disassembled, mem-
branes fixed and stained and the number of cells that had migrated to the
top of the filter in 10 high-power fields counted (a high power field is
×1000). DME supplemented with 0.1% BSA was used as a negative con-
trol to measure background resulting from random migration. All samples
Fig. 3 Decreased tumor vessels after long-term treatment with exogenous
maspin protein. Tumors were collected between 7 and 8 weeks from GST-
treated mice (aand c) and from maspin-treated mice (band d) and fixed
and stained with hematoxylin and eosin (a and b) or with antibody against
CD31 (c and d).
Table 1 Analysis of tumor volume and vessel number in mice treated with
GST or GST–maspin
GST GST-maspin P
Short Average of 28.6 ± 3.6 15.3 ± 1.8 0.001
term microvessel densityan=10 n=10
Total tumor sites 37 32
Percentage of complete 2.7% (1/37) 53% (17/32) 0.0001
Long inhibitionb
term
Average of tumor 116.3 ± 26.1, n=36 33.9 ± 6.1, n=15 0.027
volume (mm3) of sites (12.1–600) (9.0–87.5)
with tumors
Average of MVD 83.0 ±10, n=10 31.4 ±1.8, n=10 0.002
(38–143) (21–42)
aCollected from three ‘hot fields’ (400×) in each tumor. bComplete inhibition indicates no sign of tumor or
tumor size smaller than 2 mm in diameter after treatment. ±, standard error.
a b
cd
© 2000 Nature America Inc. • http://medicine.nature.com
© 2000 Nature America Inc. • http://medicine.nature.com
NATURE MEDICINE VOLUME 6 NUMBER 2 FEBRUARY 2000 199
ARTICLES
were tested in quadruplicate.
To assess mitogenesis, HMVECs were plated at a concentration of 1 ×
104cells per well in gelatinized, 96-well microtiter plates, allowed to attach
for 24 h and re-fed with endothelial cell basal medium (Life Technologies,
New York) supplemented with 2% serum and GST or GST–maspin fusion
protein and mutants at a concentration of 1 µM where indicated. The cells
were incubated at 37 °C, with 5%CO2for 5 days to allow at least one pop-
ulation doubling. Mitogenesis was assessed using CellTiter nonradioactive
proliferation assay (Promega) according to manufacturer’s instructions.
Baseline (no mitogen) reflects the value, determined before population
doubling, 24 h after seeding.
Migration assays on non-endothelial cells. Breast tumor migration as-
says (MDA-MB-435) were done in quadruplicate as described7. After sub-
traction of background, data were normalized to give percent inhibition
equating GST treated samples to 100% motility. For migration assays on
fibroblasts and keratinocytes, HFF-S1 (0.7 ×106cells/ml) and NHOK (1.2 ×
106cells/ml) were plated in serum-free basal media supplemented with
0.1%BSA on the bottom side of the microporous membrane (8-µm pore
size) in the inverted modified Boyden chamber. The cells were allowed to
attach for 1.5 h, the chambers were re-inverted and test substances in ap-
propriate serum-free basal medium were added to the other side of the
membrane. The migration was assessed, background was subtracted and
data were reported as percent inhibition, as in tumor cell invasion assay,
with bFGF induced motility normalized as 100%.
Corneal neovascularization assay. The assay was done as previously de-
scribed16. Hydron pellets containing bFGF (100 ng/ml), GST (10 µM), GST-
maspin (10 µM) or mutants (10 µM), alone in combination with bFGF
were implanted into a pocket surgically created in avascular corneas of
anesthetized female rats (Fisher 344: Harlan, Indianapolis, Indiana) 1–1.5
mm from the limbus. The compounds were used at concentrations at least
10-fold higher than in migration assay to account for the diffusion rate
from a slow release pellet. Neovascularization was observed on day 6 or 7
after implantation. Vigorous growth of the blood vessels in the direction of
the pellet was noted as a positive response. The animals were perfused
with colloidal carbon, eyes were removed and fixed, corneas excised, flat-
tened and photographed for a permanent record.
Tumor angiogenesis assay. LNCaP tumor cells were grown to 80% con-
fluence, collected and resuspended in sterile HEPES-buffered salt solution
at a concentration of 4 ×10 7cells/ml. This cell suspension was then mixed
with Matrigel (Collaborative Research, Bedford Massachusetts) at a 1:3
ratio and added in 100-µl aliquotes into Eppendorf tubes containing
maspin or GST, mixed on ice for 10 min and subsequently injected subcul-
taneously into the dorsal back of 5-week-old male athymic mice (K & K
Universal, Freemont, California). Each mouse was inoculated at two to four
sites. Ten mice were used for the initial vessel formation assay and 20 mice,
for the tumor inhibition study. Primary tumors at the site of the subcuta-
neous injection were measured using calipers, and tumor volume was cal-
culated according to the algorithm: length x [width]2×0.5 (ref. 17).
Ethylene/vinyl acetate copolymer (Evac; NEN) slow release pellets, con-
taining maspin and GST, were prepared as described18,19. A lyophilized
mixture of BSA and varied amounts of recombinant proteins was dispersed
in 0.125 ml Evac dissolved in dichloromethane (Sigma). This mixture was
frozen, dried, and cut into pellets of appropriate size. Each Evac pellet con-
tained about 210 µg GST-maspin or 70 µg GST. The follow-up treatment
of tumors was carried out by subcutaneous implantation of the pellet at
days 15 and 30 within 0.3 cm of the tumor site and the incision sealed
after implantation. At the end of each experiment (7–8 weeks), the tumors
were measured, excised and fixed in 10% neutral buffered formalin.
Samples were then embedded and sectioned to 5 µm for histology (hema-
toxylin and eosin) and immunohistochemistry. For immunohistochem-
istry, monoclonal antibodies to CD 31 (Pharmingen, Sandiego, California)
was used at 1:50 dilution at 4 °C overnight. After rinsing, slides were incu-
bated with the secondary goat antibody to rat at a dilution of 1:100 for 1 h
at room temperature. Slides were then rinsed and incubated with an
avidin–biotin–peroxidase complex (ABC kit; Vector Laboratories,
Burlingame, California), followed by DNBA (3, 3-Diaminobenzidine
Tetrahydrochloride) color development. Vessels were counted as described
20 by first scanning the sections at the low power for hot spots or high vas-
cular density (×40), and then counting the areas of microvessels at high
power (×400). Microvessel density was calculated by adding the numbers
from three hot spot fields with a ×400 objective.
Acknowledgments
The authors thank J. Rosen for his advice and equipment support, N.
Greenberg, D. Rowley, and W. Porter for discussion, W. Huss for kindly provid-
ing the protocol for CD31 and factor VIII immunostaining. This work is
supported by National Cancer Institute grants CA 52750 and CA 64239 to
N.B. and a Department of Defense grant (DAMD17-98-1-8028) to M.Z.
RECEIVED 7 SEPTEMBER; ACCEPTED 1 NOVEMBER 1999
1. Zou, Z. et al. Maspin, a serpin with tumor-suppressing activity in human mammary
epithelial cells [see comments]. Science 263, 526–529 (1994).
2. Zhang, M., Maass, N., Magit, D. & Sager, R. Transactivation through Ets and Ap1
transcription sites determines the expression of the tumor-suppressing gene
maspin. Cell Growth Differ. 8, 179–186 (1997).
3. Pemberton, P.A. et al. The tumor suppressor maspin does not undergo the stressed
to relaxed transition or inhibit trypsin-like serine proteases. Evidence that maspin is
not a protease inhibitory serpin. J. Biol. Chem. 270, 15832–15837 (1995).
4. Sheng, S. et al. Tissue-type plasminogen activator is a target of the tumor suppres-
sor gene maspin. Proc. Natl. Acad. Sci. U. S. A. 95, 499–504 (1998).
5. Zhang, M. et al. Maspin plays an important role in mammary gland development.
Dev. Biol. 215, 278–287 (1999).
6. Stringer, H.A. & Pannekoek, H. The significance of fibrin binding by plasminogen
activator inhibitor 1 for the mechanism of tissue-type plasminogen activator-medi-
ated fibrinolysis. J. Biol. Chem. 270, 11205–11208 (1995).
7. Zhang, M., Sheng, S., Maass, N. & Sager, R. mMaspin: the mouse homolog of a
human tumor suppressor gene inhibits mammary tumor invasion and motility. Mol.
Med. 3, 49–59 (1997).
8. Sheng, S., Pemberton, P.A. & Sager, R. Production, purification, and characteriza-
tion of recombinant maspin proteins. J. Biol. Chem. 269, 30988–30993 (1994).
9. Dawson, D.W. et al. CD36 mediates the In vitro inhibitory effects of throm-
bospondin-1 on endothelial cells. J. Cell Biol. 138, 707–717 (1997).
10. Lawrence, D.A., Berkenpas, M.B., Palaniappan, S. & Ginsburg, D. Localization of
vitronectin binding domain in plasminogen activator inhibitor-1. J. Biol. Chem. 269,
15223–15228 (1994).
11. Gately, S. et al. Human prostate carcinoma cells express enzymatic activity that
converts human plasminogen to the angiogenesis inhibitor, angiostatin. Cancer
Res. 56, 4887–4890 (1996).
12. Volpert, O.V. et al. Captopril inhibits angiogenesis and slows the growth of exper-
imental tumors in rats [see comments]. J. Clin. Invest. 98, 671–679 (1996).
13. Sheng, S. et al. Maspin acts at the cell membrane to inhibit invasion and motility of
mammary and prostatic cancer cells. Proc. Natl. Acad. Sci. U. S. A. 93, 11669–11674
(1996).
14. Sternlicht, M.D., Kedeshian, P., Shao, Z.M., Safarians, S. & Barsky, S.H. The human
myoepithelial cell is a natural tumor suppressor. Clin. Cancer Res. 3, 1949–1958
(1997).
15. Dawson, D.W. et al. Pigment Epithelium-Derived Factor: A Potent Inhibitor of
Angiogenesis. Science 285, 245–248 (1999).
16. Polverini, P.J., Bouck, N.P. & Rastinejad, F. Assay and purification of naturally occur-
ring inhibitor of angiogenesis. Methods Enzymol. 198, 440–450 (1991).
17. Tsujii, M. et al. Cyclooxygenase regulates angiogenesis induced by colon cancer
cells [published erratum appears in Cell 1998 Jul 24;94(2):following 271]. Cell 93,
705–716 (1998).
18. Silberstein, G.B. & Daniel, C.W. Investigation of mouse mammary ductal growth
regulation using slow- release plastic implants. J. Dairy Sci. 70, 1981–1990 (1987).
19. Talhouk, R.S., Bissell, M.J. & Werb, Z. Coordinated expression of extracellular ma-
trix-degrading proteinases and their inhibitors regulates mammary epithelial func-
tion during involution. J. Cell Biol. 118, 1271–1282 (1992).
20. Obermair, A. et al. Correlation of vascular endothelial growth factor expression and
microvessel density in cervical intraepithelial neoplasia. J. Natl. Cancer Inst. 89,
1212–1217 (1997).
© 2000 Nature America Inc. • http://medicine.nature.com
© 2000 Nature America Inc. • http://medicine.nature.com
... From the gastric cancer dataset of 80 patients, the maspin has been identified to be upregulated in 55 (69%), and downregulated in 8 (10%), while it was not detected/significant in 17 (21%) of samples (Supplementary method, Supplementary Fig. 1). Maspin has been attributed as the antitumor protein where it prevents metastasis, a hallmark of cancer, and functional loss of this protein can enhance tumor potential (Zhang et al., 2000). Normally, maspin-P176 functions as a tumor suppressor, however, the substitution of P176S can alter the functions making it a protumor protein, prompting this study to reveal the functional aspect of these polymorphic forms (Fig. 1A). ...
... The localization and cellular environment of maspin largely dictate its pro-or antitumor functions (Bernardo et al., 2015;Dzinic et al., 2017;Sakabe et al., 2021) (Zhang et al., 2000;Ngamkitidechakul et al., 2003;Dzinic et al., 2013). Recently, it has been reported that maspin can translocate to the nucleus through active and passive mechanisms, and it also harbors nuclear localization signals, spanning from 87 to 114 (s2A and hE) (Reina et al., 2019). ...
Preprint
Maspin has been known to regress tumors by inhibiting angiogenesis, however, its roles have been reported to be context and sequence-dependent. There are various proteins and cofactors that bind with maspin possibly explaining the conflicting roles of maspin. Moreover, maspin polymorphic forms have also been linked to tumor regression or survival, for instance, maspin with Ser at 176 (maspin-S176) promotes tumor while maspin with Pro at 176 (maspin-Pro176) has opposing roles in cancer pathogenesis. With the help of long molecular dynamic simulation, a possible link between polymorphic forms and tumor progression has been established. First, the maspin is dynamically stable with either amino acid at 176 position, secondly, differential contacts have been observed among various regions, thirdly, these contacts have significantly altered the electrostatic energetics of various residues, and finally, these altered electrostatics of maspin-S176 and maspin-P176 rewired the polar contacts that abolished the allosteric control in protein. By combining these factors, the altered electrostatics can substantially affect the localization, and the preference of maspin binding partners, thus, culminating in a different maspin-protein (cofactor)-interaction landscape that could have been manifested with conflicting reports in previous studies. Here, the underlying reason has been highlighted and discussed that could be helpful for better therapeutic manipulations. Significance Protein altered functions in response to mutations are well documented, however, a slight perturbation in structure can lead to dramatic effects that are being felt at longer distances are rare. Here, we have reported that the substitution of Pro to Ser at 176 th position in Maspin can substantially alter the protein electrostatic interactions that can hamper the allosteric control. This could lead to different binding partners, localization preferences, and altered cytoplasmic retention duration resulting in functions that are not associated with normal protein. Moreover, the electrostatic attraction/repulsion can immensely affect the allosteric cohesion of protein resulting in unexpected outcomes.
... HepG2-derived exosomes are known to accelerate tumor progression by increasing angiogenesis [21,22]. Maspin has anti-angiogenic functions [23]. We evaluated the anti-angiogenic response of exosomal Maspin by treating HUVECs with normal and radiation-derived exosomes secreted by HepG2. ...
Article
Full-text available
Background Tumor-derived exosomes are critical elements of the cell–cell communication response to various stimuli. This study aims to reveal that the histone deacetylase 5 (HDAC5) and p53 interaction upon radiation in hepatocellular carcinoma intricately regulates the secretion and composition of exosomes. Methods We observed that HDAC5 and p53 expression were significantly increased by 2 Gy and 4 Gy radiation exposure in HCC. Normal- and radiation-derived exosomes released by HepG2 were purified to investigate the exosomal components. Results We found that in the radiation-derived exosome, exosomal Maspin was notably increased. Maspin is known as an anti-angiogenic gene. The expression of Maspin was regulated at the cellular level by HDAC5, and it was elaborately regulated and released in the exosome. Radiation-derived exosome treatment caused significant inhibition of angiogenesis in HUVECs and mouse aortic tissues. Meanwhile, we confirmed that miR-151a-3p was significantly reduced in the radiation-derived exosome through exosomal miRNA sequencing, and three HCC-specific exosomal miRNAs were also decreased. In particular, miR-151a-3p induced an anti-apoptotic response by inhibiting p53, and it was shown to induce EMT and promote tumor growth by regulating p53-related tumor progression genes. In the HCC xenograft model, radiation-induced exosome injection significantly reduced angiogenesis and tumor size. Conclusions Our present findings demonstrated HDAC5 is a vital gene of the p53-mediated release of exosomes resulting in tumor suppression through anti-cancer exosomal components in response to radiation. Finally, we highlight the important role of exosomal Maspin and mi-151a-3p as a biomarker in enhancing radiation treatment sensitivity. Graphical Abstract Therapeutic potential of HDAC5 through p53-mediated exosome modulation in radiation treatment of hepatocellular carcinoma
... Hence, maspin may increase cell-cell adhesion, subsequently prevent tumor invasion and metastasis [14]. In addition, maspin inhibits in vivo, neoplasm-associated angiogenesis [15,16]. Maspin expression has been demonstrated in different cell types, and appeared to be down-regulated in breast, prostate, and gastric cancers [17][18][19]. ...
Article
Purpose: Maspin is a member of serpin (serine protease inhibitor) family, and is known to have tumor-suppressor function. Maspin has been reported to play a critical role in processes relevant to tumor growth and metastasis such as cell invasion, angiogenesis, and apoptosis. The aim of this study is to evaluate immunohistochemical expression of maspin and its correlation to histologic grade of salivary mucoepidermoid carcinoma. Materials and Methods: A total of 22 formalin-fixed, paraffin-embedded specimens of mucoepidermoid carcinoma were prepared for immunohistochemical staining with maspin antibodies. Maspin immunoreactivity was evaluated by calculating the proportion of positively-stained tumor cells present in 5 high-power fields. Results: All cases were expressed maspin with variable cellular localization. No significant correlation was found between maspin expression and clinicopathologic features such as patient’s age, gender, tumor site, clinical stage and lymph node metastasis (p> 0.05), whereas maspin expression showed a statistically significant correlation with grade of tumors classified in regard to Auclair system (p= 0.011), compared to that evaluated by Brandwein system (p= 0.155). Conclusion: Salivary mucoepidermoid carcinoma showed maspin immunoreactivity with variable levels and cellular localization. Maspin is a valuable biomarker to predict the prognosis of salivary mucoepidermoid carcinoma.
... When the cornea is injured, the stromal keratinocytes below the wound undergo apoptosis and their neighbors transform into fibroblasts or myofibroblasts. In this process, maspin acts directly on endothelial cells to stop their migration towards basic fibroblast growth factor(bFGF) and vascular endothelial growth factor(VEGF) (31)(32)(33). Therefore, we speculate that maspin may affect the healing of the injured cornea. ...
Article
Full-text available
Background Immunotherapy has been widely used to treat Colorectal cancer but has also observe some immune-related adverse effects. With proper treatment, most irAE can be solved and the effect of immunotherapy will not be affected by temporary immunosuppression. However, there are few reports about corneal irAE, and the current understanding of irAE is incomplete. Here we report a metastatic colorectal cancer case of immune-related keratitis caused by nivolumab and to explore the occurrence of immune-related keratitis. Case description Here we report the case of a 49-year-old man with mCRC who had no previous ocular disease but developed immune-related ulcerative keratitis after treatment with nivolumab. We summarize a large amount of literature to discuss the mechanism of immune-related keratitis. In addition, we conclude a method that may be used to detect the occurrence of immune keratitis, by monitoring MMPs and maspin in patients treated with nivolumab. We believe immune-related keratitis may be a rare complication of nivolumab in the treatment of mCRC. The effect of simple anti-infective therapy and repair-promoting drugs was not obvious, but the effect of glucocorticoid combined with autologous serum was significant. Conclusion The mechanism of immune-related keratitis is that nivolumab destroys the immune microenvironment and ACAID, and affects corneal healing. Patients who use nivolumab can prevent immune keratitis by testing MMPs and maspin. The occurrence of immune keratitis may be a good indicator of the efficacy of ICI, and further study can be done in the follow-up.
... Maspin, a protein encoded by the SERPIN5 gene in chromosome 18q21.3-q23, is classified under the mammalian serine protease inhibitor family and functions as a tumor suppressor gene (16). Although maspin has been suggested to play a role as an anti-angiogenic inhibitor in various types of malignancies (17), its role is still controversial in ovarian cancer because the relatively few reports have demonstrated conflicting results about the relationship between maspin expression and its prognosis (18)(19)(20)(21). ...
Article
Background/aim: To investigate the role of the expression of hypoxia-related genes on the prognosis of ovarian clear cell carcinoma (OCCC). Materials and methods: Basal mRNA levels of eight hypoxia-related genes were compared. Cell viability was assayed after treating ES-2 cells under hypoxic conditions. The mRNA and protein levels were evaluated after the induction of hypoxia and administration of increased doses of N-acetylcysteine (NAC). Finally, the prognostic role of their expression levels was evaluated in 61 patients with OCCC. Results: The mRNA and protein levels of maspin increased gradually with the induction of hypoxia. Maspin protein expression decreased after treatment with paclitaxel and NAC. High expression of maspin was related to poor progression-free and overall survival in patients with OCCC (adjusted hazard ratios, 3.97 and 7.47; 95% confidence intervals=1.34-11.81, and 1.98-28.13). Conclusion: High expression of maspin induced by hypoxia might be associated with poor prognosis of OCCC.
Article
Full-text available
Maspin is known to regress tumors by inhibiting angiogenesis; however, its roles have been reported to be context- and sequence-dependent. Various proteins and cofactors bind to maspin, possibly explaining its conflicting roles. Moreover, polymorphic forms of maspin have also been linked to tumor regression and survival; for instance, maspin with Ser at 176 (maspin-S176) promotes tumors, while maspin with Pro at 176 (maspin-P176) has opposing roles in cancer pathogenesis. With the help of long molecular dynamics simulations, a possible link between polymorphic forms and tumor progression has been established. First, maspin is dynamically stable with either amino acid at the 176 position. Second, differential contacts have been observed among various regions; third, these contacts have significantly altered the electrostatic energetics of various residues; finally, these altered electrostatics of maspin-S176 and maspin-P176 rewire the polar contacts that abolished the allosteric control of the protein. By combining these factors, the altered electrostatics substantially affect the localization and preference of maspin-binding partners, thus culminating in a different maspin-protein(cofactor)-interaction landscape that may have been manifested in previous conflicting reports. Here, the underlying reason has been highlighted and discussed, which may be helpful for better therapeutic manipulation.
Article
Full-text available
Over the past decades, luminal epithelial cell lineage has gained considerable attraction as the functionally milk-secreting units and as the most fruitful acreage for breast cancer launching. Recognition of the effective involvement of the myoepithelial cells in mammary gland development and in hampering tumorigenesis has renewed the interest in investigating the biological roles of this second main mammary lineage. The human breast is made up of an extensively branching ductal system intervening by copious lobular units. The ductal system is coated by a chain of luminal epithelial cells (LECs) situated on a layer of myoepithelial cells (MECs) and encompassed by a distinguished basement membrane. Ductal contractility during lactation is a well-known function delivered by the MECs however this is not the only assignment mediated by these cellular populations. It has been well appreciated that the MECs exhibit a natural paracrine power in defeating cancer development and advancement. MECs were found to express numerous proteinase inhibitors, anti-angiogenic factors, and tumour suppressors proteins. Additionally, MECs contributed effectively to maintaining the right luminal cells' polarization and further separating them from the adjacent stroma by making an integrated fence. Indeed, disruption of the MECs layer was reported to facilitate the invasion of the cancer cells to the surrounding stroma. Nonetheless, MECs were also found to exhibit cancer-promoting effects and provoke tumour invasion and dissemination by displaying distinct cancer chemokines. Herein in this review, we aimed to address the roles delivered by MECs in breast cancer progression and decipher the molecular mechanisms regulating proper MECs’ physiology, integrity, and terminal differentiation. Graphical Abstract
Article
Maspin is a novel serine protease inhibitor differentially expressed in several types of human cancers. It proved to be a key biomarker in the assessment of gastric cancer. Therefore, we design, characterize, and validate two stochastic microsensors based on graphene co-doped with N and S, and modified with α-cyclodextrin and maltodextrin, for the pattern recognition and quantification of maspin in whole blood, gastric tumor tissue, saliva, and urine. While the sensitivities were comparable with magnitude order, the variations were in the wideness of the linear concentration range, when measurements were performed at a pH of 7.40. Very low limits of quantification were recorded at both working pHs: 7.40, and 3.00. High recoveries of maspin in whole blood, gastric tissue tumor, saliva, and urine were also recorded.
Article
Full-text available
Extracellular matrix (ECM) plays an important role in the maintenance of mammary epithelial differentiation in culture. We asked whether changes in mouse mammary specific function in vivo correlate with changes in the ECM. We showed, using expression of beta-casein as a marker, that the temporal expression of ECM-degrading proteinases and their inhibitors during lactation and involution are inversely related to functional differentiation. After a lactation period of 9 d, mammary epithelial cells maintained beta-casein expression up to 5 d of involution. Two metalloproteinases, 72-kD gelatinase (and its 62-kD active form), and stromelysin, and a serine proteinase tissue plasminogen activator were detected by day four of involution, and maintained expression until at least day 10. The expression of their inhibitors, the tissue inhibitor of metalloproteinases (TIMP) and plasminogen activator inhibitor-1, preceded the onset of ECM-degrading proteinase expression and was detected by day two of involution, and showed a sharp peak of expression centered on days 4-6 of involution. When involution was accelerated by decreasing lactation to 2 d, there was an accelerated loss of beta-casein expression evident by day four and a shift in expression of ECM-remodeling proteinases and inhibitors to a focus at 2-4 d of involution. To further extend the correlation between mammary-specific function and ECM remodeling we initiated involution by sealing just one gland in an otherwise hormonally sufficient lactating animal. Alveoli in the sealed gland contained casein for at least 7 d after sealing, and closely resembled those in a lactating gland. The relative expression of TIMP in the sealed gland increased, whereas the expression of stromelysin was much lower than that of a hormone-depleted involuting gland, indicating that the higher the ratio of TIMP to ECM-degrading proteinases the slower the process of involution. To test directly the functional role of ECM-degrading proteinases in the loss of tissue-specific function we artificially perturbed the ECM-degrading proteinase-inhibitor ratio in a normally involuting gland by maintaining high concentrations of TIMP protein with the use of surgically implanted slow-release pellets. In a concentration-dependent fashion, involuting mammary glands that received TIMP implants maintained high levels of casein and delayed alveolar regression. These data suggest that the balance of ECM-degrading proteinases and their inhibitors regulates the organization of the basement membrane and the tissue-specific function of the mammary gland.(ABSTRACT TRUNCATED AT 400 WORDS)
Article
Full-text available
Plasminogen activator inhibitor type 1 (PAI-1) is the rapid physiologic inhibitor of tissue-type plasminogen activator and urokinase-type plasminogen activator (uPA). In plasma and the extracellular matrix, PAI-1 is associated with the adhesive glycoprotein vitronectin. In order to characterize the PAI-1 structural domain responsible for binding to vitronectin, the segment of the PAI-1 cDNA encoding amino acids 13-147 (nucleotides 248-650) was randomly mutagenized and subcloned into a bacterial expression vector containing the mature PAI-1 coding sequence. Recombinant PAI-1 mutants were expressed in Escherichia coli and bacterial lysates assayed in duplicate for uPA inhibitory activity and vitronectin binding. Of 190 clones screened, six consistently demonstrated decreased vitronectin binding relative to uPA inhibitory activity. DNA sequence analysis of four of these clones identified 10 unique missense mutations, all located between base pairs 298 and 641, with each clone containing between one and four substitutions. Each substitution was expressed independently by site-directed mutagenesis and again analyzed for uPA inhibitory activity and vitronectin binding. Five point mutations that selectively disrupt vitronectin binding were identified. All 5 residues are located on the exterior of the PAI-1 structure. These findings appear to define a complex binding surface that bridges alpha-helices C and E to beta-strand 1A and includes amino acids 55, 109, 110, 116, and 123. These results suggest that vitronectin binding may stabilize the active conformation of PAI-1 by restricting the movement of beta-sheet A and thereby preventing insertion of the reactive center loop.
Article
Full-text available
The specific, reversible interaction between plasminogen activator inhibitor 1 (PAI-1) and intact fibrin polymers was studied using both purified components and isolated activated platelets as a source of PAI- 1. A key reagent in these experiments is a PAI-1 mutant, having its P1 reactive center residue arginine replaced by methionine (PAI-1 R346M). The second-order association rate of PAI-1 R346M with tissue-type plasminogen activator is over 10,000-fold lower than that of wild-type PAI-1, whereas the ability of the variant to bind to fibrin is unaltered. Competition experiments demonstrated that PAI-1 R346M is equally effective as wild-type PAI-1 in displacing 125I-labeled PAI-1 from fibrin. Fibrinolysis, mediated by tissue-type plasminogen activator, is inhibited in a dose-dependent manner by purified PAI-1. The inhibition can be relieved in a dose-dependent manner by PAI-1 R346M, presumably due to displacement of wild-type PAI-1 by PAI-1 R346M. Perfusion studies, using platelet-rich clots, revealed that the incorporation of PAI-1 R346M dose dependently decreased the 50% clot lysis time. These data indicate that PAI-1 R346M displaces fibrin- bound, endogenous PAI-1 released from activated platelets. Implications to manipulate PAI-1 activity for the management of clinical complications, in particular reocclusion after thrombolytic therapy, are discussed.
Article
Full-text available
The role of tumor suppressor proteins in the development of malignancy has made the understanding of their molecular mechanisms of action of great importance. Maspin is a tumor suppressor produced by a number of cell types of epithelial origin. Exogenous recombinant maspin has been shown to block the growth, motility, and invasiveness of breast tumor cell lines in vitro and in vivo. Although belonging to the the serine proteinase inhibitor (serpin) superfamily of proteins, the molecular mechanism of maspin is currently unknown. Here we show that the reactive site loop of maspin exists in an exposed conformation that does not require activation by cofactors. The reactive site loop of maspin, however, does not act as an inhibitor of proteinases such as chymotrypsin, elastase, plasmin, thrombin, and trypsin but rather as a substrate. Maspin is also unable to inhibit tissue and urokinase type plasminogen activators. Stability studies show that maspin cannot undergo the stressed-relaxed transition typical of proteinase-inhibitory serpins, and the protein is capable of spontaneous polymerization induced by changes in pH. It is likely, therefore, that maspin is structurally more closely related to ovalbumin and angiotensinogen, and its tumor suppressor activity is independent of a latent or intrinsic trypsin-like serine proteinase-inhibitory activity.
Article
Full-text available
Maspin, a novel mammary serine protease inhibitor, was shown to have tumor suppressing activity (Zou, Z., Anisowicz, A., Hendrix, M. J. C., Thor, A., Neveu, M., Sheng, S., Rafidi, K., Seftor, E., and Sager, R. (1994) Science 263, 526-529). In this paper, we report the production of recombinant glutathione S-transferase-maspin fusion protein, expressed in the bacterium Escherichia coli, and recombinant maspin, expressed in the insect Spodoptera frugiperda cells. The fusion protein was purified by glutathione affinity chromatography. Maspin expressed in insect cells was purified by a combination of Bio-Rad AG1-2X anion exchange chromatography and heparin affinity chromatography. The recombinant maspin from insect cells was cleaved at the putative reactive center, as confirmed by protein sequencing. Both recombinant proteins demonstrated strong inhibitory effects on the invasion by two breast tumor cell lines across reconstituted basement membranes and such inhibitory effect was abolished in the presence of the polyclonal antibody made against the reactive center region of maspin. The trypsin-cleaved recombinant maspin did not inhibit invasion, indicating that the inhibitory activity requires the intact putative reactive center. This paper provides evidence that recombinant maspin protein itself inhibits invasion, and supports the role of maspin as a tumor suppressor.
Article
Myoepithelial cells, which surround ducts and acini of glandular organs, form a natural border separating proliferating epithelial cells from basement membrane and underlying stroma. Myoepithelial cells in situ and in vitro constitutively express high amounts of proteinase inhibitors that include tissue inhibitor of metalloproteinase 1, protease nexin-II, alpha-1 antitrypsin, and maspin. Human myoepithelial xenografts (HMS-X, HMS-3X, and HMS-4X), which our laboratory has established, accumulate an abundant extracellular matrix containing sequestered proteinase inhibitors. Humatrix, a gel that we have derived from HMS-X, inhibits tumor cell invasion (down to 25% +/- 10% of Matrigel control; P < 0.01), and our recently established human myoepithelial cell lines, HMS-1, HMS-3, and HMS-4, inhibit tumor cell invasion in cellular invasion (down to 42% +/- 7% of control; P < 0.05) and in conditioned media assays (down to 30% +/- 8% of control; P < 0.01). The anti-invasive effects of HMS-1, HMS-3, and HMS-4 can be enhanced by phorbol 12-myristate 13-acetate (down to 2% +/- 1% of control) by a maspin-dependent mechanism and abolished by dexamethasone (up to 95% +/- 5% of control) by a maspin-independent mechanism (P < 0.01). HMS-X, HMS-3X, HMS-4X, and Humatrix inhibit tumor invasion and metastasis in severe combined immunodeficient mice (P < 0.001). The cumulative data suggest that myoepithelial cells are natural paracrine suppressors of invasion and metastasis and may specifically inhibit the progression of precancerous disease states to invasive cancer in vivo.
Article
Extracellular matrix (ECM) plays an important role in the maintenance of mammary epithelial differentiation in culture. We asked whether changes in mouse mammary specific function in vivo correlate with changes in the ECM. We showed, using expression of beta-casein as a marker, that the temporal expression of ECM-degrading proteinases and their inhibitors during lactation and involution are inversely related to functional differentiation. After a lactation period of 9 d, mammary epithelial cells maintained beta-casein expression up to 5 d of involution. Two metalloproteinases, 72-kD gelatinase (and its 62-kD active form), and stromelysin, and a serine proteinase tissue plasminogen activator were detected by day four of involution, and maintained expression until at least day 10. The expression of their inhibitors, the tissue inhibitor of metalloproteinases (TIMP) and plasminogen activator inhibitor-1, preceded the onset of ECM-degrading proteinase expression and was detected by day two of involution, and showed a sharp peak of expression centered on days 4-6 of involution. When involution was accelerated by decreasing lactation to 2 d, there was an accelerated loss of beta-casein expression evident by day four and a shift in expression of ECM-remodeling proteinases and inhibitors to a focus at 2-4 d of involution. To further extend the correlation between mammary-specific function and ECM remodeling we initiated involution by sealing just one gland in an otherwise hormonally sufficient lactating animal. Alveoli in the sealed gland contained casein for at least 7 d after sealing, and closely resembled those in a lactating gland. The relative expression of TIMP in the sealed gland increased, whereas the expression of stromelysin was much lower than that of a hormone-depleted involuting gland, indicating that the higher the ratio of TIMP to ECM-degrading proteinases the slower the process of involution. To test directly the functional role of ECM-degrading proteinases in the loss of tissue-specific function we artificially perturbed the ECM-degrading proteinase-inhibitor ratio in a normally involuting gland by maintaining high concentrations of TIMP protein with the use of surgically implanted slow-release pellets. In a concentration-dependent fashion, involuting mammary glands that received TIMP implants maintained high levels of casein and delayed alveolar regression. These data suggest that the balance of ECM-degrading proteinases and their inhibitors regulates the organization of the basement membrane and the tissue-specific function of the mammary gland.(ABSTRACT TRUNCATED AT 400 WORDS)
Article
Mammary ductal development in the mouse is now thought to depend on an interplay of locally produced (glandular) and systemic mammogens. A novel plastic implant material, ethylene vinyl acetate copolymer (Elvax 40P), capable of the slow-release of undenatured, bioactive molecules in situ, now enables treatment of small regions of the mammary gland for extended periods with hormones and growth factors. Here we describe results obtainable with this technique. Specifically, the classical mammogens, estrogen, growth hormone, and prolactin, as well as the nontraditional mammogens, epidermal growth factor and cholera toxin, were shown to stimulate ductal growth in zones around an implant in ovariectomized animals. The possibility that these observations reflect the existence of multiple mammogenic pathways is discussed.
Article
A gene encoding a protein related to the serpin family of protease inhibitors was identified as a candidate tumor suppressor gene that may play a role in human breast cancer. The gene product, called maspin, is expressed in normal mammary epithelial cells but not in most mammary carcinoma cell lines. Transfection of MDA-MB-435 mammary carcinoma cells with the maspin gene did not alter the cells' growth properties in vitro, but reduced the cells' ability to induce tumors and metastasize in nude mice and to invade through a basement membrane matrix in vitro. Analysis of human breast cancer specimens revealed that loss of maspin expression occurred most frequently in advanced cancers. These results support the hypothesis that maspin functions as a tumor suppressor.