Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 56
Filter
Add more filters










Publication year range
1.
Biochem Biophys Res Commun ; 445(2): 375-80, 2014 Mar 07.
Article in English | MEDLINE | ID: mdl-24525132

ABSTRACT

Proteosome inhibitors such as bortezomib (BTZ) have been used to treat muscle wasting in animal models. However, direct effect of BTZ on skeletal muscle cells has not been reported. In the present study, our data showed that C2C12 cells exhibited a dose-dependent decrease in cell viability in response to increasing concentrations of BTZ. Consistent with the results of cell viability, Annexin V/PI analysis showed a significant increase in apoptosis after exposing the cells to BTZ for 24h. The detection of cleaved caspase-3 further confirmed apoptosis. The apoptosis induced by BTZ was associated with reduced expression of p-ERK. Cell cycle analysis revealed that C2C12 cells underwent G2/M cell cycle arrest when incubated with BTZ for 24h. Furthermore, BTZ inhibited formation of multinucleated myotubes. The inhibition of myotube formation was accompanied by decreased expression of Myogenin. Our data suggest that BTZ induces cell death and inhibits differentiation of C2C12 cells at clinically relevant doses.


Subject(s)
Apoptosis/drug effects , Boronic Acids/pharmacology , G2 Phase Cell Cycle Checkpoints/drug effects , M Phase Cell Cycle Checkpoints/drug effects , Muscle Fibers, Skeletal/drug effects , Proteasome Inhibitors/pharmacology , Pyrazines/pharmacology , Animals , Bortezomib , Cell Line , Cell Survival/drug effects , Down-Regulation/drug effects , Extracellular Signal-Regulated MAP Kinases/genetics , Mice , Muscle Fibers, Skeletal/cytology , Muscle Fibers, Skeletal/metabolism
2.
Circulation ; 105(10): 1220-5, 2002 Mar 12.
Article in English | MEDLINE | ID: mdl-11889017

ABSTRACT

BACKGROUND: Inflammatory mediators such as tumor necrosis factor-alpha (TNF-alpha), interleukin 1beta (IL-1beta), IL-6, and interferon gamma (IFN-gamma) may change coronary plaque integrity by altering vascular smooth muscle cell (VSMC) survival and modifying the extracellular matrix. Insulin-like growth factor-1 (IGF-1) prevents apoptosis, promotes matrix formation, and can decrease TNF-alpha or IL-1beta--induced proteoglycan degradation. METHODS AND RESULTS: To determine the effects of cytokines on the IGF-1 system, rat aortic VSMCs were exposed to TNF-alpha (10 to 500 ng/mL), IL-1beta (20 pg to 10 ng/mL), IL-6 (100 pg to 15 ng/mL), or IFN-gamma (10 to 600 U/mL). IL-1beta, IL-6, and IFN-gamma did not regulate IGF-1, IGF-1 receptor (R), or IGF binding proteins (IGFBPs). However, TNF-alpha markedly decreased IGF-1 mRNA (85% reduction at 24 hours) and increased IGFBP-3 mRNA and protein (300% increase at 24 hours). These changes were blocked by actinomycin D, consistent with a transcriptional mechanism. Experiments using TNF binding protein-1 indicated that these effects were not attributable to secretion of an autocrine factor. Anti--IGFBP-3 antibodies increased VSMC DNA synthesis 3-fold. In addition, apoptosis induced by TNF-alpha, IFN-gamma, and Fas ligand was markedly reduced by desamino-(1-3)-IGF-1. CONCLUSIONS: TNF-alpha, a cytokine that is upregulated in atherosclerotic plaques, reduces IGF-1 and increases IGFBP-3 in VSMCs, likely leading to a reduction in bioactive IGF-1. Because IGF-1 is important for growth and survival of VSMCs, its downregulation by TNF-alpha possibly plays a crucial role in acute and chronic coronary syndromes by decreasing VSMC viability and promoting plaque instability.


Subject(s)
Insulin-Like Growth Factor Binding Protein 3/metabolism , Insulin-Like Growth Factor I/metabolism , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/metabolism , Receptors, Tumor Necrosis Factor , Tumor Necrosis Factor-alpha/pharmacology , Animals , Antibodies/pharmacology , Apoptosis/drug effects , Arteriosclerosis/etiology , Arteriosclerosis/metabolism , Carrier Proteins/metabolism , Carrier Proteins/pharmacology , Cells, Cultured , Culture Media, Conditioned/pharmacology , DNA/biosynthesis , Dose-Response Relationship, Drug , Fas Ligand Protein , Gene Expression/drug effects , Insulin-Like Growth Factor Binding Protein 3/antagonists & inhibitors , Insulin-Like Growth Factor I/analogs & derivatives , Insulin-Like Growth Factor I/genetics , Interferon-gamma/pharmacology , Interleukin-1/pharmacology , Interleukin-6/pharmacology , Membrane Glycoproteins/pharmacology , Muscle, Smooth, Vascular/cytology , Nucleic Acid Synthesis Inhibitors/pharmacology , RNA, Messenger/metabolism , Rats , Receptor, IGF Type 1/genetics , Receptor, IGF Type 1/metabolism , Receptors, Tumor Necrosis Factor, Type I , Tumor Necrosis Factor Decoy Receptors , Tumor Necrosis Factor-alpha/metabolism
3.
J Mol Cell Cardiol ; 33(10): 1777-89, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11603921

ABSTRACT

Insulin-like growth factor-1 (IGF-1) plays an important role in migration, cell cycle progression and survival of vascular smooth muscle cells (VSMC). We investigated the specific localization of IGF-1 and its receptor (IGF-1R) and their association with apoptosis and the expression of apoptosis-related proteins in early and advanced atherosclerotic lesions. Human atherosclerotic plaques (n=23) from patients undergoing aortic, carotid or femoral arterial surgery were studied. Immunohistochemistry and in situ hybridization revealed significantly higher expression of IGF-1 and IGF-1R in the media than in the intima of early atherosclerotic lesions (P<0.01). Medial VSMC positive for BAX, a proapoptotic protein of the B-cell CLL/lymphoma 2 (BCL2) family, showed colocalization of IGF-1. Apoptosis, as detected by DNA in situ terminal deoxynucleotidyl transferase end labeling (TUNEL), was not present in these early lesions. In advanced atherosclerotic plaques, the expression of IGF-1 and IGF-1R was significantly lower in the intimal regions with macrophage infiltration than in those without macrophage infiltration or than in the media (P<0.01). Furthermore, IGF-1 and IGF-1R immunoreactivity was markedly lower in intimal TUNEL-positive VSMC compared with intimal BAX-positive and medial VSMC (P<0.01). We conclude that IGF-1 and IGF-1R expression are reduced in the deep intima of early atherosclerotic lesions and in areas of advanced plaques with macrophage infiltration. Since IGF-1 is a potent survival factor for VSMC, poor expression of IGF-1 and IGF-1R in intimal regions with macrophage infiltration would likely contribute to triggering VSMC apoptosis potentially leading to plaque weakening, plaque rupture and acute coronary events.


Subject(s)
Apoptosis , Arteriosclerosis/pathology , Insulin-Like Growth Factor I/biosynthesis , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/pathology , Proto-Oncogene Proteins c-bcl-2 , Aged , DNA, Complementary/metabolism , Female , Humans , Immunohistochemistry , In Situ Hybridization , In Situ Nick-End Labeling , Macrophages/metabolism , Male , Middle Aged , Proto-Oncogene Proteins/biosynthesis , Receptor, IGF Type 1/biosynthesis , bcl-2-Associated X Protein
4.
J Investig Med ; 49(3): 258-64, 2001 May.
Article in English | MEDLINE | ID: mdl-11352183

ABSTRACT

BACKGROUND: Vessel wall responses to percutaneous transluminal coronary angioplasty include neointimal proliferation and arterial remodeling. The contraction of a collagen gel is a good in vitro model of wound repair and vascular remodeling. Because irradiation is an important new therapeutic modality capable of preventing restenosis, the purpose of this study was to evaluate the effect of irradiation on the contraction of a collagen gel by smooth muscle cells (SMCs), on SMCs viability, and on DNA synthesis. METHODS: We studied the effect of different doses of gamma-irradiation (0 [control], 6, 12, and 18 Gy) on the contraction of a collagen gel seeded with SMCs (calf carotid arteries) during a period of 15 days. RESULTS: Maximal gel diameter reduction (from 35 to 6.8 mm, +/-0.5 mm in control) was markedly inhibited in the 6-, 12-, and 18-Gy groups (35 to 13.7 mm, +/-0.8 mm; 35 to 15.5 mm, +/-0.9 mm; and 35 to 16.1 mm, +/-0.9 mm, respectively; P<0.0001). The irradiated gels showed a dose-dependent reduction in the SMC proliferation rate (P<0.0001) and an increase in the number of nonviable SMCs (P<0.002) 15 days after irradiation. CONCLUSIONS: Gamma-irradiation produces a significant dose-dependent inhibition of the contraction of collagen gels seeded with arterial SMCs. This effect is related to a significant decrease in SMC viability and a decrease in SMC proliferation rate. These findings shed light on mechanisms whereby irradiation may positively affect arterial remodeling after percutaneous transluminal coronary angioplasties.


Subject(s)
Collagen/radiation effects , Muscle, Smooth, Vascular/cytology , Wound Healing/radiation effects , Animals , Cattle , Cell Survival/radiation effects , Cells, Cultured , DNA/biosynthesis , Dose-Response Relationship, Radiation , Gamma Rays , Gels/radiation effects , Models, Biological , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/radiation effects
5.
Circ Res ; 88(10): 1044-52, 2001 May 25.
Article in English | MEDLINE | ID: mdl-11375274

ABSTRACT

We have previously demonstrated that thrombin upregulation of insulin-like growth factor-1 receptor (IGF-1R) is essential for thrombin-induced mitogenic signaling. To characterize the mechanisms involved, we studied transcription of the IGF-1R gene in rat aortic smooth muscle cells. Thrombin markedly increased IGF-1R mRNA levels, peaking at 3 hours (112+/-7% above control). This effect was mimicked by the hexapeptide SFFLRN (that functions as a tethered ligand) and was blocked by the thrombin inhibitor hirudin. Nuclear run-on assays indicated that thrombin stimulated IGF-1R gene transcription by 2.1-fold, and this was confirmed with the use of actinomycin D. Thrombin-mediated upregulation of IGF-1R mRNA and protein levels was protein kinase C independent but was completely inhibited by the protein tyrosine kinase inhibitor genistein and by the antioxidants N-acetyl-L-cysteine and pyrrolidinedithiocarbamate, suggesting the involvement of reactive oxygen species. The thrombin-induced increase in IGF-1R mRNA was inhibitable by diphenyleneiodonium chloride but not by other inhibitors of cellular oxidase systems, suggesting that NAD(P)H oxidase was necessary for the increase. Furthermore, inhibitors of the epidermal growth factor receptor kinase, Janus kinase-2 kinase, and Src kinase did not block the effect. Thus, thrombin transcriptionally regulates the IGF-1R gene via a redox-sensitive protein tyrosine kinase-dependent pathway that does not require protein kinase C activation. In view of our prior data indicating that IGF-1R density is a critical determinant of vascular smooth muscle cell growth, our findings have particular relevance to understanding mechanisms whereby growth factors such as thrombin regulate vascular proliferation in vivo.


Subject(s)
Gene Expression Regulation/drug effects , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/metabolism , Proto-Oncogene Proteins , Receptor, IGF Type 1/metabolism , Thrombin/metabolism , Animals , Antioxidants/pharmacology , Aorta , Cells, Cultured , Enzyme Inhibitors/pharmacology , Genistein/pharmacology , Hirudins/pharmacology , Janus Kinase 2 , Muscle, Smooth, Vascular/cytology , NADH, NADPH Oxidoreductases/antagonists & inhibitors , NADPH Oxidases , Onium Compounds/pharmacology , Protein Kinase C/antagonists & inhibitors , Protein-Tyrosine Kinases/antagonists & inhibitors , RNA, Messenger/metabolism , Rats , Reactive Oxygen Species/metabolism , Receptor, IGF Type 1/genetics , Receptor, PAR-1 , Receptors, Thrombin/metabolism , Signal Transduction/drug effects , Signal Transduction/physiology , Thrombin/antagonists & inhibitors , Thrombin/pharmacology , Up-Regulation/drug effects , src-Family Kinases/antagonists & inhibitors
6.
Endocrinology ; 142(4): 1489-96, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11250929

ABSTRACT

We previously showed that angiotensin II (ang II) infusion in the rat produces cachexia and decreases circulating insulin-like growth factor I (IGF-I). The weight loss derives from an anorexigenic response and a catabolic effect of ang II. In these experiments we assessed potential catabolic mechanisms and the involvement of the IGF-I system in these responses to ang II. Ang II infusion caused a significant decrease in body weight compared with that of pair-fed control rats. Kidney and left ventricular weights were significantly increased by ang II, whereas fat tissue was unchanged. Skeletal muscle mass was significantly decreased in the ang II-infused rats, and a reduction in lean muscle mass was a major reason for their overall loss of body weight. In skeletal muscles, ang II did not significantly decrease protein synthesis, but overall protein breakdown was accelerated; inhibiting lysosomal and calcium-activated proteases did not reduce the ang II-induced increase in muscle proteolysis. Circulating IGF-I levels were 33% lower in ang II rats vs. control rats, and this difference was reflected in lower IGF-I messenger RNA levels in the liver. Moreover, IGF-I, IGF-binding protein-3, and IGF-binding protein-5 messenger RNAs in the gastrocnemius were significantly reduced. To investigate whether the reduced circulating IGF-I accounts for the loss in muscle mass, we increased circulating IGF-I by coinfusing ang II and IGF-I, but this did not prevent muscle loss. Our data suggest that ang II causes a loss in skeletal muscle mass by enhancing protein degradation probably via its inhibitory effect on the autocrine IGF-I system.


Subject(s)
Angiotensin II/pharmacology , Autocrine Communication/drug effects , Down-Regulation/drug effects , Insulin-Like Growth Factor I/biosynthesis , Muscle Proteins/metabolism , Muscle, Skeletal/pathology , Wasting Syndrome/chemically induced , Animals , Blotting, Northern , Blotting, Western , Body Weight/drug effects , Insulin-Like Growth Factor I/genetics , Insulin-Like Growth Factor I/pharmacology , Liver/drug effects , Liver/metabolism , Male , Muscle Proteins/biosynthesis , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Nuclease Protection Assays , Organ Size/drug effects , RNA, Messenger/biosynthesis , Radioimmunoassay , Rats , Rats, Sprague-Dawley , Wasting Syndrome/pathology
7.
J Biol Chem ; 276(8): 5668-75, 2001 Feb 23.
Article in English | MEDLINE | ID: mdl-11063741

ABSTRACT

The insulin-like growth factor I receptor (IGF-IR) is a heterotetrameric receptor mediating the effects of insulin-like growth I and other growth factors. This receptor is encoded by an mRNA containing an unusually long, G-C-rich, and highly structured 5' untranslated region. Using bicistronic constructs, we demonstrated here that the 5' untranslated region of the IGF-IR allows translation initiation by internal ribosome entry and therefore constitutes an internal ribosome entry site. In vitro cross-linking revealed that this internal ribosome entry site binds a protein of 57 kDa. Immunoprecipitation of UV cross-linked proteins proved that this protein was the polypyrimidine tract-binding protein, a well known regulator of picornavirus mRNA translation. The efficiency of translation of the endogenous IGF-IR mRNA is not affected by rapamycin, which is a potent inhibitor of cap-dependent translation. This result provides evidence that the endogenous IGF-IR mRNA is translated, at least in part, through a cap-independent mechanism. This is the first report of a growth factor receptor containing sequence elements that allow translation initiation to occur by internal initiation. Because the IGF-IR has a pivotal function in the cell cycle, this mechanism of translation regulation could play a crucial role in the control of cell proliferation and differentiation.


Subject(s)
Peptide Chain Initiation, Translational , RNA, Messenger/genetics , Receptor, IGF Type 1/biosynthesis , Ribosomes/metabolism , 5' Untranslated Regions , Animals , Base Sequence , Binding Sites , Cell Differentiation , Cell Division , Cells, Cultured , Humans , Molecular Sequence Data , Muscle, Smooth, Vascular/cytology , Nucleic Acid Conformation , Polypyrimidine Tract-Binding Protein , Protein Binding , Protein Biosynthesis/drug effects , RNA, Messenger/metabolism , RNA-Binding Proteins/metabolism , Rats , Receptor, IGF Type 1/genetics , Ribonucleoproteins/metabolism , Sirolimus/pharmacology
8.
Circulation ; 102(22): 2680-6, 2000 Nov 28.
Article in English | MEDLINE | ID: mdl-11094032

ABSTRACT

BACKGROUND: Cytotoxic oxidized LDL (oxLDL) has been shown to promote apoptosis in cultured vascular smooth muscle cells (VSMCs). We investigated the localization of oxLDL and its association with apoptosis and the expression of apoptosis-related proteins in early and advanced atherosclerotic lesions. METHODS AND RESULTS: Atherosclerotic plaques (n=23) from patients undergoing aortic, carotid, or femoral arterial surgery were studied. In early lesions, oxLDL was located predominantly in the superficial intima and in the media just beneath the internal elastic lamina. Medial VSMCs staining positive for oxLDL showed expression of BAX, a proapoptotic protein of the BCL-2 family. Apoptosis, as detected by DNA in situ terminal deoxynucleotidyl transferase end-labeling (TUNEL), was not present in these early lesions. In advanced plaques, areas of the intima positive for oxLDL showed lower alpha-smooth muscle actin immunoreactivity (P<0.01) and higher BAX immunoreactivity (P<0.05). Furthermore, these areas showed an increased number of apoptotic VSMCs (P<0.01). Western blot analysis revealed that oxLDL increases BAX expression in cultured human coronary VSMCs. CONCLUSIONS: We conclude that in early atherosclerotic lesions, oxLDL-positive VSMCs express BAX, which increases the susceptibility of these cells to undergo apoptosis. This could be important in our understanding of the transition of early lesions into advanced atherosclerotic plaques, which are characterized by regions of cell death. In advanced plaques, oxLDL-positive areas of the intima show higher BAX immunoreactivity and TUNEL-positive VSMCs, and this may contribute to plaque instability and rupture.


Subject(s)
Apoptosis , Arteriosclerosis/metabolism , Lipoproteins, LDL/analysis , Muscle, Smooth, Vascular/chemistry , Proto-Oncogene Proteins c-bcl-2 , Actins/analysis , Aged , Arteriosclerosis/pathology , Blotting, Western , Cell Line , Female , Humans , Immunohistochemistry , In Situ Nick-End Labeling , Male , Muscle, Smooth, Vascular/cytology , Proto-Oncogene Proteins/analysis , bcl-2-Associated X Protein
9.
J Biol Chem ; 275(49): 38921-8, 2000 Dec 08.
Article in English | MEDLINE | ID: mdl-10982795

ABSTRACT

Insulin-like growth factor (IGF-1) is a potent mitogen for vascular smooth muscle cells. Both IGF-1 and its receptor have been shown to be highly expressed in atherosclerotic lesions. Here we investigated whether part of the vasculoprotective properties of E(2) may be mediated by its negative regulation of the IGF-1 system. HeLa cells, which do not contain endogenous estrogen receptors (ER), were transiently transfected with IGF-1R promoter constructs with or without a plasmid encoding human ERalpha or ERbeta and treated with 100 nm 17beta-estradiol (E(2)) for 24 h. E(2) treatment decreased basal luciferase activity by 51%, and this effect was dependent on co-expression of ERalpha, whereas no repression was observed with ERbeta. A mutation within the DNA binding domain of the ERalpha abolished the repressor function of the ER receptor. Similarly, E(2) decreased IGF-1R transcription by 21% in rat aortic smooth muscle cells (RASMC), which express endogenous ER. This effect was specific for E(2), because it was inhibited by an antiestrogen and because progesterone did not have any effect on IGF-1R expression in HeLa or RASMC transfected with progesterone receptor. Accordingly, E(2) decreased IGF-1R and IGF-1 mRNA in RASMC by 47% and 33%. Western blot analysis and radioligand binding studies showed that E(2) also dose-dependently decreased IGF-1R protein expression in RASMC by 40% and 30%, respectively, and that IGF-1 protein was reduced by 43%. Repression of IGF-1R promoter activity by a combination of ERalpha and E(2) did not appear to be mediated via direct binding of ER to the IGF-1R promoter but rather by inhibition of SP1 binding to the IGF-1R promoter. Thus, E(2) down-regulates IGF-1R and IGF-1 expression in vascular smooth muscle cells. This may have important implications for the understanding of the beneficial effects of estrogen in the cardiovascular system.


Subject(s)
Estradiol/pharmacology , Estrogen Antagonists/pharmacology , Gene Expression Regulation/drug effects , Muscle, Smooth, Vascular/metabolism , Receptor, IGF Type 1/genetics , Receptor, IGF Type 2/genetics , Receptors, Estrogen/physiology , Animals , Aorta , Cells, Cultured , Estradiol/analogs & derivatives , Estrogen Receptor alpha , Estrogen Receptor beta , Gene Expression Regulation/physiology , HeLa Cells , Humans , Polyunsaturated Alkamides , Promoter Regions, Genetic/drug effects , Rats , Receptors, Estrogen/genetics , Recombinant Proteins/metabolism , Tamoxifen/analogs & derivatives , Tamoxifen/pharmacology , Transfection
10.
Coron Artery Dis ; 11(6): 495-502, 2000 Sep.
Article in English | MEDLINE | ID: mdl-10966136

ABSTRACT

BACKGROUND: Wall shear stress (WSS) is closely associated with arteriosclerosis. WSS values for various vessels and species are available, but fully in-vivo measurements in human coronary arteries have not yet been reported. OBJECTIVE: To measure WSS in undiseased coronary arteries of adult patients at rest. METHODS: We recorded the temporal average value (APV) of the instantaneous maximal blood velocity in the three vessel segments of angiographically normal coronary artery bifurcations in 21 patients undergoing cardiac catheterization to treat various diseases by means of a 0.036 cm Doppler wire (FloWire). In total, 36 bifurcations were examined. The 36 x 3 cross-sectional areas (CSA) were determined by means of a three-dimensional angiographic technique. The three flows, Q1 (inflow), Q2, and Q3 of each bifurcation were calculated according to Q=0.5 x APV x CSA. For each segment, WSS was calculated as WSS=32 eta Q/(pi D3) (where blood viscosity eta=3.5 mPa s and D is vessel diameter). Only the 54 WSS values obtained from the 18 flow triplets which satisfied the equation Q1/(Q2+Q3)=1 better than did the 18 other ones were retained. RESULTS: The 54 WSS values ranged from 0.33 to 1.24 Pa (mean 0.68 Pa, SEM, 0.027 Pa). They did not depend significantly on Q (r=0.07; P=0.60) and the CSA (r=0.24, P=0.08) but the second relationship approached significance. CONCLUSION: The obtained mean WSS value (0.68 Pa) is half the value predicted for coronary arteries from optimality principles. It is also smaller than many values reported for human carotid, renal, and femoral arteries.


Subject(s)
Coronary Vessels/physiology , Hemorheology , Aged , Blood Flow Velocity , Cardiac Catheterization , Coronary Angiography , Echocardiography, Doppler , Female , Humans , Male , Middle Aged , Predictive Value of Tests , Ultrasonography, Doppler , Ultrasonography, Interventional
11.
J Biol Chem ; 275(35): 26864-9, 2000 Sep 01.
Article in English | MEDLINE | ID: mdl-10862615

ABSTRACT

Low density lipoproteins (LDLs) play an important role in the pathogenesis of atherosclerosis. LDL has been shown to be mitogenic and proapoptotic for vascular smooth muscle cells. However, the mechanisms are poorly understood and may result from an alteration in intracellular mitogenic signaling either directly by LDL or indirectly through an autocrine effect involving growth factor secretion and/or growth factor receptor expression. Insulin-like growth factor-1 (IGF-1) is an autocrine/paracrine factor for vascular smooth muscle cells and has potent anti-apoptotic effects. Thus, we hypothesized that part of the proliferative responses to LDLs may be explained by its modulation of IGF-1 or IGF-1 receptor (IGF-1R) expression. Treatment of rat vascular smooth muscle cells with increasing doses of native LDL dose-dependently increased IGF-1 mRNA by up to 2.6-fold; however, native LDL had no effect on IGF-1R mRNA expression. In contrast, the same doses of oxidized LDL significantly reduced IGF-1 and IGF-1R mRNA by 80 and 61%, respectively, and reduced IGF-1 and IGF-1R protein expression by 63 and 46%. In addition, native and oxidized LDL significantly increased IGF-1-binding protein-2 and IGF-1-binding protein-4 expression as measured by Western ligand blot. Most interestingly, anti-IGF-1 antiserum completely inhibited LDL-induced but not serum-induced increase in (3)H-thymidine incorporation, indicating a requirement for IGF-1 in the LDL-stimulated mitogenic signaling pathway. In summary, these results suggest that native and oxidized LDLs have differential effects on IGF-1 and IGF-1R expression. Because IGF-1 is a potent survival factor for vascular smooth muscle cells, our findings suggest that moderately oxidized LDL may favor proliferation of smooth muscle cells, whereas oxidized LDL may contribute to plaque apoptosis by local depletion of IGF-1 and IGF-1R.


Subject(s)
Insulin-Like Growth Factor I/metabolism , Lipoproteins, LDL/physiology , Muscle, Smooth, Vascular/metabolism , Receptors, Somatomedin/metabolism , Animals , Cells, Cultured , DNA Replication/physiology , Gene Expression Regulation/physiology , Humans , Insulin-Like Growth Factor I/genetics , Muscle, Smooth, Vascular/cytology , RNA, Messenger/genetics , Rats , Receptors, Somatomedin/genetics
12.
Ann Endocrinol (Paris) ; 61(1): 22-6, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10790588

ABSTRACT

Experimental data suggests that growth hormone and IGF-1 have beneficial effects on myocardial function in animal models of heart failure. Preliminary evidence suggests an abnormality in the growth hormone-IGF-1 axis in heart failure with relative growth hormone resistance. Beneficial effects of growth hormone and IGF-1 include vasodilatation, stimulation of cardiac hypertrophy, increase in calcium sensitivity of cardiac myofilaments and prevention of apoptosis. Recently, cardiac cachexia has been shown to be a powerful negative predictive factor in heart failure. Cachectic patients have higher angiotensin II levels. In the rat there is an important interaction between the renin-angiotensin system and IGF-1. Thus, angiotensin II infusion causes weight loss in part through a catabolic effect. This effect results from increased protein degradation. Angiotensin II reduces circulating and skeletal muscle IGF-1 but increases IGF-1 and the IGF-IR expression in cardiac muscle. Preliminary data suggest a potential beneficial effect of growth hormone in heart failure. Further trials are necessary to test the potential beneficial effect of growth hormone and/or IGF-1 in heart failure.


Subject(s)
Heart Diseases , Human Growth Hormone/physiology , Insulin-Like Growth Factor I/physiology , Animals , Cachexia/complications , Cachexia/drug therapy , Heart Diseases/complications , Heart Diseases/drug therapy , Heart Diseases/etiology , Heart Diseases/physiopathology , Human Growth Hormone/pharmacology , Human Growth Hormone/therapeutic use , Humans , Insulin-Like Growth Factor I/pharmacology , Insulin-Like Growth Factor I/therapeutic use
14.
Arterioscler Thromb Vasc Biol ; 20(2): 370-6, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10669632

ABSTRACT

Insulin-like growth factor-I (IGF-I) is a ubiquitous peptide that regulates cellular growth and differentiation and is involved in vascular proliferative responses. The effects of IGF-I are modulated by several IGF-I binding proteins (IGFBPs), including IGFBP-4, the main IGFBP produced by vascular smooth muscle cells (VSMCs). We have previously shown that angiotensin II (Ang II)-induced and thrombin-induced mitogenesis in VSMCs is dependent on autocrine IGF-I. In addition, we have demonstrated that IGF-I and IGFBP-4 mRNA levels are upregulated in the hypertensive aorta of abdominally coarcted rats, a high-renin hypertension model. To obtain further insight into the IGF-I system and to specifically study changes in IGFBP-4, a known inhibitor of IGF-I action, VSMCs were incubated with Ang II or thrombin. Compared with control, Ang II induced an 87+/-2% downregulation of IGFBP-4 mRNA levels at 24 hours, with a 61+/-6% decrease of IGFBP-4 levels, as determined by Western ligand blot analysis. Thrombin had the same depressor effects (87+/-2% for the mRNA levels and 61+/-3% for the protein levels). Ang II and thrombin coincubation with (125)I-IGFBP-4 in the conditioned media failed to reveal any increase in fragmentation, indicating that proteolytic cleavage of IGFBP-4 was not involved in the observed effects. Exogenous recombinant human IGFBP-4 decreased thrombin-induced DNA synthesis of human aortic VSMCs by 64%, whereas anti-IGFBP-4 antibody potentiated thrombin-induced DNA synthesis. These data suggest that downregulation of IGFBP-4 expression in VSMCs may play a critical role in vascular growth response to Ang II and thrombin in normal and diseased states, by increasing the bioavailability of IGF-I for its cell-surface receptor.


Subject(s)
Angiotensin II/pharmacology , Aorta/metabolism , Insulin-Like Growth Factor Binding Protein 4/metabolism , Muscle, Smooth, Vascular/metabolism , Thrombin/pharmacology , Animals , Aorta/drug effects , Cells, Cultured , Culture Media, Conditioned , Dactinomycin/pharmacology , Endopeptidases/metabolism , Humans , Insulin-Like Growth Factor Binding Protein 4/genetics , Insulin-Like Growth Factor Binding Protein 4/pharmacology , Insulin-Like Growth Factor Binding Proteins/metabolism , Muscle, Smooth, Vascular/drug effects , Protein Synthesis Inhibitors/pharmacology , RNA, Messenger/metabolism , Rats , Recombinant Proteins/pharmacology , Thymidine/metabolism
15.
Hypertension ; 34(5): 1053-9, 1999 Nov.
Article in English | MEDLINE | ID: mdl-10567181

ABSTRACT

Angiotensin II (Ang II) is known to act as a growth factor and may be involved in cardiac remodeling. We have shown that insulin-like growth factor-I (IGF-I) is an autocrine mediator of growth responses to Ang II in vascular smooth muscle cells in vitro, and we hypothesized that IGF-I also serves as an important modulator of cardiovascular growth in vivo. To study the effect of Ang II on cardiac IGF-I, we infused rats for 3, 7, or 14 days with Ang II through osmotic minipumps. After 7 days, left ventricular mass normalized for body weight was increased by 20% (P<0.01) in Ang II rats compared with pair-fed control rats that were given a restricted amount of food identical to that eaten by the anorexic, Ang II-infused rats. Ang II increased left ventricular IGF-I mRNA levels by 1.5- to 1.8-fold compared with ad libitum-fed or pair-fed control rats (P<0.05). Cardiac IGF-I protein was increased correspondingly and was localized on the cardiomyocytes. Treatment with hydralazine abolished the induction of IGF-I mRNA, which indicates that Ang II induces cardiac IGF-I mRNA expression through a pressor-mediated mechanism. IGF-I receptor (IGF-IR) mRNA was induced 2.1-fold in Ang II rats compared with ad libitum-fed rats (P<0.01). However, this increase was also observed in pair-fed controls and is thus due to the anorexigenic effect of Ang II. We have recently shown that circulating IGF-I levels are reduced in response to Ang II infusion. Elevation of IGF-I levels by coinfusion of IGF-I and Ang II significantly increased left ventricular index by 16% compared with rats infused with Ang II alone (P<0.05). In conclusion, autocrine upregulation of cardiac IGF-I and IGF-IR mRNA by Ang II occurs through hemodynamic and nonhemodynamic mechanisms, respectively, and may modulate cardiac structural changes that occur in hypertension.


Subject(s)
Angiotensin II/pharmacology , Blood Pressure/drug effects , Eating/drug effects , Hypertrophy, Left Ventricular/etiology , Insulin-Like Growth Factor I/genetics , Myocardium/metabolism , Receptor, IGF Type 1/genetics , Animals , Antihypertensive Agents/pharmacology , Insulin-Like Growth Factor I/biosynthesis , Insulin-Like Growth Factor I/pharmacology , Male , RNA, Messenger/analysis , Rats , Rats, Sprague-Dawley
16.
Arterioscler Thromb Vasc Biol ; 19(9): 2119-26, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10479653

ABSTRACT

We have recently shown that angiotensin II activation of insulin-like growth factor 1 receptor (IGF-1R) transcription is a critical requirement for angiotensin-stimulated vascular smooth muscle cell growth; therefore, we examined the signaling pathway involved. In rat aortic smooth muscle cells, the antioxidants N-acetyl-L-cysteine (5 mmol/L) and pyrrolidine dithiocarbamate (100 micromol/L) completely inhibited angiotensin II-stimulated increases in IGF-1R mRNA and protein levels, suggesting the involvement of reactive oxygen species. Indeed, catalase abolished the Ang II-stimulated increase of IGF-1R protein expression, and accordingly, H(2)O(2) (0.2 mmol/L) or the oxidized products of linoleic acid, hydroperoxyoctadecadienoic acids (10 micromol/L), increased IGF-1R mRNA levels at 3 hours by 74+/-20% and 107+/-22% and increased receptor number at 24 hours by 51+/-6.7% and 55+/-7.4%, respectively. The protein tyrosine kinase inhibitors genistein and tyrphostin A25 also blocked angiotensin II increases in IGF-1R mRNA and protein levels and blocked the ability of hydroperoxyoctadecadienoic acids and H(2)O(2) to increase IGF-1R expression, suggesting that oxidative stress may be an early event in the angiotensin II signaling cascade. Furthermore, calcium chelation inhibited the angiotensin II effect. Transient transfection assays revealed that a (-2350)+640 IGF-1R promoter/luciferase construct was fully responsive to angiotensin II stimulation (127+/-20% increase). Ten millimoles per liter hydroperoxyoctadecadienoic acids and 0.2 mmol/L H(2)O(2) increased luciferase activity by 79+/-8.5% and 63+/-12%, respectively, and 5 mmol/L N-acetyl-L-cysteine blocked the angiotensin II-induced upregulation of luciferase activity by 70%. These data suggest that angiotensin II stimulates IGF-1R gene transcription via calcium-dependent activation of protein tyrosine kinase activity that lies downstream from an oxidant stimulus. These findings provide key insights into the signaling mechanisms whereby angiotensin II exerts its growth-promoting effects on the vasculature.


Subject(s)
Angiotensin II/pharmacology , Protein-Tyrosine Kinases/metabolism , Receptor, IGF Type 1/genetics , Acetylcysteine/pharmacology , Animals , Antioxidants/pharmacology , Calcium/physiology , Cells, Cultured , Genes, Reporter/drug effects , Hydrogen Peroxide/pharmacology , Linoleic Acids/pharmacology , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/physiology , Oxidation-Reduction , Protein-Tyrosine Kinases/physiology , Pyrrolidines/pharmacology , Rats , Thiocarbamates/pharmacology , Transcription, Genetic/drug effects , Transcription, Genetic/physiology
18.
J Biol Chem ; 274(6): 3522-30, 1999 Feb 05.
Article in English | MEDLINE | ID: mdl-9920898

ABSTRACT

Angiotensin II (Ang II) and basic fibroblast growth factor (bFGF) are important modulators of cell growth under physiological and pathophysiological conditions. We and others have previously shown that these growth factors increase insulin-like growth factor-1 receptor (IGF-1R) number and mRNA in vascular smooth muscle cells and that this effect is transcriptionally regulated. To study the mechanisms and the signaling pathways involved, IGF-1R promoter reporter constructs were transiently transfected in CHO-AT1 cells that overexpress angiotensin AT1 receptors. Our findings indicate that Ang II and bFGF significantly increased IGF-1R promoter activity up to 7- and 3-fold, respectively. The effect induced by Ang II was mediated via a tyrosine kinase-dependent mechanism, since tyrphostin A25 largely inhibited the Ang II-induced increase in promoter activity. In addition, co-transfection of dominant negative Ras, Raf, and MEK1 or pretreatment with the MEK inhibitor PD 98059 dose-dependently decreased both the Ang II- and bFGF-induced increase in IGF-1R transcription and protein expression, suggesting that the Ras-Raf-mitogen-activated protein kinase kinase pathway is required for both growth factors. Reactive oxygen species have been shown to act as second messengers in Ang II-induced signaling, and activation of the transcription factor NF-kappaB is redox-sensitive. While co-transfection of dominant negative IkappaBalpha mutant completely inhibited the Ang II-induced increase in transcription, it had no effect on the bFGF signaling. In contrast, co-transfection studies indicated that the transcription factors STAT1, STAT3, and c-Jun and the Janus kinase 2 kinase are required in the signaling pathway of bFGF, whereas only dominant c-Jun inhibited the Ang II-induced effect. In summary, these data demonstrate that Ang II and bFGF increase IGF-1R gene transcription via distinct as well as shared pathways and have important implications for understanding growth-stimulatory effects of these growth factors on vascular cells.


Subject(s)
Angiotensin II/physiology , Fibroblast Growth Factor 2/physiology , Gene Expression Regulation/physiology , Receptor, IGF Type 1/genetics , Signal Transduction , Angiotensin II/metabolism , Animals , CHO Cells , Cricetinae , Fibroblast Growth Factor 2/metabolism , Promoter Regions, Genetic , Receptor, IGF Type 1/metabolism , Transcription, Genetic
19.
Eur Heart J ; 19 Suppl G: G18-22, 1998 Jul.
Article in English | MEDLINE | ID: mdl-9717051

ABSTRACT

The accumulation of vascular smooth muscle cells plays an important role in the development of atherosclerotic plaques, and in the restenotic process occurring after balloon angioplasty. Accumulation results from the synergistic effects of various growth factors and cytokines, acting to induce both proliferation and migration. Additionally, a variety of matrix proteins and extracellular proteases modulate the growth responses and phenotype of vascular smooth muscle cells. One of the important effects of growth factors such as platelet-derived growth factors, fibroblast growth factor, angiotensin II, and thrombin is to up-regulate insulin-like growth factor (IGF)-1 receptors on vascular smooth muscle cells. Furthermore, agonists that bind to G-protein-coupled receptors, such as angiotensin II and thrombin, interact with the IGF-1 receptor signalling pathway. Thus, growth factor-stimulated up-regulation of IGF-1 receptors is critical in mediating the proliferative responses to multiple growth factors.


Subject(s)
Arteriosclerosis/physiopathology , Cell Division/physiology , Growth Substances/physiology , Muscle, Smooth, Vascular/physiopathology , Angioplasty, Balloon , Animals , Arteriosclerosis/therapy , Cytokines/physiology , Humans , Recurrence
20.
Circulation ; 96(3): 927-33, 1997 Aug 05.
Article in English | MEDLINE | ID: mdl-9264503

ABSTRACT

BACKGROUND: Estrogen protects against cardiovascular disease in both patients and animal models and regulates insulin-like growth factor-I (IGF-I), an important cell-cycle progression factor. METHODS AND RESULTS: Smooth muscle cells and tissues were harvested from male recipient rabbits that 6 weeks earlier had received a cardiac allograft transplant consisting of a donor heart and ascending aorta. Segments of the ascending aorta from the native and allograft hearts from 9 placebo-treated and 8 estradiol-treated recipients were compared by using IGF-I-stimulated [3H]thymidine incorporation. The responses of the native vessel segments were similar (175.3+/-32% and 166.9+/-41%, respectively; P>.05) whether or not the recipients had been treated for 6 weeks with estradiol. In the grafts, however, estradiol markedly inhibited vascular cell thymidine incorporation (328.04+/-56% compared with 67.3+/-11%; P<.02). Smooth muscle cells were derived from the native aorta of the placebo-treated rabbits to study the effect of estradiol in vitro. IGF-I increased cell counts in a concentration-dependent manner. In serum-starved cells estradiol further decreased cell proliferation; this effect was blocked by the specific estrogen receptor antagonist ZK-119.010. Immunohistochemistry staining for IGF-I protein in the coronary arteries and ascending aorta of the cardiac allograft from the placebo-treated recipients revealed extensive IGF-I expression in the myointima. In contrast, IGF-I protein was not expressed in the coronary arteries and ascending aorta of the cardiac allograft from the estradiol-treated recipients. The IGF-I protein was extensively expressed only in the placebo-treated graft vessels. Myointimal thickening of the coronary arteries was significantly reduced by estradiol treatment (17.9+/-1.5% versus 44.3+/-3.7%; P<.02). CONCLUSIONS: In vivo estradiol treatment abolishes both IGF-I mitogenic effects and IGF-I protein expression in the vascular wall, which may be causally related to the inhibitory effect of estradiol on transplant arteriosclerosis.


Subject(s)
Coronary Vessels/metabolism , Estradiol/pharmacology , Heart Transplantation , Insulin-Like Growth Factor I/metabolism , Insulin-Like Growth Factor I/pharmacology , Animals , Aorta/cytology , Aorta/drug effects , Arteries , Cell Division/drug effects , Coronary Vessels/cytology , Coronary Vessels/drug effects , Hyperplasia , Male , Mitogens/pharmacology , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/drug effects , Myocardium/metabolism , Rabbits , Transplantation, Homologous , Tunica Intima/metabolism , Tunica Intima/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...