Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
PLoS One ; 10(7): e0132302, 2015.
Article in English | MEDLINE | ID: mdl-26147990

ABSTRACT

BACKGROUND: Insulin-resistant subjects develop more severe and diffuse coronary artery atherosclerosis than insulin-sensitive controls but the mechanisms that mediate this atherosclerosis phenotype are unknown. RESEARCH OBJECTIVE: To determine the metabolic parameters that associate with the severity of coronary atherosclerosis in insulin resistant pigs fed a high fat/high NaCl diet. KEY METHODS: The primary endpoint was severity of coronary atherosclerosis in adult pigs (Sus scrofa, n = 37) fed a high fat diet that also contained high NaCl (56% above recommended levels) for 1 year. PRINCIPAL FINDINGS: Twenty pigs developed severe and diffuse distal coronary artery atherosclerosis (i.e., severe = intimal area as a percent medial area > 200% in at least 2 coronary artery cross sections and diffuse distal = intimal area as a percent medial area ≥ 150% over 3 sections separated by 2 cm in the distal half of the coronary artery). The other 17 pigs had substantially less coronary artery atherosclerosis. All 37 pigs had blood pressure in a range that would be considered hypertensive in humans and developed elevations in total and LDL and HDL cholesterol, weight gain, increased backfat, and increased insulin resistance (Bergman Si) without overt diabetes. Insulin resistance was not associated with atherosclerosis severity. Five additional pigs fed regular pig chow also developed increased insulin resistance but essentially no change in the other variables and little to no detectible coronary atherosclerosis. Most importantly, the 20 high fat/high NaCl diet-fed pigs with severe and diffuse distal coronary artery atherosclerosis had substantially greater increases (p< 0.05) in oxidized LDL (oxLDL) and fructosamine consistent with increased protein glycation. CONCLUSION: In pigs fed a high fat/high NaCl diet, glycated proteins are induced in the absence of overt diabetes and this degree of increase is associated with the development of severe and diffuse distal coronary artery atherosclerosis.


Subject(s)
Blood Pressure/drug effects , Coronary Artery Disease , Fructosamine/blood , Insulin Resistance , Lipoproteins, LDL/blood , Sodium Chloride, Dietary/pharmacology , Animals , Coronary Artery Disease/blood , Coronary Artery Disease/physiopathology , Disease Models, Animal , Severity of Illness Index , Sodium Chloride, Dietary/adverse effects , Sus scrofa
2.
Endocrinology ; 155(12): 4665-75, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25171599

ABSTRACT

Hyperglycemia stimulates secretion of αVß3 ligands from vascular cells, including endothelial cells, resulting in activation of the αVß3 integrin. This study determined whether blocking ligand occupancy of αVß3 would inhibit the development of diabetic nephropathy. Ten diabetic pigs received an F(ab)2 fragment of an antibody directed against the extracellular domain of the ß3-subunit, and 10 received a control IgG F(ab)2 for 18 weeks. Nondiabetic pigs excreted 115 ± 50 µg of protein/mg creatinine compared with control F(ab)2-treated diabetic animals (218 ± 57 µg/mg), whereas diabetic animals treated with the anti-ß3 F(ab)2 excreted 119 ± 55 µg/mg (P < .05). Mesangial volume/glomerular volume increased to 21 ± 2.4% in control-treated diabetic animals compared with 14 ± 2.8% (P < .01) in animals treated with active antibody. Diabetic animals treated with control F(ab)2 had significantly less glomerular podocin staining compared with nondiabetic animals, and this decrease was attenuated by treatment with anti-ß3 F(ab)2. Glomerular basement membrane thickness was increased in the control, F(ab)2-treated diabetic animals (212 ± 14 nm) compared with nondiabetic animals (170 ± 8.8 nm), but it was unchanged (159.9 ± 16.4 nm) in animals receiving anti-ß3 F(ab)2. Podocyte foot process width was greater in control, F(ab)2-treated, animals (502 ± 34 nm) compared with animals treated with the anti-ß3 F(ab)2 (357 ± 47 nm, P < .05). Renal ß3 tyrosine phosphorylation decreased from 13 934 ± 6437 to 6730 ± 1524 (P < .01) scanning units in the anti-ß3-treated group. We conclude that administration of an antibody that inhibits activation of the ß3-subunit of αVß3 that is induced by hyperglycemia attenuates proteinuria and early histologic changes of diabetic nephropathy, suggesting that it may have utility in preventing the progression of this disease complication.


Subject(s)
Diabetic Nephropathies/etiology , Integrin alphaVbeta3/metabolism , Animals , Antibodies, Monoclonal , Diabetic Nephropathies/metabolism , Diabetic Nephropathies/pathology , Glomerular Basement Membrane/pathology , Integrin alphaVbeta3/antagonists & inhibitors , Male , Mice, Inbred BALB C , Podocytes/pathology , Proteinuria/etiology , Swine
3.
Endocrinology ; 151(12): 5851-64, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20881255

ABSTRACT

The IGF-I pathway and renin-angiotensin-aldosterone axis are both involved in the pathogenesis of hypertension and atherosclerosis, but no information is available about IGF-I and aldosterone interaction or their potential synergistic effects in vascular smooth muscle cells (VSMCs). The aims of this study were to investigate whether aldosterone influences IGF-I signaling and to determine the mechanism(s) by which aldosterone affects IGF-I function. Aldosterone resulted in significant increases in the Akt (1.87 ± 0.24, P < 0.001), MAPK (1.78 ± 0.13, P < 0.001), p70S6kinase (1.92 ± 0.15, P < 0.001), IGF-I receptor (1.69 ± 0.05, P < 0.01), and insulin receptor substrate-1 (1.7 ± 0.04, P < 0.01) (fold increase, mean ± SEM, n = 3) phosphorylation responses to IGF-I compared with IGF-I treatment alone. There were also significant increases in VSMC proliferation, migration, and protein synthesis (1.63 ± 0.03-, 1.56 ± 0.08-, and 1.51 ± 0.04-fold increases compared with IGF-I alone, respectively, n = 3, P < 0.001). Aldosterone induced osteopontin (OPN) mRNA expression and activation of αVß3-integrin as well as an increase in the synthesis of IGF-I receptor. The enhancing effects of aldosterone were inhibited by eplerenone (10 µmol/liter), actinomycin-D (20 nmol/liter), and an anti-αVß3-integrin antibody that blocks OPN binding. The antioxidant N-acetylcysteine (2 mmol/liter) completely inhibited the ability of aldosterone to induce any of these changes. In conclusion, our results show that aldosterone enhances IGF-I signaling and biological actions in VSMCs through induction of OPN followed by its subsequent activation of the αVß3-integrin and by increasing IGF-I receptor. These changes are mediated in part through increased oxidative stress. The findings suggest a new mechanism by which aldosterone could accelerate the development of atherosclerosis.


Subject(s)
Aldosterone/pharmacology , Insulin-Like Growth Factor I/metabolism , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/drug effects , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/physiology , Signal Transduction/physiology , Animals , Aorta/cytology , Cell Movement , Cell Proliferation , Cells, Cultured , Integrin alphaVbeta3/genetics , Integrin alphaVbeta3/metabolism , Mitogen-Activated Protein Kinase Kinases/genetics , Mitogen-Activated Protein Kinase Kinases/metabolism , Osteopontin/genetics , Osteopontin/metabolism , Phosphorylation , Proto-Oncogene Proteins c-akt , Ribosomal Protein S6 Kinases, 70-kDa/genetics , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , Swine
4.
Am J Pathol ; 177(4): 1936-45, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20813965

ABSTRACT

Adiponectin is a hormone that lowers glucose production by increasing liver insulin sensitivity. Insulin blocks the generation of biochemical intermediates for glucose production by inhibiting autophagy. However, autophagy is stimulated by an essential mediator of adiponectin action, AMPK. This deadlock led to our hypothesis that adiponectin inhibits autophagy through a novel mediator. Mass spectrometry revealed a novel protein that we call suppressor of glucose by autophagy (SOGA) in adiponectin-treated hepatoma cells. Adiponectin increased SOGA in hepatocytes, and siRNA knockdown of SOGA blocked adiponectin inhibition of glucose production. Furthermore, knockdown of SOGA increased late autophagosome and lysosome staining and the secretion of valine, an amino acid that cannot be synthesized or metabolized by liver cells, suggesting that SOGA inhibits autophagy. SOGA decreased in response to AICAR, an activator of AMPK, and LY294002, an inhibitor of the insulin signaling intermediate, PI3K. AICAR reduction of SOGA was blocked by adiponectin; however, adiponectin did not increase SOGA during PI3K inhibition, suggesting that adiponectin increases SOGA through the insulin signaling pathway. SOGA contains an internal signal peptide that enables the secretion of a circulating fragment of SOGA, providing a surrogate marker for intracellular SOGA levels. Circulating SOGA increased in parallel with adiponectin and insulin activity in both humans and mice. These results suggest that adiponectin-mediated increases in SOGA contribute to the inhibition of glucose production.


Subject(s)
Adiponectin/blood , Adiponectin/pharmacology , Blood Glucose/metabolism , Hypoglycemic Agents/blood , Insulin/blood , Intracellular Signaling Peptides and Proteins/metabolism , Liver/metabolism , AMP-Activated Protein Kinases/metabolism , Adult , Amino Acid Sequence , Animals , Autophagy , Autophagy-Related Proteins , Cloning, Molecular , Female , Humans , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/immunology , Liver/cytology , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Obese , Mice, Transgenic , Middle Aged , Molecular Sequence Data , Peptide Fragments/immunology , Rabbits , Signal Transduction , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Young Adult
5.
Sci Transl Med ; 2(18): 18ra11, 2010 Feb 10.
Article in English | MEDLINE | ID: mdl-20371482

ABSTRACT

Atherosclerotic lesions develop and progress more rapidly in diabetic patients than in nondiabetic individuals. This may be caused by accelerated lesion formation in the high-glucose environment of diabetes. Smooth muscle cells (SMCs) cultured in high glucose are more responsive to growth factors such as insulin-like growth factor-1 (IGF-1). This enhanced response to IGF-1 is due in part to increased activation of the alpha(V)beta(3) integrin. We tested whether alpha(V)beta(3) integrin activation was increased in diabetic animals and whether an antibody to beta(3) would inhibit IGF-1 action and development of atherosclerosis. Eight male pigs were made diabetic with streptozotocin and fed a high-fat diet. A F(ab)(2) antibody fragment directed at beta(3) was infused into one femoral artery, whereas the other artery received control F(ab)(2) for 3.5 months. There was a 65 +/- 8% reduction in atherosclerotic lesion area in the arteries treated with F(ab)(2) antibody to beta(3). Phosphorylation of beta(3) was reduced by 75 +/- 18% in vessels treated with the antibody. Shc and mitogen-activated protein kinase phosphorylation, which are required for IGF-1-stimulated SMC proliferation, were also significantly reduced. We conclude that activation of IGF-1 receptor and alpha(V)beta(3)-linked signaling pathways accelerates atherosclerosis in diabetes and that administration of an antibody to beta(3) to diabetic pigs inhibits alpha(V)beta(3) activation, IGF-1-stimulated signaling, and atherosclerotic lesion development. This approach offers a potential therapeutic approach to the treatment of this disorder.


Subject(s)
Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , Atherosclerosis/complications , Atherosclerosis/prevention & control , Diabetes Mellitus, Experimental/complications , Integrin alphaVbeta3/immunology , Animals , Atherosclerosis/metabolism , Atherosclerosis/pathology , Cells, Cultured , Femoral Artery/metabolism , Femoral Artery/pathology , Immunoglobulin Fab Fragments , Insulin-Like Growth Factor I/metabolism , Male , Mice , Myocytes, Smooth Muscle/metabolism , Phosphorylation , Signal Transduction , Swine , Time Factors
6.
J Biol Chem ; 285(21): 15682-95, 2010 May 21.
Article in English | MEDLINE | ID: mdl-20207740

ABSTRACT

Vascular smooth muscle cells maintained in normal (5.6 mm) glucose respond to insulin-like growth factor-I (IGF-I) with increased protein synthesis but do not proliferate. In contrast, hyperglycemia alters responsiveness to IGF-I, resulting in increased SHPS-1 phosphorylation and assembly of a signaling complex that enhances MAPK and phosphatidylinositol 3-kinase pathways. Hyperglycemia also reduces the basal IRS-1 concentration and IGF-I-stimulated IRS-1-linked signaling. To determine if failure to down-regulate IRS-1 alters vascular smooth muscle cell (VSMC) responses to IGF-I, we overexpressed IRS-1 in VSMCs maintained in high glucose. These cultures showed reduced SHPS-1 phosphorylation, transfer of SHP-2 to SHPS-1, and impaired Shc and MAPK phosphorylation and cell proliferation in response to IGF-I. In vitro studies demonstrated that SHPS-1 was a substrate for type I IGF receptor (IGF-IR) and that IRS-1 competitively inhibited SHPS-1 phosphorylation. Exposure of VSMC cultures to a peptide that inhibited IRS-1/IGF-IR interaction showed that IRS-1 binding to IGF-IR impairs SHPS-1 phosphorylation in vivo. IRS-1 also sequestered SHP-2. Expression of an IRS-1 mutant (Y1179F/Y1229F) reduced IRS-1/SHP-2 association, and exposure of cells expressing the mutant to the inhibitory peptide enhanced SHPS-1 phosphorylation and SHP-2 transfer. This result was confirmed by expressing an IRS-1 mutant that had both impaired binding to IGF-IR and to SHP-2 IGF-I increased SHPS-1 phosphorylation, SHP-2 association with SHPS-1, Shc MAPK phosphorylation, and proliferation in cells expressing the mutant. We conclude that IRS-1 is an important factor for maintaining VSMCs in the non-proliferative state and that its down-regulation is a component of the VSMC response to hyperglycemic stress that results in an enhanced response to IGF-I.


Subject(s)
Antigens, Differentiation/metabolism , Insulin Receptor Substrate Proteins/metabolism , Insulin-Like Growth Factor I/metabolism , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Receptor, IGF Type 1/metabolism , Receptors, Immunologic/metabolism , Animals , Antigens, Differentiation/genetics , Cell Line , Cell Proliferation/drug effects , Glucose/pharmacology , Humans , Insulin Receptor Substrate Proteins/genetics , Insulin-Like Growth Factor I/genetics , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/physiology , Mice , Mutation , Phosphorylation/drug effects , Phosphorylation/physiology , Protein Binding , Protein Tyrosine Phosphatase, Non-Receptor Type 11/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 11/metabolism , Rats , Receptor, IGF Type 1/genetics , Receptors, Immunologic/genetics , Shc Signaling Adaptor Proteins , Stress, Physiological/drug effects , Stress, Physiological/physiology , Sweetening Agents/pharmacology , Swine
7.
Exp Diabetes Res ; 2009: 267107, 2009.
Article in English | MEDLINE | ID: mdl-20111736

ABSTRACT

Increased responsiveness of vascular cells to the growth factor IGF-I has been implicated in complications associated with diabetes. Here we describe the development of an assay and screening of a library of compounds for their ability to accelerate cleavage of the transmembrane protein integrin-associated protein (IAP) thereby disrupting the association between IAP and SHPS-1 which we have shown as critical for the enhanced response of vascular cells to IGF-I. The cell-based ELISA utilizes an antibody that specifically detects cleaved, but not intact, IAP. Of the 1040 compounds tested, 14 were considered active by virtue of their ability to stimulate an increase in antibody-binding indicative of IAP cleavage. In experiments with smooth muscle and retinal endothelial cell cultures in hyperglycemic conditions, each active compound was shown to accelerate the cleavage of IAP, and this was associated with a decrease in IAP association with SHPS-1 as determined by coimmunoprecipitation of the proteins from cell lysates. As a consequence of the acceleration in IAP cleavage, the compounds were shown to inhibit IGF-I-stimulated phosphorylation of key signaling molecules including Shc and ERK1/2, and this in turn was associated with a decrease in IGF-I-stimulated cell proliferation. Identification of these compounds that utilize this mechanism has the potential to yield novel therapeutic approaches for the prevention and treatment of vascular complications associated with diabetes.


Subject(s)
Hyperglycemia/physiopathology , Insulin-Like Growth Factor I/antagonists & inhibitors , Signal Transduction/physiology , Animals , CD47 Antigen/physiology , Cattle , Cell Proliferation , Cells, Cultured , Disease Models, Animal , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Endothelium, Vascular/physiopathology , Glucose/pharmacology , Insulin-Like Growth Factor I/physiology , Signal Transduction/drug effects , Swine
8.
J Cell Biochem ; 105(2): 437-46, 2008 Oct 01.
Article in English | MEDLINE | ID: mdl-18615592

ABSTRACT

The interaction between the arginine glycine and aspartic acid motif (RGD) of integrin ligands such as vitronectin and the integrin receptor alphaVbeta3 in mediating cell attachment has been well described. Similarly, the ability of disintegrins, small RGD containing peptides, to inhibit cell attachment and other cellular processes has also been studied extensively. Recently, we characterized a second site of interaction between vitronectin and its integrin partner. We determined that amino acids within the heparin-binding domain of vitronectin bind to a cysteine loop (C-loop) region of beta3 and that this interaction is required for the positive effects of alphaVbeta3 ligand occupancy on IGF-I signaling in smooth muscle cells. In this study we examine the signaling events activated following ligand binding of disintegrins to the alphaVbeta3 and the ability of these signals to be regulated by binding of the heparin-binding domain of vitronectin. We demonstrate that disintegrin ligand binding activates a series of events including the sequential activation of the tyrosine kinases c-Src and Syk. This leads to the activation of calpain and the cleavage of the beta3 cytoplasmic tail. Addition of vitronectin or a peptide homologous to the heparin-binding domain inhibited activation of this pathway. Our results suggest that the signaling events that occur following ligand binding to the alphaVbeta3 integrin reflects a balance between the effects mediated through the RGD binding site interaction and the effects mediated by the heparin binding site interaction and that for intact vitronectin the effect of the heparin-binding domain predominates.


Subject(s)
Integrin alphaVbeta3/metabolism , Signal Transduction , Vitronectin/metabolism , Animals , Binding Sites , CSK Tyrosine-Protein Kinase , Disintegrins/metabolism , Heparin , Intracellular Signaling Peptides and Proteins/metabolism , Ligands , Myocytes, Smooth Muscle/metabolism , Protein Binding , Protein-Tyrosine Kinases/metabolism , Swine , Syk Kinase , src-Family Kinases
9.
Growth Horm IGF Res ; 17(4): 265-70, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17412627

ABSTRACT

Under usual conditions, the role of IGF-I in vascular cell types is to maintain cellular protein synthesis and cell size, and even excess IGF-I does not stimulate proliferation. In pathophysiologic states, such as hyperglycemia, smooth muscle cells (SMC) dedifferentiate and change their responsiveness to IGF-I. During hyperglycemia IGF-I stimulates both SMC migration and proliferation. Our laboratory has investigated the molecular mechanism by which this change is mediated. During hyperglycemia SMC secrete increased concentrations of thrombospondin, vitronectin and osteopontin, ligands for the integrin alphaVbeta3. Activation of alphaVbeta3 stimulates recruitment of a tyrosine phosphatase, SHP-2. Exposure of SMC to IGF-I results in phosphorylation of the transmembrane protein, SHPS-1, which provides a docking site for alphaVbeta3-associated SHP-2. After IGF-I stimulation SHP-2 associates with Src kinase, which associates with the signaling protein Shc. Src phosphorylates Shc, resulting in activation of MAP kinases, which are necessary both for stimulation of cell proliferation and migration. Blocking activation of alphaVbeta3 results in an inability of IGF-I to stimulate Shc phosphorylation. Under conditions of normoglycemia, there are insufficient alphaVbeta3 ligands to recruit SHP-2, and no increase in Shc phosphorylation can be demonstrated in SMC. In contrast, if alphaVbeta3 ligands are added to cells in normal glucose, the signaling events that are necessary for Shc phosphorylation can be reconstituted. Therefore when SMC are exposed to normal glucose they are protected from excessive stimulation of mitogenesis by IGF-I. With hyperglycemia there is a marked increased in alphaVbeta3 ligands and Shc phosphorylation in response to IGF-I is sustained. These findings indicate that in SMC hyperglycemic stress leads to altered IGF-I signaling, which allows the cells to undergo a mitogenic response, and which may contribute to the development of atherosclerosis.


Subject(s)
Hyperglycemia/physiopathology , Insulin-Like Growth Factor I/pharmacology , Integrin alphaVbeta3/physiology , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/drug effects , Stress, Physiological/physiopathology , Animals , Blood Glucose/physiology , Models, Biological , Myocytes, Smooth Muscle/metabolism
10.
Mol Endocrinol ; 20(4): 881-92, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16322097

ABSTRACT

We have shown that vitronectin (Vn) binding to a cysteine loop sequence within the extracellular domain of the beta3-subunit (amino acids 177-184) of alphaVbeta3 is required for the positive effects of Vn on IGF-I signaling. When Vn binding to this sequence is blocked, IGF-I signaling in smooth muscle cells is impaired. Because this binding site is distinct from the site on beta3 to which the Arg-Gly-Asp sequence of extracellular matrix ligands bind (amino acids 107-171), we hypothesized that the region of Vn that binds to the cysteine loop on beta3 is distinct from the region that contains the Arg-Gly-Asp sequence. The results presented in this study demonstrate that this heparin binding domain (HBD) is the region of Vn that binds to the cysteine loop region of beta3 and that this region is sufficient to mediate the positive effects of Vn on IGF-I signaling. We provide evidence that binding of the HBD of Vn to alphaVbeta3 has direct effects on the activation state of beta3 as measured by beta3 phosphorylation. The increase in beta3 phosphorylation associated with exposure of cells to this HBD is associated with enhanced phosphorylation of the adaptor protein Src homology 2 domain-containing transforming protein C and enhanced activation MAPK, a downstream mediator of IGF-I signaling. We conclude that the interaction of the HBD of Vn binding to the cysteine loop sequence of beta3 is necessary and sufficient for the positive effects of Vn on IGF-I-mediated effects in smooth muscle cells.


Subject(s)
Heparin/metabolism , Insulin-Like Growth Factor I/metabolism , Vitronectin/chemistry , Vitronectin/metabolism , Amino Acid Sequence , Animals , Base Sequence , Binding Sites , DNA/genetics , Humans , In Vitro Techniques , Molecular Sequence Data , Muscle, Smooth/metabolism , Phosphorylation , Protein Structure, Tertiary , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Signal Transduction , Swine , Vitronectin/genetics
11.
Mol Endocrinol ; 20(2): 405-13, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16195248

ABSTRACT

The response of smooth muscle cells to IGF-I requires ligand occupancy of the alphaVbeta3 integrin. We have shown that vitronectin (Vn) is required for IGF-I-stimulated migration or proliferation, whereas the anti-alphaVbeta3 monoclonal antibody, LM609, which inhibits ligand binding, blocks responsiveness of these cells to IGF-I. The amino acids 177-184 ((177)CYDMKTTC(184)) within the extracellular domain of beta3 have been proposed to confer the ligand specificity of alphaVbeta3; therefore, we hypothesized that ligand binding to the 177-184 cysteine loop of beta3 may be an important regulator of the cross talk between alphaVbeta3 and IGF-I in SMCs. Here we demonstrate that blocking ligand binding to a specific amino acid sequence within the beta3 subunit of alphaVbeta3 (i.e. amino acids 177-184) blocked Vn binding to the beta3 subunit of alphaVbeta3 and correspondingly beta3 phosphorylation was decreased. In the presence of this antibody, IGF-I-stimulated Shc phosphorylation and ERK 1/2 activation were impaired, and this was associated with an inhibition in the ability of IGF-I to stimulate an increase in migration or proliferation. Furthermore, in cells expressing a mutated form of beta3 in which three critical residues within the 177-184 sequence were altered beta3 phosphorylation was decreased. This was associated with a loss of IGF-I-stimulated Shc phosphorylation and impaired smooth muscle cell proliferation in response to IGF-I. In conclusion, we have demonstrated that the 177-184 sequence of beta3 is necessary for Vn binding to alphaVbeta3 and that ligand occupancy of this site is necessary for an optimal response of smooth muscle cells to IGF-I.


Subject(s)
Insulin-Like Growth Factor I/pharmacology , Integrin alphaVbeta3/metabolism , Integrin beta3/metabolism , Myocytes, Smooth Muscle/drug effects , Vitronectin/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Amino Acid Sequence , Amino Acid Substitution , Antibodies/pharmacology , Cell Movement , Cell Proliferation , Cells, Cultured , Humans , Integrin alphaVbeta3/antagonists & inhibitors , Ligands , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Molecular Sequence Data , Myocytes, Smooth Muscle/metabolism , Phosphorylation , Shc Signaling Adaptor Proteins , Signal Transduction , Src Homology 2 Domain-Containing, Transforming Protein 1 , Vitronectin/antagonists & inhibitors
12.
J Cell Physiol ; 203(2): 328-34, 2005 May.
Article in English | MEDLINE | ID: mdl-15700281

ABSTRACT

Insulin-like growth factor binding protein-5 (IGFBP-5) and thrombospondin-1 (TS-1) are both present in extracellular matrix (ECM). Both proteins have been shown to bind to one another with high affinity. The purpose of these studies was to determine how the interaction between IGFBP-5 and TS-1 modulates IGF-I actions in porcine aortic smooth muscle cells (pSMC) in culture. The addition of increasing concentrations of TS-1 to pSMC cultures enhanced the protein synthesis and cell migration responses to IGF-I; whereas the addition of IGFBP-5 alone resulted in minimal changes. In contrast, the addition of IGFBP-5 to cultures that were also exposed to IGF-I and TS-1 resulted in inhibition of protein synthesis. When the cell migration response was assessed, the response to IGF-I plus TS-1 was also significantly inhibited by the addition of IGFBP-5, whereas 1.0 microg/ml of IGFBP-5 alone had no effect on the response to IGF-I. To determine the molecular mechanism by which this inhibition occurred, a mutant form of IGFBP-5 that does not bind to IGF-I was tested. This mutant was equipotent compared to native IGFBP-5 in its ability to inhibit both protein synthesis and cell migration responses to IGF-I plus TS-1 thus excluding the possibility that IGFBP-5 was inhibiting the response to TS-1 and IGF-I by inhibiting IGF-I binding to the IGF-I receptor. To determine if an interaction between TS-1 and IGFBP-5 was the primary determinant of the inhibitory effect of IGFBP-5, an IGFBP-5 mutant that bound poorly to TS-1 was utilized. The addition of 1.0 microg/ml of this mutant did not inhibit the protein synthesis or cell migration responses to IGF-I plus TS-1. To determine the mechanism by which IGFBP-5 binding to TS-1 inhibited cellular responses to TS-1 plus IGF-I, TS-1 binding to integrin associated protein (IAP) was assessed. The addition of IGFBP-5 (1.0 microg/ml) inhibited TS-1-IAP association. In contrast, a mutant form of IGFBP-5 that bound poorly to TS-1 had a minimal effect on TS-1 binding to IAP. Further analysis showed that IGFBP-5 addition altered the ability of TS-1 to modulate the SHPS-1/IAP interaction. When the IGFBP-5 mutant that did not bind to IGF-I was incubated with TS-1 and IGF-I, it inhibited the capacity of TS-1 to enhance the IGF-I receptor phosphorylation and MAP kinase activation in response to IGF-I. In contrast, the IGFBP-5 mutant that did not bind to TS-1 had no effect on IGF-I stimulated IGF-I receptor phosphorylation or MAP kinase activation. These results indicate that IGFBP-5 inhibits the binding of TS-1 to IAP, and this results in an alteration of the ability of TS-1 to modulate the disruption of the IAP/SHPS-1 interaction which leads to attenuation of the ability of TS-1 to enhance cellular responsiveness to IGF-I.


Subject(s)
Down-Regulation/physiology , Insulin-Like Growth Factor Binding Protein 5/metabolism , Insulin-Like Growth Factor I/metabolism , Muscle, Smooth, Vascular/metabolism , Thrombospondin 1/metabolism , Animals , Antigens, CD/metabolism , Antigens, Differentiation/metabolism , CD47 Antigen , Cell Movement/drug effects , Cell Movement/physiology , Cells, Cultured , Dose-Response Relationship, Drug , Down-Regulation/drug effects , Insulin-Like Growth Factor Binding Protein 5/pharmacology , Insulin-Like Growth Factor I/antagonists & inhibitors , Integrin alphaVbeta3/drug effects , Integrin alphaVbeta3/metabolism , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/physiology , Membrane Glycoproteins/metabolism , Muscle, Smooth, Vascular/drug effects , Mutation/genetics , Neural Cell Adhesion Molecule L1/metabolism , Phosphorylation/drug effects , Protein Binding/physiology , Receptor, IGF Type 1/metabolism , Receptors, Immunologic/metabolism , Sus scrofa , Thrombospondin 1/pharmacology
13.
J Biol Chem ; 278(44): 42886-92, 2003 Oct 31.
Article in English | MEDLINE | ID: mdl-12917428

ABSTRACT

Insulin-like growth factor (IGF)-I is a pleiotropic hormone that regulates vascular smooth muscle cell (VSMC) migration, proliferation, apoptosis, and differentiation. These actions are mediated by the IGF-I receptor. How activation of the same receptor by the same ligand leads to these diverse cellular responses is not well understood. Here we describe a novel mechanism specifying VSMC responses to IGF-I stimulation, distinctive for the pivotal roles of local IGF-binding proteins (IGFBPs). The role of local IGFBPs was indicated by comparing the activities of IGF-I and des-1-3-IGF-I, an IGF-I analog with reduced binding affinity to IGFBPs. Compared with IGF-I, des-1-3-IGF-I was more potent in stimulating DNA synthesis but much less potent in inducing directed migration of VSMCs. When the effects of individual IGFBPs were tested, IGFBP-2 and IGFBP-4 were found to inhibit IGF-I-stimulated DNA synthesis and migration. IGFBP-5 had an inhibitory effect on IGF-I-stimulated DNA synthesis, but it strongly potentiated IGF-I-induced VSMC migration. By using a non-IGF-binding IGFBP-5 mutant and an IGF-I-neutralizing antibody, it was demonstrated that IGFBP-5 also stimulates VSMC migration in an IGF-independent manner. This effect of IGFBP-5 was inhibited by soluble heparin and by treating cells with heparinase. Mutation of the heparin-binding motif of IGFBP-5 reduced its migration promoting activity. These findings suggest that local IGFBPs are important determinants of cellular responses to IGF-I stimulation, and a key player in this paradigm is IGFBP-5. IGFBP-5 not only modulates IGF-I actions, but it also stimulates cell migration by interacting with cell-surface heparan sulfate proteoglycans.


Subject(s)
Insulin-Like Growth Factor I/metabolism , Muscle, Smooth, Vascular/cytology , Animals , Cell Membrane/metabolism , Cell Movement , Chemotaxis , DNA/metabolism , Dose-Response Relationship, Drug , Glycosaminoglycans/metabolism , Heparan Sulfate Proteoglycans/metabolism , Heparin Lyase/metabolism , Insulin-Like Growth Factor Binding Protein 2/metabolism , Insulin-Like Growth Factor Binding Protein 4/metabolism , Insulin-Like Growth Factor Binding Protein 5/metabolism , Ligands , Mutation , Protein Binding , Swine/metabolism , Time Factors , Transfection
14.
Endocrinology ; 144(6): 2489-95, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12746311

ABSTRACT

IGF binding protein-5 (IGFBP-5) is an important trophic factor for controlling the actions of IGF-I in human dermal fibroblasts and porcine aortic smooth muscle cells. When IGFBP-5 is associated with extracellular matrix, it acts to enhance the cell growth response to IGF-I. The amount of IGFBP-5 within the extracellular matrix is related in part to the amount that is present in conditioned medium, which is related to its rate of synthesis and degradation. A serine protease that degrades IGFBP-5 is present in the conditioned medium of both of these cell types. Because the IGFBP-5 protease activity that is secreted by fibroblasts has been shown to be due to the complement components C1r and C1s, these studies were undertaken to determine whether smooth muscle cells also secreted these proteases and to identify some of the factors that regulate their secretion by both cell types. Both smooth muscle cells and human fibroblasts were shown to release C1r and C1s into conditioned medium. Both C1r and C1s were detected as activated forms, as determined by SDS-PAGE using reducing conditions. The addition of increasing concentrations of either IL-1beta or TNFalpha resulted in increased synthesis of C1s by fibroblasts and smooth muscle cells, and they each increased C1r release. TNFalpha (50 ng/ml) and IL-1beta (20 ng/ml) resulted in maximum stimulation of release of both proteases. In contrast dexamethasone (10(-7) M) had no effect on C1s release and stimulated C1r release only by smooth muscle cells. To determine the physiological significance of this increase in C1r and C1s, the amount of IGFBP-5 protease activity that was present in conditioned medium was determined before and after exposure to TNFalpha, IL-beta, and dexamethasone. All three compounds resulted in an increase in the amount of IGFBP-5 proteolytic activity. Dexamethasone inhibited the release of C(1) inhibitor from fibroblasts, and this contributed to the net increase in proteolytic activity. TNFalpha inhibited the smooth muscle cell DNA synthesis response to IGF-I, but the effect of IGF-I was partially restored by the addition of C1 inhibitor. In conclusion, both C1r and C1s are released by cultured fibroblasts, and the release of each into fibroblast or porcine smooth muscle cells medium is stimulated by TNFalpha and IL-1beta. This increase results in a net increase in IGFBP-5 proteolysis, which has the potential to modify IGF-I and IGFBP-5 actions.


Subject(s)
Fibroblasts/metabolism , Insulin-Like Growth Factor Binding Protein 5/genetics , Insulin-Like Growth Factor Binding Protein 5/metabolism , Muscle, Smooth, Vascular/metabolism , Animals , Aorta/cytology , Cells, Cultured , Complement C1r/metabolism , Complement C1s/metabolism , Culture Media, Conditioned/metabolism , Culture Media, Conditioned/pharmacology , Extracellular Matrix/metabolism , Fibroblasts/cytology , Fibroblasts/drug effects , Gene Expression/physiology , Humans , Insulin-Like Growth Factor I/pharmacology , Muscle, Smooth, Vascular/cytology , Serine Endopeptidases/metabolism , Skin/cytology , Swine
15.
Arthritis Rheum ; 46(3): 694-703, 2002 Mar.
Article in English | MEDLINE | ID: mdl-11920405

ABSTRACT

OBJECTIVE: The complement component C1s is present in dog joint fluid in an activated state. Since C1s degrades insulin-like growth factor binding protein 5 (IGFBP-5), we undertook to determine whether inhibiting C1s in joint fluid would result in an increase in the amount of intact IGFBP-5 and IGF-1 in cartilage and joint fluid, and whether C1s inhibition would be associated with a reduction in cartilage destruction during the development of osteoarthritis (OA). METHODS: Twenty-two dogs were randomized to 3 treatment groups. All dogs underwent anterior cruciate ligament transection and were exercised. Dogs received 1 of 3 treatments: buffer alone (controls; n = 6); PB-145, a peptide derived from the sequence of antithrombin III (n = 9); and pentosan polysulfate (PPS; n = 7). PB-145 or saline was injected into the joint space 3 times per week for 3 weeks. PPS was injected intramuscularly weekly for 3 weeks. RESULTS: Joint histology showed preservation of chondrocytes and a smooth joint surface in the animals treated with PB-145 and PPS. Mankin scoring showed statistically significant reductions in joint destruction with PB-145 and PPS treatments (P < 0.01) compared with buffer control. Mean active collagenase concentrations were decreased by these two treatments. Immunoblotting of joint fluid showed that both treatments increased concentrations of intact IGFBP-5. Direct analysis of IGFBP-3 and IGFBP-5 protease activity showed that IGFBP-5 was degraded more rapidly and that PB-145 and PPS inhibited the degradation of both proteins. Total IGF-1 concentrations in joint fluid were increased 5.6-5.8-fold by these two treatments. Analysis showed that C1s was being activated in joint fluid and that its activation was inhibited by the addition of PB-145 or PPS. CONCLUSION: The findings suggest that direct inhibition of the serine protease C1s results in increased concentrations of intact IGFBP-5 and that proteolysis of IGFBP-3 is also inhibited, probably by the inhibition of some other protease. This increase in concentrations of intact IGFBP-3 and IGFBP-5 leads to an increase in IGF-1 which is associated with an improvement in joint architecture during the development of OA.


Subject(s)
Cartilage, Articular/metabolism , Complement C1s/metabolism , Insulin-Like Growth Factor Binding Protein 5/metabolism , Insulin-Like Growth Factor I/metabolism , Osteoarthritis/metabolism , Osteoarthritis/pathology , Synovial Fluid/metabolism , Animals , Antithrombin III/pharmacology , Complement C1s/drug effects , Dogs , Enzyme Inhibitors/pharmacology , Female , Hindlimb , Insulin-Like Growth Factor Binding Protein 3/metabolism , Osmolar Concentration , Pentosan Sulfuric Polyester/pharmacology , Peptide Fragments/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...