ArticlePDF Available

Expression of colony-stimulating factor 1 receptor during prostate development and prostate cancer progression

Authors:
  • Dokkyo Medical University Koshigaya Hospital, Japan, Koshigaya

Abstract and Figures

Colony-stimulating factor-1 receptor (CSF-1R) is the major regulator of macrophage development and is associated with epithelial cancers of the breast and ovary. Immunohistochemistry analysis of murine prostate development demonstrated epithelial expression of CSF-1R during the protrusion of prostatic buds from the urogenital sinus, during the prepubertal and androgen-driven proliferative expansion and branching of the gland, with a decline in older animals. Models of murine prostate cancer showed CSF-1R expression in areas of carcinoma- and tumor-associated macrophages. Several human prostate cancer cell lines and primary cultures of human prostate epithelial cells had low but detectable levels of CSF-1R. Human prostatectomy samples showed low or undetectable levels of receptor in normal glands or benign prostatic hypertrophy specimens. Staining was strongest in areas of prostatic intraepithelial neoplasia or carcinoma of Gleason histological grade 3 or 4. The activated form of the receptor reactive with antibodies specific for phosphotyrosine modified peptide sequences was observed in samples of metastatic prostate cancer. Immunohistochemistry showed strong expression of CSF-1R by macrophage lineage cells, including villous macrophages and the syncytiotrophoblast layer of placenta, Kupper cells in the liver, and histiocytes infiltrating near prostate cancers. These observations correlate CSF-1R expression with changes in the growth and development of the normal and neoplastic prostate.
Content may be subject to copyright.
Expression of colony-stimulating factor 1 receptor
during prostate development and prostate
cancer progression
Hisamitsu Ide*, David B. Seligson
, Sanaz Memarzadeh
, Li Xin*, Steve Horvath
§¶
, Purnima Dubey
, Maryann B. Flick
,
Barry M. Kacinski
**
††
, Aarno Palotie
†¶
, and Owen N. Witte*
‡,‡‡
*Howard Hughes Medical Institute, Departments of Pathology and Laboratory Medicine, Microbiology, Immunology, and Molecular Genetics,
§Biostatistics, and Human Genetics, University of California, Los Angeles, CA 90095; and Departments of Therapeutic Radiology,
**Dermatology, and ††Obstetrics and Gynecology, Yale University School of Medicine, New Haven, CT 06520-8040
Contributed by Owen N. Witte, September 4, 2002
Colony-stimulating factor-1 receptor (CSF-1R) is the major regulator of
macrophage development and is associated with epithelial cancers of
the breast and ovary. Immunohistochemistry analysis of murine
prostate development demonstrated epithelial expression of CSF-1R
during the protrusion of prostatic buds from the urogenital sinus,
during the prepubertal and androgen-driven proliferative expansion
and branching of the gland, with a decline in older animals. Models
of murine prostate cancer showed CSF-1R expression in areas of
carcinoma- and tumor-associated macrophages. Several human pros-
tate cancer cell lines and primary cultures of human prostate epithelial
cells had low but detectable levels of CSF-1R. Human prostatectomy
samples showed low or undetectable levels of receptor in normal
glands or benign prostatic hypertrophy specimens. Staining was
strongest in areas of prostatic intraepithelial neoplasia or carcinoma
of Gleason histological grade 3 or 4. The activated form of the
receptor reactive with antibodies specific for phosphotyrosine mod-
ified peptide sequences was observed in samples of metastatic
prostate cancer. Immunohistochemistry showed strong expression of
CSF-1R by macrophage lineage cells, including villous macrophages
and the syncytiotrophoblast layer of placenta, Kupper cells in the
liver, and histiocytes infiltrating near prostate cancers. These obser-
vations correlate CSF-1R expression with changes in the growth and
development of the normal and neoplastic prostate.
Perturbation of protein tyrosine kinase signaling is frequently
associated with malignant transformation (1). Tyrosine kinase
receptors and their ligands have been implicated in prostate de-
velopment and cancer, including transforming growth factor
,
epidermal growth factor, insulin-like growth factor 1 (IGF-1),
fibroblast growth factors, hepatocyte growth factor (HGF), plate-
let-derived growth factor (PDGF), and nerve growth factors (re-
viewed in refs. 2 and 3). Strategies attacking EGF and PDGF
receptors are currently being evaluated in prostate cancer (4–7).
We defined the tyrosine kinase expression profile of normal
prostatic epithelial cells during a phase of rapid growth in a
relatively low androgen environment from day 10 murine prostate.
CD44 was used as a marker of early progenitor cells within prostatic
epithelium and is expressed by actively proliferating epithelia at
sites of epithelial–mesenchymal interaction (8, 9). Embryonic for-
mation of the prostate occurs through epithelial budding from the
urogenital sinus. Elongation and branching of the ducts begin
prenatally and are extensive during the first 21 days after birth.
Although ductal morphogenesis of the prostate is androgen-
dependent, the early postnatal period is marked by low levels of
circulating androgen (10, 11). Mice with loss-of-function mutations
in the homeobox genes NKX 3.1 or Hox D13 show mild defects in
prostate development (10). A more severe block in prostate de-
velopment is seen in P63mice, which do not develop a
recognizable prostate (12).
We prepared cDNA libraries from highly enriched CD44
prostate cells from day 10 mice. A PCR-based strategy targeting
highly conserved tyrosine kinase catalytic domain sequences was
used (13, 14). One of the most frequently recovered tyrosine
kinases was the colony-stimulating factor-1 receptor (CSF-1R).
CSF-1R is encoded by the cellular homolog of the retroviral
oncogene v-fms (15) and is the major regulator of development
and response for all cells belonging to the mononuclear phago-
cyte lineage (16 –18).
In osteoclastogenesis, one of the critical factors produced by
bone stromal cells is CSF-1 (19, 20). The Csf1
op
Csf1
op
mouse
has inactivated the CSF-1 gene. These mice are osteopetrotic,
toothless, and have low fecundity and reduced macrophage
numbers (21, 22). CSF-1R null mutation (Csf1r
Csf1r
) mice
have a very similar but more severe phenotype (23). Prostate
histology and function have not been well characterized in either
Csf1
op
Csf1
op
or Csf1r
Csf1r
mice.
CSF-1R is expressed in testis, uterus, ovary, placenta, and
mammary glands (21, 24). Elevated expression of CSF-1R has been
seen in breast, ovarian, and uterine cancers, and the extent of
expression in these tumors correlates with high grade and poor
prognosis (24, 25). High circulating levels of CSF-1 correlate with
active disease in ovarian and endometrial cancers and with meta-
static breast and prostate cancer (24 –26). In this article, we show
that CSF-1R is expressed during the early phases of murine prostate
development and prostate cancer progression in mouse and human.
Materials and Methods
Animal and Cell Lines. Prostate cancer cell lines LNCaP (27), PC-3
(28) and DU145 (29), and breast cancer line BT-20 (30) were
obtained from American Type Culture Collection (Rockville,
MD). BT-20 cells were incubated in medium with 1
dexa-
methasone (synthetic glucocorticoid, Sigma) (31); LAPC-4 was
provided by R. Reiter [University of California, Los Angeles
(UCLA); ref. 32]; and basaloid PrEC prostate cells were from
Clonetics (Walkersville, MD). Mouse prostate (C57BL6) was
fixed with 10% buffered formalin and embedded in paraffin wax.
Prostate tumors were from transgenic adenocarcinoma mouse
prostate (TRAMP) (33) and phosphatase and tensin homolog
deleted from chromosome 10 (PTEN) mice (34, 35). Hong
Wu (UCLA) and Norman Greenberg (Baylor College of Med-
icine) kindly provided P TENand TRAMP mice, respec-
tively. E. Richard Stanley and Xu-Ming Dai (Albert Einstein
College of Medicine) kindly provided tissue sections of Csfr
Csf1r
mice. The M-NFS-60 murine macrophage line was used
as a control in some experiments (36). Csf1r
op
Csf1r
op
mice were
from The Jackson Laboratory.
Abbreviations: IGF-1, insulin-like growth factor 1; HGF, hepatocyte growth factor; PDGF,
platelet-derived growth factor; CSF-1R, colony stimulating factor-1 receptor; TRAMP,
transgenic adenocarcinoma mouse prostate; PTEN, phosphatase and tensin homolog
deleted from chromosome 10; TMA, tissue microarray analysis; IHC, immunohistochemis-
try; PrEC, prostate-derived basaloid epithelial cell populations; PIN, prostatic intraepithelial
neoplasia; UCLA, University of California, Los Angeles.
‡‡To whom correspondence should be addressed. E-mail: owenw@microbio.ucla.edu.
14404–14409
PNAS
October 29, 2002
vol. 99
no. 22 www.pnas.orgcgidoi10.1073pnas.222537099
Prostate tissues were minced and digested with collagenase I
(Sigma) at a concentration of 1,500 unitsml for 1 hr at 37°C.
Single-cell suspensions were analyzed with FACS Vantage (Bec-
ton Dickinson). For depletion of macrophages from mouse
prostate tissues, cells were incubated with mouse monoclonal
antibody CD11b for 30 min at 4°C, washed three times, and
incubated with sheep anti-mouse IgG Dynabeads M-450 (Dynal,
Oslo) for 30 min at 4°C, then separated by magnetic column. The
retained CD11b-positive and CD11b-negative cells that flow
through the column were tested.
RNA Expression Analysis. Total RNAs were extracted by the RNeasy
Mini kit (Qiagen, Chatsworth, CA). Each portion of total RNAs
from the cells was reverse-transcribed by oligo(dT) primer and
SuperScript reverse transcriptase (Life Technologies, Gaithers-
burg, MD) in a volume of 25
l after DNase I treatment. The
resulting cDNA was subjected to PCR. DNA sequences of the
primer pairs used are as follows: human CSF-1R, 5-ACACTA-
AGCTCGCAATCCC-3and 5-GTATCGAAGGGTGAGCT-
CAAA-3; mouse CSF-1R, 5-GACCTGCTCCACTTCTC-
CAG-3and 5-GGGTTCAGACCAAGCGAGAAG-3; human
CSF-1, 5-GGAGTGGACACCTGCAGTCT-3and 5-TGTG-
CAGGGGCTGCTCACCA-3; prostate-specific antigen, 5-
GGTCGGCACAGCCTGTTTCA-3and 5-CCACGATGGT-
GTCCTTGATC-3;
-actin, 5-GACTACCTCATGAAGAT-
CCT-3and 5-GCGGATGTCCACGTCACACT-3.
Immunoblot Analysis. For Western blot analyses, the cells were
resuspended in boiling sample buffer, and each sample was sepa-
rated on a 412% gradient SDS Tris Glycine Gel (NOVEX, San
Diego). Proteins were transferred onto a nitrocellulose membrane
(Micron Separations, Westboro, MA) and visualized with a chemi-
luminescence kit (Amersham Pharmacia). Rabbit anti-human
CSF-1R antibody (Santa Cruz Biotechnology, lot K130, 1:200
dilution) was used for Western blot analysis. Anti-ABL Western
blots were probed with 2124 mouse monoclonal antibodies as an
internal loading control, as described (37). Goat anti-mouse IgG
and goat anti-rabbit antibody conjugated by horseradish peroxidase
(Bio-Rad) were used as secondary antibodies.
Tissue Microarray Analysis (TMA). Archival formalin-fixed paraffin-
embedded tissue samples were provided through the Depart-
ment of Pathology at the UCLA Medical Center under Institu-
tional Review Board approval. Primary radical prostatectomy
cases from 1984 to 1995 were randomly selected from the
pathology database to represent a wide spectrum of tumor
grades and primary disease stages. Three TMAs were con-
structed by the method of Kononen et al. (38), encompassing a
total of 246 individual patients. Patients treated preoperatively
with neoadjuvant hormones were then excluded from the anal-
ysis (n20). An additional 11 cases were uninformative due to
missing spots or lack of target tissues. Overall, 215 prostatectomy
cases encompassing 1,109 informative tissue spots on three
TMAs were used for analysis. Included were 214 radical pros-
tatectomies and one cystoprostatectomy. The median age at
diagnosis of this cohort was 65 years. Semiquantitative assess-
ment of antibody staining on the TMAs was performed by one
pathologist (D.B.S.). Target tissue for scoring included only the
glandular elements of the prostate tissue. The maximal intensity
of diaminobenzidine brown chromogen staining was graded on
a02 scale (0 negative, 1 weak, and 2 strong staining).
Immunohistochemistry (IHC). Serial 4-
m-thick sections were depar-
affinized in three changes of xylene and rehydrated through a
10070% descending series of ethanol, immersed in citrate buffer
(pH 6.0) in a 95°C water bath for 25 min, and then placed in 3%
H
2
O
2
in methanol for 1020 min at room temperature to block
endogenous peroxidase activity. After the blocking of nonspecific
protein binding by incubation for 30 min to1hwith5%goat serum,
the whole-tissue sections were incubated with each primary anti-
body against either monoclonal mouse anti-human CD68 (DAKO),
polyclonal rabbit anti-human CSF-1R (Santa Cruz Biotechnology,
lot K130, 1:200 dilution), or polyclonal rabbit anti-human phos-
phorylated tyrosine 723 peptide (PY723) of CSF-1R (1:50 dilution)
overnight at 4°C (39). Subsequently, sections were processed for
IHC by using the EnVision
System (DAKO), StreptABComplex
HRP kit (DAKO), or Vector ABC Elite (Vector Laboratories),
as described (34). For PY723 of CSF-1R staining, we used the
EnVision
(DAKO) and tyramide signal amplification (NEN)
systems, according to the manufacturersguidelines. For double
staining of CD68 and CSF-1R, we used the EnVision Doublestain
system (DAKO), according to the protocol recommended by the
manufacturer. Specificity of stain ing was confirmed by replacing the
primary antibody with mouse IgG for CD68 or rabbit IgG for other
antibodies at the same concentration and by blocking of positive
staining using excess purified peptides.
Results
CSF-1R Expression in Mouse Prostate Development and Cancer. Lim-
ited data are available on the expression of CSF-1R in rodent
prostate. One group has reported that a rat prostate cell line is
positive for CSF-1R by microarray analysis, and the level of
expression was found to increase with added androgen (40). RT-
PCR analysis for CSF-1R on a panel of mouse prostate-derived cell
lines showed a low frequency of positive lines (Fig. 7, which is
published as supporting information on the PNAS web site, www.
pnas.org). The midgestational budding of epithelium from the
urogenital sinus first defines the mouse embryonic prostate. IHC
for CSF-1R with a commercial rabbit anti-CSF-1R peptide anti-
serum (Santa Cruz Biotechnology) strongly and uniformly stains
these structures (Fig. 1). Because the gland actively proliferates
after birth, the level of CSF-1R remains high at 3 and 10 days (Fig.
1). Postpuberty at 8 weeks of age, limited cell division is occurring,
and the expression of CSF-1R is reduced to background levels
(Fig. 1). The staining on day 10 from a wild-type mouse is blocked
with competing peptide. Sections of prostate from a day 10
Csf1r
Csf1r
animal show a much lower level of staining. We
also examined liver sections from 8-week-old wild-type and
CSF-1R mice by IHC. Positive strong staining was detected
in Kupper cells of wild-type mice but not in CSF-1Rmice (Fig.
1). Strong staining for CSF-1R was seen in day 10 sections of
prostate harvested from Csf1r
op
Csf1r
op
mice (Fig. 8, which is
published as supporting information on the PNAS web site).
TRAMP is a transgenic mouse model in which the promoter
of the probasin gene controls simian virus 40 T antigen expres-
sion. These mice develop prostatic intraepithelial neoplasia by
1218 and cancer by 1925 weeks of age (33). Immunohisto-
chemical analysis of tissue sections from TRAMP tumors har-
vested between 23 and 30 weeks of age showed moderate to high
expression of CSF-1R in 2025% of the cancerous glands (Fig.
2A). The staining is largely intracellular, in concert with previous
analyses of the membrane versus internal pools of CSF-1R (41).
Background staining was detected in normal prostate glands
from age-matched control mice. After preincubation with the
immunizing peptide, staining for CSF-1R is negative (Fig. 2A).
To verify the specificity of CSF-1R staining in prostate cancer
epithelial cells in TRAMP tumors, single-cell suspensions were
prepared, and CD11b-positive tissue macrophages were de-
pleted by adherence to antibody-conjugated magnetic beads
(Dynabeads; see Materials and Methods). The per-cell level of
CSF-1R was, as expected, much higher in the CD11b-positive
macrophage cell fraction by immunoblotting, as normalized to
the c-abl protein levels (Fig. 2B) or PCR analysis normalized to
-actin levels (Fig. 2C). Interestingly, the CD11b-negative cell
fraction is enriched for expression of the slower migrating
mature glycosylated form (165 kDa) of CSF-1R compared with
Ide et al. PNAS
October 29, 2002
vol. 99
no. 22
14405
MEDICAL SCIENCES
the macrophage-enriched CD11b-positive fraction, which has
roughly equal levels of the 165-kDa form and the immature
mannose-rich 140-kDa form of the receptor (42). A macrophage
cell line, NFS60, predominately shows the mature form.
CSF-1R expression was analyzed in prostate tumors from
PTENmice from 12 to 18 months of age. Moderate diffuse
expression of CSF-1R was detected in the prostate tumors of
these mice compared with age-matched controls (Fig. 9, which
is published as supporting information on the PNAS web site).
Expression of CSF-1R in Human Prostate Cancer Cell Lines and in
Human Prostate Cancer Tissue. Several papers have noted increased
levels of CSF-1R in human breast, ovarian, and uterine cancers and
representative cell lines like BT-20 (24, 25). There are reports of the
expression of CSF-1R in human prostate cancer cell lines PC-3,
DU-145, and LNCaP by RT-PCR (43). Gene expression databases
using microarray data (Unigene, www.ncbi.nlm.nih.govUniGene
Hs.Home.html), EST analyses (GeneCards, Weizmann Institute of
Science, http:兾兾bioinformatics.weizmann.ac.ilcards), or serial
analysis of gene expression (National Center for Biotechnology
Information, www.ncbi.nlm.nih.govSAGE) show moderate to
high levels of expression of CSF-1R sequences in human prostate
cancer-derived samples. It is difficult to evaluate the contribution of
expression from the prostate epithelial component of the tumor
versus the stromal component, which can include tissue macro-
phage-derived cells by such techniques. We evaluated prostate
cancer-derived clonal cell lines and commercially available pros-
tate-derived basaloid epithelial cell populations (PrEC). We could
detect low levels of CSF-1R mRNA expression in LNCaP, DU145,
and PrEC cells by RT-PCR analysis but not in the PC3 or LAPC4
cell lines (Fig. 3A). BT-20 cells are derived from a human breast
Fig. 1. Immunohistochemical analysis of CSF-1R in mouse developing pros-
tate. Tissue sections from 17 days past conception as well as 3 or 10 days and
8 weeks of age after birth of wild-type mice (C57BL6) stained with polyclonal
rabbit anti-CSF-1R antibody (Santa Cruz Biotechnology, lot K130, 1:200 dilu-
tion) based on the ABC method [DAKO or EnVison System (DAKO)]. The
specificity of this antibody was confirmed by serial sections stained with the
antibody after preincubation with immunizing peptide. Ten-day prostate and
liver sections from Csf1rCsf1rmice (23) were also stained with rabbit
anti-CSF-1R antibody at the same dilution.
Fig. 2. CSF-1Rexpression analyses in the prostate tumors of TRAMP mice. (A)
CSF-1R expression in age-matched control mouse prostate and in the prostate
tumor of a TRAMP mouse was examined by IHC with polyclonal rabbit anti-
CSF-1R antibody (Santa Cruz Biotechnology, lot K130, 1:200 dilution) or after
preincubation with immunizing peptide developed with the EnVison System
(DAKO). (B) Immunoblot and (C) RT-PCR analyses of CSF-1R expression in
single-cell suspensions prepared from a TRAMP tumor (23 weeks) before and
after depletion of CD11b-positive macrophages by Dynabead magnetic bead
technology as described in Materials and Methods. The M-NFS-60 cell line was
derived from a myelogenous leukemia (36).
14406
www.pnas.orgcgidoi10.1073pnas.222537099 Ide et al.
cancer and are used as a positive control of CSF-1R expression (30).
All three of the prostate cell preparations positive for CSF-1R
expression were also positive for the CSF-1 ligand RNA, raising the
possibility of an autocrine or paracrine type of stimulation in some
situations. Western blot analysis of extracts from this panel of cell
lines (Fig. 3B) demonstrates the CSF-1R mature-sized protein (165
kDa) in BT-20, DU145, and PrEC cells, with lower levels in LNCaP
and borderline to undetectable levels in the PC3 and LAPC4.
Human prostate cancer tissue specimens were examined by
IHC for CSF-1R expression with rabbit anti-CSF-1R peptide
antiserum (Santa Cruz Biotechnology). Reactivity, shown as red
in cancerous glands, was much higher than in noncancerous
glands in the same field (Fig. 4A). Control rabbit IgG (Fig. 4B)
did not stain serial sections, and preincubation with the immu-
nizing peptide blocked reactivity of the antiserum (Fig. 10, which
is published as supporting information on the PNAS web site).
CSF-1R staining was heterogeneous and predominately cyto-
plasmic, with some membranous staining in prostate cancer (Fig.
4A) and prostatic intraepithelial neoplasia (PIN) (Fig. 4C),
which is consistent with prior reports of breast cancer staining
(44). The presence of CSF-1R was also detected in macrophages
near prostate cancers and within the lumens of some glands. The
level of expression in macrophages was much higher than
prostate cancer cells on a per-cell basis (Fig. 10). We confirmed
the identity of macrophages stained with anti-CSF-1R by double
staining with CD68 (Fig. 4D) (45). The CSF-1R antibody stained
placental syncytiotrophoblasts and villous macrophages at the
same dilution (Figs. 4Eand 10) and was inhibited with the
immunizing peptide (Fig. 4F). This antibody stained tumors
derived from xenographs of PC-3 cells engineered to express
CSF-1R (Fig. 11, which is published as supporting information
on the PNAS web site). A series of rat anti-human CSF-1R
monoclonal antibodies reactive with the ectodomain of the
native receptor (46) were evaluated for IHC but did not recog-
nize the antigen after tissue fixation.
IHC Analysis of CSF-1R Expression in Primary Prostate Cancers Using
Tissue Microarrays. We examined samples from 215 informative
cases, which had not received neoadjuvant therapy, by IHC
analysis using tissue microarrays. Specimen cores with normal
histology (230), benign prostatic hypertrophy (118), PIN (45),
and tumor (716) were evaluated. Sections were scored as unde-
Fig. 3. Expression analyses of CSF-1R in human prostate cancer cell lines. (A)
RT-PCR analysis. The PCR products at representative cycles were shown: CSF-1R,
CSF-1, and prostate-specic antigen, cycle 40 and
-actin, cycle 25. The primers
sequences are listed in Materials and Methods.(B) Immunoblotting analysis.
Protein lysates of each 2 105cells were subjected to SDS gel electrophoresis and
polyclonal rabbit anti-human CSF-1R antibodies (1:250 dilution) were used for
this analysis, as described in Materials and Methods. Human breast cancer cell line
BT-20; human prostate cancer cell lines PC3, DU-145, LNCap, and LAPC4, and
normal basaloid PrEC prostate cells were used in these analyses. PC-3 infected
with human CSF-1R expressing retrovirus was used as a control. Fig. 4. CSF-1R immunostaining in human primary prostate cancer with poly-
clonal rabbit anti-human CSF-1R antibodies (Santa Cruz Biotechnology) based on
the ABC method (DAKO) or the EnVisionSystem (DAKO). (A) Stronger staining
in tumor cells was seen compared with adjacent normal glands. (B) Control IgG
staining on a serial section. (C) Specic immunostaining was observed in the
cytoplasm of PIN as well as malignant epithelial tumor cells. (D) Macrophage
origin dened by CD68 staining (arrow: luminal macrophage in prostatic gland)
using the double-staining method as described in Materials and Methods. CD68
staining used DAB (brown) and CSF-1R staining used Fast red (red purple) as
substrates. (E) CSF-1R staining was detected in brush border of placental syncy-
tiotrophoblasts (arrows) and villous macrophages (arrowheads). (F) After prein-
cubation with the immunizing peptide, the staining for CSF-1R was blocked.
Ide et al. PNAS
October 29, 2002
vol. 99
no. 22
14407
MEDICAL SCIENCES
tectable, weak, or strong-staining. The majority of all specimens
(whether normal or malignant) showed some staining, but a clear
and important trend is the higher percentage of cases of PIN and
prostate cancer with strong staining (Fig. 5). Further analysis will
be needed to accurately correlate such staining results with
clinical and pathological variables. We have noted that the most
intense and uniform staining for lesions within the prostate
occurred in areas of PIN or carcinoma of histological Gleason
grade 3 or 4. Examples of representative tissue microarray IHC
results are shown in Fig. 12, which is published as supporting
information on the PNAS web site.
IHC Staining of Activated CSF-1R in Metastatic Sites by Tyrosine
723-Phosphospecific Peptide Antibody. Prostate cancer can spread to
distant sites, including lymph nodes, soft tissues, and, most com-
monly, bone and bone marrow (47). We used antibodies recogniz-
ing CSF-1R and specialized activation-specific reagents raised
against the phosphorylated tyrosine 723 peptide (PY723) of
CSF-1R (39). Phosphorylation of this tyrosine is important for the
CSF-1R-driven phenotypic traits of anchorage-independent growth
and metastasis (42, 48). Specimens of metastases were obtained
that stained positive for prostate-specific antigen, confirming that
they were of prostate cancer origin (data not shown). In each of the
specimens, carcinoma cell areas showed staining for the CSF-1R
and the PY723 modification of CSF-1R, which was blocked by the
cognate peptide (Fig. 6; also see Fig. 13, which is published as
supporting information on the PNAS web site).
Discussion
Our results correlate the expression of the CSF-1 receptor with
murine prostate development, proliferation, and cancer progres-
sion. Murine prostate carcinoma, initiated by distinct oncogenic
signals, demonstrated enhanced expression of this receptor.
Several cultured human prostate cancer-derived cell lines, pri-
mary cultures of prostate epithelium, and a large proportion of
human prostate cancer specimens, most prominent in PIN
lesions and carcinomas of histological grades 3 and 4, were
positive for CSF-1R expression. The mechanisms regulating this
pattern remain unknown.
Role of the CSF-1 Receptor in Steroid-Regulated Epithelial Develop-
ment. Mammary gland development in Csf1
op
Csf1
op
mice revealed
a lactational defect secondary to incomplete ductal growth, with
precocious development of the lobuloalveolar system (49). Crossing
Csf1
op
Csf1
op
mice with a mammary cancer-susceptible strain of
mice did not affect the incidence or growth of primar y tumors but
delayed their development to invasive metastatic carcinomas asso-
ciated with lessened infiltration and function of tumor-associated
macrophages (50). CSF-1 signaling provides a critical function for
mammary gland development during pregnancy, lactation, and
cancer progression (24).
Prostate development depends on steroid sex hormones, includ-
ing androgen. Androgen may not directly stimulate the prolifera-
tion of normal prostate epithelial cells (10, 11). Paracrine factors,
which are produced by the mesenchyme and regulated by steroid
hormones, play a critical role. Several growth factors that act as
paracrine mediators have been identified, including IGF-I, fibro-
blast growth factor (FGF)-7, FGF-10, HGF, and transforming
growth factor-
(11, 51). Our observations suggest that CSF-1 is a
candidate for such a paracrine factor.
CSF-11R and Bone Metastasis. The most favored site of metastasis
of prostate cancer is bone (47). The ability of prostate cancer to
incite an osteoblastic reaction suggests there are bidirectional
pathways between prostate cancer cells and osteoblasts and
between osteoclasts and bone stromal cells. Prostate cancer
metastases in bone have an extensive bone resorption ability, and
Fig. 5. Immunohistochemical staining distribution of prostate by anti-CSF1R
antibody on tissue microarrays. Two hundred fteen prostatectomy cases
encompassing 1,109 informative tissue spots on three TMAs were used for
analysis grading on a 02 scale (0 negative, 1 weak, and 2 strong
staining). Immunostaining conditions were as described in Materials and
Methods. NL, normal glands; BPH, benign prostatic hyperplasia; PIN, prostatic
intraepithelial neoplasia; PCA, prostate cancer. Tissue spot histology and
grading were conrmed on hematoxylineosin slides.
Fig. 6. Immunohistochemical analysis of CSF-1R and activated CSF-1R expres-
sion in metastatic prostate cancer. Tissue sections from primary and metastatic
prostate cancer (aorta) were stained with polyclonal rabbit anti-CSF-1R (Santa
Cruz Biotechnology) antibody using the EnVisionSystem (DAKO) and polyclonal
rabbit anti-PY723 CSF-1R antibody using both the EnVisionsystem and the
tyramide signal amplication system (NEN). Coincidence of CSF-1R with activated
CSF-1R in some sections was conrmed by serial sections. The specicity of
anti-PY723 CSF-1R antibody was conrmed after preincubation with immunizing
PY723 peptide (10
M) blocked reactivity (39).
14408
www.pnas.orgcgidoi10.1073pnas.222537099 Ide et al.
osteoclastogenesis may play a role in the establishment of bone
metastasis (52).
CSF-1 is an important regulator of hematopoiesis and bone
resorption (19, 20). Studies performed with Csf1
op
Csf1
op
mice
established that CSF-1 function is essential for proliferation and
differentiation of osteoclasts and is locally synthesized by bone
marrow stromal fibroblasts and osteoblasts. In addition to effects
on cell proliferation, differentiation, and invasion, the mem-
brane-bound CSF-1 may mediate adhesion (53).
Other Receptor Tyrosine Kinases Expressed in Prostate Cancer. Sev-
eral protein tyrosine kinases, such as PDGF receptor (PDGF-R),
c-met, HER-2neu, and IGF-1R, are expressed in prostate cancer,
including metastatic sites (2). CSF-1R is more closely related
structurally to the receptors for PDGF than to other PTK receptor
subfamilies (54). Immunohistochemical analyses show moderate to
strong PDGF-R
expression in PIN and primary prostate cancers
but weak or absent expression in nonneoplastic prostate epithelium
and stroma (13). The expression of PDGF-R
protein by metastatic
prostate cancer was also confirmed by IHC in a series of bone
marrow metastases (13). HER-2neu overexpression in primary
prostate cancer and in metastatic sites of prostate tumors was noted
before and after androgen-deprivation therapy (55). HGF is over-
expressed in prostate cancer, and its receptor c-met expression in
prostate cancer has been linked to higher histologic-grade and
-stage disease (56). The finding of activated CSF-1R in metastatic
tumors supports a role for CSF-1CSF-1R signaling in prostate
cancer progression.
We thank Norman Greenberg (Baylor College of Medicine) for providing
TRA MP mice; Dr. Hong Wu (UCLA) for providing PTENmice; Drs.
E. Richard Stanley and Xu-Ming Dai (Albert Einstein College of Medicine)
for Csf1rCsf1rtissues; Martine Roussel (Department of Tumor Cell
Biology, St. Jude Childrens Research Hospital) and Charles Sherr (Howard
Hughes Medical Institute at St. Jude Childrens Research Hospital) for
reagents and valuable advice; J. C. White for preparation of the manuscript
and figures; Yoon Kim, Shirley Quan, Benjamin Rafii, Adam Gottesfeld,
Gregory Ferl, Sheila Tze, and Gregg Kanter for excellent technical assis-
tance; and Drs. Tetsuro Watabe, Robert E. Reiter, and members of the
Witte laboratory for helpful discussions. We thank the members of the
Human Tissue Research Center at UCLA for processing and sectioning of
tissues. We thank Dr. Arie Belldegrun (Department of Urology, UCLA) for
help in providing specimens used in the constr uction of the tissue microarray
used in these studies. Portions of this study were supported by a CaP CURE
grant (to O.N.W.), National Institutes of Health Grant GM08243-13 (to
D.B.S.), and funds from the Tina and Richard Carolan Prostate Research
Fund (to A.P.). O.N.W. is an Investigator and H.I. an Associate of the
Howard Hughes Medical Institute.
1. Blume-Jensen, P. & Hunter, T. (2001) Nature 411, 355365.
2. Djakiew, D. (2000) Prostate 42, 150160.
3. Ware, J. L. (1999) Cancer Metastasis Rev. 17, 443447.
4. Gibbs, J. B. (2000) J. Clin. Invest. 105, 913.
5. Schwaab, T., Lewis, L. D., Cole, B. F., Deo, Y., Fanger, M. W., Wallace, P.,
Guyre, P. M., Kaufman, P. A., Heaney, J. A., Schned, A. R., et al. (2001)
J. Immunother. 24, 7987.
6. Ko, Y. J., Small, E. J., Kabbinavar, F., Chachoua, A., Taneja, S., Reese, D.,
DePaoli, A., Hannah, A., Balk, S. P. & Bubley, G. J. (2001) Clin. Cancer Res.
7, 800805.
7. Bergan, R. C., Waggle, D. H., Carter, S. K., Horak, I., Slichenmyer, W. &
Meyers, M. (2001) Urology 57, 7780.
8. Yu, Q. & Toole, B. P. (1997) Dev. Dyn. 208, 110.
9. Gakunga, P., Frost, G., Shuster, S., Cunha, G., Formby, B. & Stern, R. (1997)
Development (Cambridge, U.K.) 124, 39873997.
10. Abate-Shen, C. & Shen, M. M. (2000) Genes Dev. 14, 2410 2434.
11. Hayward, S. W. & Cunha, G. R. (2000) Radiol. Clin. North Am. 38, 114.
12. Signoretti, S., Waltregny, D., Dilks, J., Isaac, B., Lin, D., Garraway, L., Yang, A.,
Montironi, R., McKeon, F. & Loda, M. (2000) Am. J. Pathol. 157, 1769 1775.
13. Chott, A., Sun, Z., Morganstern, D., Pan, J., Li, T., Susani, M., Mosberger, I.,
Upton, M. P., Bubley, G. J. & Balk, S. P. (1999) Am. J. Pathol. 155, 12711279.
14. Robinson, D. R., Wu, Y. M. & Lin, S. F. (2000) Oncogene 19, 5548 5557.
15. Sherr, C. J., Rettenmier, C. W., Sacca, R., Roussel, M. F., Look, A. T. &
Stanley, E. R. (1985) Cell 41, 665676.
16. Roth, P. & Stanley, E. R. (1992) Curr. Top. Microbiol. Immunol. 181, 141167.
17. Cecchini, M. G., Dominguez, M. G., Mocci, S., Wetterwald, A., Felix, R.,
Fleisch, H., Chisholm, O., Hofstetter, W., Pollard, J. W. & Stanley, E. R. (1994)
Development (Cambridge, U.K.) 120, 13571372.
18. Pollard, J. W. & Stanley, E. R. (1996) Adv. Dev. Biochem. 4, 153193.
19. Cecchini, M. G., Hofstetter, W., Halasy, J., Wetterwald, A. & Felix, R. (1997)
Mol. Reprod. Dev. 46, 7583.
20. Ryan, G. R., Dai, X. M., Dominguez, M. G., Tong, W., Chuan, F., Chisholm,
O., Russell, R. G., Pollard, J. W. & Stanley, E. R. (2001) Blood 98, 7484.
21. Pollard, J. W. (1997) Mol. Reprod. Dev. 46, 5461.
22. Yoshida, H., Hayashi, S. I., Kunisada, T., Ogawa, M., Nishikawa, S., Okamura,
H., Sudo, T., Shultz, L. D. & Nishikawa, S. I. (1990) Nature 345, 442444.
23. Dai, X. M., Ryan, G. R., Hapel, A. J., Dominguez, M. G., Russell, R. G., Kapp,
S., Sylvestre, V. & Stanley, E. R. (2002) Blood 99, 111120.
24. Sapi, E. & Kacinski, B. M. (1999) Proc. Soc. Exp. Biol. Med. 220, 18.
25. Kacinski, B. M. (1997) Mol. Reprod. Dev. 46, 7174.
26. McDermott, R. S., Deneux, L., Mosseri, V., Vedrenne, J., Clough, K.,
Fourquet, A., Rodriguez, J., Cosset, J. M., Sastre, X., Beuzeboc, P., et al. (2002)
Eur. Cytokine Network 13, 121137.
27. Horoszewicz, J. S., Leong, S. S., Chu, T. M., Wajsman, Z. L., Friedman, M.,
Papsidero, L., Kim, U., Chai, L. S., Kakati, S., Arya, S. K. & Sandberg, A. A.
(1980) Prog. Clin. Biol. Res. 37, 115132.
28. Kaighn, M. E., Narayan, K. S., Ohnuki, Y., Lechner, J. F. & Jones, L. W. (1979)
Invest. Urol. 17, 16 23.
29. Stone, K. R., Mickey, D. D., Wunderli, H., Mickey, G. H. & Paulson, D. F.
(1978) Int. J. Cancer 21, 274281.
30. Filder man, A. E., Bruckner, A., Kacinski, B. M., Deng, N. & Remold, H. G.
(1992) Cancer Res. 52, 36613666.
31. Sapi, E., Flick, M. B., Gilmore-Hebert, M., Rodov, S. & Kacinski, B. M. (1995)
Oncogene 10, 529542.
32. Klein, K. A., Reiter, R. E., Redula, J., Moradi, H., Zhu, X. L., Brothman, A. R.,
Lamb, D. J., Marcelli, M., Belldegrun, A., Witte, O. N.,etal.(1997) Nat . Med.
3, 402408.
33. Greenberg, N. M., DeMayo, F., Finegold, M. J., Medina, D., Tilley, W. D.,
Aspinall, J. O., Cunha, G. R., Donjacour, A. A., Matusik, R. J. & Rosen, J. M.
(1995) Proc. Natl. Acad. Sci. USA 92, 34393443.
34. Dubey, P., Wu, H., Reiter, R. E. & Witte, O. N. (2001) Cancer Res. 61,
32563261.
35. Stiles, B., Gilman, V., Khanzenzon, N., Lesche, R., Li, A., Qiao, R., Liu, X. &
Wu, H. (2002) Mol. Cell. Biol. 22, 38423851.
36. Nakoinz, I., Lee, M. T., Weaver, J. F. & Ralph, P. (1990) J. Immunol. 145, 860 864.
37. Era, T. & Witte, O. N. (2000) Proc. Natl. Acad. Sci. USA 97, 17371742.
38. Kononen, J., Bubendorf, L., Kallioniemi, A., Barlund, M., Schraml, P.,
Leighton, S., Torhorst, J., Mihatsch, M. J., Sauter, G. & Kallioniemi, O. P.
(1998) Nat. Med. 4, 767768.
39. Flick, M. B., Sapi, E., Perrott a, P. L., Maher, M. G., Halaban, R., Carter, D.
& Kacinski, B. M. (1997) Oncogene 14, 25532561.
40. Whitacre, D. C., Chauhan, S., Davis, T., Gordon, D., Cress, A. E. & Miesfeld,
R. L. (2002) Cell Growth Differ. 13, 111.
41. Guilbert, L. J. & Stanley, E. R. (1986) J. Biol. Chem. 261, 40244032.
42. Roussel, M. F., Downing, J. R. & Sherr, C. J. (1990) Oncogene 5, 2530.
43. Savarese, D. M., Valinski, H., Quesenberry, P. & Savarese, T. (1998) Prostate
34, 8091.
44. Maher, M. G., Sapi, E., Turner, B., Gumbs, A., Perrotta, P. L., Carter, D.,
Kacinski, B. M. & Haffty, B. G. (1998) Clin. Cancer Res. 4, 18511856.
45. Pulford, K. A., Sipos, A., Cordell, J. L., Stross, W. P. & Mason, D. Y. (1990)
Int. Immunol. 2, 973980.
46. Ashmun, R. A., Look, A. T., Roberts, W. M., Roussel, M. F., Seremetis, S.,
Ohtsuka, M. & Sherr, C. J. (1989) Blood 73, 827837.
47. Scher, H. I. & Yagoda, A. (1987) Am. J. Med. 82, 628.
48. Roussel, M. F., Dull, T. J., Rettenmier, C. W., Ralph, P., Ullrich, A. & Sherr,
C. J. (1987) Nature 325, 549552.
49. Pollard, J. W. & Hennighausen, L. (1994) Proc. Natl. Acad. Sci. USA 91,
93129316.
50. Lin, E. Y., Nguyen, A. V., Russell, R. G. & Pollard, J. W. (2001) J. Exp. Med.
193, 727740.
51. Stein, J., Borzillo, G. V. & Rettenmier, C. W. (1990) Blood 76, 1308 1314.
52. Zhang, J., Dai, J., Qi, Y., Lin, D. L., Smith, P., Strayhorn, C., Mizokami, A.,
Fu, Z., Westman, J. & Keller, E. T. (2001) J. Clin. Invest. 107, 12191220.
53. Uemura, N., Ozawa, K., Takahashi, K., Tojo, A., Tani, K., Harigaya, K., Suzu,
S., Motoyoshi, K., Matsuda, H. & Yagita, H. (1993) Blood 82, 26342640.
54. Hamilton, J. A. (1997) J. Leukocyte Biol. 62, 145155.
55. Osman, I., Scher, H. I., Drobnjak, M., Verbel, D., Morris, M., Agus, D., Ross,
J. S. & Cordon-Cardo, C. (2001) Clin. Cancer Res. 7, 26432647.
56. Humphrey, P. A., Zhu, X., Zarnegar, R., Swanson, P. E., Ratliff, T. L., Vollmer,
R. T. & Day, M. L. (1995) Am. J. Pathol. 147, 386396.
Ide et al. PNAS
October 29, 2002
vol. 99
no. 22
14409
MEDICAL SCIENCES
... In normal mouse prostate, CSF-1R expression is restricted to early development. Though, CSF-1R expression has been reported in mouse and human prostate cancer tissues and cell lines [23,24]. This reexepression in adult cancer cells suggested a potential role of CSF-1R in prostate carcinogenesis. ...
... CSF-1 protein was detected in the culture medium conditioned by C2H parental cells and its three subclones, demonstrating that these cell populations produced and secreted CSF-1R ligand ( Figure 1B). Although the C2H cell line derived from TRAMP tumors that express CSF-1R protein [23] and that Fms transcript was detected in these cells ( Figure 1A), CSF-1R protein was not detected in these cell populations, neither by flow cytometric ( Figure 1C) nor by western blot analyses ( Figure 1D). Thus, murine c-fms cDNA was transduced in the H9, N10, and N18 subclones using a retrovirus vector [29]. ...
... Few studies have investigated the expression of CSF-1R and its ligand in prostate cancer. Ide et al. [23] described CSF-1R expression in the adenoma of the prostate of the TRAMP mouse model of prostate cancer [27] and in human prostate cancer cell lines. Using a panel of the human prostate cancer tissue specimen, they showed by immunohistochemistry that CSF-1R is expressed by almost all specimens with the most intense signal in prostatic intraepithelial neoplasia (PIN) and carcinomas of histological Gleason grade three or four. ...
Article
Full-text available
Prostate cancer is a major public health concern and one of the most prevalent forms of cancer worldwide. The definition of altered signaling pathways implicated in this complex disease is thus essential. In this context, abnormal expression of the receptor of Macrophage Colony-Stimulating Factor-1 (M-CSF or CSF-1) has been described in prostate cancer cells. Yet, outcomes of this expression remain unknown. Using mouse and human prostate cancer cell lines, this study has investigated the functionality of the wild-type CSF-1 receptor in prostate tumor cells and identified molecular mechanisms underlying its ligand-induced activation. Here, we showed that upon CSF-1 binding, the receptor autophosphorylates and activates multiple signaling pathways in prostate tumor cells. Biological experiments demonstrated that the CSF-1R/CSF-1 axis conferred significant advantages in cell growth and cell invasion in vitro. Mouse xenograft experiments showed that CSF-1R expression promoted the aggressiveness of prostate tumor cells. In particular, we demonstrated that the ligand-activated CSF-1R increased the expression of spp1 transcript encoding for osteopontin, a key player in cancer development and metastasis. Therefore, this study highlights that the CSF-1 receptor is fully functional in a prostate cancer cell and may be a potential therapeutic target for the treatment of prostate cancer.
... The same mechanism was observed in ovarian cancer, where the blockade of this autocrine loop reversed the malignant phenotype [19]. Ide et al. reported the involvement of CSF-1R in prostate cancer carcinogenesis [20]. According to the study, low levels of CSF-1R and CSF-1 mRNA were detected in human prostate cancer cell lines, indicating the autocrine activation of the receptor. ...
... According to the study, low levels of CSF-1R and CSF-1 mRNA were detected in human prostate cancer cell lines, indicating the autocrine activation of the receptor. Further analysis indicated the expression of the receptor in prostatic intraepithelial neoplasia (PIN) and metastatic sites, suggesting the role of CSF-1R in prostate tumor development [20]. IHC analysis of prostatic adenocarcinoma indicated that CSF-1R expression was higher in metastatic tissue compared to non-metastatic controls, and the receptor was expressed by both cancer and stromal cells [21]. ...
Article
Full-text available
Simple Summary Several studies have highlighted the importance of the myeloid receptor CSF-1R in the context of tumors as a key player in the generation of an immunosuppressive microenvironment. Since recent research has demonstrated its expression also on the surface of cancer cells, the relevance of CSF-1R in this field has increased. Why might myeloid receptors be expressed by tumor cells? What advantages does CSF-1R expression provide to cancer cells? The aim of this review is to gather available data on CSF-1R expression in cancer cells to provide a new way to consider this receptor. Although previous works demonstrated the pro-tumoral role of CSF-1R in cancer cells in different tumor types, the precise mechanisms regulating its expression in cancer cells are still unknown and need further investigation in order to identify novel tumoral markers and the possible candidate for therapeutic intervention. Abstract Colony-stimulating factor 1 receptor (CFS-1R) is a myeloid receptor with a crucial role in monocyte survival and differentiation. Its overexpression is associated with aggressive tumors characterized by an immunosuppressive microenvironment and poor prognosis. CSF-1R ligands, IL-34 and M-CSF, are produced by many cells in the tumor microenvironment (TME), suggesting a key role for the receptor in the crosstalk between tumor, immune and stromal cells in the TME. Recently, CSF-1R expression was reported in the cell membrane of the cancer cells of different solid tumors, capturing the interest of various research groups interested in investigating the role of this receptor in non-myeloid cells. This review summarizes the current data available on the expression and activity of CSF-1R in different tumor types. Notably, CSF-1R⁺ cancer cells have been shown to produce CSF-1R ligands, indicating that CSF-1R signaling is positively regulated in an autocrine manner in cancer cells. Recent research demonstrated that CSF-1R signaling enhances cell transformation by supporting tumor cell proliferation, invasion, stemness and drug resistance. In addition, this review covers recent therapeutic strategies, including monoclonal antibodies and small-molecule inhibitors, targeting the CSF-1R and designed to block the pro-oncogenic role of CSF-1R in cancer cells.
... The same mechanism was observed in ovarian cancer, where blockade of this autocrine loop reversed the malignant phenotype [19]. Ide et al. reported the involvement of CSF-1R in prostate cancer carcinogenesis [20]. IHC analysis of prostatic adenocarcinoma indicated that CSF-1R expression was higher in metastatic tissue compared to nonmetastatic controls and the receptor was expressed by both cancer and stromal cells [21]. ...
Preprint
Full-text available
Colony-stimulating factor 1 receptor (CFS-1R) is a myeloid receptor with a crucial role in monocyte survival and differentiation. Its overexpression is associated with aggressive tumor characterized by an immunosuppressive microenvironment and poor prognosis. CSF-1R ligands, IL-34 and M-CSF, are produced by many cells in the tumor microenvironment (TME), suggesting a key role for the receptor in the crosstalk between tumor, immune and stromal cells in the TME. Recently, CSF-1R expression was reported at the cell membrane of cancer cell from different solid tumors, capturing the interest of various research group interested in investigating the role of this receptor in non-myeloid cells. This review summarizes current data available on the expression and activity of CSF-1R in different tumor types. Notably, CSF-1R+ cancer cells have been shown to produce CSF-1R ligands, indicating that CSF-1R signaling is positively regulated in autocrine manner in cancer cells. Recent research demonstrated that CSF-1R signaling enhances cell transformation by supporting tumor cell proliferation, invasion, stemness and drug resistance. In addition, this review covers recent therapeutic strategies, including monoclonal antibodies and small molecule inhibitors, targeting the CSF-1R and designed to block the pro-oncogenic role of CSF-1R in cancer cell.
... A variety of growth factor ligands are upregulated in the glioma microenvironment and facilitate cross-talk between glioma cells and TAMs. Colony Stimulating Factor-1 (CSF-1) has been shown to be especially important in mediating this interaction within glioma tumors and blockade of CSF-1/CSF-1R pathway has been very effective preventing metastatic progression in many different preclinical cancer models [10,[12][13][14][15][16][17][18][19]. Using the GL261 model cell line, we ...
Article
Full-text available
Glioblastoma multiforme (GBM) is the most aggressive form of adult brain tumor which is highly resistant to conventional treatment and therapy. Glioma cells are highly motile resulting in infiltrative tumors with poorly defined borders. Another hallmark of GBM is a high degree of tumor macrophage/microglia infiltration. The level of these tumor-associated macrophages/microglia (TAMs) correlates with higher malignancy and poorer prognosis. We previously demonstrated that inhibition of TAM infiltration into glioma tumors with the CSF-1R antagonist pexidartinib (PLX3397) can inhibit glioma cell invasion in-vitro and in-vivo. In this study, we demonstrate an important role for the chemokine receptor CCR1 in mediating microglia/TAM stimulated glioma invasion. Using two structurally distinct CCR1 antagonists, including a novel inhibitor “MG-1-5”, we were able to block microglial activated GL261 glioma cell invasion in a dose dependent manner. Interestingly, treatment of a murine microglia cell line with glioma conditioned media resulted in a strong induction of CCR1 gene and protein expression. This induction was attenuated by inhibition of CSF-1R. In addition, glioma conditioned media treatment of microglia resulted in a rapid upregulation of gene expression of several CCR1 ligands including CCL3, CCL5, CCL6 and CCL9. These data support the existence of tumor stimulated autocrine loop within TAMs which ultimately mediates tumor cell invasion.
... Mounting data suggest that CSF-1R is highly expressed in immune cells, particularly macrophages (30,36). In order to verify whether CSF-1R is mainly expressed in macrophages in COAD, we performed immunofluorescence analysis to investigate the co-localization between CSF-1R and macrophage marker CD68. ...
Article
Full-text available
Background Colony stimulating factor 1 receptor (CSF-1R) is a single channel III transmembrane receptor tyrosine kinase (RTK) and plays an important role in immune regulation and the development of various cancer types. The expression of CSF-1R in colon adenocarcinoma (COAD) and its prognostic value remain incompletely understood. Therefore, we aim to explore the prognostic value of CSF-1R in COAD and its relationship with tumor immunity. Methods CSF-1R expression in a COAD cohort containing 103 patients was examined using immunohistochemistry (IHC). The relationship between CSF-1R expression and clinicopathological parameters and prognosis was evaluated. Dual immunofluorescence staining was conducted to determine the localization of CSF-1R in COAD tissues. Univariate and multivariate Cox regression analysis were performed to evaluate independent prognostic factors. Transcriptomic profiles of CSF-1Rhigh and CSF-1Rlow tumor-associated macrophages (TAMs) were investigated. Gene enrichment analysis was used to explore the signal pathways related to CSF-1R. In addition, the relationship between CSF-1R in tumor microenvironment (TME) and tumor immunity was also studied. Results IHC analysis showed that CSF-1R was overexpressed in COAD, and higher expression was associated with shorter overall survival (OS). Immunofluorescence staining showed that CSF-1R was co-localized with macrophage marker CD68. Univariate and multivariate Cox regression analysis showed that CSF-1R was an independent prognostic factor for COAD. The results of gene enrichment analysis showed that CSF-1R was involved in tumor immune response and regulation of TME. In addition, CSF-1R was significantly correlated with TME, immune cell infiltration, TMB, MSI, Neoantigen, and immune checkpoint molecules. Conclusion CSF-1R can serve as an independent prognostic factor of COAD and promising immunotherapeutic target of COAD.
... Therefore, the epithelial phenotype displayed in the Pb-Csf1 model is induced by Csf1 indirectly via the immune cells. However, Csf1R was also shown expressed by breast and prostate cancer cells and its expression level in cancer cells is correlated with poor prognosis [46,47]. Csf1R is also reported to be expressed by cancer associated fibroblast cells (CAFs). ...
Article
Full-text available
Macrophages are increased in human benign prostatic hyperplasia and prostate cancer. We generate a Pb-Csf1 mouse model with prostate-specific overexpression of macrophage colony-stimulating factor (M-Csf/Csf1). Csf1 overexpression promotes immune cell infiltration into the prostate, modulates the macrophage polarity in a lobe-specific manner, and induces senescence and low-grade prostatic intraepithelial neoplasia (PIN). The Pb-Csf1 prostate luminal cells exhibit increased stem cell features and undergo an epithelial-to-mesenchymal transition. Human prostate cancer patients with high CSF-1 expression display similar transcriptional alterations with the Pb-Csf1 model. P53 knockout alleviates senescence but fails to progress PIN lesions. Ablating epithelial Gp130 but not Il1r1 substantially blocks PIN lesion formation. The androgen receptor (AR) is downregulated in Pb-Csf1 mice. ChIP-Seq analysis reveals altered AR binding in 2482 genes although there is no significant widespread change in global AR transcriptional activity. Collectively, our study demonstrates that increased macrophage infiltration causes PIN formation but fails to transform prostate cells.
Article
Full-text available
Binding affinity is an important factor in drug design to improve drug-target selectivity and specificity. In this study, in silico techniques based on molecular docking followed by molecular dynamics (MD) simulations were utilized to identify the key residue(s) for CSF1R binding affinity among 14 pan-tyrosine kinase inhibitors and 15 CSF1R-specific inhibitors. We found tryptophan at position 550 (W550) on the CSF1R binding site interacted with the inhibitors' aromatic ring in a π–π way that made the ligands better at binding. Upon W550-Alanine substitution (W550A), the binding affinity of trans -(−)-kusunokinin and imatinib to CSF1R was significantly decreased. However, in terms of structural features, W550 did not significantly affect overall CSF1R structure, but provided destabilizing effect upon mutation. The W550A also did not either cause ligand to change its binding site or conformational changes due to ligand binding. As a result of our findings, the π–π interaction with W550's aromatic ring could be still the choice for increasing binding affinity to CSF1R. Nevertheless, our study showed that the increasing binding to W550 of the design ligand may not ensure CSF1R specificity and inhibition since W550-ligand bound state did not induce significantly conformational change into inactive state.
Article
Full-text available
Tumor-associated macrophages (TAMs) constitute a significant component of the tumor microenvironment. This work reviewed the latest progress in comprehending the function of TAMs and their strategies for cancer therapy. TAMs are highly heterogeneous and plastic and exhibit different functional phenotypes in response to different signal stimuli. The emergence of single-cell technologies allows us to revisit their diversity in cancer. When their pro-inflammatory function is activated, antitumor TAMs support and activate adaptive immune cells to eliminate cancer cells through T cell-mediated killing. In the context of cancer, anti-inflammatory TAMs play a variety of pro-tumor functions, such as releasing cytokines to promote the recruitment of bone marrow cells, promoting tumor angiogenesis, and inhibiting cytotoxic T cell function. The plasticity of TAMs makes them a potential tumor therapeutic target, so finally, we updated strategies for targeting TAMs and the TAM-targeting agents currently being evaluated in clinical trials.
Article
Macrophage colony-stimulating factor (M-CSF, also known as CSF1) in tumor tissues stimulates tumor growth and tumor-induced angiogenesis through an autocrine and paracrine action on CSF1 receptor (CSF1R). In the present study, novel bioisosteres of pexidartinib (1) were synthesized and evaluated their inhibitory activities against CSF1R kinase and tumor growth. Among newly synthesized bioisosteres, compound 3 showed the highest inhibition (95.1%) against CSF1R tyrosine kinase at a fixed concentration (1 μM). The half maximal inhibitory concentration (IC50) of pexidartinib (1) and compound 3 was 2.7 and 57.8 nM, respectively. Unlike pexidartinib (1), which cross-reacts to three targets with structural homology, such as CSF1R, c-KIT, and FLT3, compound 3 inhibited CSF1R, c-KIT, but not FLT3, indicating compound 3 may be a more selective CSF1R inhibitor than pexidartinib (1). The inhibitory effect of compound 3 on the proliferation of various cancer cell lines was the strongest in U937 cells followed by THP-1 cells. In the case of cancer cell lines derived from solid tumors, the anti-proliferative activity of compound 3 was weaker than pexidartinib (1), except for Hep3B. However, compound 3 was safer than pexidartinib (1) in terminally differentiated normal cells such as macrophages. Pexidartinib (1) and compound 3 suppressed the production of CSF1 in Hep3B liver cancer cells as well as in the co-culture of Hep3B cells and macrophages. Also, pexidartinib (1) and compound 3 decreased the population ratio of the M2/M1 phenotype and inhibited their migration. Importantly, compound 3 preferentially inhibited M2 phenotype over M1, and the effect was about 4 times greater than that of pexidartinib (1). In addition, compound 3 inhibited maintenance of cancer stem cell population. In a chick chorioallantoic membrane (CAM) tumor model implanted with Hep3B cells, tumor growth and tumor-induced angiogenesis were significantly blocked by compound 3 to a similar extent as pexidartinib (1). Overall, compound 3, a bioisostere of pexidartinib, is an effective dual inhibitor to block CSF1R kinase and CSF1 production, resulting in significant inhibition of tumor growth.
Article
BACKGROUND The predeliction for prostate carcinoma cells to metastasize to bone suggests the hypothesis that bone and/or bone marrow‐derived factors may promote prostate carcinoma cell growth or survival, or serve as chemoattractants for these cells. METHODS We screened three prostate carcinoma cell lines, DU‐145, PC‐3, and LNCaP, for the expression of several hematopoiesis‐associated colony‐stimulating factors (CSFs) and their receptors using RT‐PCR (reverse transcriptase‐polymerase chain reaction) and immunohistochemical methods, and examined their functional effects. RESULTS All of these cell lines express granulocyte‐macrophage colony‐stimulating factor (GM‐CSF) and macrophage colony‐stimulating factor (M‐CSF), and the DU‐145 and PC‐3 lines express stem‐cell factor (SCF), as determined by RT‐PCR and ELISA. Each of these cell lines expresses the receptors for SCF, GM‐CSF, M‐CSF, and granulocyte colony‐stimulating factor (G‐CSF). M‐CSF enhanced the soft‐agar clonogenicity of PC‐3 and DU‐145 cells, and GM‐CSF stimulated all three cell lines. SCF stimulated the clonogenic growth of DU‐145 cells. G‐CSF marginally abrogated the induction of cell death in the PC‐3 and LNCaP cell lines under serum‐free conditions. GM‐CSF and M‐CSF stimulated modest chemotaxis of PC‐3, DU‐145, and LNCaP cells (most prominently in PC‐3 cells). CONCLUSIONS These data suggest that 1) CSFs may be part of a network of paracrine and autocrine loops that modulate prostate carcinoma cell activity, and 2) the growth‐stimulatory, survival‐enhancing, and/or chemotactic actions of bone marrow‐derived CSFs on prostate carcinoma cells may explain in part why bone is a preferential site of prostatic carcinoma metastases. Prostate 34:80–91, 1998. © 1998 Wiley‐Liss, Inc.
Article
The first monoclonal antibodies (MoAbs) to epitopes in the extracellular domain of the human c-fms proto-oncogene product (receptor for the macrophage colony stimulating factor, CSF-1) were used with flow cytometric techniques to study receptor expression on normal human peripheral blood monocytes, bone marrow cells, and leukemic blasts. On normal cells CSF-1 receptors were restricted in their expression to cells of the mononuclear phagocyte lineage. CSF-1 receptors were detected on leukemic blasts from 15 (30%) of 50 children with acute myeloid leukemia, compared with four (15%) of 26 adults. By contrast, detectable CSF-1 receptors were uniformly absent on blasts from 19 children with acute lymphoblastic leukemia. CSF-1 receptors on normal monocytes and myeloid leukemia cells could be induced to downmodulate by incubation with either human recombinant CSF-1 or phorbol esters, confirming that the receptors had functional ligand- binding sites and responded to transmodulation by inducers of protein kinase C. The numbers of receptors per cell and the percentage of positive cases were highest for leukemic blasts with cytochemical and morphological features of monocytes. However, CSF-1 receptors were also detected on a subset of leukemic blast cells with features of granulocytic differentiation (FAB subtypes M1 through M3). Southern blotting analyses of DNA from 47 cases of acute myeloid leukemia demonstrated no rearrangements within the 32 kb of genomic sequences that contain CSF-1 receptor coding exons or in the 50 kb upstream of the first coding exon. Analysis of the upstream region of the c-fms locus revealed that sequences representing the terminal 112 untranslated nucleotides of c-fms mRNA map 26 kb 5′ to the first coding exon, suggesting that at least one c-fms promoter is separated from the receptor coding sequences by a very long intron. Whereas expression of the CSF-1 receptor in myeloid leukemic blasts is not restricted to cells with monocytic characteristics, the apparently aberrant pattern of receptor synthesis in a subset of cases with granulocytic features appears not to be due to chromosomal rearrangements within 50 kb upstream of sequences encoding the receptor.
Article
Secreted forms of macrophage colony-stimulating factor (M-CSF or CSF-1) are generated by proteolytic cleavage of membrane-bound glycoprotein precursors. Alternatively spliced transcripts of the human CSF-1 gene encode at least two different transmembrane precursors that are differentially processed in mammalian expression systems. The larger precursor rapidly undergoes proteolysis to yield the secreted growth factor and does not give rise to forms of CSF-1 detected on the cell surface. By contrast, the smaller human CSF-1 precursor is stably expressed on the plasma membrane where it is inefficiently cleaved to release a soluble molecule. To determine whether the smaller precursor is biologically active on the cell surface, mouse NIH-3T3 fibroblasts expressing the different forms of human CSF-1 were killed by chemical fixation and tested for their ability to support the proliferation of cells that require this growth factor. Only fixed cells expressing human CSF-1 precursors on their surface stimulated the growth in vitro of a murine macrophage cell line or normal mouse bone marrow-derived mononuclear phagocytes. The ability of these nonviable fibroblasts to induce the proliferation of CSF-1-dependent cells was not mediated by release of soluble growth factor, required direct contact with the target cells, and was blocked by neutralizing antiserum to CSF-1. These results demonstrate that the cell surface form of the human CSF-1 precursor is biologically active and indicate that plasma membrane- bound growth factors can functionally interact with receptor-bearing targets by direct cell-cell contact.
Article
Colony-stimulating factor-1 (CSF-1) or macrophage-CSF (M-CSF) is a growth factor involved in the proliferation, differentiation, and activation of cells of the monocyte/macrophage lineage. Its receptor is the homodimeric, tyrosine kinase product of the c-fms proto-oncogene, which contains a so-called kinase insert domain. This review focuses mainly on recent studies of signal transduction events that are initiated on interaction of CSF-1 and its receptor. A summary is given of the tyrosine autophosphorylation sites on c-Fms identified to date, including their interaction with various substrates and their possible significance for signal transduction and cellular function. In addition, the signal transduction pathways that have been identified to lie downstream of activated c-Fms are reviewed. Although it is apparent that there have been many recent significant developments in our understanding of CSF-1 signaling, a number of examples are mentioned of significant discrepancies in the literature, some possible reasons for which can sometimes be offered. It is also apparent that any particular biochemical response or signal transduction pathway, even though widespread in other ligand receptor/cellular systems, including those with similar receptor structures to c-Fms, may not be relevant to CSF-1 signaling. The relevance of any potentially important molecular signaling pathway activated by CSF-1 in cells in vitro will ultimately have to be related to the functions of monocytes/macrophages in vivo.
Chapter
This chapter discusses the pleiotropic roles for colony-stimulating factor-1 (CSF-1) in development defined by the mouse mutation osteopetrotic (op). The mouse mutation op is an inactivating mutation in the CSF-1 gene. Thus, op/op mice have been invaluable in establishing pleiotropic roles for CSF-1 in vivo. Studies with the op/op mouse have established the central role for CSF-1 in osteoclastogenesis and confirmed its central roles in the regulation of mononuclear phagocyte proliferation, survival, and differentiation. Colony stimulating factors (CSFs) induce the proliferation and differentiation of hematopoietic progenitor cells to granulocytes and/or macrophages. Apart from the CSF that specifically stimulates the production of granulocytes, there are three which cause differentiation to macrophages. All of the effects of CSF-1 are mediated by a high affinity cell surface receptor tyrosine kinase that is encoded by the c-fms proto-oncogene product.
Article
The goal of this study was to evaluate, in patients with prostate cancer, the toxicity profle acid biologic activity of the bispecific antibody MDXH210, which has specificity for the non-ligand-binding site of the high-affinity immunoglobulin G receptor (Fc gamma RI) and the extracellular. domain of the HER-2/neu photo-oncogene product. Patients with prostate cancer that expressed HER-2/neu were entered into a phase I dose-escalation trial of MDXH210. Patients received an intravenous infusion MDXH210 during a period of 2 h three times per week for 2 weeks and were monitored for toxicity. Pharmacokinetic and pharmacodynamic parameters were measured and included the biologic end points of monocyte-bound MDXH210, cytokine production, and clinical response. Seven patients were treated with MDXH210 doses ranging from 1 to 8 mg/m(2). In general, MDXH210 was well tolerated, with only mild infusion-related malaise, fever, chills, and myalgias. No dose-limiting toxic effects were observed. Biologic effects included induction of low plasma concentrations of tumor necrosis factor-alpha and interleukin-6 observed immediately after MDXH210 infusion and 70% saturation of circulating monocyte-associated Fc gamma RT with MDXH210 at a dose level of 4 to 8 mg/m(2). Five of six patients had stable prostate-specific antigen levels during the course of 40 days or more. Circulating plasma HER-2/neu levels decreased by 80% at days 12 and 29 (p = 0.03 and 0.06, respectively, by the Wilcoxon signed rank test). MDXH210 can be given safely to patients with HER-2/neu-positive prostate cancer in doses of at least 8 mg/m(2). At the doses studied, biologic activity was demonstrated and characterized by binding of MDXH210 to circulating monocytes, release of monocyte-derived cytokines, a decrease in circulating HER-2/neu, and short-term stabilization of prostate-specific antigen levels.
Article
Colony-stimulating factor 1 (CSF-1) regulates the survival, proliferation, and differentiation of mononuclear phagocytes. It is expressed as a secreted glycoprotein or proteoglycan found in the circulation or as a biologically active cell-surface glycoprotein. To investigate tissue CSF-1 regulation, CSF-1–nullCsf1op/Csf1opmice expressing transgenes encoding the full-length membrane-spanning CSF-1 precursor driven by 3.13 kilobases of the mouse CSF-1 promoter and first intron were characterized. Transgene expression corrected the gross osteopetrotic, neurologic, weight, tooth, and reproductive defects ofCsf1op/Csf1opmice. Detailed analysis of one transgenic line revealed that circulating CSF-1, tissue macrophage numbers, hematopoietic tissue cellularity, and hematopoietic parameters were normalized. Tissue CSF-1 levels were normal except for elevations in 4 secretory tissues. Skin fibroblasts from the transgenic mice secreted normal amounts of CSF-1 but also expressed some cell-surface CSF-1. Also, lacZ driven by the same promoter/first intron revealed β-galactosidase expression in hematopoietic, reproductive, and other tissue locations proximal to CSF-1 cellular targets, consistent with local regulation by CSF-1 at these sites. These studies indicate that the 3.13-kilobase promoter/first intron confers essentially normal CSF-1 expression. They also pinpoint new cellular sites of CSF-1 expression, including ovarian granulosa cells, mammary ductal epithelium, testicular Leydig cells, serous acinar cells of salivary gland, Paneth cells of the small intestine, as well as local sites in several other tissues.
Article
Colony stimulating factor (CSF-1) and its receptor (CSF-1R, product of c-fms proto-oncogene) were initially implicated as essential for normal monocyte development as well as for trophoblastic implantation. However, recent findings have suggested that CSF-1 and CSF-1R might have additional roles in mammary gland development during pregnancy and lactation. Studies with osteopetrotic (op-/op-) mice, which bear a specific mutation that inactivates the CSF-1 gene, demonstrated that op-/op- mothers are incapable of normal milk production due to the incomplete development of their mammary glands during pregnancy. Also, significant increases in the levels of CSF-1 and CSF-1R proteins are observed in the epithelial cells of mammary gland during pregnancy and lactation. In vitro studies investigating the effect of the three major lactogenic hormones (prolactin, insulin, and glucocorticoids) on the expression of CSF-1 and CSF-1R have demonstrated that expression of CSF-1 can be regulated by prolactin and insulin whereas CSF-1R expression is regulated by glucocorticoids. This apparent role for CSF-1/CSF-1R in normal mammary gland development is very intriguing because this receptor/ligand pair has also been found to be important in the biology of breast cancer, where they regulate tumor cell invasion by a urokinase-dependent mechanism. This review aims to summarize recent findings on the role of CSF-1 and its receptor in normal and neoplastic mammary development which may elucidate potential relationships of growth factor-induced biological changes in the breast during pregnancy and tumor progression.