WO1992021363A1 - Inhibition of smooth muscle cell proliferation by vitronectin - Google Patents

Inhibition of smooth muscle cell proliferation by vitronectin Download PDF

Info

Publication number
WO1992021363A1
WO1992021363A1 PCT/GB1992/000958 GB9200958W WO9221363A1 WO 1992021363 A1 WO1992021363 A1 WO 1992021363A1 GB 9200958 W GB9200958 W GB 9200958W WO 9221363 A1 WO9221363 A1 WO 9221363A1
Authority
WO
WIPO (PCT)
Prior art keywords
vitronectin
smooth muscle
smc
pdgf
muscle cell
Prior art date
Application number
PCT/GB1992/000958
Other languages
French (fr)
Inventor
Errol Wijelath
Catherine Demoliou-Mason
Sibylle Hess
Klaus Theodore Preissner
Vijay Vir Kakkar
Original Assignee
Thrombosis Research Institute
Max Planck Gesellschaft
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Thrombosis Research Institute, Max Planck Gesellschaft filed Critical Thrombosis Research Institute
Publication of WO1992021363A1 publication Critical patent/WO1992021363A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/39Connective tissue peptides, e.g. collagen, elastin, laminin, fibronectin, vitronectin, cold insoluble globulin [CIG]

Definitions

  • This invention relates to the inhibition of smooth muscle cell proliferation and particularly to substances and compositions for use therein.
  • SMC smooth muscle cells
  • vascular smooth muscle cells have prompted research towards the identification of negative modulators that may regulate normal SMC growth in vivo.
  • Vascular SMC in culture are highly responsive to the chemotactic and mitogenic properties of platelet-derived growth factor (PDGF) and it is believed that the paracrine and autocrine activity of this factor may play a major role in SMC migration from the media into the intima and in intimal SMC proliferation in atheroma development [3,4].
  • PDGF platelet-derived growth factor
  • Recent studies [8] have shown that in atherosclerotic lesions there occurs deposition of a particular adhesion protein, namely vitronectin. Vitronectin is one example of a so-called adhesion protein.
  • Vitronectin is involved particularly in focal adhesion and cell spreading, processes which are essential for wound healing and cellular movement [5,6,7].
  • Vitronectin shares the unique recognition sequence Arg-Gly-Asp (RGD) with the group of substrate adhesion proteins such as fibrinogen, fibronectin, van Willebrand factor and laminin.
  • RGD Arg-Gly-Asp
  • a common characteristic in binding of these proteins to cell surfaces is via the integrins [2], a family of heterodimeric membrane receptors which enable cells to anchor, migrate and make contact with other cells or extracellular matrix components, processes which are important for cell growth and tissue integrity.
  • vitronectin acts as a growth inhibitory modulator of vascular smooth muscle cell proliferation.
  • vitronectin The binding and effect of vitronectin on the response of SMC to PDGF and other mitogens have been characterised.
  • the antagonistic effects of vitronectin on cell proliferation induced by a variety of known growth factors notably that contained in 2% Fetal Calf Serum (FCS) and also PDGF and insulin ⁇ like growth factor (IGF-I), have been identified.
  • FCS Fetal Calf Serum
  • IGF-I insulin ⁇ like growth factor
  • the present invention provides a method of inhibiting smooth muscle cell proliferation, especially vascular smooth muscle cell proliferation, comprising administration of an effective amount of vitronectin.
  • the invention provides vitronectin for use as an antiproliferative agent.
  • the invention also provides pharmaceutical compositions comprising vitronectin together with a pharmaceutically acceptable diluent and/or carrier.
  • vitronectin may exist in either a folded ("native" - as present circulating in plasma) or an unfolded ("extended") conformation.
  • the "extended” form is especially preferred, because of its superior antiproliferative activity.
  • vitronectin As an antiproliferative agent in accordance with the invention, therefore, it will usually be the extended form which is supplied.
  • the extended form of vitronectin is preferably multimeric, e.g. greater than the dimer and more preferably greater than the tetramer. This feature is preferred because of the superior antiproliferative activity of the higher molecular weight species.
  • vitronectin in its native form is naturally present in the body, it is within the scope of the present invention to generate the extended form of vitronectin in situ by the action of a suitable agent which affects the equilibrium of the multimeric vitronectin species, for example proteoglycans, glycosaminoglycans and similarly acting agents and/or agents which are generated upon activation of the coagulation/fibrinolysis cascade.
  • a suitable agent which affects the equilibrium of the multimeric vitronectin species, for example proteoglycans, glycosaminoglycans and similarly acting agents and/or agents which are generated upon activation of the coagulation/fibrinolysis cascade.
  • a method of inhibiting smooth muscle cell proliferation comprising generating the extended form of vitronectin in situ by administration of an agent which converts native vitronectin to its extended form.
  • the known vitronectin structure may vary by substitution, deletion or addition at one or more amino acid residues, so long as it retains essentially the same biological activity.
  • Vitronectin for use in accordance with the invention may be provided in any suitable form appropriate to the protocol of administration and/or the needs of a patient.
  • compositions comprising vitronectin together with one or more pharmaceutically acceptable carriers and/or diluents.
  • Suitable carriers/diluents are well known in the art and include saline or other sterile aqueous media, optionally including additional components such as buffer salts and preservatives.
  • vitronectin for administration may be provided in lyophilised or freeze dried solid forms.
  • Administration may be via any suitable protocol.
  • administration is by intravenous injection or infusion, and may be systemic or topical.
  • vitronectin as an antiproliferative agent is in such an amount as to give the desired effective result of inhibiting SMC proliferation at the intended site.
  • a quantity which constitutes an "effective" amount may depend upon various parameters, such as body weight of the patient, degree of inhibition required, intended site of activity, all of which will be well understood and appreciated by persons skilled in the art.
  • an amount of vitronectin will be administered which gives a concentration in plasma of from about 1 to about 100 mg ml" - , more preferably from about 1 to about 10 mg ml" - .
  • the present invention provides vitronectin for use in kits and assays, as well as such kits and assays, for example for screening individuals for those with a high risk of atherosclerosis. This may be achieved for example by exploiting the inhibitory effect of vitronectin on PDGF- induced SMC proliferation and observing the antagonistic effect of blood components, e.g. fibrinogen, on the degree of cell proliferation inhibition by vitronectin. (Such blood components bind vitronectin, thereby preventing it from performing its inhibitory function. )
  • blood components e.g. fibrinogen
  • a further example of the use of the present invention as a diagnostic tool is in the in vitro testing for whether a patient will react to antiproliferative treatment with vitronectin.
  • Figure la illustrates the inhibitory effect of vitronectin on vascular SMC proliferation induced by various growth factors
  • Figure lb illustrates the anti-mitogenic effect of vitronectin on PDGF-stimulated SMC when pre-added and post-added at various times;
  • Figure lc illustrates the differences in SMC antiproliferative activity of the native and extended forms of vitronectin
  • Figure 2a shows the dose-response profile of vitronectin on PDGF-induced DNA synthesis
  • Figure 2b shows the electrophoresis profiles of native and extended vitronectin in the presence or absence of SDS
  • Figure 2c is a plot of total binding of radioisotope-labelled vitronectin (native and extended forms) to SMC monolayers;
  • Figure 2d illustrates the inhibition of binding of radioisotope-labelled vitronectin (extended form) to SMC in suspension by various peptides
  • Figure 3a illustrates vitronectin inhibition of PDGF-induced DNA synthesis in human vascular SMC
  • Figure 3b shows a Northern blot illustrating the absence of vitronectin inhibition of PDGF-induced mRNA expression of the c-fos proto-oncogene.
  • Vascular SMC were isolated from sections of aortic or vein tissue removed during surgery and transferred into DMEM supplemented with 100 IU ml" * ⁇ penicillin, lOO ⁇ g ml" 1 streptomycin sulphate, 100 ⁇ g ml" l neomycin and 2.5 ⁇ g ml" • ⁇ fungizone (Gibco). Dissected medial tissue was finely minced, rinsed and digested under constant agitation (18-20 h, 37 ⁇ C) in DMEM 15 mM Hepes (Flow), 10% FCS with 0.1% collagenase Type II (Worthington) and 0.05% elastase Type I (Sigma).
  • Figure 1(b) illustrates SMC stimulation with 10 ng ml" 1 PDGF-BB in serum-free medium in the presence of 0.1 uM vitronectin (VN) added at the indicated times before (pre-addition) or after (post-addition) PDGF.
  • VN vitronectin
  • vitronectin had no effect on mitogen- induction of DNA synthesis. This indicates that the response to vitronectin may depend on the differential expression of vitronectin receptor(s) and/or of other cellular components) with cell growth.
  • vitronectin Although the concentration of native (i.e. folded) vitronectin in serum is high, serum-vitronectin is not anti-mitogenic suggesting that it is in an inactive state or associated with other serum components that may neutralize its anti-mitogenic activity. Only the extended (i.e. unfolded) form of vitronectin which is considered to be similar to that generated upon activation of the coagulation/fibrinolysis cascade or to that associated with extracellular matrix (ECM) components [9,10], appeared to have the preferential conformational state required for the transmission of this inhibitory effect. Purified human "native" vitronectin which is believed to be in the folded conformation [11] and constitutes the major form present in plasma [1], was considerably less active.
  • ECM extracellular matrix
  • PDGF-BB (10 ng ml" - ) 1200 ⁇ 100
  • the minimum concentration of cRGD that could be used without causing the cells to detach within the time course required to measure DNA synthesis was 1.0 ⁇ M. It could not be evaluated, therefore, whether the cRGD peptide is exerting its antagonistic effect by acting as a competitive inhibitor as in the case of cell adhesion to vitronectin [13].
  • the growth inhibitory effect of vitronectin may be transmitted via secondary interactions through other domains in addition to the RGD site, the kinetics of which may differ.
  • vitronectin Since none of the other adhesive proteins tested, including fibronectin, collagen or fibrinogen were able to inhibit PDGF-induced DNA synthesis in confluent, quiescent SMC, it would suggest that the anti-mitogenic activity of vitronectin is transmitted via receptor(s) that recognize, in addition to the RGD site, specific regions of the vitronectin molecule such as the heparin binding domain.
  • Figure 2(a) illustrates the dose-dependent inhibition of PDGF-BB-induced DNA synthesis in SMC by vitronectin.
  • SMC were stimulated with 10 ng ml" - PDGF-BB in the presence of the indicated concentrations of vitronectin.
  • PDGF-induced [ 3 H]thymidine uptake in the absence of vitronectin was 1000 +_ 110 dpm.
  • Vitronectin alone had no effect on basal DNA synthesis.
  • the dose-response profile of vitronectin (0.1 nM- 0.1 ⁇ M) was biphasic, as shown in Figure 2(a).
  • This biphasic response may reflect concentration-dependent differences in the anti-mitogenic or adhesive properties of the conformational states of oligomeric species formed as a result of exposure of epitopes in the extended form of vitronectin in agrement with the conformational lability/flexibility of this molecule [15,16].
  • Figure 2(c) illustrates the time-dependent differential association of extended (•) and native (o) [ 125 I]vitronectin 3.5-6.0 nM with SMC monolayers at 37 ⁇ C. While native [ 125 1]vitronectin did not demonstrate appreciable binding, extended (multimeric) [ 125 i]vitronectin binding to cell surface receptors was considerably higher and appeared to reach steady state within 2 h. Since approximately 50% of extended [ i25 1]vitronectin binding to SMC monolayer was due to binding to ECM, the effects of cRGD and heparin on vitronectin binding to cell surface were assessed with cells in suspension.
  • Figure 2(d) shows the inhibition of [ 125 I]vitronectin (extended form) (6.0 nM) binding to SMC in suspension at 37 ⁇ C in the absence (A) or presence (B) of 100-fold molar excess unlabelled vitronectin; 1 ⁇ m cRGD (C) or 10 ⁇ g ml" *• unfractionated heparin (Sigma) (D).
  • FIG. 2(d) shows, one hundred-fold excess of unlabelled vitronectin suppressed [ 12 I]vitronectin binding to SMC surface by more than 60%, indicating the specificity of the vitronectin-SMC interactions. Furthermore, [ 125 I]vitronectin binding was suppressed by 34% using the cRGD peptide (1 ⁇ M) or by 50% using unfractionated heparin (10 ⁇ g ml" * - ), suggesting the involvement of both the RGD and heparin binding domains in the interactions of vitronectin with SMC cell surface receptors.
  • the inhibition of [- 2 - I]vitronectin binding by heparin may be due to heparin binding and changes in the vitronectin conformation and also the monomer-oligomer equilibrium which may effect the concentration of active vitronectin species.
  • Our preliminary studies have shown that the two molecules may neutralize each other's anti-mitogenic activity.
  • vitronectin and heparin may be of importance in the regulation of cell growth, especially since heparin has been shown to inhibit mitogen and, in particular, PDGF-induced SMC proliferation [19,20] .
  • vitronectin inhibits dose-dependent PDGF-induction of DNA synthesis in human vascular smooth muscle cells without affecting c-fos mRNA expression.
  • Figure 3(b) shows a Northern blot of equal amounts (20 ⁇ g) of total cellular RNA hybridized with a 2 kb c-fos probe or 0.53 kb G6PD probe.
  • Vitronectin was able to inhibit DNA synthesis over a range of • concentrations of PDGF-BB (0.1-100 ng ml" x ), and at all concentrations the extent of inhibition did not vary (Fig. 3a). This effect was not due to binding and depletion of PDGF since vitronectin did not inhibit the induction of mRNA expression of the c-fos proto-oncogene by PDGF (Fig. 3b). These data also indicate that vitronectin may inhibit growth by affecting mechanisms other than those involved in the expression of early biochemical events induced by growth factors.
  • Vitronectin may prevent growth factor- induced medial-SMC proliferation either directly via integrin/proteoglycan receptor activation or indirectly by preventing access of growth factors and chemo- attractants to SMC as well as cell migration via inhibition of ECM degradation by proteases.
  • Generation of the extended form of circulating vitronectin at sites of vascular injury upon activation of the coagulation/fibrinolysis cascade may be of physiological significance to limit the early proliferative stages of atheroma development or in intimal hyperplasia, since both of these events involve vascular SMC migration and proliferation.

Abstract

Vitronectin, an adhesion protein, acts as a growth inhibitory modulator of vascular smooth muscle cell proliferation, a phenomenon which is known to cause atherosclerosis. The invention provides a method of inhibiting smooth muscle cell proliferation comprising administration of vitronectin, as well as vitronectin for such use and pharmaceutical compositions containing vitronectin. Preferably, the vitronectin is the extended form thereof.

Description

INHIBITION OF SMOOTH MUSCLE CELL PROLIFERATION BY VITRONECTIN
BACKGROUND OF THE INVENTION
This invention relates to the inhibition of smooth muscle cell proliferation and particularly to substances and compositions for use therein.
Background literature material, the disclosures of which are incorporated herein by reference, are referred to herein by parenthetical numerical citation in the text to the appended bibliography.
The hardening of human arteries caused by rapid and uncontrolled division of smooth muscle cells (hereinafter SMC) in the middle to outer- arterial layers ( "atherosclerosis") is a well recognised phenomenon and leads to narrowing of the arteries and increased chances of blood clot formation. This effect is a major cause of myocardial infarction in coronary patients and can for example lead also to gangrene in the case of lower limbs.
The association between atherosclerosis and growth factor-induced proliferation of vascular smooth muscle cells has prompted research towards the identification of negative modulators that may regulate normal SMC growth in vivo. Vascular SMC in culture are highly responsive to the chemotactic and mitogenic properties of platelet-derived growth factor (PDGF) and it is believed that the paracrine and autocrine activity of this factor may play a major role in SMC migration from the media into the intima and in intimal SMC proliferation in atheroma development [3,4]. Recent studies [8] have shown that in atherosclerotic lesions there occurs deposition of a particular adhesion protein, namely vitronectin. Vitronectin is one example of a so-called adhesion protein. It is a multifunctional glycoprotein present in blood and in various body tissues and plays a major role in cellular adhesion, immune defense mechanisms and heamostasis [1] . Vitronectin is involved particularly in focal adhesion and cell spreading, processes which are essential for wound healing and cellular movement [5,6,7].
Vitronectin shares the unique recognition sequence Arg-Gly-Asp (RGD) with the group of substrate adhesion proteins such as fibrinogen, fibronectin, van Willebrand factor and laminin. A common characteristic in binding of these proteins to cell surfaces is via the integrins [2], a family of heterodimeric membrane receptors which enable cells to anchor, migrate and make contact with other cells or extracellular matrix components, processes which are important for cell growth and tissue integrity.
As a result of in vitro studies to assess whether vitronectin may affect SMC growth, we have now discovered that vitronectin acts as a growth inhibitory modulator of vascular smooth muscle cell proliferation.
This inhibitory effect of vitronectin extends also to the proliferation of 3T3-fibroblast cells.
The binding and effect of vitronectin on the response of SMC to PDGF and other mitogens have been characterised. In particular, the antagonistic effects of vitronectin on cell proliferation induced by a variety of known growth factors, notably that contained in 2% Fetal Calf Serum (FCS) and also PDGF and insulin¬ like growth factor (IGF-I), have been identified.
SUMMARY OF THE INVENTION
Accordingly, in one aspect the present invention provides a method of inhibiting smooth muscle cell proliferation, especially vascular smooth muscle cell proliferation, comprising administration of an effective amount of vitronectin.
In another aspect, the invention provides vitronectin for use as an antiproliferative agent.
The invention also provides pharmaceutical compositions comprising vitronectin together with a pharmaceutically acceptable diluent and/or carrier.
As described and discussed more fully hereinbelow, vitronectin may exist in either a folded ("native" - as present circulating in plasma) or an unfolded ("extended") conformation. For use in the present invention, the "extended" form is especially preferred, because of its superior antiproliferative activity.
When administering vitronectin as an antiproliferative agent in accordance with the invention, therefore, it will usually be the extended form which is supplied.
In accordance with this preferred aspect of the invention, the extended form of vitronectin is preferably multimeric, e.g. greater than the dimer and more preferably greater than the tetramer. This feature is preferred because of the superior antiproliferative activity of the higher molecular weight species.
However, because vitronectin in its native form is naturally present in the body, it is within the scope of the present invention to generate the extended form of vitronectin in situ by the action of a suitable agent which affects the equilibrium of the multimeric vitronectin species, for example proteoglycans, glycosaminoglycans and similarly acting agents and/or agents which are generated upon activation of the coagulation/fibrinolysis cascade.
Accordingly, in a further aspect of the present invention there is provided a method of inhibiting smooth muscle cell proliferation, comprising generating the extended form of vitronectin in situ by administration of an agent which converts native vitronectin to its extended form.
Within the scope of this invention, the known vitronectin structure may vary by substitution, deletion or addition at one or more amino acid residues, so long as it retains essentially the same biological activity.
Vitronectin for use in accordance with the invention may be provided in any suitable form appropriate to the protocol of administration and/or the needs of a patient.
Particularly preferred is a composition comprising vitronectin together with one or more pharmaceutically acceptable carriers and/or diluents. Suitable carriers/diluents are well known in the art and include saline or other sterile aqueous media, optionally including additional components such as buffer salts and preservatives.
Alternatively, vitronectin for administration, together with or without pharmaceutically acceptable adjuncts as mentioned above, may be provided in lyophilised or freeze dried solid forms.
Administration may be via any suitable protocol. Preferably, administration is by intravenous injection or infusion, and may be systemic or topical.
In accordance with the present invention, administration of vitronectin as an antiproliferative agent is in such an amount as to give the desired effective result of inhibiting SMC proliferation at the intended site. Thus, a quantity which constitutes an "effective" amount may depend upon various parameters, such as body weight of the patient, degree of inhibition required, intended site of activity, all of which will be well understood and appreciated by persons skilled in the art.
Generally, an amount of vitronectin will be administered which gives a concentration in plasma of from about 1 to about 100 mg ml" - , more preferably from about 1 to about 10 mg ml" - .
In a further aspect the present invention provides vitronectin for use in kits and assays, as well as such kits and assays, for example for screening individuals for those with a high risk of atherosclerosis. This may be achieved for example by exploiting the inhibitory effect of vitronectin on PDGF- induced SMC proliferation and observing the antagonistic effect of blood components, e.g. fibrinogen, on the degree of cell proliferation inhibition by vitronectin. (Such blood components bind vitronectin, thereby preventing it from performing its inhibitory function. )
A further example of the use of the present invention as a diagnostic tool is in the in vitro testing for whether a patient will react to antiproliferative treatment with vitronectin.
The various aspects of the present invention will be further described by and better understood from the following detailed description, taken in conjunction with the accompanying drawings.
BRIEF DESCRIPTION OF THE DRAWINGS
Figure la illustrates the inhibitory effect of vitronectin on vascular SMC proliferation induced by various growth factors;
Figure lb illustrates the anti-mitogenic effect of vitronectin on PDGF-stimulated SMC when pre-added and post-added at various times;
Figure lc illustrates the differences in SMC antiproliferative activity of the native and extended forms of vitronectin;
Figure 2a shows the dose-response profile of vitronectin on PDGF-induced DNA synthesis;
Figure 2b shows the electrophoresis profiles of native and extended vitronectin in the presence or absence of SDS;
Figure 2c is a plot of total binding of radioisotope-labelled vitronectin (native and extended forms) to SMC monolayers;
Figure 2d illustrates the inhibition of binding of radioisotope-labelled vitronectin (extended form) to SMC in suspension by various peptides;
Figure 3a illustrates vitronectin inhibition of PDGF-induced DNA synthesis in human vascular SMC;
Figure 3b shows a Northern blot illustrating the absence of vitronectin inhibition of PDGF-induced mRNA expression of the c-fos proto-oncogene.
DETAILED DESCRIPTION OF THE INVENTION
In relation to Figures 1(a), 1(b) and 1(c), the following experimental procedures were used.
Experimental No. 1
Vascular SMC were isolated from sections of aortic or vein tissue removed during surgery and transferred into DMEM supplemented with 100 IU ml"*■ penicillin, lOOμg ml"1 streptomycin sulphate, 100 μg ml" l neomycin and 2.5 μg ml"•■ fungizone (Gibco). Dissected medial tissue was finely minced, rinsed and digested under constant agitation (18-20 h, 37βC) in DMEM 15 mM Hepes (Flow), 10% FCS with 0.1% collagenase Type II (Worthington) and 0.05% elastase Type I (Sigma). Cells were cultured in L-glutamine supplemented DMEM/ Hepes, 20% FCS plus antibiotics at 37βC in 10% C02 humidified incubators. FCS was reduced to 10% after the second passage. Cells grown to confluency were dissociated with 0.05% trypsin/0.02% EDTA and subcultured at a split ratio of 1:3. Experiments were carried out between passage 2-8 with cells characterized as SMC by their morphology and SMC specific <*<-actin antigen expression. For DNA synthesis assays SMC were plated in 96-well plates at lxlO4 cells/well in DMEM/Hepes supplemented with 10% FCS and antibiotics. 72 h later, cells were growth-arrested for 48 h by reducing FCS to 0.5%. Induction of DNA synthesis by growth activators was subsequently measured 24 h later (32 h for thrombin) in serum-free DMEM/Hepes by pulse labelling with 1 μCi [3H]thymidine (Amersha ) 6h prior to harvesting cells on a Brandel cell harvester using GF/B (Whatman) filters. Thrombin-specific activity was 3,000 U ml"1. The extended form of human vitronectin was obtained after extensive dialysis of 6 M urea-treated "native" vitronectin [12]. Unless indicated otherwise cells were treated with vitronectin for 1 h prior to mitogen addition.
As shown in Figure la, treatment of growth- arrested, confluent human vascular SMC with O.luM human vitronectin inhibited DNA synthesis induced by various growth factors, namely: foetal calf serum (FCS) 2%; transforming growth factor -beta ( TGF- S ) 10 ng ml' - ; PDGF-BB 10 ng ml" - ; basic fibroblast growth factor (bFGF) 10 ng ml"1 (all from Bachem); or -<-thrombin 5.0 U ml"1 (from Sigma). "-" represents in the absence of vitronectin; "+" represents in the presence thereof. This inhibition occurred at a concentration of vitronectin (0.1 uM) well below the physiological range (3-6 μM; 0.25-0.45 mg/ml of plasma) in circulation, and was greater than 50% (see the Figure for the exact percentage inhibition with respect to each growth factor) for all the mitogens tested. Similar results were obtained with aortic SMC. Vitronectin on its own had no effect on basal DNA synthesis.
Figure 1(b) illustrates SMC stimulation with 10 ng ml"1 PDGF-BB in serum-free medium in the presence of 0.1 uM vitronectin (VN) added at the indicated times before (pre-addition) or after (post-addition) PDGF. The arrow indicates addition of PDGF and vitronectin simultaneously.
The anti- itogenic response was observed only if vitronectin was added to the cells before or together with the growth factor, but not after the cells had been activated by the mitogen, suggesting a close association between mitogen-induced intracellular signal transduction and regulation of the cellular mechanisms involved in the anti-mitogenic response to vitronectin.
In sparsely plated, quiescent SMC, it was also found that vitronectin had no effect on mitogen- induction of DNA synthesis. This indicates that the response to vitronectin may depend on the differential expression of vitronectin receptor(s) and/or of other cellular components) with cell growth.
Figure 1(c) illustrates the results of SMC incubation in serum-free medium (A) or stimulation with 10 ng ml-1 PDGF-BB (B) in the presence of 0.1 uM "extended" vitronectin (C) or 0.1 uM "native" vitronectin (D). Values represent the mean + SEM (n=4) of one of four representative experiments.
Although the concentration of native (i.e. folded) vitronectin in serum is high, serum-vitronectin is not anti-mitogenic suggesting that it is in an inactive state or associated with other serum components that may neutralize its anti-mitogenic activity. Only the extended (i.e. unfolded) form of vitronectin which is considered to be similar to that generated upon activation of the coagulation/fibrinolysis cascade or to that associated with extracellular matrix (ECM) components [9,10], appeared to have the preferential conformational state required for the transmission of this inhibitory effect. Purified human "native" vitronectin which is believed to be in the folded conformation [11] and constitutes the major form present in plasma [1], was considerably less active.
The specificity of the growth-inhibitory effect of vitronectin was confirmed by the attenuation of its activity in the presence of an anti-vitronectin antibody [12]. The results are shown in Table 1 below. Further, the transmission of this effect via RGD-dependent interactions with receptor(s) on the cell surface was indicated in the inhibition by the cyclic RGD (cRGD)- peptide which specifically recognizes the vitronectin integrin receptor [13].
Table 1. Inhibition of the antiproliferative effect of vitronectin*
DNA synthesis Condition [3H]thymidine incorporation (dpm)
No addition 180 + 15
PDGF-BB (10 ng ml" - ) 1200 ± 100
PDGF-BB + VN (0.1 μM) 420 +.30
PDGF-BB + VN + cRGD (2 μM) 780 + 38
PDGF-BB + VN + VN antibody (1/10,000) 982 ± 45
*SMC were stimulated with PDGF-BB in the presence of vitronectin (VN) and either the cyclic GPenGRGDSPCA peptide [13] (cRGD) or a vitronectin specific polyclonal antibody [12]. DNA synthesis was measured as in Fig. 1. Neither the cRGD peptide nor the vitronectin antibody had an effect on basal or PDGF-induced [3H]thymidine incorporation. Results are the mean SEM (n=5) of a single experiment representative of two others.
The minimum concentration of cRGD that could be used without causing the cells to detach within the time course required to measure DNA synthesis was 1.0 μM. It could not be evaluated, therefore, whether the cRGD peptide is exerting its antagonistic effect by acting as a competitive inhibitor as in the case of cell adhesion to vitronectin [13]. Alternatively, the growth inhibitory effect of vitronectin may be transmitted via secondary interactions through other domains in addition to the RGD site, the kinetics of which may differ.
Such interactions that lead to a highly active vitronectin-integrin receptor association have been suggested recently from competition studies of [- - - 1]vitronectin binding to purified integrin receptor (άv /$3 ) and may explain the partial inhibition by the cRGD peptide observed here [14] .
Since none of the other adhesive proteins tested, including fibronectin, collagen or fibrinogen were able to inhibit PDGF-induced DNA synthesis in confluent, quiescent SMC, it would suggest that the anti-mitogenic activity of vitronectin is transmitted via receptor(s) that recognize, in addition to the RGD site, specific regions of the vitronectin molecule such as the heparin binding domain.
In relation to Figures 2(a) to 2(d), the following experimental procedures were used. Experimental No. 2
For the time course studies, human vein SMC plated in 48-well plates (3 x 104 cells per well) were growth arrested when confluent as in Fig. 1. After incubation with either form of [125 I]vitronectin (1.0 x 106 cpm ml"1 ) in 175 μl serum-free DMEM medium/0.25% BSA for the time period indicated, cells were washed 3 times with DMEM/0.25% BSA, solubilized with IN NaOH and counted. For [1 5I]vitronectin binding to cells in suspension, confluent, growth-arrested cells were trypsinized as in Fig. 1 and incubated (2.5 x 104 cells per vial) with [1251]vitronectin plus or minus competitor for 30 min. Binding was measured by filtration of cells on 3.0% BSA-presoaked filters as in Fig. 1. Values represent the mean +_ SEM (n = 3) of one of three representative experiments. Both forms of vitronectin (native and extended) were radiolabelled and their specific activity (1.5-2.5 μCi μg-1 ) determined according to methods described previously [28] .
Figure 2(a) illustrates the dose-dependent inhibition of PDGF-BB-induced DNA synthesis in SMC by vitronectin. SMC were stimulated with 10 ng ml" - PDGF-BB in the presence of the indicated concentrations of vitronectin. PDGF-induced [3H]thymidine uptake in the absence of vitronectin was 1000 +_ 110 dpm. Vitronectin alone had no effect on basal DNA synthesis. The dose-response profile of vitronectin (0.1 nM- 0.1 μM) was biphasic, as shown in Figure 2(a). This biphasic response may reflect concentration-dependent differences in the anti-mitogenic or adhesive properties of the conformational states of oligomeric species formed as a result of exposure of epitopes in the extended form of vitronectin in agrement with the conformational lability/flexibility of this molecule [15,16].
The presence of such non-covalently linked multimers in the extended form was confirmed by the different electrophoresis profiles of native and extended vitronectin obtained under SDS conditions as compared to those in the absence of SDS. This is illustrated in Figure 2(b).
In this experiment native (lanes 1 and 3) and extended (lanes 2 and 4) [-■ 251]vitronectin were electrophoresed in 8% polyacrylamide gels in the presence (lef panel) or absence (right panel) of 0.1% SDS (sodium dodecyl sulphate). Bars indicate the position of protein standards (Mr x 10"3 ). Arrows indicate the top of the separation gel. Samples (1 x 10s cpm) in 50 μl Tris-buffer saline, 0.2% BSA with or without 1.0% SDS were loaded on each lane.
Figure 2(c) illustrates the time-dependent differential association of extended (•) and native (o) [125I]vitronectin 3.5-6.0 nM with SMC monolayers at 37βC. While native [1251]vitronectin did not demonstrate appreciable binding, extended (multimeric) [125i]vitronectin binding to cell surface receptors was considerably higher and appeared to reach steady state within 2 h. Since approximately 50% of extended [i251]vitronectin binding to SMC monolayer was due to binding to ECM, the effects of cRGD and heparin on vitronectin binding to cell surface were assessed with cells in suspension. This is illustrated in Figure 2(d), which shows the inhibition of [125I]vitronectin (extended form) (6.0 nM) binding to SMC in suspension at 37βC in the absence (A) or presence (B) of 100-fold molar excess unlabelled vitronectin; 1 μm cRGD (C) or 10 μg ml"*• unfractionated heparin (Sigma) (D).
As Figure 2(d) shows, one hundred-fold excess of unlabelled vitronectin suppressed [12 I]vitronectin binding to SMC surface by more than 60%, indicating the specificity of the vitronectin-SMC interactions. Furthermore, [125I]vitronectin binding was suppressed by 34% using the cRGD peptide (1 μM) or by 50% using unfractionated heparin (10 μg ml"*- ), suggesting the involvement of both the RGD and heparin binding domains in the interactions of vitronectin with SMC cell surface receptors. In view of recent evidence on the role of heparan sulphate proteoglycans in the binding and cellular responses to bFGF or TGF-^ [17,18], it will be of interest to know whether the vitronectin effect is mediated via a similar dual receptor system involving a high-affinity RGD-dependent receptor and a low-affinity glycosaminoglycan-type binding site.
Alternatively, the inhibition of [- 2 - I]vitronectin binding by heparin may be due to heparin binding and changes in the vitronectin conformation and also the monomer-oligomer equilibrium which may effect the concentration of active vitronectin species. Our preliminary studies have shown that the two molecules may neutralize each other's anti-mitogenic activity.
The relationship between vitronectin and heparin may be of importance in the regulation of cell growth, especially since heparin has been shown to inhibit mitogen and, in particular, PDGF-induced SMC proliferation [19,20] .
As illustrated in Figures 3(a) and 3(b), vitronectin inhibits dose-dependent PDGF-induction of DNA synthesis in human vascular smooth muscle cells without affecting c-fos mRNA expression. Figure 3(a) shows the results of measurement of induction of DNA synthesis in SMC with increasing concentrations of PDGF- BB in serum-free medium in the absence (•) or presence (o) of 0.1 μM vitronectin as in Figure 1. The results represent the mean +_ SEM (n=4) of one of four representative experiments. Figure 3(b) shows a Northern blot of equal amounts (20 μg) of total cellular RNA hybridized with a 2 kb c-fos probe or 0.53 kb G6PD probe. Growth-arrested, confluent SMC preincubated (1 h) without (lanes 1 and 2) or with (lane 3) vitronectin (0.1 μM) in serum-free medium, were exposed for 30 min to 50 ng ml" - PDGF-BB prior to harvesting the cells for total RNA extraction.
Vitronectin was able to inhibit DNA synthesis over a range ofconcentrations of PDGF-BB (0.1-100 ng ml"x ), and at all concentrations the extent of inhibition did not vary (Fig. 3a). This effect was not due to binding and depletion of PDGF since vitronectin did not inhibit the induction of mRNA expression of the c-fos proto-oncogene by PDGF (Fig. 3b). These data also indicate that vitronectin may inhibit growth by affecting mechanisms other than those involved in the expression of early biochemical events induced by growth factors.
It has been shown recently that binding of plasminogen activator inhibitor-1 to vitronectin on ECM stabilizes its activity and prevents tissue or urokinase-type plasminogen activator from generating plasmin and subsequently matrix degradation [28] . Vitronectin, therefore, may prevent growth factor- induced medial-SMC proliferation either directly via integrin/proteoglycan receptor activation or indirectly by preventing access of growth factors and chemo- attractants to SMC as well as cell migration via inhibition of ECM degradation by proteases. Generation of the extended form of circulating vitronectin at sites of vascular injury upon activation of the coagulation/fibrinolysis cascade, may be of physiological significance to limit the early proliferative stages of atheroma development or in intimal hyperplasia, since both of these events involve vascular SMC migration and proliferation.
BIBLIOGRAPHY [1]. Preissner, K.T. Ann. Rev. Cell Biol. 7_, 275-310
(1991). [2]. Hynes, R. Cell 48_, 549-554 (1987). [3]. Heldin, C-H. & Westermark, B. Cell Regul. 1, 555-
566 (1990). [4]. Ross, R. New Engl. J. Med. 314, 448-500 (1986). [5]. Hayman, E.G.. et al. Exp. Cell Res. 160, 245-258
(1985). [6]. Underwood, P.A., & Bennett, F.A. J. Cell Sci. 93_,
641-649 (1989). [7]. Horton, M. Int. J. Exp. Pathol. 71, 741-759
(1990). [8]. Niculescu, F., Rus, H.G. & Vlaicu, R.
Atherosclerosis 65, 1-11 (1987). [9]. Izumi, M., Yamada, K.M. & Hayashi, M. Biochim.
Biophys. Acta 990, 101-108 (1989). [10]. Preissner, K.T. et al. J. Biol. Chem. 265, 18490-
18498 (1990). [11]. Preissner, K.T. & Mϋller-Berghaus, G. J. Biol.
Chem. 262, 12247-12253 (1987). [12]. Preissner, K.T., Wassmuth, R. & Mϋller-Berghaus,
G. Biochem. J. 231, 349-355 (1985). [13]. Pierschbacher, M.D. & Ruoslahti, E. J. Biol.
Chem. 262, 17294-17298 (1987). [14]. Orlando, R.A. & Cheresh, D.A. J. Biol. Chem. 266, 19543-19550 (1991). [15]. Tomasini, B. & Mosher, D.F. Blood 72, 903-912
(1988). [16]. Tomasini, B.R., Mosher, D.F., Owen, M.C. &
Fenton, J.W.II. Biochemistry 28, 832-842 (1989). [17]. Klagsbrun, M. & Baird, A. Cell 6>7, 229-231
(1991). [18]. Wang, X-F. et al. Cell 67, 797-805 (1991). [19]. Castellot, J. et al. J. Cell Physiol. 124, 13-20
(1985). [20]. Reilly, C.F., Fritze, L.M.S. & Rosenberg, R.D. J.
Cell Physiol. 129, 11-19 (1986). [21]. Belkin, V.M., Belkin, A.M. & Koteliansky, V.E. J.
Cell Biol. Ill, 2159-2170 (1990). [22]. Horton, M. Int. J. Exp. Pathol. 71, 741-759
(1990). [23]. Charo, I.F. et al. Proc. Natl. Acad. Sci. USA 83_,
8351-8355 (1986). [24], Ruoslahti, E. & Pierschbacher, M.D. Science 238,
491-497 (1987). [25], Vogel, D.E., Tarone, G., Giancotti, F.G., Gailit,
J. & Ruoslahti, E. J. Biol. Chem. 265, 5934-5937
(1990). [26]. Smith, J.W., Vestal, D.J., Irwin, S.V., Burke,
T.A. & Cheresh, D.A. J. Biol. Chem. 265, 11008-
11013 (1990). [27]. Wayner, E.A., Orlando, R.A. & Cheresh. D.A. J.
Cell. Biol. 113, 919-929 (1991). [28]. Ciambrone, G.J., McKeown-Longo, P.J. J. Cell
Biol. Ill, 2183-2195 (1990).

Claims

1. A method of inhibiting smooth muscle cell proliferation, comprising administration of an effective amount of vitronectin.
2. A method according to claim 1, wherein the smooth muscle cells are vascular smooth muscle cells.
3. A method according to claim 1 or claim 2, wherein the vitronectin is the extended form thereof.
4. A method according to claim 3, wherein the extended form of vitronectin is multimeric.
5. A method according to any one of claims 1 to 4, wherein the vitronectin is administered in an amount such as to give a concentration in plasma of from 1 to 100 mg ml" - .
6. A method according to claim 5, wherein the administration is in an amount such as to give a concentration in plasma of from 1 to 10 mg ml" - .
7. A method according to any preceding claim, wherein the administration is by intravenous injection or infusion.
8. A method of inhibiting smooth muscle cell proliferation, comprising generating vitronectin in its extended form in situ by administration of an agent which converts native vitronectin to its extended form.
9. A method according to claim 8, wherein said agent is selected from proteoglycans and glycosaminoglycans.
10. Vitronectin for use as an antiproliferative agent.
11. Vitronectin according to claim 10 which is in its extended form.
12. A pharmaceutical composition comprising vitronectin together with a pharmaceutically acceptable diluent and/or carrier.
13. A pharmaceutical composition according to claim 12, wherein the vitronectin is the extended form thereof.
14. Use of vitronectin in the preparation of a medicament for inhibiting smooth muscle cell proliferatio .
PCT/GB1992/000958 1991-05-28 1992-05-27 Inhibition of smooth muscle cell proliferation by vitronectin WO1992021363A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
GB919111439A GB9111439D0 (en) 1991-05-28 1991-05-28 Inhibition of vascular smooth muscle cell proliferation
GB9111439.7 1991-05-28

Publications (1)

Publication Number Publication Date
WO1992021363A1 true WO1992021363A1 (en) 1992-12-10

Family

ID=10695684

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/GB1992/000958 WO1992021363A1 (en) 1991-05-28 1992-05-27 Inhibition of smooth muscle cell proliferation by vitronectin

Country Status (6)

Country Link
EP (1) EP0586451A1 (en)
AU (1) AU1763792A (en)
GB (2) GB9111439D0 (en)
IE (1) IE921697A1 (en)
WO (1) WO1992021363A1 (en)
ZA (1) ZA923901B (en)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5545569A (en) * 1993-05-13 1996-08-13 Neorx Corporation Prevention and treatment of pathologies associated with abnormally proliferative smooth muscle cells
US6171609B1 (en) 1995-02-15 2001-01-09 Neorx Corporation Therapeutic inhibitor of vascular smooth muscle cells
US6268390B1 (en) * 1991-09-27 2001-07-31 Neorx Corporation Therapeutic inhibitor of vascular smooth muscle cells

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE102006014190A1 (en) 2006-03-24 2007-09-27 Henkel Kgaa Single component, hot-setting reactive composition, useful e.g. as an adhesive in automobile construction, comprises e.g. a liquid polyene, a block copolymer with a polyene block and a saturated block and/or a vulcanization system

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0292663A1 (en) * 1987-05-25 1988-11-30 Research Development Corporation Of Japan A method for producing vitronectin
EP0341006A2 (en) * 1988-05-03 1989-11-08 The Board Of Trustees Of The Leland Stanford Junior University Smooth muscle cell growth inhibitor
EP0410006A1 (en) * 1989-02-13 1991-01-30 Nisshin Flour Milling Co., Ltd. Eye drops for healing wound of corneal epithelium

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0292663A1 (en) * 1987-05-25 1988-11-30 Research Development Corporation Of Japan A method for producing vitronectin
EP0341006A2 (en) * 1988-05-03 1989-11-08 The Board Of Trustees Of The Leland Stanford Junior University Smooth muscle cell growth inhibitor
EP0410006A1 (en) * 1989-02-13 1991-01-30 Nisshin Flour Milling Co., Ltd. Eye drops for healing wound of corneal epithelium

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
Journal of cellular Physiology, vol. 124, no. 1, July 1985, J.J. CASTELLOT et al.: "Effect of heparin on vascular smooth muscle cells. I. Cell metabolism", pages 21-28, see page 21, left-hand column, paragraph 2 - right-hand column, paragraph 2; page 23, right-hand column, paragraph 2 *

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6268390B1 (en) * 1991-09-27 2001-07-31 Neorx Corporation Therapeutic inhibitor of vascular smooth muscle cells
US5545569A (en) * 1993-05-13 1996-08-13 Neorx Corporation Prevention and treatment of pathologies associated with abnormally proliferative smooth muscle cells
US6171609B1 (en) 1995-02-15 2001-01-09 Neorx Corporation Therapeutic inhibitor of vascular smooth muscle cells

Also Published As

Publication number Publication date
IE921697A1 (en) 1992-12-02
ZA923901B (en) 1993-03-08
GB9120748D0 (en) 1991-11-13
AU1763792A (en) 1993-01-08
EP0586451A1 (en) 1994-03-16
GB9111439D0 (en) 1991-07-17

Similar Documents

Publication Publication Date Title
Ashizawa et al. Osteopontin is produced by rat cardiac fibroblasts and mediates A (II)-induced DNA synthesis and collagen gel contraction.
Hajjar et al. Tumor necrosis factor-mediated release of platelet-derived growth factor from cultured endothelial cells.
JP2961708B2 (en) Growth hormone and insulin-like growth factor-1 combination enhances growth
Nyberg et al. Endogenous inhibitors of angiogenesis
EP0835115B1 (en) 4-(2-(n-2-carboxamidoindole)aminoethyl)-benzenesulfonamides or sulfonylureas as pdgf antagonists
DE69911609T2 (en) PROCESS FOR ACCELERATING THE GROWTH AND HEALING OF BONES AND CARTILES
KR0137017B1 (en) Use of gamma interferon in pharmaceutical preparations for the treatment of vascular stenosis
JPH07504650A (en) Inhibition of transforming growth factor β to prevent extracellular matrix accumulation
Shebuski et al. Demonstration of Ac-Arg-Gly-Asp-Ser-NH2 as an antiaggregatory agent in the dog by intracoronary administration
US20090110709A1 (en) Elastin protective polyphenolics and methods of using the same
US6432706B1 (en) Method for inhibiting angiogenesis and tumors with the isolated NC1 α6 chain monomer of type IV collagen
US5155038A (en) Use of thrombospondin to promote wound healing
CN101300269A (en) Mecano growth factor peptides and their use
WO1992021363A1 (en) Inhibition of smooth muscle cell proliferation by vitronectin
US7858577B2 (en) Use of the long pentraxin PTX3 for the treatment of diseases caused by an altered activation of the growth factor FGF-2
CA2318432C (en) The use of isolated domains of type iv collagen to modify cell and tissue interactions
JP2002539267A (en) Methods for limiting scarring and adhesion formation
JP2014043461A (en) Bmp-7 for use in treating vascular sclerosis
US7517654B2 (en) Peptide and a derivative thereof promoting cell adhesion and spreading
Cechowska Pasko et al. Alterations in glycosaminoglycans in wounded skin of diabetic rats. A possible role of IGF-I, IGF-binding proteins and proteolytie activity
AU2005222085B2 (en) Treating or preventing extracellular matrix build-up
Guth et al. Antagonism of the GPIIb/IIIa receptor with the nonpeptidic molecule BIBU52: inhibition of platelet aggregation in vitro and antithrombotic efficacy in vivo
JPH07316066A (en) Wound healing agent
CA2036592C (en) Therapeutic agent for diabetic gangrene
JPH09151137A (en) Medicine for inhibiting multiplication of smooth muscle cell

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AT AU BB BG BR CA CH CS DE DK ES FI GB HU JP KP KR LK LU MG MN MW NL NO PL RO RU SD SE US

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): AT BE BF BJ CF CG CH CI CM DE DK ES FR GA GB GN GR IT LU MC ML MR NL SE SN TD TG

DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
WWE Wipo information: entry into national phase

Ref document number: 1992910615

Country of ref document: EP

ENP Entry into the national phase

Ref country code: US

Ref document number: 1994 142351

Date of ref document: 19940128

Kind code of ref document: A

Format of ref document f/p: F

WWP Wipo information: published in national office

Ref document number: 1992910615

Country of ref document: EP

REG Reference to national code

Ref country code: DE

Ref legal event code: 8642

WWW Wipo information: withdrawn in national office

Ref document number: 1992910615

Country of ref document: EP

NENP Non-entry into the national phase

Ref country code: CA