Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 116
Filter
1.
J Clin Invest ; 93(5): 2286-90, 1994 May.
Article in English | MEDLINE | ID: mdl-7514194

ABSTRACT

IGF Binding Protein-3 (IGFBP-3), the major IGF carrier in the blood, undergoes limited proteolysis which reduces its affinity for IGFs, thus facilitating dissociation. The functional effects of this at the cellular level were studied by comparing two serum pools, one from healthy adults, one from women during late pregnancy when IGFBP-3 proteolysis is increased. Sera were mixed to yield identical IGF-I and IGF-II concentrations in the two pools. Western ligand and immunoblotting gave the characteristic IGFBP patterns for the two types of serum. Both pools dose-dependently stimulated DNA synthesis in cultured chick embryo fibroblasts. Stimulation by pregnancy serum was twice that by normal serum at 0.05-0.2% concentrations (P < 0.001). In the presence of excess monoclonal anti-IGF-I and -II antibodies, stimulation by both (0.1-0.2%) pools was 70-80% reduced and residual stimulation was similar. Addition of recombinant human (rh) IGFBP-3 dose-dependently depressed both pools' activity, more so for normal serum at 25 and 50 ng/ml, equally for each at 100 ng/ml. At the latter concentration, slight proteolysis of the rhIGFBP-3 was detectable in the presence of 0.2% pregnancy serum, but at 25 ng/ml, proteolysis was absent. These results suggest that IGFs are released more readily from pregnancy serum, accounting for the weaker inhibitory effect of low rhIGFBP-3 concentrations. For identical IGF concentrations, pregnancy serum's greater biological activity therefore reflects greater IGF availability to the cells. This study demonstrates the functional consequences at cellular level of serum IGFBP-3 proteolysis, underlining its significance in regulating serum IGF bioavailability.


Subject(s)
Carrier Proteins/metabolism , Insulin-Like Growth Factor II/metabolism , Insulin-Like Growth Factor I/metabolism , Adult , Animals , Biological Availability , Carrier Proteins/blood , Carrier Proteins/drug effects , Chick Embryo , Female , Humans , Insulin-Like Growth Factor Binding Proteins , Male , Pregnancy , Pregnancy Trimester, Third/metabolism , Serine Endopeptidases/pharmacology
2.
J Clin Invest ; 86(3): 952-61, 1990 Sep.
Article in English | MEDLINE | ID: mdl-1697608

ABSTRACT

UNLABELLED: Insulin-like growth factors (IGFs) in blood form two complexes with specific binding proteins (BPs): a large, growth hormone (GH)-dependent complex with restricted capillary permeability, and a smaller complex, inversely related to GH, with high turnover of its IGF pool and free capillary permeability. The distribution of BPs and of IGFs I and II between these complexes was studied in sera from healthy adults treated with IGF I or/and GH and from patients with extrapancreatic tumor hypoglycemia. Like GH, IGF I administration raises IGF I and two glycosylation variants of IGFBP-3 in the large complex, but unlike GH drastically reduces IGF II. During IGF I infusion, IGFBP-3 appears in the small complex whose IGFBP-2 and IGF I increase three- to fivefold and fivefold, respectively. GH treatment, associated with elevated insulin levels, suppresses IGFBP-2 and inhibits its increase owing to infused IGF I. The small complex of tumor sera contains increased amounts of IGFBP-2 and -3, and two- to threefold elevated IGF II. CONCLUSIONS: low GH and/or insulin during IGF I infusion and in extrapancreatic tumor hypoglycemia enhance expression of IGFBP-2 and favor partition of IGFBP-3 into the small complex. Free capillary passage and high turnover of its increased IGF I or II pools may contribute to compensate for suppressed insulin secretion during IGF I infusion or to development of tumor hypoglycemia.


Subject(s)
Carrier Proteins/metabolism , Hypoglycemia/metabolism , Insulin-Like Growth Factor II/metabolism , Insulin-Like Growth Factor I/pharmacology , Neoplasms/complications , Somatomedins/metabolism , Somatomedins/pharmacology , Carrier Proteins/analysis , Growth Hormone/pharmacology , Humans , Insulin-Like Growth Factor Binding Proteins , Insulin-Like Growth Factor I/metabolism , Macromolecular Substances , Molecular Weight
3.
Endocrinology ; 134(3): 1254-62, 1994 Mar.
Article in English | MEDLINE | ID: mdl-7509737

ABSTRACT

In a variety of physio-pathological conditions, but also in the normal state, calcium-dependent serine protease(s) partially proteolyze proportions of circulating insulin-like growth factor binding protein-3 (IGFBP-3). This occurs without disrupting the 150-kilodalton (kDa) complexes in which IGFBP-3 carries of most of the IGF-I and IGF-II in serum. In this work we show that the 150-kDa complex is functionally altered during pregnancy, which is when the largest proportion of IGFBP-3 is proteolyzed. After preincubation at 4 C with [125I]IGF-I or -II with or without 1 microgram unlabeled IGF-I or -II, pooled plasma samples were gel filtered at pH 7.4. Larger proportions of labeled IGFs were found in the 150-kDa complexes of the third trimester pregnancy plasma pool than in those of the normal plasma pool, suggesting increased binding capacity. Nevertheless, competitive binding experiments using [125I]IGF-I and -II and IGFBP-3 preparations from each of the plasma pools showed that the competitive potencies of IGF-II and especially IGF-I were lower in pregnancy IGFBP-3 than in normal IGFBP-3. Scatchard analysis revealed a 2-fold loss of affinity for IGF-II and a 10-fold loss for IGF-I compared with that for normal plasma IGFBP-3. In studies at 37 C of the kinetics of dissociation of [125I]IGF-I and -II bound to IGFBP-3, IGF-II was dissociated 6 times faster, and IGF-I 10 times faster from pregnancy plasma IGFBP-3 than from normal plasma IGFBP-3. After incubation of individual plasma samples at 37 C and gel filtration at room temperature (in the presence of EDTA), IGFs were assayed in the three circulating pools (150-kDa and 40-kDa complexes and free IGFs), revealing a redistribution of pregnancy plasma IGFs. The proportion of total IGF-I in free form was nearly three times greater in pregnancy than in normal plasma (11.4% vs. 4.1%, P < 0.005), whereas that of free IGF-II was slightly smaller (1.5% vs. 2%). In the 150-kDa complexes, the proportion of total IGF-I was significantly smaller in pregnancy than in normal plasma, and that of IGF-II was greater. In the 40-kDa complexes, the proportion of total IGF-II was significantly smaller. The mean ratios of molar concentrations of free IGF-I/IGF-II were 0.43 in normal plasma and 2.23 in pregnancy plasma (P < 0.005).(ABSTRACT TRUNCATED AT 400 WORDS)


Subject(s)
Carrier Proteins/metabolism , Pregnancy/metabolism , Somatomedins/metabolism , Adult , Binding, Competitive , Carrier Proteins/blood , Female , Humans , Insulin-Like Growth Factor Binding Proteins , Insulin-Like Growth Factor I/metabolism , Insulin-Like Growth Factor II/metabolism , Pregnancy/blood
4.
Endocrinology ; 142(1): 108-13, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11145572

ABSTRACT

Insulin-like growth factor binding proteins (IGFBPs) regulate the cellular actions of the IGFs owing to their strong affinities, which are equal to or stronger than the affinity of the type 1 IGF receptor (IGF-IR), the mediator of IGF signal transduction. We recently found that IGFBP-3 modulates IGF-I binding to its receptor via a different mechanism possibly involving conformational alteration of the receptor. We have now investigated the effects of IGFBP-3 on the initial steps in the IGF signaling pathway. MCF-7 breast carcinoma cells were preincubated with increasing concentrations of IGFBP-3 and then stimulated with IGF-I, des(1-3)IGF-I, or [Q(3)A(4)Y(15)L(16)]-IGF-I, the latter two being IGF-I analogs with intact affinity for the type 1 IGF receptor, but weak or virtually no affinity for IGFBPs. Stimulation of autophosphorylation of the receptor and its tyrosine kinase activity was dose-dependently depressed. At 2.5 nM, IGFBP-3 provoked more than 50% inhibition of the stimulation induced by 3 nM des(1-3)IGF-1 and, at 10 nM, more than 80% inhibition. Similar results were obtained with [Q(3)A(4)Y(15)L(16)]-IGF-I. Cross-linking experiments using iodinated or unlabeled IGFBP-3 and anti-IGF-IR antibodies indicated that the inhibitory effects do not involve direct interaction between IGFBP-3 and IGF-IR. The inhibition appeared to be specific to IGFBP-3, because IGFBP-1 and IGFBP-5 at 10 nM had no significant effect. Also, inhibition was restricted to the IGF receptor, because IGFBP-3 failed to inhibit the tyrosine kinase activity of the insulin receptor stimulated by physiological concentrations of insulin. Our results provide the first demonstration that IGFBP-3 can specifically modulate the IGF-I signaling pathway independently of its IGF-I-binding ability. They also reveal a regulatory mechanism specific to the type 1 IGF receptor, with no effect on insulin receptor activation.


Subject(s)
Insulin-Like Growth Factor Binding Protein 3/pharmacology , Insulin-Like Growth Factor I/pharmacology , Receptor, IGF Type 1/metabolism , Breast Neoplasms , Female , Humans , Insulin-Like Growth Factor Binding Protein 1/pharmacology , Insulin-Like Growth Factor Binding Protein 5/pharmacology , Insulin-Like Growth Factor I/analogs & derivatives , Phosphorylation , Protein-Tyrosine Kinases/metabolism , Receptor, IGF Type 1/drug effects , Recombinant Proteins/pharmacology , Signal Transduction/drug effects , Tumor Cells, Cultured
5.
Endocrinology ; 138(7): 3069-72, 1997 Jul.
Article in English | MEDLINE | ID: mdl-9202255

ABSTRACT

We previously reported that a 16-kDa proteolytic fragment of IGF Binding Protein-3 (IGFBP-3), which is devoid of affinity for IGFs, inhibits the mitogenic effects of IGF-I on chick embryo fibroblasts. Here, we set out to determine if the fragment had biological effects on fibroblasts from mouse embryos homozygous for a targeted disruption of the Type 1 IGF receptor gene. In the cell clone used, bFGF (but not IGF, EGF or PDGF) was mitogenic in serum-free medium, increasing 14C-thymidine uptake by a factor of 10-15 within 24 hours and doubling cell proliferation. The 16-kDa fragment, isolated by HPLC following limited proteolysis of recombinant human (rh) IGFBP-3 by plasmin, in both assays dose-dependently (20 to 100 ng/ml) inhibited (up to 100%) maximal stimulation induced by 25 ng/ml bFGF, whereas intact IGFBP-3 had virtually no effect. Similar results were obtained with control wild-type cells. In the latter, the mitogenic activity of 1% fetal calf serum (equal to that of 25 ng/ml bFGF) was inhibited by only 25-30% by 100 ng/ml 16-kDa fragment or 200 ng/ml rhIGFBP-3. This agrees with an antagonistic action, affecting the mitogenic activity of serum that is attributable to IGFs. The 16-kDa IGFBP-3 fragment therefore appears to be a potent inhibitor of mitogenic signals resulting from activation of both the type 1 IGF and FGF receptors.


Subject(s)
Fibroblast Growth Factors/antagonists & inhibitors , Insulin-Like Growth Factor Binding Protein 3/pharmacology , Peptide Fragments/pharmacology , Receptor, IGF Type 1/genetics , Animals , Cells, Cultured , Fibroblast Growth Factor 2/pharmacology , Fibroblasts/drug effects , Humans , Mice , Molecular Weight , Thymidine/metabolism
6.
Endocrinology ; 138(2): 683-90, 1997 Feb.
Article in English | MEDLINE | ID: mdl-9003003

ABSTRACT

Insulin-like growth factor (IGF) binding proteins (IGFBPs) modulate IGF action at cellular level through inhibition or, alternatively, potentiation, where their limited proteolysis is a contributory mechanism. Under basal conditions, neuroblastoma cells secrete IGFs (essentially IGF-II), IGFBPs (IGFBP-4 and predominantly IGFBP-2 that is partially proteolysed), and proteases, including tissue-type plasminogen (PLG) activator, whose activity is inhibited by PLG activator inhibitor-1. Neuroblastoma cells were used to investigate the influence of the plasmin system, transforming growth factor-beta retinoic acid on cell growth and the IGF system. In cells treated with 5 micrograms/ml PLG, proliferation was stimulated, an effect that was inhibited in the presence of either alpha IR-3 (which blocks the type 1 IGF receptor) or anti-IGF-II antibodies. There was a parallel increase in IGFBP-2 proteolysis, which resulted in a 5-fold loss of affinity for IGF-II. In the presence of 1 ng/ml transforming growth factor-beta, PLG-induced mitogenesis and IGFBP-2 proteolysis were reduced, and Northern blot analysis revealed increased PLG activator inhibitor-1 mRNA. Conversely, with 2 microM retinoic acid, the mitogenic effect of PLG, IGFBP-2 proteolysis, and tissue-type PLG activator mRNAs were increased. Therefore, IGF-II mediates autocrine proliferation in neuroblastoma cells under the control of IGFBPs secreted by the cells, its bioavailability being enhanced as a result of plasmin-induced IGFBP-2 proteolysis.


Subject(s)
Cell Division , Insulin-Like Growth Factor Binding Protein 2/metabolism , Neuroblastoma/pathology , Transforming Growth Factor beta/pharmacology , Tretinoin/pharmacology , Blotting, Northern , Fibrinolysin/metabolism , Humans , Immunoblotting , Plasminogen Activator Inhibitor 1/genetics , RNA, Messenger/metabolism , Tissue Plasminogen Activator/pharmacology , Tumor Cells, Cultured
7.
Endocrinology ; 136(12): 5305-10, 1995 Dec.
Article in English | MEDLINE | ID: mdl-7588275

ABSTRACT

We previously showed that when added to fresh medium, insulin-like growth factor (IGF)-II stimulates glycogen synthesis in cultured 18-day-old fetal rat hepatocytes. In the present study, we investigated the influence of 24-h culture-conditioned medium on IGF-II- and insulin-induced glycogenesis. The stimulatory effect of IGF-II (2.9-fold) on [14C]glucose incorporation into glycogen over 3 h was dose dependently inhibited by conditioned medium, whereas that of insulin (3.2-fold) was unaffected. Western ligand and immunoblot analysis of the conditioned media revealed IGF binding protein (IGFBP)-1, IGFBP-2, and IGFBP-4, with a predominance of IGFBP-1. IGFBP-3 was not detected. Preincubation of conditioned medium with IGF-II for 4 h at 4 C restored the glycogenic effect of newly added IGF-II. Preincubation of fresh medium with recombinant IGFBP-1 or IGFBP-3 in the presence of IGF-II suppressed IGF-II stimulation of glycogen synthesis. IGFBPs alone had no effect on glycogenesis. Des(1-6)IGF-II and R6IGF-II, structural analogs of IGF-II that have weak affinity for the IGFBPs, elicited maximal stimulation, near that of IGF-II (2.8-fold and 3.1-fold vs. 3.0-fold for IGF-II), whether tested in fresh or conditioned medium. The inhibitory effect of conditioned medium on IGF-II-induced glycogenesis is therefore mediated by the IGFBPs via sequestration of IGF-II. This suggests that the IGFBPs, particularly IGFBP-1, produced by fetal rat hepatocytes in culture may play a role in regulating glycogenesis.


Subject(s)
Fetus/metabolism , Glycogen/biosynthesis , Insulin-Like Growth Factor Binding Proteins/physiology , Insulin-Like Growth Factor II/pharmacology , Liver/metabolism , Animals , Cells, Cultured , Insulin/pharmacology , Liver/cytology , Rats , Rats, Sprague-Dawley
8.
Endocrinology ; 136(12): 5485-92, 1995 Dec.
Article in English | MEDLINE | ID: mdl-7588299

ABSTRACT

PC-3 cells, whose growth is androgen-independent, were shown to be capable of slow proliferation in serum-free medium and in the absence of added growth factor for 7 days. They secreted insulin-like growth factor (IGF)-II but no detectable IGF-I. This IGF-II, although produced in small amounts, plays a role in their proliferation because growth could be inhibited dose dependently by up to 80% in the presence of monoclonal antibodies directed against IGFs or the type 1 IGF receptor. PC-3 cells also secreted IGF binding proteins (IGFBPs) -2, -3, -4, and -6. Immunoblot analysis revealed selective proteolysis of IGFBP-3, yielding fragments of the same molecular size as those generated from IGFBP-3 in vivo. With the addition to the culture medium of a serine protease inhibitor, 4-(2-aminoethyl)-benzenesulfonyl fluoride (Pefabloc-SC), at concentrations < 0.2 mM that were nontoxic to the cells, cell proliferation was dose dependently inhibited up to 80% and, at the same time, proteolysis of the IGFBP-3 secreted by the cells was depressed. Urokinase activity detected in the conditioned media was depressed by Pefabloc, suggesting that the urokinase-type plasminogen activator was involved in the proteolysis of IGFBP-3. In addition, 0.01-5 micrograms/ml plasminogen induced a dose-dependent increase in both proliferation and the proportions of proteolysed IGFBP-3 in the media. The stimulation of proliferation was totally blocked in the presence of anti-type 1 IGF receptor antibody. Recombinant human IGF-II (5-200 ng/ml) added to cell-free medium conditioned by 48 h of culture dose dependently stimulated PC-3 cell proliferation. At concentrations < or = 100 ng/ml, its mitogenic action was potentiated when medium had been conditioned by cells cultured in the presence of plasminogen but inhibited when medium had been conditioned by cells cultured in the presence of Pefabloc. We conclude from these results 1) that IGF-II is involved in the autocrine control of PC-3 cell proliferation via the type 1 IGF receptor; and 2) that this proliferation is directly dependent on IGF-II bioavailability that itself is modulated by the limited IGFBP-3 proteolysis induced, at least in part, by urokinase-type plasminogen activator and plasmin.


Subject(s)
Endopeptidases/physiology , Insulin-Like Growth Factor Binding Protein 3/metabolism , Prostatic Neoplasms/pathology , Somatomedins/physiology , Cell Division , Humans , Insulin-Like Growth Factor II/pharmacology , Male , Receptor, IGF Type 1/physiology , Recombinant Proteins/pharmacology , Tumor Cells, Cultured , Urokinase-Type Plasminogen Activator/physiology
9.
Endocrinology ; 138(12): 5645-8, 1997 Dec.
Article in English | MEDLINE | ID: mdl-9389554

ABSTRACT

With a view to determining the mechanisms by which insulin-like growth factor binding protein-3 (IGFBP-3) and its proteolytic fragments modulate IGF action, we used a fibroblast cell line to investigate the possibility of an interaction with the type 1 IGF receptor (IGFR-1). In competitive binding experiments, IGFBP-3 was as potent as unlabelled IGF-I in displacing its truncated analogue, 125I-des(1-3)IGF-I, which has weak affinity for IGFBPs, from its binding to the cell surface. None of the proteolytic fragments of IGFBP-3 tested affected this binding. IGFBP-3 had no effect on insulin binding to its receptor. At 10 nM, IGFBP-1 was ineffective where IGFBP-3 provoked 90% displacement of 125I-des(1-3)IGF-I, but it was equally potent in displacing 125I-IGF-I. At the same concentration, binding of 125I-des(1-3)IGF-I to free IGFBP-3 in the supernatant was only 2%. After pre-incubation of the cells with 125I-des(1-3)IGF-I, low concentrations of IGFBP-3 were as potent as IGF-I in dissociating IGFR-1-bound ligand. After pre-incubation of cells with IGFBP-3, washing and then incubation with 125I-des(1-3)IGF-I, inhibition by low IGFBP-3 concentrations was suppressed, but some degree of inhibition by high concentrations persisted. At these high concentrations, addition of IGF-I restored binding owing to uptake of cell-associated IGFBP-3. The present results provide the first evidence that IGFBP-3 may inhibit IGF binding to IGFR-1, and hence limit IGF action via a cellular mechanism that is different from the extracellular mechanism of sequestration.


Subject(s)
Insulin-Like Growth Factor Binding Protein 3/pharmacology , Insulin-Like Growth Factor I/metabolism , Receptors, Somatomedin/metabolism , Animals , Binding, Competitive , Cricetinae , Cricetulus , Humans , Insulin-Like Growth Factor I/antagonists & inhibitors , Insulin-Like Growth Factor I/physiology , Ligands , Peptide Fragments/metabolism , Peptide Fragments/pharmacology
10.
Endocrinology ; 137(8): 3206-12, 1996 Aug.
Article in English | MEDLINE | ID: mdl-8754741

ABSTRACT

Limited proteolysis of insulin-like growth factor binding protein-3 (IGFBP-3) is increasingly becoming recognized as an essential mechanism in the regulation of insulin-like growth factor (IGF) bioavailability, both in the bloodstream and at cellular level. Plasmin generated on contact with various cell types provokes proteolytic cleavages that are similar to those induced in vivo by (as yet unidentified) IGFBP-3 proteases. Experimental conditions were determined to achieve plasmin-induced limited proteolysis of recombinant human nonglycosylated IGFBP-3. Two major fragments of 22/25 kilodaltons (kDa) and one of 16 kDa were identified by Western immunoblotting and isolated by reverse-phase chromatography. The 22/25-kDa fragments correspond to the major approximately 30-kDa glycosylated fragment of IGFBP-3 in serum and the 16-kDa fragment, to one of the same size, that is nonglycosylated. Western ligand blot analysis, affinity cross-linking, and competitive binding experiments using radiolabeled IGF and unlabeled IGF-I or -II showed that in the high performance liquid chromatography eluate containing the 16-kDa fragment, all affinity for IGFs had been lost, whereas the affinity of the 22/25-kDa fragments was considerably reduced. Scatchard analysis of the data indicated a 20-fold loss of affinity for IGF-II and an 50-fold loss for IGF-I compared with that of recombinant human IGFBP-3. In a chick embryo fibroblast assay in which DNA synthesis was stimulated both by IGF-I and by insulin (at 100-fold concentrations, so that interaction with the Type 1 IGF receptor would occur), IGFBP-3 was found to inhibit IGF-I-induced stimulation almost totally. It had no effect on stimulation by insulin, which has no affinity for the IGFBPs. With the 22/25-kDa fragments, barely 50% inhibition of IGF-I stimulation was achieved and no inhibition of insulin stimulation. Unexpectedly, with the fraction containing the 16-kDa fragment (despite the total lack of affinity for IGF-I), IGF-I-induced stimulation was inhibited to nearly the same extent as with intact IGFBP-3. In addition, insulin-induced stimulation was inhibited with similar potency. IGFBP-3 proteolysis therefore generates two types of fragment with different activities. One has weak affinity for IGF-I and is only a weak antagonist of IGF action. The other lacks affinity for the IGFs, but nevertheless inhibits IGF-stimulated mitogenesis, thus acting by a mechanism that is independent of the IGFs.


Subject(s)
Insulin Antagonists/pharmacology , Insulin-Like Growth Factor Binding Protein 3/pharmacology , Insulin-Like Growth Factor I/antagonists & inhibitors , Peptide Fragments/pharmacology , DNA/antagonists & inhibitors , DNA/biosynthesis , Electrophoresis, Polyacrylamide Gel , Fibrinolysin/pharmacology , Glycosylation , Humans , Insulin-Like Growth Factor Binding Protein 3/chemistry , Insulin-Like Growth Factor Binding Protein 3/metabolism , Mitogens/antagonists & inhibitors , Peptide Fragments/isolation & purification , Peptide Fragments/metabolism , Peptide Hydrolases/metabolism , Somatomedins/metabolism
11.
Endocrinology ; 135(6): 2318-26, 1994 Dec.
Article in English | MEDLINE | ID: mdl-7527330

ABSTRACT

Limited proteolysis in vivo of insulin-like growth factor-binding protein-3 (IGFBP-3) by as yet unidentified serine proteases plays a key role in controlling the bioavailability of IGFBP-3-associated insulin-like growth factors (IGFs). Both the IGF system and the system of plasminogen activators (PAs) and their inhibitors (PAIs) are involved in bone remodeling, and plasmin has been shown to provoke dissociation of IGFBP-IGF complexes in cultured MG-63 human osteoblasts. The aim of this work was to investigate interactions between IGF-I and the PA/PAI system and their influence on IGFBP-3 production and proteolysis in this cell model. At confluency, MG-63 cells maintained for 3 days in serum-free medium constitutively secreted IGFBP-2 and small amounts of IGFBP-3 and IGFBP-4. As shown by Western ligand and immunoblot analyses of the culture medium and Northern blot analysis of IGFBP-3 messenger RNA, production of these IGFBPs, and of IGFBP-3 in particular, was dose dependently stimulated by the addition of 12.5-100 ng/ml recombinant human (rh) IGF-I. Increasing concentrations of plasminogen (0.05-3.5 micrograms/ml) added during the final 12 h of culture reduced the amounts of IGFBP detectable by Western ligand blotting, especially IGFBP-3. This reduction reflected proteolysis, as shown by immunoblotting, which revealed 30-, 20-, and 16-kilodalton fragments of IGFBP-3. In the presence of 25 ng/ml IGF-I, which stimulated IGFBP-3 production, proteolysis was reduced by approximately half. Incubation of glycosylated [125I]rh-IGFBP-3 as substrate in cell-free conditioned medium gave the same results. Addition of 50 ng/ml rhIGF-I to conditioned medium (to promote IGFBP-3-rhIGF-I complex formation) failed to diminish plasmin-induced proteolysis of IGFBP-3. Urokinase PA activity in the conditioned medium decreased significantly when the cells were cultured with rhIGF-I, indicating a direct action of IGF-I on urokinase PA and/or PAI production. Our results support the notion of a regulation loop whereby IGF-I controls its own bioavailability via its action on both IGFBP-3 production and the PA/PAI system, which regulates IGFBP-3 proteolysis. The proteolytic cleavages of IGFBP-3 caused by plasmin were the same as those caused in vivo by serine protease acting on this IGFBP.


Subject(s)
Carrier Proteins/metabolism , Insulin-Like Growth Factor I/physiology , Osteosarcoma/metabolism , Peptide Hydrolases/metabolism , Plasminogen Activators/physiology , Plasminogen Inactivators/physiology , Fibrinolysin/physiology , Humans , Insulin-Like Growth Factor Binding Proteins , Insulin-Like Growth Factor I/pharmacology , Osteosarcoma/pathology , Somatomedins/metabolism , Tumor Cells, Cultured
12.
Endocrinology ; 132(6): 2586-92, 1993 Jun.
Article in English | MEDLINE | ID: mdl-7684981

ABSTRACT

Insulin-like growth factor binding proteins (IGFBPs) are essential mediators of the bioavailability and biological effects of the IGFs. Liver expression of the rat (r) IGFBP-1 and rIGFBP-2 genes has been characterized between day 16 in utero (16 diu) and 16 days postnatally (+16 dpn). Run-on experiments showed transcriptional activity of the rIGFBP-1 and rIGFBP-2 genes at birth (B) to be 25 and 5 times that at 16 diu, respectively. After B, transcriptional activity of the rIGFBP-1 gene remained high (140% B at +6 dpn), but that of the rIGFBP-2 gene dropped to 70% B by +6 dpn. Northern blot analysis done simultaneously showed rIGFBP-1 messenger RNA (mRNA) levels to increase approximately 50-fold between 16 diu and B, whereas rIGFBP-2 mRNA increased only 5- to 10-fold. rIGFBP-1 mRNA levels decreased after birth, reaching about 20% B by +6 dpn; rIGFBP-2 mRNA, however, remained stable (about 80% B) at least up to +6 dpn. Parallel Western ligand blot and immunoblot analyses of serum rIGFBPs revealed rIGFBP-1 and rIGFBP-2 concentrations to be increased 3- and 2-fold, respectively between 20 diu and B. Maximal expression of rIGFBP-1 was at +1 dpn (220% B), and of rIGFBP-2, at B. Both rIGFBPs then decreased, reaching about 5% B at adulthood. All these data indicate that increased transcriptional activity of the rIGFBP-1 and rIGFBP-2 genes at birth would determine the increased synthesis in the liver and circulating levels of these proteins. In addition, it would seem that post-transcriptional events (reduced half-life of the rIGFBP-1 messenger after birth, translation efficiency of the rIGFBP-2 messenger) modulate transcriptional regulation.


Subject(s)
Carrier Proteins/genetics , Fetus/physiology , Gene Expression , Aging/physiology , Animals , Animals, Newborn , Carrier Proteins/blood , Fetus/metabolism , Immunoblotting , Insulin-Like Growth Factor Binding Protein 1 , Insulin-Like Growth Factor Binding Protein 2 , Liver/embryology , Liver/physiology , RNA, Messenger/metabolism , Rats , Rats, Wistar , Somatomedins/metabolism , Transcription, Genetic
13.
Endocrinology ; 124(5): 2365-72, 1989 May.
Article in English | MEDLINE | ID: mdl-2539977

ABSTRACT

Articular chondrocytes from 2- to 3-month-old rabbits were cultured in serum-free medium supplemented with fibroblast growth factor. The effects were studied of GH, insulin-like growth factors (IGFs), and insulin on the production of IGF-I, IGF-II, and their binding proteins (BPs) and on cell multiplication. In the control culture medium, IGF-I levels were about one fifth those of IGF-II. Western blot analysis of the BPs revealed a predominant 30K form and 24K and 20K forms which appeared inconsistently and in small quantities. Ten to 100 ng/ml human GH had no mitogenic effect, and even had a slightly inhibitory effect. IGF-I at 10 ng/ml stimulated cell multiplication above the control level by 41% and at 50 ng/ml by 74%, whereas the mean increase obtained with IGF-II (10 and 50 ng/ml) was only 19%. At the same doses, insulin had no effect, but at 5 micrograms/ml it stimulated cell multiplication by a mean of 67%. There was a positive correlation between cell number and release into the medium of both IGF-I (r = 0.86) and IGF-II (r = 0.77). Neither IGF-I nor IGF-II production was affected by GH. Insulin (5 micrograms/ml) increased IGF-I production by a factor of 2.6, but increased IGF-II production by a factor of only 1.4. Under the various conditions of culture with different doses of GH and insulin, cell multiplication, relative to the control value was positively correlated to the IGF-I/IGF-II production ratio (r = 0.77). It would, therefore, seem that IGF-I secreted by the chondrocytes may stimulate their own proliferation. When IGFs or insulin were added to the culture medium, changes in the electrophoretic profiles of the BPs included an increase in the 30K form and an increase in or the appearance of the 24K and 20K forms. Ten and 50 ng/ml IGF-I or IGF-II had effects equal to or greater than those induced by 5 micrograms/ml insulin. These results indicate that the syntheses of BPs and IGFs are coordinated and that IGFs may be implicated in the control of the synthesis of their BPs.


Subject(s)
Cartilage, Articular/metabolism , Receptors, Cell Surface/biosynthesis , Somatomedins/biosynthesis , Animals , Cartilage, Articular/cytology , Cell Division/drug effects , Cells, Cultured , Growth Hormone/pharmacology , Insulin/pharmacology , Rabbits , Receptors, Somatomedin , Somatomedins/pharmacology
14.
Endocrinology ; 125(2): 773-7, 1989 Aug.
Article in English | MEDLINE | ID: mdl-2473891

ABSTRACT

The insulin-like growth factors (IGFs) appear to be secreted into interstitial fluids by many cell types, along with specific, high affinity binding proteins (IGF-BPs). These proteins, therefore, have the potential to bind IGF-I and -II and modify their ability to interact with specific cell surface receptors In these studies we report the detection of high mol wt, multimeric forms of one form of IGF-BP that has been purified from human amniotic fluid. The multimeric forms, which are either not or barely detectable in native amniotic fluid, are the result of intermolecular disulfide bond formation and can be reduced to a monomeric form by exposure to dithiothreitol. After reduction, the multimers are reduced to either monomeric or dimeric forms, as detected by Western blotting. The multimers can be separated from monomeric and dimeric forms by gel filtration chromatography. The purified multimers were fully biologically active in potentiating the effect of IGF-I on porcine aortic smooth muscle cell DNA synthesis. The monomeric form was also bioactive. No significant differences in the affinity of the monomeric and multimeric forms for IGF-I or -II could be detected. In summary, multimeric forms of this form of IGF-BP are detected during purification. The formation of these multimers is through intermolecular disulfide bonds and does not disrupt IGF binding or potentiation of the cellular growth response to IGF-I. These findings indicate that these higher mol wt forms may be fully active in biological test systems.


Subject(s)
Amniotic Fluid/analysis , Carrier Proteins/analysis , Animals , Carrier Proteins/isolation & purification , Carrier Proteins/metabolism , Cells, Cultured , Electrophoresis, Polyacrylamide Gel , Insulin-Like Growth Factor Binding Proteins , Insulin-Like Growth Factor I/metabolism , Insulin-Like Growth Factor II/metabolism , Muscle, Smooth/cytology , Muscle, Smooth/metabolism , Swine
15.
Endocrinology ; 141(7): 2557-66, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10875258

ABSTRACT

The insulin-like growth factor (IGF) system is a major regulator of somatic growth in vertebrates. Both ligands (IGF-I and IGF-II) signal via the same IGF receptor (IGF-IR). Classical IGF-IR invalidation is lethal at birth, so that conditional models are needed to study the postnatal role of this receptor. To establish a genetically inducible invalidation of IGF-IR, we targeted the IGF-IR gene using a construct that introduced a neomycin resistance cassette into intron 2, leaving the rest of the gene intact. This neomycin resistance cassette interfered with the processing of the primary transcript, resulting in there being 12% fewer IGF-binding sites at the cell surface in heterozygous mice and 41% fewer in homozygous mice. Hetero- and homozygous offspring grew more slowly than their wild-type littermates. This difference was noticeable from 4 weeks after birth and was significant from 5 weeks after birth in males. In females, the effect on postnatal growth of insertion of the neo cassette was not significant. In males, IGF-I levels increased moderately (+26%) but significantly, indicating effective feedback regulation of the IGF system. IGF-binding protein-4 (IGFBP-4) levels, estimated by Western ligand blotting, were low in homozygotes (-38%), whereas IGFBP-1, -2, and -3 levels were unaffected. In females, IGF-I and IGFBP-1, -2, -3, and -4 levels did not differ significantly among heterozygous, homozygous, and wild-type animals. We investigated the molecular mechanism involved and characterized two RNA-splicing events that could account for the decrease in IGF-IR. The phenotype of these mice developed exclusively postnatally, and body proportions were maintained. IGF-IRneo mice constitute a new model for human postnatal growth deficiency.


Subject(s)
Animals, Newborn/genetics , Gene Targeting , Growth Disorders/genetics , RNA Splicing/genetics , Receptor, IGF Type 1/genetics , Animals , Animals, Newborn/growth & development , Anti-Bacterial Agents , Base Sequence/genetics , DNA Transposable Elements , Directed Molecular Evolution , Drug Resistance, Microbial/genetics , Female , Humans , Introns/genetics , Male , Mice , Molecular Sequence Data , Neomycin , Phenotype , Somatomedins/metabolism
16.
Endocrinology ; 138(7): 2937-47, 1997 Jul.
Article in English | MEDLINE | ID: mdl-9202238

ABSTRACT

Study of the in vivo functions of the insulin-like growth factor binding proteins (IGFBPs) is complicated by their variety (six molecular species) and the differences in their expression related to tissue of origin and stage of development. To investigate the physiological role of IGFBP-1 in the bloodstream, we induced hepatic overexpression of IGFBP-1 in transgenic mice, placing human IGFBP-1 (hIGFBP-1) cDNA under the control of the alpha1-antitrypsin promoter so as to obtain liver-specific expression. Five transgenic founder mice were raised, only two of which (lines 124 and 149) produced transgenic offspring. Northern blotting revealed transgene expression exclusively in the liver during fetal life and unchanged through to adulthood, whereas expression of the endogenous gene was undetectable beyond 10-15 days postnatally. hIGFBP-1 was detected by western immunoblotting in the plasma of transgenic mice and IRMAs yielded mean concentrations of 2.41 +/- 0.33 ng/ml and 13.69 +/- 1.42 ng/ml in homozygous animals of lines 124 and 149, respectively. In the latter, IGFBP-1 levels were distinctly higher than in heterozygotes (2.99 +/- 0.39 ng/ml), P < 0.0001. These levels remained stable in each given animal and did not change with age. Plasma concentrations of IGF-I measured in line 149 exhibited the well-known profile of an increase from birth up to puberty. Values for heterozygotes were similar to those for wild-type mice, with adult levels (544 +/- 98 ng/ml) slightly below those of controls (630 +/- 56 ng/ml), P < 0.05. In homozygotes they were distinctly lower, with adult levels of 370 +/- 75 ng/ml, P = 0.001. In heterozygous and homozygous adults, there was a negative correlation between IGF-I and IGFBP-1 concentrations (r = 0.8, P < 0.0001), suggesting a link between transgene expression and IGF-I levels. Study of body weight gain in line 149 revealed growth retardation within the first weeks after birth, which was marked in homozygous males and females (P < 0.001) but also present in heterozygous males (P = 0.002), indicating some relationship with transgene expression. In addition, body weight in adult mice was negatively correlated to plasma concentrations of IGFBP-1 (r = 0.7, P < 0.0001). Reproductive function also appeared to be severely affected, especially in homozygous females: mating that failed to result in pregnancy in half of the homozygous females crossed with nontransgenic males, suggestive of impaired fertilization or implantation; interrupted or prolonged pregnancies with fetal and neonatal death. Litter size was reduced in transgenic females (by about half in homozygotes) and in nontransgenic females mated with homozygous males, resulting from pre- or neonatal mortality. Moreover, deaths occurred within the first 5 days of life, with an incidence of approximately 50% in the litters of homozygous females, 12-18% among heterozygotes mated with nontransgenic or heterozygous males, respectively, and 30% among those mated with homozygous males. These results, suggesting that fetal transgene expression largely accounted for ante- and perinatal mortality, were confirmed by the predominance of homozygotes among those that could be analyzed genetically. Similarly impaired reproductive function was seen in line 124, but to a lesser degree. Although the mechanisms responsible for these disorders remain to be determined, our results indicate that permanent and uncontrolled hepatic expression of IGFBP-1, even at low levels, affects fertility in females and both ante- and postnatal development.


Subject(s)
Insulin-Like Growth Factor Binding Protein 1/genetics , Liver/metabolism , Animals , Animals, Newborn/growth & development , Female , Fetal Death , Gene Expression , Humans , Insulin-Like Growth Factor Binding Protein 1/blood , Insulin-Like Growth Factor Binding Protein 1/pharmacology , Insulin-Like Growth Factor I/metabolism , Litter Size , Liver/drug effects , Male , Mice , Mice, Transgenic/genetics , Mice, Transgenic/growth & development , Pregnancy , Promoter Regions, Genetic , Reproduction , Transgenes/genetics , Weight Gain , alpha 1-Antitrypsin/genetics
17.
Endocrinology ; 140(9): 4175-84, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10465290

ABSTRACT

The regulation of insulin-like growth factor binding protein (IGFBP)-4 proteolytic degradation by insulin-like growth factors (IGFs) has been largely studied in vitro, but not in vivo. The aim of this study was to investigate the involvement of IGFs, IGFBP-2, IGFBP-3, and IGFBP-3 proteolytic fragments in the regulation of IGFBP-4 proteolytic activity in ovine ovarian follicles. Follicular fluid from preovulatory follicles contains proteolytic activity degrading exogenous IGFBP-4. The addition of an excess of IGF-I enhanced IGFBP-4 proteolytic degradation, whereas addition of IGFBP-2 or -3 or monoclonal antibodies against IGF-I and -II dose dependently inhibited IGFBP-4 proteolytic degradation. IGF-I and IGF-II, but not LongR3-IGF-I, reversed this inhibition in a dose-dependent manner. C-terminal, but not N-terminal, proteolytic fragments derived from IGFBP-3 (aa 161-264), as well as heparin-binding domain-containing peptides derived from the C-terminal domain of IGFBP-3 and -5 also induced the inhibition of IGFBP-4 proteolytic degradation. Other heparin-binding domain-containing peptides derived from the connective tissue growth factor (CTGF) and from proteins not related to IGFBP, heparan/heparin interacting protein (HIP) and vitronectin, but not from p36 subunit of annexin II tetramer, inhibited IGFBP-4 degradation. Furthermore, IGFBP-3, mutated on its heparin-binding domain, was not able to inhibit IGFBP-4 proteolytic degradation. So, in ovine preovulatory follicles, IGFBP-4 proteolytic degradation both 1) depends on IGFs, and 2) is inhibited by IGFBP-3 via its C-terminal heparin-binding domain as well as by heparin-binding domain containing peptides. These data suggest that in early atretic follicles, the increase in IGFBP-2 participates in the decrease in IGFBP-4 degradation. In late atretic follicles, the increase in the levels of C-terminal IGFBP-3 proteolytic fragments, generated by IGFBP-3 degradation, as well as the increase in IGFBP-5 expression would strengthen the inhibition of IGFBP-4 degradation. This inhibition might be partly mediated by direct interaction of IGFBP-4 proteinase(s) and heparin-binding domain within the C-terminal region from IGFBP-3 and -5.


Subject(s)
Follicular Phase/physiology , Insulin-Like Growth Factor Binding Protein 4/metabolism , Ovarian Follicle/metabolism , Peptide Hydrolases/metabolism , Animals , Female , Heparin/metabolism , Insulin-Like Growth Factor Binding Protein 3/genetics , Insulin-Like Growth Factor Binding Protein 3/metabolism , Insulin-Like Growth Factor Binding Protein 3/pharmacology , Insulin-Like Growth Factor I/pharmacology , Mutation/physiology , Peptide Fragments/pharmacology , Peptides/chemical synthesis , Peptides/pharmacology , Sheep , Somatomedins/physiology
18.
J Clin Endocrinol Metab ; 67(3): 509-14, 1988 Sep.
Article in English | MEDLINE | ID: mdl-2457595

ABSTRACT

The extent to which the association between insulin-like growth factors (IGFs) and their specific binding proteins (BPs) prevents their crossing the capillary barrier was studied by comparing their distribution in serum with that in samples of lymph collected from the lower leg of five subjects undergoing radiographical investigation of the lymphatic system. The IGF concentrations in lymph were 10-30% of the corresponding serum levels, and in each subject the ratios of IGF-I and IGF-II in the lymph to those in the serum were similar. Western blot analysis of the BPs revealed that the five molecular forms identified in serum also were present in lymph, but in significantly smaller quantities. The 41.5K and 38.5K forms, which constitute the binding units of the large complex (approximately 150K) of serum and are also capable of binding IGFs in monomeric form, were present in smaller amounts than the 34K, 30K and 24K forms, which belong specifically to the small complex (approximately 40K) of serum. The BPs extracted from lymph were similar to those of the small complex, with a preferential affinity for IGF-II and only half of the affinity for IGF-I of the BPs extracted from serum. With neutral pH gel filtration of lymph, more than 90% of IGFs and binding activity eluted with the material in the area of the 40K zone. These data indicate that the 150K IGF-BP complexes do not cross the capillary barrier, whereas the 40K complexes do. The function of the former may be to provide a reservoir and buffering action of the IGFs, whereas the latter may be involved in the transport of the IGFs to their target cells.


Subject(s)
Carrier Proteins/analysis , Lymph/analysis , Somatomedins/analysis , Adult , Binding, Competitive , Carrier Proteins/blood , Chromatography, Gel , Female , Humans , Insulin-Like Growth Factor Binding Proteins , Male , Middle Aged , Somatomedins/blood
19.
J Clin Endocrinol Metab ; 86(3): 1260-6, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11238518

ABSTRACT

Limited proteolysis of insulin-like growth factor binding protein-3 (IGFBP-3) is a fundamental mechanism in the regulation of IGF-I bioavailability in the bloodstream. Its measurement by Western immunoblotting provides only semiquantitative estimation. We have developed a ligand immunofunctional assay (LIFA) for quantifying human (h) intact IGFBP-3 in biological fluids. IGFBP-bound IGFs are dissociated and separated by acid pH ultrafiltration, and a monoclonal antibody specific to the first 160 amino acids of IGFBP-3 is used to capture hIGFBP-3 in a solid-phase assay. The complex is then incubated with (125)I-IGF-I, which binds to intact IGFBP-3 but not to its proteolytic fragments. Binding specificity was demonstrated in competition experiments with unlabeled IGF. Nonspecific binding was 1.4%. The fragments comprising residues 1-160 and 1-95 of recombinant hIGFBP-3 [corresponding to the major proteolytic fragments of approximately 30 kDa and (glycosylated) 20 or (nonglycosylated) 16 kDa detected in serum by Western immunoblotting, respectively] fail to bind (125)I-IGF-I when complexed with the monoclonal antibody. Similarly, no binding of (125)I-IGF-I was obtained in the LIFA when applied to plasmas from pregnant women during the final 3 months of pregnancy, where the characteristic 42- to 39-kDa doublet of intact IGFBP-3 is undetectable. The standard curve was established using a pool of plasmas (EDTA) from healthy adults, for which standardization with glycosylated recombinant hIGFBP-3 yielded an intact IGFBP-3 content of 2 microg/mL. The dynamic range of the LIFA was 0.50-3.75 microL equivalent of the plasma pool in a total volume of 300 microL per assay tube, with a sensitivity threshold of approximately 1 ng intact IGFBP-3. Unknown plasma samples were studied at three concentrations. Intra- and interassay variations were 3.6% and 4%, respectively. In 31 healthy adults, the mean plasma concentration of intact IGFBP-3 was 2.24 +/- 0.08 (SEM) mg/L, and that of total IGFBP-3 measured by immunoradiometric assay was 3.27 +/- 0.14 mg/L. The calculated mean proportion of proteolysed IGFBP-3 was 29.4 +/- 1.9%. In these subjects, a close correlation was found between intact and total IGFBP-3 (r = 0.71, P = 0.0001). The LIFA for IGFBP-3, therefore, provides accurate and sensitive measurement of intact IGFBP-3, the form with the functional capacity to sequester IGF-I in the bloodstream by association with the acid-labile subunit in 140-kDa complexes. In combination with total IGFBP-3 and IGF-I assays, the LIFA opens new perspectives in investigating the regulation of IGFBP-3 proteolysis and IGF-I bioavailability in man.


Subject(s)
Immunoassay/methods , Insulin-Like Growth Factor Binding Protein 3/blood , Ligands , Acromegaly/blood , Adult , Antibodies, Monoclonal , Antibody Specificity , Blotting, Western , Chromatography, High Pressure Liquid , Female , Human Growth Hormone/deficiency , Humans , Hydrogen-Ion Concentration , Insulin-Like Growth Factor I , Iodine Radioisotopes , Male , Peptide Fragments/blood , Quality Control , Recombinant Proteins , Reference Values , Sensitivity and Specificity
20.
J Clin Endocrinol Metab ; 60(4): 781-5, 1985 Apr.
Article in English | MEDLINE | ID: mdl-2579089

ABSTRACT

Serum insulin-like growth factor (IGF) and IGF-binding protein (IGF BP) levels were determined in 13 insulin-dependent diabetic patients (30-60 yr of age) during an episode of severe metabolic decompensation and the recovery phase. After separation by acidic gel filtration, the samples were assayed for IGF using a protein-binding assay (which measures mainly IGF I-related peptides) and for IGF BP by measuring the binding activity, in both assays using IGF I as tracer. The reference standard was a pool of normal adult serum with an assigned potency of 1 U IGF and 1 U IGF BP per ml. The mean IGF level in the uncontrolled state, 0.55 +/- 0.05 (SEM) U/ml, was about half that of normal subjects (1.03 +/- 0.03 U/ml, P less than 0.001). With treatment, IGF levels reached the normal range within 3 days. The pattern of changes in IGF BP levels was roughly similar, although the values in the uncontrolled state were less depressed (0.78 +/- 0.04 U/ml vs. 0.98 +/- 0.04 in normal subjects, P less than 0.01). Highly significant correlations (P less than 0.001) were found between IGF levels and the biological parameters reflecting control of the diabetes: glycosuria (r = -0.60), glycemia (r = -0.52), ketonemia (r = -0.65), and HCO3- (r = 0.58). Similar but less significant correlations were found for IGF BP. The mean GH level during the period of metabolic decompensation (9.0 +/- 1.5 ng/ml) was elevated compared to that after recovery (2.9 +/- 0.8 ng/ml) (P less than 0.025). There was a negative correlation between GH values and IGF levels (r = -0.67, P less than 0.001). The correlation with IGF BP was much less significant (r = -0.38, P less than 0.05). The results clearly reflect the role of insulin and nutritional factors in the control of IGF levels. They also support the notion that the biosynthesis of IGF and IGF BP is not regulated in the same way.


Subject(s)
Carrier Proteins/blood , Diabetes Mellitus, Type 1/blood , Insulin/blood , Peptides/blood , Somatomedins/blood , Adult , Female , Humans , Insulin-Like Growth Factor Binding Proteins , Male , Middle Aged , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL