Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Am J Physiol Heart Circ Physiol ; 319(4): H730-H743, 2020 10 01.
Article in English | MEDLINE | ID: mdl-32795184

ABSTRACT

Insulin-like growth factor-1 (IGF-1) decreases atherosclerosis in apolipoprotein E (Apoe)-deficient mice when administered systemically. However, mechanisms for its atheroprotective effect are not fully understood. We generated endothelium-specific IGF-1 receptor (IGF1R)-deficient mice on an Apoe-deficient background to assess effects of IGF-1 on the endothelium in the context of hyperlipidemia-induced atherosclerosis. Endothelial deficiency of IGF1R promoted atherosclerotic burden, when animals were fed on a high-fat diet for 12 wk or normal chow for 12 mo. Under the normal chow feeding condition, the vascular relaxation response to acetylcholine was increased in the endothelial IGF1R-deficient aorta; however, feeding of a high-fat diet substantially attenuated the relaxation response, and there was no difference between endothelial IGF1R-deficient and control mice. The endothelium and its intercellular junctions provide a barrier function to the vasculature. In human aortic endothelial cells, IGF-1 upregulated occludin, claudin 5, VE-cadherin, JAM-A, and CD31 expression levels, and vice versa, specific IGF1R inhibitor, picropodophyllin, an IGF1R-neutralizing antibody (αIR3), or siRNA to IGF1R abolished the IGF-1 effects on junction and adherens proteins, suggesting that IGF-1 promoted endothelial barrier function. Moreover, endothelial transwell permeability assays indicated that inhibition of IGF-1 signaling elevated solute permeability through the monolayer of human aortic endothelial cells. In summary, endothelial IGF1R deficiency increases atherosclerosis, and IGF-1 positively regulates tight junction protein and adherens junction protein levels and endothelial barrier function. Our findings suggest that the elevation of the endothelial junction protein level is, at least in part, the mechanism for antiatherogenic effects of IGF-1.NEW & NOTEWORTHY Endothelial insulin-like growth factor-1 (IGF-1) receptor deficiency significantly elevated atherosclerotic burden in apolipoprotein E-deficient mice, mediated at least in part by downregulation of intercellular junction proteins and, thus, elevated endothelial permeability. This study revealed a novel role for IGF-1 in supporting endothelial barrier function. These findings suggest that IGF-1's ability to promote endothelial barrier function may offer a novel therapeutic strategy for vascular diseases such as atherosclerosis.


Subject(s)
Aortic Diseases/metabolism , Atherosclerosis/metabolism , Capillary Permeability , Endothelial Cells/metabolism , Receptor, IGF Type 1/deficiency , Animals , Antigens, CD/metabolism , Aortic Diseases/genetics , Aortic Diseases/pathology , Atherosclerosis/genetics , Atherosclerosis/pathology , Cadherins/metabolism , Disease Models, Animal , Disease Progression , Endothelial Cells/pathology , Humans , Mice, Inbred C57BL , Mice, Knockout, ApoE , Plaque, Atherosclerotic , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Receptor, IGF Type 1/genetics , Receptor, IGF Type 1/metabolism , THP-1 Cells , Tight Junction Proteins/metabolism , Tight Junctions/metabolism
2.
Arterioscler Thromb Vasc Biol ; 38(10): 2306-2317, 2018 10.
Article in English | MEDLINE | ID: mdl-30354209

ABSTRACT

Objective- IGF-1 (insulin-like growth factor 1) is a major autocrine/paracrine growth factor, which promotes cell proliferation, migration, and survival. We have shown previously that IGF-1 reduced atherosclerosis and promoted features of stable atherosclerotic plaque in Apoe-/- mice-an animal model of atherosclerosis. The aim of this study was to assess effects of smooth muscle cell (SMC) IGF-1 signaling on the atherosclerotic plaque. Approach and Results- We generated Apoe-/- mice with IGF1R (IGF-1 receptor) deficiency in SMC and fibroblasts (SM22α [smooth muscle protein 22 α]-CreKI/IGF1R-flox mice). IGF1R was decreased in the aorta and adventitia of SM22α-CreKI/IGF1R-flox mice and also in aortic SMC, embryonic, skin, and lung fibroblasts isolated from SM22α-CreKI/IGF1R-flox mice. IGF1R deficiency downregulated collagen mRNA-binding protein LARP6 (La ribonucleoprotein domain family, member 6) and vascular collagen, and mice exhibited growth retardation. The high-fat diet-fed SM22α-CreKI/IGF1R-flox mice had increased atherosclerotic burden and inflammatory responses. α-SMA (α-smooth muscle actin)-positive plaque cells had reduced proliferation and elevated apoptosis. SMC/fibroblast-targeted decline in IGF-1 signaling decreased atherosclerotic plaque SMC, markedly depleted collagen, reduced plaque fibrous cap, and increased plaque necrotic cores. Aortic SMC isolated from SM22α-CreKI/IGF1R-flox mice had decreased cell proliferation, migration, increased sensitivity to apoptosis, and these effects were associated with disruption of IGF-1-induced Akt signaling. Conclusions- IGF-1 signaling in SMC and in fibroblast is a critical determinant of normal vascular wall development and atheroprotection.


Subject(s)
Aortic Diseases/metabolism , Atherosclerosis/metabolism , Microfilament Proteins/genetics , Muscle Proteins/genetics , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Plaque, Atherosclerotic , Promoter Regions, Genetic , Receptor, IGF Type 1/deficiency , Actins/metabolism , Animals , Aorta/metabolism , Aorta/pathology , Aortic Diseases/genetics , Aortic Diseases/pathology , Apoptosis , Atherosclerosis/genetics , Atherosclerosis/pathology , Autoantigens/metabolism , Cell Movement , Cell Proliferation , Cells, Cultured , Collagen/metabolism , Disease Models, Animal , Female , Fibroblasts/metabolism , Fibrosis , Male , Mice, Inbred C57BL , Mice, Knockout, ApoE , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/pathology , Proto-Oncogene Proteins c-akt/metabolism , Receptor, IGF Type 1/genetics , Ribonucleoproteins/metabolism , Signal Transduction , SS-B Antigen
3.
Circulation ; 133(23): 2263-78, 2016 Jun 07.
Article in English | MEDLINE | ID: mdl-27154724

ABSTRACT

BACKGROUND: We have previously shown that systemic infusion of insulin-like growth factor-1 (IGF-1) exerts anti-inflammatory and antioxidant effects and reduces atherosclerotic burden in apolipoprotein E (Apoe)-deficient mice. Monocytes/macrophages express high levels of IGF-1 receptor (IGF1R) and play a pivotal role in atherogenesis, but the potential effects of IGF-1 on their function are unknown. METHODS AND RESULTS: To determine mechanisms whereby IGF-1 reduces atherosclerosis and to explore the potential involvement of monocytes/macrophages, we created monocyte/macrophage-specific IGF1R knockout (MΦ-IGF1R-KO) mice on an Apoe(-/-) background. We assessed atherosclerotic burden, plaque features of stability, and monocyte recruitment to atherosclerotic lesions. Phenotypic changes of IGF1R-deficient macrophages were investigated in culture. MΦ-IGF1R-KO significantly increased atherosclerotic lesion formation, as assessed by Oil Red O staining of en face aortas and aortic root cross-sections, and changed plaque composition to a less stable phenotype, characterized by increased macrophage and decreased α-smooth muscle actin-positive cell population, fibrous cap thinning, and decreased collagen content. Brachiocephalic artery lesions of MΦ-IGF1R-KO mice had histological features implying plaque vulnerability. Macrophages isolated from MΦ-IGF1R-KO mice showed enhanced proinflammatory responses on stimulation by interferon-γ and oxidized low-density lipoprotein and elevated antioxidant gene expression levels. Moreover, IGF1R-deficient macrophages had decreased expression of ABCA1 and ABCG1 and reduced lipid efflux. CONCLUSIONS: Our data indicate that macrophage IGF1R signaling suppresses macrophage and foam cell accumulation in lesions and reduces plaque vulnerability, providing a novel mechanism whereby IGF-1 exerts antiatherogenic effects.


Subject(s)
Aorta/metabolism , Aortic Diseases/metabolism , Apolipoproteins E/deficiency , Atherosclerosis/metabolism , Macrophages/metabolism , Plaque, Atherosclerotic , Receptor, IGF Type 1/deficiency , ATP Binding Cassette Transporter 1/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 1/metabolism , Animals , Aorta/drug effects , Aorta/pathology , Aortic Diseases/genetics , Aortic Diseases/pathology , Aortic Diseases/prevention & control , Apolipoproteins E/genetics , Atherosclerosis/genetics , Atherosclerosis/pathology , Atherosclerosis/prevention & control , Cell Plasticity , Cells, Cultured , Disease Models, Animal , Foam Cells/metabolism , Foam Cells/pathology , Genetic Predisposition to Disease , Inflammation Mediators/metabolism , Inflammation Mediators/pharmacology , Interferon-gamma/pharmacology , Lipoproteins, LDL/pharmacology , Macrophages/drug effects , Macrophages/pathology , Mice, Knockout , Phenotype , Receptor, IGF Type 1/genetics , Rupture, Spontaneous
4.
Atherosclerosis ; 238(2): 313-20, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25549319

ABSTRACT

OBJECTIVE: We have shown that insulin-like growth factor I (IGF-1) infusion in Apoe(-/-) mice decreased atherosclerotic plaque size and plaque macrophage and lipid content suggesting that IGF-1 suppressed formation of macrophage-derived foam cells. Since 12/15-lipoxygenase (12/15-LOX) plays an important role in OxLDL and foam cell formation, we hypothesized that IGF-1 downregulates 12/15-LOX, thereby suppressing lipid oxidation and foam cell formation. APPROACH AND RESULTS: We found that IGF-1 decreased 12/15-LOX plaque immunopositivity and serum OxLDL levels in Apoe(-/-) mice. IGF-1 reduced 12/15-LOX protein and mRNA levels in cultured THP-1 macrophages and IGF-1 also decreased expression of STAT6 transcription factor. IGF-1 reduction in macrophage 12/15-LOX was mediated in part via a PI3 kinase- and STAT6-dependent transcriptional mechanism. IGF-1 suppressed THP-1 macrophage ability to oxidize lipids and form foam cells. IGF-1 downregulated 12/15-LOX in human blood-derived primary macrophages and IGF-1 decreased LDL oxidation induced by these cells. IGF-1 reduced LDL oxidation and formation of foam cells by wild type murine peritoneal macrophages, however these effects were completely blocked in 12/15-LOX-null macrophages suggesting that the ability of IGF-1 to reduce LDL oxidation and foam cells formation is dependent on its ability to downregulate 12/15-LOX. CONCLUSIONS: Overall our data demonstrate that IGF-1 reduces lipid oxidation and foam cell formation via downregulation of 12/15-LOX and this mechanism may play a major role in the anti-atherosclerotic effects of IGF-1.


Subject(s)
Arachidonate 12-Lipoxygenase/metabolism , Arachidonate 15-Lipoxygenase/metabolism , Atherosclerosis/prevention & control , Foam Cells/drug effects , Insulin-Like Growth Factor I/administration & dosage , Lipoproteins, LDL/metabolism , Macrophages, Peritoneal/drug effects , Animals , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Arachidonate 12-Lipoxygenase/genetics , Arachidonate 15-Lipoxygenase/genetics , Atherosclerosis/blood , Atherosclerosis/enzymology , Atherosclerosis/genetics , Atherosclerosis/pathology , Cell Line , Disease Models, Animal , Dose-Response Relationship, Drug , Down-Regulation , Foam Cells/enzymology , Foam Cells/pathology , Gene Expression Regulation, Enzymologic , Lipoproteins, LDL/blood , Macrophages, Peritoneal/enzymology , Macrophages, Peritoneal/pathology , Mice, Inbred C57BL , Mice, Knockout , Phosphatidylinositol 3-Kinase/metabolism , RNA, Messenger/metabolism , STAT6 Transcription Factor/metabolism , Signal Transduction/drug effects
5.
Front Horm Res ; 43: 107-24, 2014.
Article in English | MEDLINE | ID: mdl-24943302

ABSTRACT

The process of vascular aging encompasses alterations in the function of endothelial (ECs) and vascular smooth muscle cells (VSMCs) via oxidation, inflammation, cell senescence and epigenetic modifications, increasing the probability of atherosclerosis. Aged vessels exhibit decreased endothelial antithrombogenic properties, increased reactive oxygen species generation, inflammatory signaling and migration of VSMCs to the subintimal space, impaired angiogenesis and increased elastin degradation. The key initiating step in atherogenesis is subendothelial accumulation of apolipoprotein B-containing low-density lipoproteins resulting in activation of ECs and recruitment of monocytes. Activated ECs secrete 'chemokines' that interact with cognate chemokine receptors on monocytes and promote directional migration. Recruitment of immune cells establishes a proinflammatory status, further causing elevated oxidative stress, which in turn triggers a series of events including apoptotic or necrotic death of vascular and nonvascular cells. Increased oxidative stress is also considered to be a key factor in mechanisms of aging-associated changes in tissue integrity and function. Experimental evidence indicates that insulin-like growth factor-1 exerts antioxidant, anti-inflammatory and pro-survival effects on the vasculature, reducing atherosclerotic plaque burden and promoting features of atherosclerotic plaque stability.


Subject(s)
Aging/physiology , Atherosclerosis/physiopathology , Endothelial Cells/physiology , Insulin-Like Growth Factor I/metabolism , Muscle, Smooth, Vascular/physiology , Myocytes, Smooth Muscle/physiology , Adaptor Proteins, Signal Transducing/physiology , Animals , Anti-Inflammatory Agents/pharmacology , Apolipoproteins E/deficiency , Cell Movement , Cellular Senescence , Endothelin-1/physiology , Humans , Hypertension/physiopathology , Lipoproteins, LDL , Mice , Monocytes , Oxidative Stress , Plaque, Atherosclerotic/prevention & control , Receptor, IGF Type 1/metabolism , Regeneration
6.
J Biol Chem ; 289(11): 7264-74, 2014 Mar 14.
Article in English | MEDLINE | ID: mdl-24469459

ABSTRACT

Collagen content in atherosclerotic plaque is a hallmark of plaque stability. Our earlier studies showed that insulin-like growth factor-1 (IGF-1) increases collagen content in atherosclerotic plaques of Apoe(-/-) mice. To identify mechanisms we investigated the effect of IGF-1 on the la ribonucleoprotein domain family member 6 (LARP6). LARP6 binds a stem-loop motif in the 5'-UTR of the mRNAs encoding the collagen type I α-subunits (α1(I) and α2(I)), and coordinates their translation into the heterotrimeric collagen type I molecule. In human aortic smooth muscle cells (SMCs), IGF-1 rapidly increased LARP6 expression and the rate of collagen synthesis and extracellular accumulation. IGF-1 increased both LARP6 and collagen type I expression via a post-transcriptional and translation-dependent mechanism involving PI3K/Akt/p70S6k-signaling. Immunoprecipitation of LARP6, followed by qPCR indicated that IGF-1 increased the level of COL1a1 and COL1a2 mRNA bound to LARP6. Mutation of the 5' stem-loop of Col1a1 mRNA, which inhibits binding of LARP6, abolished the ability of IGF-1 to increase synthesis of collagen type I. Furthermore, overexpression of a 5' stem-loop RNA molecular decoy that sequesters LARP6, prevented the ability of IGF-1 to increase pro-α1(I) and mature α1(I) expression in cultured medium. IGF-1 infusion in Apoe(-/-) mice increased expression of LARP6 and pro-α1(I) in aortic lysates, and SMC-specific IGF-1-overexpression robustly increased collagen fibrillogenesis in atherosclerotic plaque. In conclusion, we identify LARP6 as a critical mediator by which IGF-1 augments synthesis of collagen type I in vascular smooth muscle, which may play an important role in promoting atherosclerotic plaque stability.


Subject(s)
Autoantigens/metabolism , Collagen Type I/metabolism , Gene Expression Regulation , Insulin-Like Growth Factor I/metabolism , Ribonucleoproteins/metabolism , Animals , Aorta/cytology , Atherosclerosis/metabolism , Cells, Cultured , Collagen Type I, alpha 1 Chain , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Muscle, Smooth/cytology , Mutation , Myocytes, Smooth Muscle/metabolism , Oxidative Stress , Plaque, Atherosclerotic/metabolism , RNA, Messenger/metabolism , Signal Transduction , SS-B Antigen
7.
J Gerontol A Biol Sci Med Sci ; 67(6): 626-39, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22491965

ABSTRACT

Insulin-like growth factor 1 (IGF-1) is an endocrine and autocrine/paracrine growth factor that circulates at high levels in the plasma and is expressed in most cell types. IGF-1 has major effects on development, cell growth and differentiation, and tissue repair. Recent evidence indicates that IGF-1 reduces atherosclerosis burden and improves features of atherosclerotic plaque stability in animal models. Potential mechanisms for this atheroprotective effect include IGF-1-induced reduction in oxidative stress, cell apoptosis, proinflammatory signaling, and endothelial dysfunction. Aging is associated with increased vascular oxidative stress and vascular disease, suggesting that IGF-1 may exert salutary effects on vascular aging processes. In this review, we will provide a comprehensive update on IGF-1's ability to modulate vascular oxidative stress and to limit atherogenesis and the vascular complications of aging.


Subject(s)
Aging/metabolism , Atherosclerosis/metabolism , Insulin-Like Growth Factor I/metabolism , Aging/drug effects , Animals , Apoptosis/drug effects , Apoptosis/physiology , Atherosclerosis/drug therapy , Disease Models, Animal , Female , Humans , Inflammation/drug therapy , Inflammation/physiopathology , Insulin-Like Growth Factor I/pharmacology , Longevity/drug effects , Longevity/physiology , Male , Mice , Oxidative Stress/drug effects , Oxidative Stress/physiology , Plaque, Atherosclerotic/drug therapy , Plaque, Atherosclerotic/metabolism , Rats
8.
FEBS Lett ; 585(19): 3065-72, 2011 Oct 03.
Article in English | MEDLINE | ID: mdl-21872589

ABSTRACT

We have shown previously that insulin like-growth factor I (IGF-1) suppressed atherosclerosis in Apoe(-/-) mice and activated endothelial nitric oxide (NO) synthase. To determine whether IGF-1-induced atheroprotection depends on NO, IGF-1- or saline-infused mice were treated with l-NAME, the pan-NO synthase inhibitor or with d-NAME (control). IGF-1 reduced atherosclerosis in both the d-NAME and l-NAME groups suggesting that IGF-1's anti-atherogenic effect was NO-independent. IGF-1 increased plaque smooth muscle cells, suppressed cell apoptosis and downregulated lipoprotein lipase and these effects were also NO-independent. On the contrary, IGF-1 decreased oxidative stress and suppressed TNF-α levels and these effects were blocked by l-NAME. Thus IGF-1's anti-oxidant effect is dependent on its ability to increase NO but is distinct from its anti-atherosclerotic effect which is NO-independent.


Subject(s)
Antioxidants/metabolism , Apoptosis/physiology , Atherosclerosis/physiopathology , Insulin-Like Growth Factor I/metabolism , Nitric Oxide Synthase Type III/metabolism , Nitric Oxide/metabolism , Animals , Aorta/metabolism , Aorta/pathology , Apolipoproteins E/genetics , Apolipoproteins E/metabolism , Atherosclerosis/pathology , Cell Line , Enzyme Inhibitors/metabolism , Gene Expression Profiling , Humans , Lipoprotein Lipase/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , NG-Nitroarginine Methyl Ester/chemistry , NG-Nitroarginine Methyl Ester/metabolism , Oxidative Stress , Tumor Necrosis Factor-alpha/metabolism
9.
Am J Physiol Heart Circ Physiol ; 300(5): H1898-906, 2011 May.
Article in English | MEDLINE | ID: mdl-21335474

ABSTRACT

Some clinical studies have suggested that lower IGF-I levels may be associated with an increased risk of ischemic heart disease. We generated atherosclerosis-prone apolipoprotein E-deficient (ApoE(-/-)) mice with 6T alleles (6T/ApoE(-/-) mice) with a 20% decline in circulating IGF-I and fed these mice and control ApoE(-/-) mice with normal chow or a Western diet for 12 wk to evaluate the effect of low serum IGF-I on atherosclerosis progression. We found that the 6T/ApoE(-/-) phenotype was characterized by an increased atherosclerotic burden, elevated plaque macrophages, and increased proinflammatory cytokine TNF-α levels compared with ApoE(-/-) controls. 6T/ApoE(-/-) mice had similar body weight, blood pressure, serum total cholesterol levels, total plaque and smooth muscle cell apoptosis rates, and circulating levels of endothelial progenitor cells as ApoE(-/-) mice. 6T/ApoE(-/-) mice fed with normal chow had reduced vascular endothelial nitric oxide synthase mRNA levels and a trend to increased aortic expression of chemokine (C-C motif) receptor (CCR)1, CCR2, and monocyte chemoattractant protein-1/chemokine (C-C motif) ligand 2. Western diet-fed 6T/ApoE(-/-) mice had a trend to increased expression of macrophage scavenger receptor-1/scavenger receptor-A, osteopontin, ATP-binding cassette (subfamily A, member 1), and angiotensin-converting enzyme and elevated circulating levels of the neutrophil chemoattractant chemokine (C-X-C motif) ligand 1 (KC). Our data establish a link between lower circulating IGF-I and increased atherosclerosis that has important clinical implications.


Subject(s)
Apolipoproteins E/deficiency , Atherosclerosis/epidemiology , Atherosclerosis/physiopathology , Insulin-Like Growth Factor I/metabolism , Animals , Apolipoproteins E/genetics , Apolipoproteins E/physiology , Blood Pressure/physiology , Body Weight/physiology , Cholesterol/blood , Disease Models, Animal , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Knockout , Rats , Risk Factors , Tumor Necrosis Factor-alpha/blood
10.
Arterioscler Thromb Vasc Biol ; 30(10): 1916-24, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20671230

ABSTRACT

OBJECTIVE: Growth factors may play a permissive role in atherosclerosis initiation and progression, in part via their promotion of vascular smooth muscle cell (VSMC) accumulation in plaques. However, unstable human plaques often have a relative paucity of VSMC, which has been suggested to contribute to plaque rupture and erosion and to clinical events. Insulin-like growth factor-1 (IGF-1) is an endocrine and autocrine/paracrine growth factor that is a mitogen for VSMC, but when infused into Apoe(-/-) mice it paradoxically reduces atherosclerosis burden. METHODS AND RESULTS: To determine the effect of stimulation of VSMC growth on atherosclerotic plaque development and to understand mechanisms of IGF-1's atheroprotective effect, we assessed atherosclerotic plaques in mice overexpressing IGF-1 in smooth muscle cells (SMC) under the control of the α-smooth muscle actin promoter, after backcrossing to the Apoe(-/-) background (SMP8/Apoe(-/-)). Compared with Apoe(-/-) mice, these SMP8/Apoe(-/-) mice developed a comparable plaque burden after 12 weeks on a Western diet, suggesting that the ability of increased circulating IGF-1 to reduce plaque burden was mediated in large part via non-SMC target cells. However, advanced plaques in SMP8/Apoe(-/-) mice displayed several features of plaque stability, including increased fibrous cap area, α-smooth muscle actin-positive SMC and collagen content, and reduced necrotic cores. CONCLUSIONS: These findings indicate that stimulation of VSMC IGF-1 signaling does not alter total atherosclerotic plaque burden and may improve atherosclerotic plaque stability.


Subject(s)
Apolipoproteins E/deficiency , Atherosclerosis/pathology , Atherosclerosis/physiopathology , Insulin-Like Growth Factor I/physiology , Myocytes, Smooth Muscle/pathology , Myocytes, Smooth Muscle/physiology , Animals , Aorta/pathology , Aorta/physiopathology , Apolipoproteins E/genetics , Atherosclerosis/genetics , Cells, Cultured , Disease Models, Animal , Gene Expression , Humans , Hypertrophy , Insulin-Like Growth Factor I/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Oxidative Stress , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
11.
Trends Endocrinol Metab ; 21(4): 245-54, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20071192

ABSTRACT

Atherosclerosis is a chronic inflammatory disease in which early endothelial dysfunction and subintimal modified lipoprotein deposition progress to complex, advanced lesions that are predisposed to erosion, rupture and thrombosis. Oxidative stress plays a crucial role not only in initial lesion formation but also in lesion progression and destabilization. Although most growth factors are thought to promote vascular smooth muscle cell proliferation and migration, thereby increasing neointima, recent animal studies indicate that insulin-like growth factor (IGF)-1 exerts both pleiotropic anti-oxidant effects and anti-inflammatory effects, which together reduce atherosclerotic burden. This review discusses the effects of IGF-1 in models of vascular injury and atherosclerosis, emphasizing the relationship between oxidative stress and potential atheroprotective actions of IGF-1.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Antioxidants/therapeutic use , Atherosclerosis/drug therapy , Insulin-Like Growth Factor I/therapeutic use , Oxidative Stress , Animals , Apoptosis , Atherosclerosis/pathology , Atherosclerosis/prevention & control , Endothelium, Vascular/cytology , Insulin-Like Growth Factor I/metabolism , Models, Animal , Muscle, Smooth, Vascular/pathology , Stem Cells/physiology
12.
Circ Res ; 102(9): 1109-17, 2008 May 09.
Article in English | MEDLINE | ID: mdl-18340010

ABSTRACT

The dystrophin-glycoprotein complex is a large complex of membrane-associated proteins linking the cytoskeleton to the extracellular matrix in muscle. Transmembrane heterodimeric (alphabeta) integrins serve also as cellular adhesion molecules and mechanotransducers. In the animal model for Duchenne muscular dystrophy, the mdx mouse, loss of dystrophin causes more severe abnormalities in skeletal than in cardiac muscle. We hypothesized that ablation of cardiac myocyte integrins in the mdx background would lead to a severe cardiomyopathic phenotype. Mdx mice were crossed to ones with cardiac myocyte-specific deletion of beta1 integrin (beta1KO) to generate beta1KOmdx. Unstressed beta1KOmdx mice were viable and had normal cardiac function; however, high mortality was seen in peri- and postpartum females by 6 months of age, when severe myocardial necrosis and fibrosis and extensive dystrophic calcification was seen. Decreased ventricular function and blunted adrenergic responsiveness was found in the beta1KOmdx mice compared with control (Lox/Lox, no Cre), beta1KO, and mdx. Similarly, adult beta1KOmdx males were more prone to isoproterenol-induced heart failure and death compared with control groups. Given the extensive calcification, we analyzed transcript levels of genes linked to fibrosis and calcification and found matrix gamma-carboxyglutamic acid protein, decorin, periostin, and the osteoblast transcription factor Runx2/Cbfa1 significantly increased in beta1KOmdx cardiac muscle. Our data show that combined deficiency of dystrophin and integrins in murine cardiac myocytes results in more severe cardiomyopathic changes in the stressed myocardium than reduction of either dystrophin or integrins alone and predisposes to myocardial calcification.


Subject(s)
Calcinosis/metabolism , Cardiomyopathies/metabolism , Dystrophin/metabolism , Integrin beta1/metabolism , Myocytes, Cardiac/metabolism , Ventricular Function , Adrenergic beta-Agonists/administration & dosage , Animals , Calcinosis/genetics , Calcinosis/physiopathology , Calcium-Binding Proteins/metabolism , Cardiomyopathies/genetics , Cardiomyopathies/pathology , Cardiomyopathies/physiopathology , Cell Adhesion Molecules/metabolism , Core Binding Factor Alpha 1 Subunit/metabolism , Decorin , Disease Models, Animal , Dobutamine/administration & dosage , Dystrophin/deficiency , Dystrophin/genetics , Extracellular Matrix Proteins/metabolism , Female , Fibrosis , Heart Failure/chemically induced , Heart Failure/genetics , Heart Failure/metabolism , Heart Failure/physiopathology , Integrin beta1/genetics , Isoproterenol/administration & dosage , Male , Mice , Mice, Inbred mdx , Mice, Knockout , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/pathology , Necrosis , Phenotype , Pregnancy , Proteoglycans/metabolism , RNA, Messenger/metabolism , Severity of Illness Index , Time Factors , Up-Regulation , Ventricular Function/drug effects , Matrix Gla Protein
13.
Mol Cell Biol ; 28(2): 794-802, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17984225

ABSTRACT

The largest subgroup of integrins is that containing the beta1 subunit. beta1 integrins have been implicated in a wide array of biological processes ranging from adhesion to cell growth, organogenesis, and mechanotransduction. Global deletion of beta1 integrin expression results in embryonic death at ca. embryonic day 5 (E5), a developmental time point too early to determine the effects of this integrin on vascular development. To elucidate the specific role of beta1 integrin in the vasculature, we conditionally deleted the beta1 gene in the endothelium. Homozygous deletion of beta1 integrins in the endothelium resulted in failure of normal vascular patterning, severe fetal growth retardation, and embryonic death at E9.5 to 10, although there were no overt effects on vasculogenesis. Heterozygous endothelial beta1 gene deletion did not diminish fetal or postnatal survival, but it reduced beta1 subunit expression in endothelial cells from adult mice by approximately 40%. These mice demonstrated abnormal vascular remodeling in response to experimentally altered in vivo blood flow and diminished vascularization in healing wounds. These data demonstrate that endothelial expression of beta1 integrin is required for developmental vascular patterning and that endothelial beta1 gene dosing has significant functional effects on vascular remodeling in the adult. Understanding how beta1 integrin expression is modulated may have significant clinical importance.


Subject(s)
Embryo, Mammalian/blood supply , Embryo, Mammalian/metabolism , Endothelium, Vascular/metabolism , Gene Expression Regulation, Developmental , Integrin beta1/metabolism , Animals , Animals, Newborn , Embryo Loss/genetics , Embryo Loss/metabolism , Embryo, Mammalian/embryology , Endothelium, Vascular/embryology , Endothelium, Vascular/growth & development , Gene Deletion , Homozygote , Integrin beta1/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Wound Healing
14.
Arterioscler Thromb Vasc Biol ; 27(12): 2684-90, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17916769

ABSTRACT

OBJECTIVE: Whereas growth factors, via their ability to stimulate vascular smooth muscle cell (VSMC) proliferation and migration, have been thought to play a permissive role in atherosclerosis initiation and progression, the role of insulin-like growth factor-1 (IGF-1) is unknown. Here we report for the first time that IGF-1 infusion decreased atherosclerotic plaque progression in ApoE-deficient mice on a Western diet. METHODS AND RESULTS: ApoE-null mice (8 weeks) were infused with vehicle or recombinant human IGF-1 and fed a high-fat diet for 12 weeks. Analysis of aortic sinuses revealed that IGF-1 infusion decreased atherosclerotic plaque progression and macrophage infiltration into lesions. Furthermore, IGF-1 decreased vascular expression of the proinflammatory cytokines interleukin-6 and tumor necrosis factor-alpha, reduced aortic superoxide formation and urinary 8-isoprostane levels, and increased aortic pAkt and eNOS expression and circulating endothelial progenitor cells, consistent with an antiinflammatory, antioxidant, and prorepair effect on the vasculature. CONCLUSIONS: Our data indicate that an increase in circulating IGF-1 reduces vascular inflammatory responses, systemic and vascular oxidant stress and decreases atherosclerotic plaque progression. These findings have major implications for the treatment of atherosclerosis.


Subject(s)
Anti-Inflammatory Agents/metabolism , Antioxidants/metabolism , Apolipoproteins E/metabolism , Atherosclerosis/prevention & control , Inflammation/prevention & control , Insulin-Like Growth Factor I/metabolism , Oxidative Stress , Animals , Anti-Inflammatory Agents/administration & dosage , Antioxidants/administration & dosage , Aorta/metabolism , Aorta/pathology , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Atherosclerosis/genetics , Atherosclerosis/metabolism , Atherosclerosis/pathology , Cells, Cultured , Dietary Fats/administration & dosage , Dinoprost/analogs & derivatives , Dinoprost/urine , Disease Models, Animal , Disease Progression , Endothelial Cells/metabolism , Endothelial Cells/pathology , Humans , Inflammation/genetics , Inflammation/metabolism , Inflammation/pathology , Insulin-Like Growth Factor I/administration & dosage , Interleukin-6/metabolism , Macrophages/metabolism , Macrophages/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Nitric Oxide Synthase Type II/metabolism , Nitric Oxide Synthase Type III , Oxidative Stress/drug effects , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , RNA, Messenger/metabolism , Recombinant Proteins/metabolism , Stem Cells/metabolism , Stem Cells/pathology , Superoxides/metabolism , Tumor Necrosis Factor-alpha/metabolism
15.
Circ Res ; 99(2): 191-200, 2006 Jul 21.
Article in English | MEDLINE | ID: mdl-16778134

ABSTRACT

Glyceraldehyde-3-phosphate dehydrogenase (GAPDH) is a classical glycolytic enzyme that is involved in cellular energy production and has important housekeeping functions. We used the natural prooxidant and proatherogenic molecule oxidized low-density lipoprotein (OxLDL) to determine a potential link between OxLDL-promoted oxidative stress, GAPDH expression, and smooth muscle cell energy metabolism. OxLDL but not native LDL (nLDL) produced a 60% to 100% dose- and time-dependent reduction of GAPDH protein. OxLDL increased reactive oxygen species (ROS) formation, including rapid elevation of H2O2 levels. OxLDL decreased intracellular catalase expression, likely contributing to the increase in H2O2. Antioxidants, anti-CD36 receptor antibody, NADPH oxidase, or lipoxygenase blockers decreased OxLDL-specific ROS and prevented GAPDH downregulation. 12/15-Lipoxygenase or p47phox deficiency resulted in attenuation of GAPDH downregulation, but 5-lipoxygenase suppression had no effect. OxLDL or exogenous H2O2 oxidized GAPDH thiols, decreasing GAPDH protein half-life and increasing GAPDH sensitivity to proteasome-mediated protein degradation in vitro. OxLDL- or small interfering RNA-specific downregulation of GAPDH resulted in 65% reduction in glycolysis rate and 82% decrease in ATP levels. In conclusion, our data demonstrate that OxLDL downregulated GAPDH via a H2O2-dependent decrease in protein stability. GAPDH protein damage resulted in marked depletion of cellular ATP levels. Our data have important implications for understanding the metabolic effect of OxLDL on the vessel wall and mechanism of atherogenesis.


Subject(s)
Adenosine Triphosphate/metabolism , Energy Metabolism , Glyceraldehyde 3-Phosphate Dehydrogenase (NADP+)/metabolism , Lipoproteins, LDL/physiology , Muscle, Smooth, Vascular/cytology , Myocytes, Smooth Muscle/metabolism , Aorta/cytology , Atherosclerosis/etiology , Catalase , Cells, Cultured , Down-Regulation , Humans , Hydrogen Peroxide , Oxidative Stress , Reactive Oxygen Species/metabolism
16.
Histochem Cell Biol ; 118(6): 431-9, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12483308

ABSTRACT

Integrins are heterodimeric cell-surface receptors that link the extracellular matrix and the intracellular cytoskeleton and function as mechanotransducers. Signaling through integrins is important for cell growth, migration, and survival. Extracellular matrix is altered in the myocardium during hypertrophic induction and the transition to heart failure. The role of integrins in this process is poorly understood. Recently, integrin subunits have been identified that are dominantly expressed in striated muscle. We tested the hypothesis that since integrins are mechanotransducers, their expression and signaling would be modulated with murine left ventricular hemodynamic loading. The acute and chronic effects of pressure overload on changes in the expression of integrins, as well as related integrin-mediated signaling events were studied. Acute pressure loading increased phosphorylation of focal adhesion kinase, p42 and p44 extracellular signal-regulated kinase. Chronic loading: (1) increased expression of alpha1, alpha5, and beta1 integrin transcripts and (2) increased protein expression of integrin subunits which are dominantly expressed in striated muscle (alpha7 and beta1D) both by western blotting and immunofluorescent microscopy. These results show that adaptive responses of the myocardium to pressure overload include acute modulation of integrin-related signaling molecules and more chronic changes effect expression of integrin subunits, including ones dominantly expressed in muscle.


Subject(s)
Heart Ventricles/metabolism , Integrins/metabolism , Mitogen-Activated Protein Kinases/metabolism , Protein-Tyrosine Kinases/metabolism , Animals , Aorta/surgery , Atrial Natriuretic Factor/genetics , Atrial Natriuretic Factor/metabolism , Blotting, Northern , Blotting, Western , Cardiomegaly/etiology , Cardiomegaly/genetics , Cardiomegaly/metabolism , Constriction, Pathologic/complications , Focal Adhesion Kinase 1 , Focal Adhesion Protein-Tyrosine Kinases , Gene Expression , Heart Ventricles/pathology , In Vitro Techniques , Integrins/genetics , Mice , Mice, Inbred C57BL , Phosphorylation , Pressure , Signal Transduction
18.
Circ Res ; 90(4): 458-64, 2002 Mar 08.
Article in English | MEDLINE | ID: mdl-11884376

ABSTRACT

Integrins link the extracellular matrix to the cellular cytoskeleton and serve important roles in cell growth, differentiation, migration, and survival. Ablation of beta1 integrin in all murine tissues results in peri-implantation embryonic lethality. To investigate the role of beta1 integrin in the myocardium, we used Cre-LoxP technology to inactivate the beta1 integrin gene exclusively in ventricular cardiac myocytes. Animals with homozygous ventricular myocyte beta1 integrin gene excision were born in appropriate numbers and grew into adulthood. These animals had 18% of control levels of beta1D integrin protein in the heart and displayed myocardial fibrosis. High-fidelity micromanometer-tipped catheterization of the intact 5-week-old beta1 integrin knockout mice showed depressed left ventricular basal and dobutamine-stimulated contractility and relaxation (LV dP/dt(max) and LV dP/dt(min)) as compared with control groups (n=8 to 10 of each, P<0.01). Hemodynamic loading imposed by 7 days of transverse aortic constriction showed that the beta1 integrin knockout mice were intolerant of this stress as they had 53% survival versus 88% in controls (n=15 each). By 6 months of age, mice with depressed ventricular expression of beta1 integrin developed a dilated cardiomyopathy that was not evident in any control animals and had patchy decrease in glucose metabolism as determined by positron emission tomography. Myocyte membrane integrity as determined via Evan's blue dye staining was disrupted in the beta1 integrin knockout mice. This model provides strong evidence for the importance of beta1 integrin in cardiac form and function and indicates that integrins can be linked to development of cardiomyopathies.


Subject(s)
Fibrosis/genetics , Heart Failure/genetics , Integrin beta1/genetics , Myocardium/metabolism , Animals , Cardiomyopathy, Dilated/complications , Cardiomyopathy, Dilated/genetics , Cardiomyopathy, Dilated/pathology , Cell Membrane/metabolism , Cell Membrane/pathology , Cell Membrane Permeability , Disease Progression , Fibrosis/complications , Fibrosis/pathology , Gene Targeting , Glucose/metabolism , Heart Failure/complications , Heart Failure/pathology , Heart Ventricles/pathology , Homozygote , Integrases , Integrin beta1/metabolism , Mice , Mice, Knockout , Myocardium/pathology , Myocardium/ultrastructure , Organ Specificity , Talin/metabolism , Viral Proteins
SELECTION OF CITATIONS
SEARCH DETAIL
...