Transcript

~ ) Pergamon Progress in Growth Factor Research, Vol. 6. Nos. 2-4, pp. 503-512, 1995

Copyright © 1996 Elsevier Science Ltd. All rights reserved Printed in Great Britain.

0955-2235/95 $29.00 + .00

0955--2235(95)00025-9

ANTIPROLIFERATIVE ACTIONS OF INSULIN-LIKE GROWTH FACTOR BINDING PROTEIN (IGFBP)-3

IN HUMAN BREAST CANCER CELLS

Youngman Oh,* Zoran Gucev, Lilly Ng, Hermann L. Mi~ller and Ron G. Rosenfeld

Department of Pediatrics, School of Medicine, Oregon Health Sciences University, Portland, OR 97201-3042, U.S.A.

A number of lines of evidence suggest that IGFs are important mitogens in human breast cancer: (1) IGFs are the most potent growth factor in human breast cancer cells; (2) estrogen stimulates expression of IGF-H and the type 1 IGF receptor; and (3) stromal cells express IGFs, which may act in a paracrine manner. Numerous studies have demonstrated that IGFBPs modulate the mitogenic effects o f IGFs in the local environment. In particular, we have recently demonstrated that IGFBP-3 inhibits the growth of Hs578T and MDA-MB-231 human breast cancer cells in an IGF-inde- pendent manner. Further studies revealed the existence of cell surface-associated IGFBP-3 receptors. Receptor binding and the subsequent antiproliferative action of IGFBP-3 was inhibited by IGFs, owing to the formation o f an IGF-IGFBP-3 complex that prevents the binding of lGFBP-3 to its receptors. In addition, exogeneously added soluble heparin or heparan sulfate inhibited the binding of IGFBP-3 to the cell surface in a dose-dependent manner. However, when heparin and heparan sulfate linkages of glycosaminoglycans on the cell surface were enzymatically removed, IGFBP-3 binding was only minimally affected. These data suggest that soluble heparin or heparan sulfate forms a complex with IGFBP-3, thereby inhibiting receptor binding of IGFBP-3, rather than competing with cell-surface glycosaminoglycans for binding of 1GFBP-3.

Additionally, the role of IGFBP-3 in the antiproliferative effects of transforming growth factor (TGF)-~ and retinoic acid (RA) is supported by our observations that: (1) inhibition of lGFBP-3 gene expression using an IGFBP-3 antisense oligodeoxynu- cleotide not only blocks TGF-fl and RA simulation o f IGFBP-3 production by up to 90%, but also inhibits their antiproliferative effects by 40--60%; and (2) treatment with IGF-H and IGF-H analogs diminish TGF-~ effects by blocking TGF-fl induced binding of IGFBP-3 to the cell surface.

Taken together, our results support the hypothesis that IGFBP-3 is an important antiproliferative factor in human breast cancer, acting in an IGF-independent manner in addition to its ability to modulate the binding of lGF peptides to IGF receptors.

Keywords: IGFBP-3, breast cancer, antiproliferation, IGFBP-3 receptor, glycosaminoglycan,

*Correspondence to: Youngman Oh, Ph.D., Tel: (503) 494-1930; Fax: (503) 494-1933.

503

504 Youngman Oh et al.

INTRODUCTION

Breast tumors express variable levels of receptors for steroids [1-3] and growth factors [4--6] and it has been proposed that the ligands for these receptors act through a variety of autocrine and paracrine mechanisms. Studies in vivo and in cell culture systems have implicated a number of steroids and polypeptide growth factors in the regulation of human breast cell replication [7-10] but the relative contribution of each growth factor and the precise mechanism by which each acts are not known. At the present time, several growth factors have also been identified as playing a major role in the regulation of mammary growth, including members of the epidermal growth factor/transforming growth factor a (EGF/TGF-a) family, IGFs, heparin-binding growth factors (fibroblast growth factors (FGFs) and others), and the TGF-/3 family.

THE IGF AXIS IN THE HUMAN MAMMARY SYSTEM

The IGFs have been recognized as major regulators of mammary epithelial cell and breast cancer cell growth [5, 6, 11, 12]. Nevertheless, limited information is currently available on the IGF axis (IGFs, IGFBPs and IGF receptors) in breast cancer. Both IGF-I and IGF-II have been shown to be potent mitogens for a number of breast cancer cell lines in vitro [5, 6, 12-14], and IGF-I and IGF-II, mRNA is detectable in normal and malignant human mammary cells [15, 16]. In addition, studies of the type 1 and type 2 IGF receptors and insulin receptors in breast tumor specimens and breast cancer cell lines, employing both mRNA expres- sion and ligand binding assays, have demonstrated that virtually all the specimens examined express and produce all three receptors [17-20]. Using estrogen-depen- dent breast cancer cells, it has been further demonstrated that the mitogenic effects of both IGF-I and IGF-II are mediated by the type 1 IGF receptor [5, 6].

Breast cancer cells also secrete various types of IGFBPs. The molecular mecha- nisms and biological functions involved in the interaction of the IGFBPs with the IGFs remain unclear, but these molecules appear to regulate the availability of free IGFs for interaction with IGF receptors [21]. The predominant secreted IGFBP appears to correlate with the estrogen receptor status of the cell [22, 23]. Estrogen- non-responsive (ER-negative) cells predominantly secrete IGFBP-3 and IGFBP-4 as major species, and IGFBP-6 as a minor binding protein, whereas estrogen- responsive (ER-positive) cells secrete IGFBP-2 and IGFBP-4 as major species, and IGFBP-3 and IGFBP-5 as minor proteins. These different patterns of IGFBP secre- tion in two different classes of breast cancer cells imply that the IGF system in breast cancer is complex, and that the biological significance and determination of the cellular response of IGFBPs to autocrine, paracrine, or endocrine-derived IGFs may be significantly different, depending on estrogen responsiveness.

IGFBP-3 is the principal IGFBP in adult serum, where it circulates as a 150 kDa complex consisting of IGFBP-3, an acid-labile subunit, and IGF peptide [24, 25]. Its principal role has been postulated to be the transport of IGFs, protecting them from rapid clearance and/or degradation [26, 27]. In human breast cancer cells, expression of IGFBP-3 is hormonally regulated: (1) estrogen inhibits expression of IGFBP-3 in ER-positive cells, whereas antiestrogens stimulate production of

Inhibition of Breast Cancer Growth by IGFBP-3 505

IGFBP-3 [28]; and (2) TGF-fl and RA stimulate IGFBP-3 production [29, 30]. Furthermore, post-translational modification of IGFBP-3 has been observed; specifically, IGFBP-3 can be proteolyzed by proteases such as cathepsin D, prostate-specific antigen (PSA) and plasmin, all of which have been detected in human breast cancer cells [31-36]. In general, IGFBP-3 proteases are postulated to play a role in altering tissue IGF availability by lowering the affinity of IGFBP-3 for its ligand, thereby increasing the availability of IGFs to cell-membrane recep- tors. PSA, for example, has been shown to reverse the inhibitory effect of IGFBP- 3 on stimulated prostate cell growth by cleaving IGFBP-3 and generating IGFBP-3 fragments that exhibit a lower affinity for IGFs [34]. However, the biological sig- nificance and the mechanisms involved in IGFBP-3 proteolysis in human breast cancer cells are unclear.

IGF-INDEPENDENT ACTION OF IGFBP-3 IN THE HUMAN BREAST CANCER CELLS

Series of our previous studies have demonstrated the presence of cell surface- associated IGFBP-3 on Hs578T ER-negative human breast cancer cells [37], and that exogeneous IGFBP-3 specifically binds to the cell surface and inhibits cell monolayer growth, by itself, through an IGF-independent mechanism [38]. Binding of [125I]-IGFBP-3E- cot~ was specific and could not be displaced by unlabeled IGFBP- 1 or fibronectin. As IGFBP-3 possesses a putative 'heparin-binding motif' near its C-terminus, we tested whether the binding of IGFBP-3 to the Hs578T cell surface resulted in interaction with heparin-containing glycosaminoglycans. Exogenously added soluble heparin and heparan sulfate inhibited [125I]-IGFBP-3E.c°Ii binding to the cell surface in a dose-dependent manner with half maximal inhibition at l0 and >_.250 pg m1-1 respectively, whereas chondroitin sulfate A showed no inhibitory effect at concentrations up to 250 /.tg m1-1 (Fig. 1). However, when heparin and heparan sulfate linkages of glycosaminoglycans on the cell surface were enzymati- cally removed by pretreatment with heparinase or heparitinase for 5 h at 37°C, IGFBP-3 binding was minimally affected (Fig. 1). These data suggest that soluble heparin or heparan sulfate forms a complex with IGFBP-3, thereby inhibiting binding of IGFBP-3 to cell-surface protein(s) specific to IGFBP-3, rather than competing with cell-surface glycosaminoglycans for binding of IGFBP-3.

These findings suggested the existence of specific cell-surface association proteins or receptors for IGFBP-3 on Hs578T human breast cancer cells. Furthermore, IGF-I and IGF-II can attenuate the inhibitory effect of IGFBP-3 by forming IGF- IGFBP-3 complexes, thereby preventing cell-surface binding of IGFBP-3 [38]. In addition, we have recently demonstrated the presence of putative IGFBP-3 specific receptors that are capable of mediating the direct inhibitory effect of IGBBP-3 on human breast cancer cell growth [39]. These 20, 26, and 50 kDa putative IGFBP-3 receptors have been further purified by IGFBP-3-anti-IGFBP-3 antibody immuno- affinity membranes.

506 Youngman Oh et al.

120 "0 c 100' 0

"Q~ 80. ? n o ~= 60

m o Z 4O

in

• " 20

Chondroitin sulfate A

~ ~ Heparinase ~ / ~ , I ~ . . . . (pU/ml)(n=3,

~ Heparin (n=41

0 : , ........ ,,,,,,, : : ,,,,,. , I ,,,,,,, I ['""",,,,,,, l : ,,,,,.~ 0.1 1 10 100 1000 10000

Concentrations (l*g/ml)

FIGURE 1. Effect of heparin and heparan sulfate on [I~I]-IGFBP-3 e'c°ti binding to Hs578T monolayers. Cells were grown to confluence 24-multiwell plates and maintained in serum-free media. Cells were incubated for 3 h at 15°C with 50,000 cpm [125I]-IGFBP-3e.c°n in the presence of various concentrations of heparin, heparan sulfate and chondroitin sulfate A. Ceils were then washed, and ceil-associated radio- activity determined. To enzymatically remove heparin and heparan sulfate linkages of glycosaminoglycaus on the cell surface, cells were pretreated with various concentrations of heparinase and heparitinase (data not shown) for 5 h, 27°C.

THE ROLE OF IGFBP-3 IN TGF-~- AND RA-INDUCED GROWTH INHIBITION IN BREAST CANCER CELLS

TGF-fl and RA have been identified as potent antiproliferative factors in human breast cancer cells in vitro. However, TGF-fl and RA have been found to stimulate IGFBP-3 production in different cell systems in which they inhibit proliferation, suggesting that IGFBP-3 may act as a mediator of their antiproliferative effects in human breast cancer cells. In estrogen receptor-negative human breast cancer cells (Hs578T and MDA-MB-231), several lines of evidence support this hypothesis. Our studies have demonstrated that TGF-fl and RA stimulate IGFBP-3 gene expression (2- fold) and production (2-3-fold) prior to their inhibition of cell growth in Hs578T and MDA-MB-231 cells (data not shown). To test more directly whether IGFBP-3 medi- ates TGF-fl and RA action, we employed two different strategies to block the IGFBP- 3 effect: (1) blocking TGF-fl induced IGFBP-3 expression with an IGFBP-3 antisense oligodeoxynucleotide (ODN); and (2) preventing IGFBP-3 binding to the cell surface by treatment with IGFs and IGF-II analogs, thereby blocking IGFBP-3 mediated action [40]. Cells were treated with antisense and sense ODN corresponding to nucleotide positions 2021 to 2040 of the human IGFBP-3 cDNA sequence. Treatment with 10/.tg ml -~ antisense, but not sense, ODN reduced basal IGFBP-3 protein concen- trations by 80% and IGFBP-3 mRNA levels by 60% in Hs578T [40] and MDA-MB- 231 cells (data not shown). Further experiments revealed that inhibition of IGFBP-3 gene expression using an IGFBP-3 antisense O D N not only blocks TGF-fl and RA stimulation of IGFBP-3 production but also inhibits their antiproliferative effects. In Hs578T cells, TGF-fl2-induced IGFBP-3 production was inhibited by as much as 80%, and this was correlated with 70% inhibition of TGF-fl2-induced IGFBP-3 mRNA expression in the presence of 20 pg ml -~ antisense ODN (Fig. 2). Similar effects

Inhibition of Breast Cancer Growth by IGFBP-3 507

IGFBP-3 protein

IGFBP-3 mRNA

actin

+ TGF- ~2 (ng/ml) 0 0 5 5 5 5 5 5 5 + Antisense (ng/ml) + Sense 0 0 0 1 10 20 1 10 20

FIGURE 2. Effect of IGFBP-3 antisense ODN on IGFBP-3 protein and mRNA levels in TGF-~2 - treated Hs578T cells. Upper panel: Western ligand blots of Hs578T CM from cells treated with 1 to 20 #g m1-10DN on day 0 and again on day 1.5 without changing media in the presence of 5 ng m1-1 of TGF-~2. CM was harvested after 3 days of treatment from triplicate wells within each experiment, pooled, and elec- trophoresed. Lower panel: Northern blots demonstrating the effects of ODN on IGFBP-3 mRNA levels in TGF-~2 treated Hs578T cells. Cells were grown until 90% confluent and pretreated with 1 to 20 Ilg ml -I ODN for 4 h. Cells were then washed and treated with ODN in the presence of 5 ng m1-1 of TGF-[12 for 12 h. A representative gel from one of two experiments is shown [40].

of the IGFBP-3 antisense ODN were observed in MDA-MB-231 cells; treatment with 20/zg ml -t antisense ODN resulted in >90% inhibition on TGF-/32-induced IGFBP-3 production and mRNA expression. In addition, RA-induced IGFBP-3 production and mRNA expression were inhibited by as much as 90% in the presence of 1 /zg ml -t IGFBP-3 antisense ODN in MDA-MB-231 cells (data not shown).

After initially ascertaining the ability of the IGFBP-3 antisense ODN to suppress TGF-fl2 and RA-induced IGFBP-3 production, we next investigated the effect of the IGFBP-3 antisense ODN on TGF-132- and RA-induced growth inhibition by treating cells with antisense or sense ODN for 5 days. No significant growth effect was observed by IGFBP-3 antisense ODN in the absence of TGF-fl2 or RA despite inhibition of IGFBP-3 expression, indicating that basal IGFBP-3 levels were not sufficient to exert a growth-inhibitory effect in both cell lines (data not shown). However, when antisense, but not sense, ODN were employed in the presence of TGF-fl2 (Hs578T cells) or RA (MDA-MB-231 cells), antisense ODN attenuated the TGF-fl2- and RA-induced cell growth inhibition by approximately 60% and 40%, respectively (P<0.05) (Fig. 3).

As demonstrated previously, blocking the interaction of exogenously added IGFBP-3 with IGFBP-3 receptors by the addition of IGF peptides resulted in atten- uation of IGFBP-3 inhibition of Hs578T cell growth. Similar effects of IGF peptides on TGF-fl induced cell growth inhibition were observed in Hs578T cells (Fig. 4). Whereas TGF-fl2 alone resulted in a 40% inhibition of cell growth, when coincubated with IGF-II, the inhibitory effect was blocked in a dose-dependent

508 Youngman Oh et al.

A 120'

=100 ' :T_. O :E_- Z 80,

60'

P. O 40, O

2 0

B

.t=

Hs578T cells

I ........................................... * ; ; ; ; ......

iniii 1 5 10 20 30 50 0 1 5 10 20 30 50

+ Antisense (uglml) + Sense (uglml) ]

+ TGF-1~2 (5 ng/ml)

MDA-MB-231 cells

"R

P o~

q . .

o

0 0.1 0.5 1 5 10 20 0 0.1 0.5 1 5 10 20

j + Antisense (pglml) + Sense (pg/ml) [

+ RA 10.5 pM) FIGURE 3. Effect of IGFBP-3 antisense ODN on TGF-~2- and RA-induced growth inhibition. Treatment of Hs578T cells with 5 ng m1-1 TGF-~2 (A) or MDA-MB-231 cells with 0.5 p M RA (B) in the presence of antisense or sense ODN at concentrations ranging from 1 to 50 pg ml -l. The inhibitory effect of TGF-~2 and RA was determined by cell counts after 5 days of treatment. Results represent means _+S.E. of two separate experiments performed in triplicate. *, P<0.05.

manner, with a 90% attenuation of the TGF-fl2 effect at an IGF-II concentration of 200 ng ml -l (P<0.05). Similar results were observed using [LeuZ7]IGF-II which has full affinity for IGFBPs, but significantly reduced affinity for the IGF-I recep- tor [40]. In contrast, co-incubation with similar concentrations of [Gln 6, Ala 7, Tyr 18, Leu 19, Leu 27] IGF-II ([QAYL-L]IGF-II), an IGF analog with 100-fold reduced affinity for both IGFBPs and the IGF-I receptor, did not result in attenuation of the TGF-fl2 inhibitory effect. These results suggest that the attenuating effect of

Inhibition of Breast Cancer Growth by IGFBP-3 509

120 T IGF-II [L27]IGF-II [QAYL-L ] IGF- I I

• 'oolm ...................................... ,.o., . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .

80 'k

o 40 ~ 'k

0~ 21

I I I IGF-II ~ ]IGF-II ~ [QAYL-L]IGF-II

,,' + TGF-p2 ,,' + TGF-p2 ,,' ~ ~ + TGF-p2

FIGURE 4. Effect of IGF-II and IGF-II analogs on TGF-~2 action. Cells were treated with IGF-I1 [Leu27]IGF-II or [QAYL-L]IGF-II at concentrations ranging from 5 to 200 ng ml -l for 5 days [40]. Results represent means +S.E. of three separate experiments performed in triplicate. *, P<0.05.

IGF-II and [Leu27]IGF-II on TGF-fl2 inhibition of cell growth was not mediated through the IGF-I receptor, but rather, reflects the ability of IGF peptides to prevent IGFBP-3 binding to the cell surface.

Our studies indicate that endogenously produced IGFBP-3 is an important direct inhibitor of proliferation of human breast cancer cells, and importantly, that a major mechanism in the inhibitory actions of TGF-fl and RA on these cells appears to be their stimulation of cellular production of IGFBP-3, which then exerts direct antiproliferative effects via interaction with IGFBP-3 specific receptors.

In summary, we have: (1) demonstrated specific and high-affinity association of IGFBP-3 with human breast cancer cell surface; (2) demonstrated that binding of IGFBP-3 to breast cancer cells results in a major IGF-independent antiproliferative effect; (3) shown that transcriptional regulation of IGFBP-3 explains, at least in part, the antiproliferative effects of TGF-fl and RA in ER-negative human breast cancer cells.

Taken together with other studies on ER-positive breast cancer cells that demon- strate that the cell growth effects of estrogen and antiestrogens are also mediated, in part, through modulation of IGFBP-3 expression [42], we hypothesize that IGFBP-3 inhibits human breast cancer growth by acting through its specific recep- tor, in addition to regulating the binding of IGF peptides to classical IGF receptors in human breast cancer cells (Fig. 5). Furthermore, we believe that the IGFBP- 3/IGFBP-3 receptor system exists widely as a growth-suppressing factor in both normal and malignant breast cancer cells and that abnormalities of the IGFBP- 3/IGFBP-3 receptor axis may contribute to the etiology of breast cancer. Therefore, a fuller understanding of IGFBP-3 and the IGFBP-3-receptor interaction will allow us to design IGFBP-3 agonists that have potential therapeutic value. In particular,

510 Youngman Oh et al.

IGFBP-3 Q Q Q (4) IGFBP'3 PrOt'as-

<2) (

g ' I +I a t

retinoic acid

FIGURE 5. Schematic diagram of mechanisms for inhibitory action of IGFBP-3. IGFBP-3 inhibits human breast cancer cell growth by: (1) acting through its own receptor; (2) blocking the binding of IGF peptides to the Type I IGF receptor; (3) acting as a major element for anti-proliferation growth factors, such as TGF-~, retinoic acid; (4) acting as a substrate for IGFBP-3 proteases; and (5) mediating antie- strogen-induced growth inhibition in ER-positive breast cancer cells.

t r e a t m e n t wi th I G F B P - 3 agon i s t s m a y be espec ia l ly v a l u a b l e in E R - n e g a t i v e o r

m u l t i - d r u g res i s tan t b reas t cancers .

R E F E R E N C E S

1. Beatson GT. On the treatment of inoperable cases of carcinoma of the mamma; suggestions for a new method of treatment with illustrative cases. Lancet. 1896; 2: 104-107, 162-165.

2. Osborne CK, Yachinowiz MG, Knight WA, McGuire WL. The value of estrogen and progesterone receptors in the treatment of breast cancer. Cancer. 1980; 46: 2884-2888.

3. Wittliff JL. Steroid-hormone receptors in breast cancer. Cancer. 1984; 53: 630-643. 4. Cullen K J, Yee D, Bates SE, Brunner N, Clarke RE, Dickson RE, Huff KK, Paik S, Rosen N,

Valverius E, Zugmaier G, Lippman ME. Regulation of human breast cancer by secreted growth factors. Acta Oncol. 1989; 28: 835-839.

5. Osborne CK, Coronado EB, Kitten L J, Arteaga CI, Fuqua SAW, Ramashara K, Marshall M, Li CH. Insulin-like growth factor II (IGF-II): a potential autocrine/paracrine growth factor for human breast cancer acting via the IGF-I receptor. Mol Endocrinol. 1989; 3: 1701-1709.

6. De Leon DD, Wilson DM, Powers M, Rosenfeld RG. Effects of insulin-like growth factors (IGFs) and IGF receptors antibodies on the proliferation of human breast cancer cells. Growth Factors 1992; 6: 327-336.

7. Leung BS, Potter AH. Mode of oestrogen action on cell proliferation in CAMA-I cells: sensitivity of GI phase population. J. Cell Biochem. 1987; 34: 213-225.

8. Lippman ME, Dickson RB, Gelmann EP, Rosen N, Knabbe C, Bates S, Bronzert D, Huff KK, Kasid A. Growth regulatory peptide production by human breast carcinoma cells. J Steroid Biochem. 1988; 30:53-61.

9. May FEB, Westley BR. Identification and characterization of estrogen-regulated RNAs in human breast cancer cells. J Biol Chem. 1988; 263: 12901-12908.

Inhibition o f Breast Cancer Growth by IGFBP-3 511

10. Clarke R, Brunner N, Katz D, Glanz P, Dickson RB, Lippman ME, Kern FG. The effects of constitutive expression of transforming growth factor u on the growth of MCF-7 human breast cancer cells in vitro and in vivo. Mol Endocrinol. 1989; 3: 372-380.

11. Furlanetto RW, DiCarlo JN. Somatomedin C receptors and growth effects in human breast cancer cells maintained in long-term culture. Cancer Res. 1984; 44: 2122-2128.

12. Huff KK, Kaufman D, Gabbay KH, Spencer EM, Lippman ME, Dickson RB. Human breast cancer cells secrete an insulin-like growth factor-l-related polypeptide. Cancer Res. 1986; 46: 4613--4619.

13. Westley B, May FEB. IGFs and control of cell proliferation in human breast and other cancers. Rev Endocrine-Related Cancer. 1991; 39: 29-34.

14. Baxter RC, Maitland JE, Raisur RL, Reddel R, Sutherland RL. High molecular weight somatomedin- C (IGF-I) from T47D human mammary carcinoma cells: immunoreactivity and bioactivity. In: Spencer EM, ed. Insulin-like growthfactors/somatomedins. Berlin: De Gruyter; 1983: 615-618.

15. Cullen K J, Smith HS, Hill S, Rosen N, Lippman ME, Yee D. Growth factor mRNA expression by human breast fibroblast from benign and malignant lesions. Cancer Res. 1991; 51: 4978~,985.

16. Paik S. Expression of IGF-I and IGF-II mRNA in breast tissue. Breast Cancer Res Treat. 1992; 22: 31-38.

17. Cullen KJ, Yee D, Sly WS, Perdue J, Hampton B, Lippman ME, Rosen N. Insulin-like growth factor receptor expression and function in human breast cancer. Cancer Res. 1990; 50: 48-53.

18. Bonneterre J, Peyrat JP, Beuscart R, Demaille A. Prognostic significance of insulin-like growth factor receptors in human breast cancer. Cancer Res. 1990; 50: 6931~935.

19. Peyrat JP, Bonneterre J. The type I IGF receptor in human breast diseases. Breast Cancer Res Treat. 1992; 22: 59~57.

20. De Leon DD, Bakker B, Wilson DM, Hintz RL, Rosenfeld RG. Demonstration of insulin-like growth factor (IGF-I and -II) receptors and binding proteins in human breast cancer cell lines. Biochem Biophys Res Commun. 1988; 152: 398-405.

21. Lamson G, Giudice LC, Rosenfeld RG. Insulin-like growth factor binding proteins. Structural and molecular relationships. Growth Factors. 1991; 5: 19-28.

22. Clemmons DR, Camacho-Hubner C, Coronado E, Osborne CK. Insulin like growth factor binding protein secretion by breast cancer cell line: correlation with estrogen receptor status. Endocrinology. 1990; 127: 2679-2685.

23. Figueroa JA, Jackson JG, McGuire WL, Krywicki RF, Yee D. Expression of insulin-like growth factor binding proteins in human breast cancer correlates with estrogen receptor status. J Cell Biochem 1993; 52: 196-205.

24. Rosenfeld RG. Somatomedin action and tissue growth-factor receptors. In: Robbins RJ, Melmed S, eds. Acromegaly. New York: Plenum Press; 1987: 45-53.

25. Oh Y, Muller HL, Neely EK, Lamson G, Rosenfeld RG. New concepts in insulin-like growth factor receptor physiology. Growth Regul. 1993; 3: 113-123.

26. Hintz RL, Liu F. Demonstration of specific plasma protein binding sites for somatomedin. J Clin Endocrinol Metab. 1977; 45: 988-982.

27. Baxter RC, Martin JL. Binding proteins for the insulin-like growth factors: structure, regulation, and function. Prog Growth Factor Res. 1989; 1: 49-56.

28. Pratt SE, Pollak MN. Estrogen and antiestrogen modulation of MCF-7 human breast cancer cell prliferation is associated with specific alterations in accumulation of insulin-like growth factor- binding proteins in conditioned media. Cancer Res. 1993; 53: 5193-5198.

29. Fontana JA, Burrows-Meszu A, Clemmons DR, LeRoith D. Retinoid modulation of insulin-like growth factor binding proteins and inhibition of breast carcinoma proliferation. Endocrinology. 1991; 128: 1115-1122.

30. Martin JL, Coverley JA, Pattison ST, Baxter RC. Insulin-like growth factor-binding protein-3 production by MCF-7 breast cancer cells: Stimulation by retinoic acid and cyclic adenosine monophosphate and differential effects of estradiol. Endocrinology. 1995; 136: 1219-1226.

31. Capony F, Rougeot C, Montcourrier P, Cavailles V, Salazar G, Rochefort H. Increased secretion, altered processing and glycosylation of pro-cathepsin D in human mammary cancer cells. Cancer Res. 1989; 49: 3904-3909.

32. Conover CA, De Leon DD. Acid-activated insulin-like growth factor-binding protein 3-proteolysis in normal and transformed cells. Role of cathepsin D. J Biol Chem. 1994; 269: 7076-7080.

512 Youngman Oh et al.

33. Yu H, Diamandis EP, Sutherland DJA. lmmunoreactive prostate-specific antigen levels in female and male breast tumors and its association with steroid hormone receptors and patient age. Clin Biochem. 1994; 27: 75-79.

34. Cohen P, Graves HCB, Peehl DM, Kamarei M, Giudice LC, Rosenfeld RG. Prostate specific antigen (PSA) is an IGF binding protein-3 (IGFBP-3) protease found in seminal plasma. J Clin Endocrinol Metab. 1993; 75: 1046-1053.

35. Schmitt M, Goretzki L, Janicke F, Calvete J, Eulitz M, Kobayashi H, Chucholowski N, Graeff H. Biological and clinical relevance of the urokinase-type plasminogen activator in breast cancer. Biomed Biochem Acta. 1991; 50: 731-741.

36. Lee DY, Park SK, Yorgin PD, Cohen P, Oh Y, Rosenfeld RG. Alteration of insulin-like growth factor-binding proteins (IGFBPs) and IGFBP-3 protease activity in serum and urine from acute and chronic renal failure. J Clin Endocrinol Metab. 1994; 79: 1376-1382.

37. Oh Y, Muller HL, Pham HM, Lamson G, Rosenfeld RG. Non-receptor mediated, post-transcrip- tional regulation of insulin-like growth factor binding protein (IGFBP)-3 in Hs578T human breast cancer cells. Endocrinology 1992; 131: 3123-3125.

38. Oh Y, Muller HL, Lamson G, Rosenfeld RG. Insulin-like growth factor (IGF)-independent action of IGF-binding protein-3 in Hs578T human breast cancer cells. J Biol Chem. 1993; 268: 14964-14971.

39. Oh Y, Muller HL, Pham HM, Rosenfeld RG. Demonstration of receptors for insulin-like growth factor binding protein-3 on Hs578T human breast cancer cells. J Biol Chem. 1993; 268: 26045-25048.

40. Oh Y, MOiler HL, Ng L, Rosenfeld RG. TGF-/~ induced cell growth inhibition in human breast cancer cells is mediated through IGFBP-3 action. J Biol Chem. 1995; 270: 13589-13592.

41. Oh Y, Muller HL, Zhang H, Ling N, Rosenfeld RG. Synthesis and characterization of IGF- analgos: Applications in the evaluation of IGF receptor function and IGF-independent actions of IGFBPs. In ReRoith D, Raizada MK, eds. Current directions in insulin-like growth factor research. Advances in E.,cperimental Medicine and Biology. New York: Plenum Press; Vol. 343; 1994: 41-54.

42. Pratt SE, Pollak MN. Insulin-like growth factor binding protein 3 (IGFBP-3) inhibits estrogen- stimulated breast cancer cell proliferation. Biochem Biophys Res Commun. 1994; 198: 292-297.


Top Related