Skip to main content
  • American Heart Association
  • Science Volunteer
  • Warning Signs
  • Advanced Search
  • Donate

  • Home
  • About this Journal
    • Editorial Board
    • General Statistics
    • Circulation Doodle
      • Doodle Gallery
      • Circulation Cover Doodle
        • → Blip the Doodle
    • Information for Advertisers
    • Author Reprints
    • Commercial Reprints
    • Customer Service and Ordering Information
    • Subscribe to AHA Journals
  • All Issues
  • Subjects
    • All Subjects
    • Arrhythmia and Electrophysiology
    • Basic, Translational, and Clinical Research
    • Critical Care and Resuscitation
    • Epidemiology, Lifestyle, and Prevention
    • Genetics
    • Heart Failure and Cardiac Disease
    • Hypertension
    • Imaging and Diagnostic Testing
    • Intervention, Surgery, Transplantation
    • Quality and Outcomes
    • Stroke
    • Vascular Disease
  • Browse Features
    • AHA Guidelines and Statements
      • Recently Published Guidelines
    • Bridging Disciplines
    • Circulation at Major Meetings
    • Special Themed Issues
    • Global Impact of the 2017 ACC/AHA Hypertension Guidelines
    • Circulation Supplements
    • Cardiovascular Case Series
    • ECG Challenge
    • Hospitals of History
      • Brigham and Women's Hospital
      • Hartford Hospital
      • Hospital Santa Maria del Popolo, Naples, Italy
      • Instituto do Coração-INCOR (São Paulo, Brasil)
      • Minneapolis City Hospital
      • Parkland Hospital: Dallas, Texas
      • Pennsylvania Hospital, Philadelphia
      • Pitié-Salpêtrière Hospital
      • Royal Infirmary of Edinburgh, Scotland
      • Tufts Medical Center
      • University of Michigan
      • Uppsala University Hospital
      • Vassar Brothers Medical Center (Poughkeepsie, NY)
      • Wroclaw Medical University
      • Women's College Hospital, Toronto, Canada
      • Henry Ford Hospital, Detroit, Michigan
      • Instituto Nacional de Cardiología Ignacio Chávez – INCICh México City, México
      • Kuang-Tien General Hospital (Taichug, Taiwan)
    • On My Mind
    • Podcast Archive
    • → Subscribe to Circulation on the Run
    • →Circulation FIT Podcast 2018
    • → #FITFAVs
  • Resources
    • Instructions for Authors
      • Accepted Manuscripts
      • Revised Manuscripts
    • → Article Types
    • → General Preparation Instructions
    • → Research Guidelines
    • → How to Submit a Manuscript
    • Journal Policies
    • Permissions and Rights Q&A
    • Submission Sites
    • Circulation CME
    • AHA Journals RSS Feeds
    • International Users
    • AHA Newsroom
  • AHA Journals
    • AHA Journals Home
    • Arteriosclerosis, Thrombosis, and Vascular Biology (ATVB)
    • Circulation
    • → Circ: Arrhythmia and Electrophysiology
    • → Circ: Genomic and Precision Medicine
    • → Circ: Cardiovascular Imaging
    • → Circ: Cardiovascular Interventions
    • → Circ: Cardiovascular Quality & Outcomes
    • → Circ: Heart Failure
    • Circulation Research
    • Hypertension
    • Stroke
    • Journal of the American Heart Association
  • Facebook
  • Twitter

  • My alerts
  • Sign In
  • Join

  • Advanced search

Header Publisher Menu

  • American Heart Association
  • Science Volunteer
  • Warning Signs
  • Advanced Search
  • Donate

Circulation

  • My alerts
  • Sign In
  • Join

  • Facebook
  • Twitter
  • Home
  • About this Journal
    • Editorial Board
    • General Statistics
    • Circulation Doodle
    • Information for Advertisers
    • Author Reprints
    • Commercial Reprints
    • Customer Service and Ordering Information
    • Subscribe to AHA Journals
  • All Issues
  • Subjects
    • All Subjects
    • Arrhythmia and Electrophysiology
    • Basic, Translational, and Clinical Research
    • Critical Care and Resuscitation
    • Epidemiology, Lifestyle, and Prevention
    • Genetics
    • Heart Failure and Cardiac Disease
    • Hypertension
    • Imaging and Diagnostic Testing
    • Intervention, Surgery, Transplantation
    • Quality and Outcomes
    • Stroke
    • Vascular Disease
  • Browse Features
    • AHA Guidelines and Statements
    • Bridging Disciplines
    • Circulation at Major Meetings
    • Special Themed Issues
    • Global Impact of the 2017 ACC/AHA Hypertension Guidelines
    • Circulation Supplements
    • Cardiovascular Case Series
    • ECG Challenge
    • Hospitals of History
    • On My Mind
    • Podcast Archive
    • → Subscribe to Circulation on the Run
    • →Circulation FIT Podcast 2018
    • → #FITFAVs
  • Resources
    • Instructions for Authors
    • → Article Types
    • → General Preparation Instructions
    • → Research Guidelines
    • → How to Submit a Manuscript
    • Journal Policies
    • Permissions and Rights Q&A
    • Submission Sites
    • Circulation CME
    • AHA Journals RSS Feeds
    • International Users
    • AHA Newsroom
  • AHA Journals
    • AHA Journals Home
    • Arteriosclerosis, Thrombosis, and Vascular Biology (ATVB)
    • Circulation
    • → Circ: Arrhythmia and Electrophysiology
    • → Circ: Genomic and Precision Medicine
    • → Circ: Cardiovascular Imaging
    • → Circ: Cardiovascular Interventions
    • → Circ: Cardiovascular Quality & Outcomes
    • → Circ: Heart Failure
    • Circulation Research
    • Hypertension
    • Stroke
    • Journal of the American Heart Association
Original Article

Insulin-Like Growth Factor-1 Receptor Deficiency in Macrophages Accelerates Atherosclerosis and Induces an Unstable Plaque Phenotype in Apolipoprotein E–Deficient MiceCLINICAL PERSPECTIVE

Yusuke Higashi, Sergiy Sukhanov, Shaw-Yung Shai, Svitlana Danchuk, Richard Tang, Patricia Snarski, Zhaohui Li, Patricia Lobelle-Rich, Meifang Wang, Derek Wang, Hong Yu, Ronald Korthuis, Patrice Delafontaine
Download PDF
https://doi.org/10.1161/CIRCULATIONAHA.116.021805
Circulation. 2016;133:2263-2278
Originally published May 6, 2016
Yusuke Higashi
From Departments of Medicine (Y.H., S.S., S.D., P.S., Z.L., P.D.) and Medical Pharmacology and Physiology (Y.H., S.S., M.W., D.W., H.Y., R.K.), University of Missouri School of Medicine, Columbia; and Department of Medicine, Tulane University School of Medicine, New Orleans, LA (S.-Y.S., R.T., P.L.-R.).
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Sergiy Sukhanov
From Departments of Medicine (Y.H., S.S., S.D., P.S., Z.L., P.D.) and Medical Pharmacology and Physiology (Y.H., S.S., M.W., D.W., H.Y., R.K.), University of Missouri School of Medicine, Columbia; and Department of Medicine, Tulane University School of Medicine, New Orleans, LA (S.-Y.S., R.T., P.L.-R.).
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Shaw-Yung Shai
From Departments of Medicine (Y.H., S.S., S.D., P.S., Z.L., P.D.) and Medical Pharmacology and Physiology (Y.H., S.S., M.W., D.W., H.Y., R.K.), University of Missouri School of Medicine, Columbia; and Department of Medicine, Tulane University School of Medicine, New Orleans, LA (S.-Y.S., R.T., P.L.-R.).
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Svitlana Danchuk
From Departments of Medicine (Y.H., S.S., S.D., P.S., Z.L., P.D.) and Medical Pharmacology and Physiology (Y.H., S.S., M.W., D.W., H.Y., R.K.), University of Missouri School of Medicine, Columbia; and Department of Medicine, Tulane University School of Medicine, New Orleans, LA (S.-Y.S., R.T., P.L.-R.).
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Richard Tang
From Departments of Medicine (Y.H., S.S., S.D., P.S., Z.L., P.D.) and Medical Pharmacology and Physiology (Y.H., S.S., M.W., D.W., H.Y., R.K.), University of Missouri School of Medicine, Columbia; and Department of Medicine, Tulane University School of Medicine, New Orleans, LA (S.-Y.S., R.T., P.L.-R.).
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Patricia Snarski
From Departments of Medicine (Y.H., S.S., S.D., P.S., Z.L., P.D.) and Medical Pharmacology and Physiology (Y.H., S.S., M.W., D.W., H.Y., R.K.), University of Missouri School of Medicine, Columbia; and Department of Medicine, Tulane University School of Medicine, New Orleans, LA (S.-Y.S., R.T., P.L.-R.).
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Zhaohui Li
From Departments of Medicine (Y.H., S.S., S.D., P.S., Z.L., P.D.) and Medical Pharmacology and Physiology (Y.H., S.S., M.W., D.W., H.Y., R.K.), University of Missouri School of Medicine, Columbia; and Department of Medicine, Tulane University School of Medicine, New Orleans, LA (S.-Y.S., R.T., P.L.-R.).
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Patricia Lobelle-Rich
From Departments of Medicine (Y.H., S.S., S.D., P.S., Z.L., P.D.) and Medical Pharmacology and Physiology (Y.H., S.S., M.W., D.W., H.Y., R.K.), University of Missouri School of Medicine, Columbia; and Department of Medicine, Tulane University School of Medicine, New Orleans, LA (S.-Y.S., R.T., P.L.-R.).
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Meifang Wang
From Departments of Medicine (Y.H., S.S., S.D., P.S., Z.L., P.D.) and Medical Pharmacology and Physiology (Y.H., S.S., M.W., D.W., H.Y., R.K.), University of Missouri School of Medicine, Columbia; and Department of Medicine, Tulane University School of Medicine, New Orleans, LA (S.-Y.S., R.T., P.L.-R.).
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Derek Wang
From Departments of Medicine (Y.H., S.S., S.D., P.S., Z.L., P.D.) and Medical Pharmacology and Physiology (Y.H., S.S., M.W., D.W., H.Y., R.K.), University of Missouri School of Medicine, Columbia; and Department of Medicine, Tulane University School of Medicine, New Orleans, LA (S.-Y.S., R.T., P.L.-R.).
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Hong Yu
From Departments of Medicine (Y.H., S.S., S.D., P.S., Z.L., P.D.) and Medical Pharmacology and Physiology (Y.H., S.S., M.W., D.W., H.Y., R.K.), University of Missouri School of Medicine, Columbia; and Department of Medicine, Tulane University School of Medicine, New Orleans, LA (S.-Y.S., R.T., P.L.-R.).
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Ronald Korthuis
From Departments of Medicine (Y.H., S.S., S.D., P.S., Z.L., P.D.) and Medical Pharmacology and Physiology (Y.H., S.S., M.W., D.W., H.Y., R.K.), University of Missouri School of Medicine, Columbia; and Department of Medicine, Tulane University School of Medicine, New Orleans, LA (S.-Y.S., R.T., P.L.-R.).
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Patrice Delafontaine
From Departments of Medicine (Y.H., S.S., S.D., P.S., Z.L., P.D.) and Medical Pharmacology and Physiology (Y.H., S.S., M.W., D.W., H.Y., R.K.), University of Missouri School of Medicine, Columbia; and Department of Medicine, Tulane University School of Medicine, New Orleans, LA (S.-Y.S., R.T., P.L.-R.).
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • Article
  • Figures & Tables
  • Supplemental Materials
  • Info & Metrics
  • eLetters

Jump to

  • Article
    • Abstract
    • Introduction
    • Methods
    • Results
    • Discussion
    • Acknowledgments
    • Sources of Funding
    • Disclosures
    • Footnotes
    • References
  • Figures & Tables
  • Supplemental Materials
  • Info & Metrics
  • eLetters
Loading

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.

  • atherosclerosis
  • inflammation
  • insulin-like growth factor 1
  • macrophages

Introduction

Insulin-like growth factor -1 (IGF-1) is a pleiotropic factor that is produced and acts locally (ie, via autocrine or paracrine effects) or circulates in blood and exerts endocrine effects. Circulating IGF-1 levels reach a peak during the pubertal growth phase, eventually declining with the progression of age. Although its role in developmental and pubertal growth as the major mediator of the effects of growth hormone is well documented, the physiological roles of IGF-1 in aged subjects are not understood. Aging is a major independent risk factor for coronary heart disease; in fact, there is increased cardiovascular and coronary heart disease prevalence with age in both sexes. In light of the aging-dependent decline in circulating IGF-1 levels, a potential link between IGF-1 levels and the elevated prevalence of cardiovascular diseases has been suggested. Indeed, epidemiological data have suggested that low IGF-1 levels are an important predictor of coronary events in aged subjects.1–3 In an animal model of atherosclerosis, we have shown that low levels of circulating IGF-1 are associated with more atherosclerosis4 and, vice versa, that an increase in circulating IGF-1 decreases atherosclerotic burden.5

Clinical Perspective on p 2278

The pathogenesis of atherosclerosis is complicated, involving multiple cell types, including vascular endothelial cells, smooth muscle cells, and proinflammatory cells such as macrophages. To determine the potential target(s) whereby IGF-1 reduces atherosclerosis, we previously investigated whether smooth muscle–specific overexpression of IGF-1 alters atherosclerosis.6 Intriguingly, overexpression of IGF-1 in smooth muscle cells did not alter atherosclerotic burden6 but increased features of plaque stability,6,7 suggesting that other cellular components were a potential target for the antiatherogenic effects of IGF-1. Macrophages play a pivotal role in the pathogenesis of atherosclerosis by modulating inflammatory status and by scavenging and accumulating excess lipid to become foam cells. Thus, regulation of macrophage functions in terms of inflammation and phagocytosis is key to comprehending the disease process. Because macrophages have a predominant role in the inflammatory status of atherosclerotic lesions, it is critical to determine whether IGF-1 regulates macrophage function, particularly inflammatory activation and phagocytic activity. Thus far, there is very limited information on the potential link between IGF-1 effects and macrophage function, particularly in relation to vascular disease. IGF-1 has been reported to enhance chemotactic macrophage migration,8 to stimulate tumor necrosis factor-α (TNFα) expression,8 and to enhance low-density lipoprotein (LDL) uptake and cholesterol esterification.9 There are also reports from clinical investigations providing indirect evidence of anti-inflammatory effects of IGF-1. For example, there is an inverse relation between serum interleukin (IL)-6 and IGF-1 levels10; IGF-1/insulin-like growth factor–binding protein 3 administration to patients with severe burn injury induced an anti-inflammatory effect and reduced IL-6 and TNFα11,12; and low IGF-1 and high IL-6 and TNFα levels are associated with higher mortality in elderly patients.13,14 In this study, we used a well-established animal model of atherosclerosis to examine the role of macrophage IGF-1 signaling in atherosclerosis development and progression.

Methods

A detailed Materials and Methods section is available in the online-only Data Supplement.

Animals

All animal experiments were performed according to protocols approved by the institutional animal care and use committee. Monocyte/macrophage–targeted IGF-1 receptor–null mice on Apoe−/− background (MΦ-IGF1R-KO) and control mice (IGF1R-flox) were generated as described in the online-only Data Supplement. Eight-week old mice were fed a high-fat diet for 8 weeks before atherosclerosis was assessed.

Atherosclerotic Burden and Plaque Composition

Atherosclerotic burden was quantified with the use of en face preparations of whole aorta stained with Oil Red O and in cross sections of aortic root. Plaque composition was assessed in cross sections of aortic root by immunostaining for Mac-3 (macrophage) and α-smooth muscle actin (smooth muscle cell), and Masson’s Trichrome staining was used for collagen. Brachiocephalic artery lesions were analyzed by the Carstairs method as described by Gough et al.15

Tracing Recruitment of Circulating Monocytes

Circulating Ly6Chi or Ly6Clo monocytes were labeled in vivo with polychromatic red microspheres as described elsewhere,16 and numbers of red microsphere–positive cells were counted in a plaque and normalized to the efficiency of labeling in circulating monocytes (percent labeled/total number) and assessed by flow cytometry.

Intravital Fluorescence Microscopy

Intravital fluorescence microscopy was performed as described elsewhere.17

Macrophage Culture, Western Blot Analysis, and Quantitative Real-Time Reverse Transcription–Polymerase Chain Reaction

Thioglycolate-elicited macrophages were obtained from MΦ-IGF1R-KO and IGF1R-flox mice.18 Western blot analysis,19 total RNA extraction,20 and real-time reverse transcription–polymerase chain reaction20 were performed as previously described.

Statistical Analysis

All numeric data are expressed as mean±SEM. Statistical analyses were performed with GraphPad PRISM (version 6.07) software. Data sets were first assessed for residuals distribution with the D’Agostino-Pearson omnibus normality test and for equal variances with the Levene test for equality of variances. Differences in outcomes were determined by ANOVA and Bonferroni multiple-comparisons test, Kruskal– Wallis test, unpaired Student t test with or without the Welch correction, or Mann– Whitney U test, accordingly with the normality of residuals distribution. Differences were considered significant at P<0.05. The Fisher’s exact test was used to compare frequency of observed indexes of plaque vulnerability (Table).

View this table:
  • View inline
  • View popup
Table.

MΦ-IGF1R-KO-Induced Features of Ruptured Plaques in the Brachiocephalic Artery

Results

Generation of MΦ-IGF1R-KO/Apoe−/− Mice

MΦ-IGF1R-KO were generated on Apoe−/− background (LyzCre/+/Igf1rflox/flox/Apoe−/−: MΦ-IGF1R-KO) by crossing LyzCre/+ mice into Igf1rflox/flox/Apoe−/− (IGF1R-flox, served as a control). Genotype segregation in the offspring followed the expected mendelian frequency, and we did not recognize any developmental/morphological abnormalities. IGF1R deficiency was confirmed by lack of IGF1R protein detection by Western blot and IGF-1–dependent phosphorylation on Akt in peritoneal macrophages (Figure IA in the online-only Data Supplement). Consistent with myeloid-selective Cre expression,21–23 we found that the exon 3 of Igf1r was also excised in neutrophils (Figure IB in the online-only Data Supplement). However, we were unable to detect IGF1R protein in IGF1R-flox neutrophils, indicating extremely low levels of IGF1R protein expression in neutrophils (Figure IC in the online-only Data Supplement).

IGF1R and insulin receptor (InsR) can form a hybrid receptor (heterotetramer consisting of α+β subunits of IGF1R and α+β subunits of InsR),24–26 which binds IGF-1 with high affinity but not insulin.27,28 Depletion of IGF1R in endothelial cells is reported to allow InsR to form a holotetramer, thereby enhancing insulin sensitivity.29 Because insulin signaling in macrophages has significant effects on atherosclerosis (there are contradictory reports showing antiatherogenic30 or proatherogenic31 effects), we assessed insulin signaling activity in MΦ-IGF1R-KO macrophages. IGF1R deficiency did not alter InsR expression levels (Figure ID in the online-only Data Supplement). In IGF1R-flox macrophages, immunoprecipitation of InsR pulled down 100% of IGF1R (Figure IE, left, in the online-only Data Supplement), whereas anti-IGF1R immunoprecipitation pulled down 50% of InsR (Figure IE, right, in the online-only Data Supplement), suggesting that the ratio of IGF1R/InsR-hybrid receptors to InsR-holoreceptors was 2:1 without the presence of IGF1R-holoreceptors. IGF1R deficiency did not alter insulin-induced dose-dependent phosphorylation of Akt (Figure IF in the online-only Data Supplement).

Animals were assessed for circulating leukocyte counts (Table I in the online-only Data Supplement), cholesterol levels, and cytokine levels (Table II in the online-only Data Supplement). As has been reported,32 Western diet feeding for 8 weeks was associated with elevated monocyte count; however, IGF1R deficiency did not result in a significant difference in white blood cell count or circulating monocyte count (Table I in the online-only Data Supplement). A subpopulation of circulating monocytes as defined by CD11b+/CD90−/B220−/CD49b−/NK1.1−/Ly6G−/Ly6Chigh cells (Ly6Chi monocytes) has been reported to be proinflammatory and to be increased under atherogenic conditions such as hyperlipidemia.33 We did not observe a significant difference in Ly6Chi monocyte levels between MΦ-IGF1R-KO and IGF1R-flox mice (Table I in the online-only Data Supplement). MΦ-IGF1R-KO did not significantly alter circulating IGF-1, proinflammatory cytokine (IL-6, TNFα, monocyte chemoattractant protein-1 [MCP-1]), or cholesterol levels (Table II in the online-only Data Supplement).

Atherosclerosis Was Enhanced by MΦ-IGF1R-KO

Atherosclerotic lesion formation was assessed after 8 weeks of high-fat diet feeding. En face Oil Red O staining of aortas revealed a significant ≈64% increase in Oil Red O–positive lesion area, and there was a consistent ≈34% increase in plaque size at the aortic root in MΦ-IGF1R-KO mice (Figure 1). The effect was confirmed in each sex, indicating that there was no sex-specific effect of IGF1R deficiency (Figure II in the online-only Data Supplement). Plaque composition with regard to Mac3 (macrophage)-positive cells, α-smooth muscle actin–positive cells, and collagen content (Masson’s trichrome stain) was significantly altered by MΦ-IGF1R-KO (Figure 2); there was a 49% increase in Mac3 detection (Figure 2A) and a 31% decrease in α-smooth muscle actin detection (Figure 2B). Plaques in MΦ-IGF1R-KO mice had a thinner smooth muscle cap than IGF1R-flox mice (Figure 2B) and decreased collagen content (Figure 2C). Because we observed a decrease in collagen content in plaques from MΦ-IGF1R-KO mice, we hypothesized that this could be attributable to enhanced collagen degradation by matrix metalloproteinases (MMPs). Cultured peritoneal macrophages from MΦ-IGF1R-KO animals expressed higher levels of MMP-1, -2, -8, -9, -12, -13, and -14 than those from IGF1R-flox animals, suggesting enhanced MMP activity (Figure 2D). The MMP protein levels in tissue lysates of ascending aortas after 2 months of Western diet feeding (Figure IIIA and IIIB in the online-only Data Supplement) showed a significant increase in the expression of MMP-1, -2, -8, and -9 in MΦ-IGF1R-KO animals. These observations are consistent with enhanced lesion formation in MΦ-IGF1R-KO mice with a phenotypic shift toward increased features of plaque vulnerability.

Figure 1.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 1.

Monocyte/macrophage insulin-like growth factor-1 receptor (IGF1R) deficiency aggravated atherosclerosis. Atherosclerotic lesion formation was assessed in monocyte/macrophage–specific (MΦ)–IGF1R knockout (KO) and IGF1R-flox mice (control) fed a high-fat diet for 2 months. A, Oil Red O staining of en face aortas; plaque covered area was determined and expressed as percent of total area (C; **P<0.0001 by the Mann–Whitney U test; n=25 for IGF1R-flox and n=23 for MΦ-IGF1R-KO). B, Plaque size was determined in hematoxylin and eosin–stained cross sections of aortic root; *P<0.05 by the Mann–Whitney U test; n=18 for IGF1R-flox and n=19 for MΦ-IGF1R-KO (D).

Figure 2.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 2.

Monocyte/macrophage insulin-like growth factor-1 receptor (IGF1R) deficiency alters atherosclerotic plaque composition. Atherosclerotic plaque composition was assessed in cross sections of aortic root. A, Macrophage content was assessed by immunostaining against Mac3 antigen. *P<0.05 by the Student t test; n=18 for IGF1R-flox and n=17 for monocyte/macrophage–specific (MΦ)–IGF1R knockout (KO). B, Smooth muscle cell (SMC) content assessed as α-smooth muscle actin (αSMA)–positive cells by immunostaining (representative images shown in top left). *P<0.05 by the Student t test; n=16 for IGF1R-flox and n=18 for MΦ-IGF1R-KO. Thickness of SMC-positive fibrous cap was also determined by immunostaining against αSMA (representative images shown in bottom left). **P<0.001 by the Student t test; n=12 for IGF1R-flox and n=14 for MΦ-IGF1R-KO. C, Collagen content determined by Masson’s trichrome stain. **P<0.01 by the Mann–Whitney U test; n=17 for IGF1R-flox and n=16 for MΦ-IGF1R-KO. D, Western blot analysis of matrix metalloproteinases (MMPs) in cultured peritoneal macrophages isolated from IGF1R-flox (Flox) and MΦ-IGF1R-KO (Cre+) mice. Representative results of 3 independent experiments are shown.

Plaque Destabilization in MΦ-IGF1R-KO Mice

Because our findings suggested plaque destabilization in MΦ-IGF1R-KO mice, we assessed indicators of vulnerability in brachiocephalic artery plaques (Figure IV in the online-only Data Supplement) by staining cross sections of brachiocephalic artery with the Carstairs34 and Verhoeff–Van Gieson methods.35 Cross sections (5 μm thick) were made every 50 μm along the artery, for a total of 10 sections per artery. If 3 consecutive sections indicated a fibrous cap disruption, intraplaque hemorrhage, fibrin deposition, or medial elastin break, the artery was considered positive for signs of plaque vulnerability. After 8 weeks on a high fat diet, MΦ-IGF1R-KO animals had increased features of vulnerable plaques compared with IGF1R-flox animals, as determined by the presence of intraplaque hemorrhage (IGF1R-flox, 3 positive in 22 animals versus MΦ-IGF1R-KO, 8 positive in 17 animals; P=0.03; Table) and medial elastin breaks (IGF1R-flox, no positive in 22 animals versus MΦ-IGF1R-KO, 4 positive in 17 animals; P=0.03; Table), suggesting that plaques in MΦ-IGF1R-KO mice are more unstable than in IGF1R-flox mice.

Proinflammatory Responses Are Enhanced in IGF1R-Deficient Macrophages

Thioglycolate-elicited peritoneal macrophages from the MΦ-IGF1R-KO mice were assessed for proinflammatory cytokine and chemokine secretion (Figure 3). Among tested cytokines and chemokines, IL-1β production was below detectable levels, whereas IL-1α, IL-6, TNFα, MCP-1, and fractalkine were detected (Figure 3A–3F). IGF1R-deficient macrophages secreted significantly higher levels of all the tested cytokines and chemokines except fractalkine, which showed a strong trend toward an increase (P=0.0508). Interferon-γ (IFNγ) significantly enhanced TNFα (Figure 3C) and MCP-1 (Figure 3D) production in IGF1R-flox cells but not in MΦ-IGF1R-KO cells. Intriguingly, IFNγ did not enhance fractalkine production in IGF1R-flox macrophages, whereas in MΦ-IGF1R-KO cells, it decreased fractalkine production (Figure 3E). In a setting of exposure to IFNγ, MΦ-IGF1R-KO cells secreted higher levels of IL-1α, IL-6, and MCP-1 compared with IGF1R-flox. We tested nuclear factor-κB (NFκB) DNA binding activity in these cells (Figure 3F). There was significantly higher NFκB DNA binding activity in MΦ-IGF1R-KO, which was further enhanced by IFNγ, consistent with higher cytokine/chemokine production. To further assess NFκB involvement, we exposed cells to NFκB inhibitors (BMS-345541 and parthenolide, Figure 3G–3J). BMS-345541 completely abolished enhanced TNFα and IL-6 production in MΦ-IGF1R-KO cells (Figure 3G and 3H), and parthenolide recapitulated the BMS-345541 effect (Figure 3I and 3J), supporting the importance of NFκB-dependent cytokine production in MΦ-IGF1R-KO macrophages.

Figure 3.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 3.

Proinflammatory cytokine and chemokine production by cultured macrophages isolated from insulin-like growth factor-1 receptor (IGF1R)–flox and monocyte/macrophage–specific (MΦ)–IGF1R knockout (KO) mice. Interleukin (IL)-1α (A), IL-6 (B), tumor necrosis factor-α (TNFα; C), monocyte chemoattractant protein-1 (MCP-1; D), and fractalkine (E) secretion by peritoneal macrophages was assessed by respective ELISA. Macrophages were isolated by peritoneal lavage from IGF1R-flox (open column) and MΦ-IGF1R-KO (closed column) mice and allowed to adhere on culture plates overnight. After removal of nonadhering cells, cells were primed by interferon-γ (IFNγ), and conditioned media were collected after 24 hours. *P<0.05, **P<0.01 vs IGF1R-flox; ##P<0.01 vs IGF1R-flox+IFNγ; $P<0.01 vs MΦ-IGF1R-KO. Statistical significance was assessed by 2-way ANOVA and subsequent post hoc analysis with the Bonferroni multiple-comparisons test; n=6. F, Nuclear factor-κB (NFκB) DNA binding activity was assessed (expressed as relative light units [RLU]) in cell lysates of IGF1R-flox and MΦ-IGF1R-KO macrophages with or without priming by IFNγ for 3 hours. *P<0.05 vs IGF1R-flox by 2-way ANOVA; n=3. G through J, Effects of NFκB inhibitors on TNFα (G and H) and IL-6 (I and J) secretion by peritoneal macrophages. IGF1R-flox (open column) and MΦ-IGF1R-KO (closed column) macrophages were exposed to the indicated dose of BMS-345541 or parthenolide for 1 hour before activation by IFNγ or lipopolysaccharide (LPS). Conditioned media were collected after 24 hours and assessed for cytokine concentration with the respective ELISA kit (R&D Systems). ##P<0.05 vs IGF1R-flox+IFNγ, #P<0.05 vs IGF1R-flox+LPS by the Mann–Whitney U test; n=4.

Macrophage Polarization

Because IGF1R-deficient macrophages manifested enhanced proinflammatory responses, we examined whether IGF1R deficiency influenced macrophage polarization. Classic activation (M1) was induced by exposure to IFNγ and subsequently to lipopolysaccharide (Figure 4A–4E). M1 marker gene (Tnf, Nos2, Il6, Ccl2, and Ccl5) expression levels were highly induced by exposure to IFNγ-LPS, and IGF1R deficiency further enhanced the expression, implying enhanced M1 activation in MΦ-IGF1R-KO macrophages. On the other hand, IL-4–induced gene expression levels (ie, M2 activation markers; Arg1, Mrc1, and Pparg) were not influenced by IGF1R deficiency (Figure 4F–4H). It has been reported that oxidatively modified LDL (oxLDL) and that oxidized lipid components of oxLDL alter macrophage activation status or induce a distinctive activation status.36–39 Hemeoxygenase-1 (Hmox1) and thioredoxin reductase-1 (Txnrd1) are signature genes that have been shown to be upregulated in macrophages exposed to oxidized phospholipid, leading to a polarization status of Mox.39 In fact, oxLDL exposure enhanced Hmox1 and Txnrd1 expression significantly (Figure 5A and 5B). Intriguingly, IGF1R deficiency did not influence Txnrd1 mRNA levels (Figure 5B) but significantly upregulated Hmox1 mRNA levels (Figure 5A). We further assessed whether IGF1R deficiency influences the effect of oxLDL on expression levels of macrophage activation marker genes (Figure 5). With regard to M1 activation markers, oxLDL by itself did not alter mRNA levels in IGF1R-flox cells (Tnf, Nos2, Il6, Ccl2, and Ccl5: Figure 5C–5G); however, in MΦ-IGF1R-KO cells, in which these mRNA levels were significantly elevated compared with IGF1R-flox cells, oxLDL significantly further upregulated Tnf, Nos2, Il6, and Ccl2 mRNA levels (Figure 5C–5F), whereas Ccl5 mRNA was suppressed (Figure 5G). On the other hand, of the M2 activation markers, Mrc1 mRNA levels were moderately upregulated by oxLDL (Figure 5I), whereas Arg1 and Pparg were not affected by oxLDL (Figure 5H and 5J). Although there were modestly lower Arg1 mRNA levels in IL-4/oxLDL–treated MΦ-IGF1R-KO cells than in IGF1R-flox cells, overall, oxLDL or IGF1R deficiency did not robustly alter M2 marker levels. These data suggested that IGF1R deficiency influenced macrophage polarization, namely enhancing the proinflammatory M1 phenotype as evoked by IFNγ and oxLDL stimulation; however, the effect is not entirely classic activation, as implicated by Ccl5 downregulation (Figure 5G). These results prompted us to examine whether Mox marker gene expression was altered by IGF1R deficiency in IFNγ-primed macrophages. IFNγ suppressed Hmox1 and Txnrd1 mRNA levels in IGF1R-flox macrophages (Figure 5A and 5B); however, in MΦ-IGF1R-KO cells, IFNγ exerted the opposite effect, leading to an upregulation of Hmox1 mRNA (Figure 5A). IFNγ suppressed Txnrd1 mRNA levels in IGF1R-deficient macrophages but to a lesser extent than in IGF1R-flox cells, resulting in higher expression levels compared with IGF1R-flox cells (Figure 5B).

Figure 4.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 4.

mRNA expression levels representing classic activation (M1) are elevated in monocyte/macrophage–specific–insulin-like growth factor-1 receptor knockout (MΦ IGF1R-KO) macrophages. Classic activation marker (Tnf, A; Nos2, B; Il6, C; Ccl2, D; Ccl5, E) and alternative activation marker (Arg1, F; Mrc1, G; Pparg, H) mRNA levels were assessed by quantitative reverse transcription–polymerase chain reaction. After isolation and adhesion to culture plates, macrophages remained untreated (NA) or were exposed to interferon-γ (IFNγ) and lipopolysaccharide (LPS) to induce classic (ie, M1) activation or exposed to interleukin (IL)-4 to induce alternative (ie, M2) activation (IGF1R-flox, open column; MΦ-IGF1R-KO, closed column). Representative data from 4 independent experiments are shown. **P<0.01 or *P<0.05 vs IGF1R-flox by 2-way ANOVA and Bonferroni multiple-comparisons test, n=3.

Figure 5.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 5.

Insulin-like growth factor-1 receptor (IGF1R) deficiency skews macrophage activation induced by oxidized low-density lipoprotein (oxLDL). Macrophages isolated from IGF1R-flox (open column) and monocyte/macrophage–specific (MΦ)–IGF1R knockout (KO; closed column) mice were primed with interferon-γ (IFNγ) for 6 hours and exposed to oxLDL for 18 hours. Total RNA was extracted and subjected to quantitative reverse transcription–polymerase chain reaction, testing oxidized lipid-induced activation (Mox) marker (Hmox1, A; Txnrd1, B), M1 activation marker (Tnf, C; Nos2, D; Il6, E; Ccl2, F; Ccl5, G), and M2 activation marker (Arg1, H; Mrc1, I; Pparg, J) mRNA levels. Representative data from 3 independent experiments are shown. ##P<0.01 vs respective condition without oxLDL, $P<0.01 vs IGF1R-flox of the respective condition, ¶P<0.01 vs respective condition without IFNγ, **P<0.01 vs IFNγ-treated MΦ-IGF1R-KO, #P<0.05 vs IL-4– and oxLDL-treated IGF1R-flox, *P<0.05 vs respective condition without oxLDL by 2-way ANOVA and Bonferroni multiple-comparisons test; n=3.

Efferocytosis, a process by which apoptotic cells are removed by phagocytosis, is considered a significant mechanism involved in the resolution of inflammation. We evaluated expression levels of genes involved in efferocytosis (Figure V in the online-only Data Supplement). M1 activation suppressed the expression of efferocytosis-related genes (Anxa1, Gas6, C1qa, and Mfge8), and M2 activation had no effect (Anxa1, C1qa, Mertk; Figure V in the online-only Data Supplement) or downregulated (Gas6, Mfge8) gene expression levels. IGF1R deficiency did not alter these mRNA levels in either activated or nonactivated cells. OxLDL by itself did not induce obvious effects except for an upregulation of Mertk mRNA levels (Figure VD in the online-only Data Supplement). IGF1R deficiency upregulated Anxa1 and Mfge8 in the presence of oxLDL (Figure VA and VE in the online-only Data Supplement), but it caused a trend to downregulation of C1qa (Figure VC in the online-only Data Supplement). Thus, IGF1R deficiency did not induce changes suggesting enhanced/reduced efferocytosis.

Lipid Internalization and Efflux

Macrophage internalization and accumulation of modified LDL lead to foam cell formation, which is a hallmark of atheroma formation. Macrophage lipid internalization was assessed by exposing cells to oxLDL and acetylated LDL (Figure 6A and 6B). Macrophages were exposed to IFNγ/lipopolysaccharide (M1 activation) or IL-4 (M2 activation) and then tested for lipid incorporation after exposure to modified LDL for 48 hours. M1 cells internalized far smaller amounts of lipid compared with the nonactivated cells or M2 cells (Figure 6A and 6B), whereas M2-activated cells incorporated more lipid than untreated cells when they were exposed to acetylated LDL (Figure 6A). IGF1R deficiency did not alter lipid incorporation (Figure 6A and 6B), regardless of activation status and exposure to acetylated LDL or oxLDL.

Figure 6.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 6.

Cholesterol efflux from macrophages was adversely altered by insulin-like growth factor-1 receptor (IGF1R) deficiency. A and B, Peritoneal macrophages were incubated with interferon-γ+lipopolysaccharide (IFNγ+LPS) to induce M1 and interleukin (IL)-4 to induce M2 activation status, subsequently exposed to acetylated low-density lipoprotein (acLDL; A) or oxidized LDL (oxLDL; B) for 48 hours, and assessed for lipid uptake by Oil Red O stain. Fluorescent images of stained cells were assessed with ImagePro software. Oil Red O staining intensity was normalized to cell number and expressed as relative fluorescent unit (RFU)/ cell number. C and D, Peritoneal macrophages were not activated (NA) or were activated to M1 and M2 and then loaded with lipid by exposure to acLDL and 3H-cholesterol for 24 hours. Subsequently, apolipoprotein AI (ApoAI)–dependent (C) or high-density lipoprotein (HDL)–dependent (D) efflux of 3H-cholesterol for the subsequent 24 hours were assessed. IGF1R-flox, open column; monocyte/macrophage–specific (MΦ)–IGF1R knockout (KO), closed column. Statistical significance was assessed by Mann–Whitney U test; n=8.

Cholesterol efflux from cells to extracellular lipid acceptors also contributes to lipid accumulation and thus foam cell formation. Thus, activated and lipid-laden macrophages were tested for cholesterol efflux activity (Figure 6C and 6D). IGF1R deficiency markedly reduced apolipoprotein AI–dependent cholesterol efflux in M1-activated cells (Figure 6C). We measured HDL-dependent cholesterol efflux in nonactivated, M1-activated, and M2-activated cells (Figure 6D) and found that IGF1R deficiency caused a small but significant reduction in cholesterol efflux to HDL (Figure 6D). To gain insights into potential mechanisms, we assessed expression levels of ABCA1, ABCG1, and SRB1, which are major lipid transporters responsible for cholesterol efflux. They were differentially regulated by IGF1R deficiency (Figure 7); ABCG1 expression levels were downregulated by ≈50% in IGF1R-deficient macrophages, regardless of activation status (Figure 7B and 7D), whereas ABCA1 and SRB1 were not regulated. To investigate ABCG1 expression in MΦ-IGF1R-KO cells, we exposed the cells to ligands of LXR, which is a major regulator of ABCG1 gene expression.40 LXR agonist GW3965 or T0901317 induced ABCG1 and ABCA1 expression in IGF1R-flox macrophages but not in MΦ-IGF1R-KO cells (Figure 7E). Thus, LXR-dependent regulation of ABCG1 expression is compromised by IGF1R deficiency, potentially accounting for downregulation of ABCG1 in MΦ-IGF1R-KO cells.

Figure 7.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 7.

Insulin-like growth factor-1 receptor (IGF1R) deficiency downregulated ABCG1 expression levels in acetylated low-density lipoprotein (acLDL)–loaded macrophages. Peritoneal macrophages were not activated (NA) or were activated to M1 and M2 and then exposed to acLDL for 24 hours. ABCA1 (A), ABCG1 (B), and SRB1 (C) expression levels were assessed by Western blot analysis (representative measurements and blots [D] from 3 independent experiments are shown). IGF1R-flox, open column; monocyte/macrophage–specific (MΦ)–IGF1R knockout (KO), closed column. *P<0.05 vs IGF1R-flox cells by 2-way ANOVA. E, Western blot analysis for ABCG1 expression, assessing effects of LXR agonists. AcLDL-loaded macrophages were exposed to LXR agonists (1 µmol/L GW3965, 1 µmol/L T0901317) for 48 hours and assessed for ABCG1 expression levels. Representative results from 3 independent experiments are shown. *P<0.05 vs IGF1R-flox, **P<0.01 vs IGF1R-flox by the Student t test; n=3.

Monocyte Recruitment, Macrophage Proliferation, and Apoptosis in MΦ-IGF1R-KO Mice

Because MΦ-IGF1R-KO caused an increase in Mac3 positivity within plaques, we evaluated monocyte recruitment to lesions, macrophage proliferation,41 and macrophage apoptosis, 3 major determinants of macrophage number in lesions, in plaques.42 To evaluate monocyte recruitment, we labeled circulating monocytes in vivo by intravenous administration of fluorescent microspheres.43 The microspheres are not capable of penetrating into tissue interstitial space; thus, red fluorescence–positive cells (identified by DAPI positivity) within plaque are considered recruited and infiltrated cells. We evaluated red microsphere labeling by flow cytometry. As described previously,16 intravenous injections of red microspheres labeled the Ly6Clo population among circulating monocytes (Figure VIA in the online-only Data Supplement), whereas clodronate administration 1 day before the injection of red microspheres introduced the label to Ly6Chi monocytes (Figure VIB in the online-only Data Supplement). As shown in Figure 8, we detected a significantly larger number of red fluorescence–positive cells in plaques from MΦ-IGF1R-KO mice both without (Figure 8C) and with (Figure 8D) clodronate administration. It is noteworthy that more red microsphere–positive cells are detected after clodronate administration, indicating that Ly6Chi monocytes are the dominant subpopulation to be recruited to lesions.33 To assess leukocyte adhesion and rolling on the luminal side of the endothelium in vivo, we performed intravital microscopy to detect CD11b-positive cells in the mesenteric circulation. MΦ-IGF1R-KO significantly increased CD11b+ leukocyte adhesion (Figure 8E) and rolling (Figure 8F) on the luminal surface of the endothelium, consistent with increased recruitment of CD11b+ cells, that is, monocytes and neutrophils. Because IGF1R expression was undetectable in neutrophils (Figure I in the online-only Data Supplement), it is unlikely that IGF1R gene deletion in neutrophils contributed significantly to these findings.

Figure 8.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 8.

Monocyte/macrophage insulin-like growth factor-1 receptor (MΦ-IGF1R) deficiency increased monocyte recruitment and infiltration into atherosclerotic plaques. MΦ-IGF1R knockout (KO) and IGF1R-flox mice were fed a high-fat diet for 12 weeks. Red microspheres were administered via intravenous injection at 3 and 7 days before death (C) or clodronate liposomes were administered intravenously 1 day before the red microsphere administration to introduce label to the Ly6Chi monocyte population (D). Serial frozen sections were obtained at aortic root and stained with Oil Red O (A) or DAPI (B). Images (B) are magnified to view the corresponding serial sections of (A) at specified areas (rectangle). L indicates lumen; blue shows the DAPI-stained nuclei. Arrows indicate red microsphere–positive cells within the lesion. The number of red microsphere–positive cells was counted per plaque (C, without clodronate; D, with clodronate), and results are shown by box-and-whiskers plot in which a box extends from the 25th to 75th percentiles with the median indicated by a horizontal line and whiskers indicating the minimum and maximum. *P<0.05, **P<0.01 by Mann–Whitney U test; n=6 for IGF1R-flox and n=5 for MΦ-IGF1R-KO. E and F, Intravital microscopy assessment of CD11b+ cell adhesion (E) and rolling (F) on the luminal surface of the mesenteric circulation endothelium. Results are shown by box-and-whiskers plot as described above. *P<0.05 and **P<0.01 by the Mann–Whitney U test; n=4 for MΦ-IGF1R-KO and n=6 for IGF1R-flox.

To assess proliferation activity of macrophages in plaques, we assessed Ki67 and proliferating cell nuclear antigen gene expression levels44–46 (Figure VIC in the online-only Data Supplement). Macrophage Marker (SC-101447; a monoclonal antibody raised against isolated macrophages of mouse origin; Santa Cruz)47,48 –positive plaque area was laser dissected from aortic root for RNA isolation, followed by quantitative reverse transcription–polymerase chain reaction. Equal amplification of CD68 was confirmed between IGF1R-flox and MΦ-IGF1R-KO lesions. Macrophage-rich regions from MΦ-IGF1R-KO plaques expressed the same levels of Ki67 or proliferating cell nuclear antigen compared with IGF1R-flox (Figure VIC in the online-only Data Supplement), suggesting no difference in macrophage proliferation in plaques. Terminal deoxynucleotidyl transferase dUTP nick-end labeling/Mac3 double-positive cell numbers were not different (Figure VID in the online-only Data Supplement). Taken together, our data suggest that IGF1R deficiency enhances monocyte recruitment to lesions, thereby increasing the macrophage cell population.

OxLDL Downregulates IGF1R in Macrophages

OxLDL plays a critical role in atherogenesis, and we have previously shown that oxLDL downregulates IGF1R in vascular smooth muscle cells.49 To determine potential regulation of macrophage IGF1R expression by oxLDL, we used the human monocytic cell line THP-1, which was differentiated into macrophages and exposed to 100 µg/mL oxLDL (Figure VII in the online-only Data Supplement). OxLDL exposure for 24 hours downregulated IGF1R levels by 80%.

Discussion

IGF-1 production has been documented in macrophages8,9,50,51; however, precise effects of IGF-1 in macrophages in relation to the pathogenesis of atherosclerosis have not been elucidated. In this study, we generated macrophage/monocyte–specific IGF1R-deficient mice and discovered pivotal roles of IGF-1 in the regulation of inflammatory responses and lipid handling in macrophages, which are relevant to the antiatherogenic effects of IGF-1. Our data indicate that IGF1R deficiency in macrophages enhances proinflammatory activation (ie, M1 polarization), thereby promoting proinflammatory cytokine production (Figures 3 and 4), and enhances lipid accumulation as a result of reduced efflux (Figures 6 and 7). These phenotypic changes resulted in increased recruitment of macrophages to atherosclerotic plaques and increased atherosclerotic burden in MΦ-IGF1R-KO mice (Figures 1 and 2). Moreover, histological evaluation of brachiocephalic arteries indicated that the MΦ-IGF1R-KO induced features of unstable plaques (Table).

There is growing interest in the role of IGF-1 in cardiovascular disease. Low circulating IGF-1 levels have been associated with cardiovascular disease risk factors,52–56 and in particular, there is growing evidence for a role for IGF-1 deficiency in the pathogenesis of metabolic syndrome.57 Acromegaly (ie, excessive growth hormone and IGF-1) or otherwise growth hormone and IGF-1 deficiency have been linked to cardiovascular complications.58,59 However, epidemiological studies linking IGF-1 with cardiovascular disease report mixed results. Some cross-sectional and prospective studies60–65 suggest a positive association between IGF-1 (and in some cases insulin-like growth factor–binding protein 3) and atherosclerosis, but others have found that low IGF-1 is a predictor of ischemic heart disease and mortality, consistent with the potential anti-apoptotic, antioxidant, and plaque stabilization effects of IGF-1.1–3,13,66–72 Methodological constraints could explain these contradictions because measurement of total IGF-1 levels represents only a crude estimate of the biologically active IGF-1. Thus, an IGF-1–specific kinase receptor activation assay may better reflect IGF-1 bioactivity.73 In fact, it has been reported that higher IGF-1 bioactivity is associated with significantly longer survival in subjects with a high inflammatory risk profile or history of cardiovascular disease.74 Additionally, polymorphisms in the IGF-1 gene promoter region that influence circulating IGF-1 levels have been reported.75–77 The alleles that indicate lower circulating IGF-1 levels have been associated with increased risk for type 2 diabetes mellitus, myocardial infarction, left ventricular hypertrophy, higher carotid intima-media thickness, higher aortic pulse-wave velocity, and lower endothelium-dependent vasodilation.75–77 Intriguingly, IGF-1 resistance in the endothelium was reported in an animal model of obesity, thereby blunting the vasodilatory response to IGF-1 via attenuated endothelial nitric oxide synthase phosphorylation and nitric oxide production.78 Our present finding that reduced macrophage IGF-1 signaling is highly proinflammatory and increases atherosclerotic burden is consistent with the growing evidence that decreased IGF-1 action may be a significant contributor to the pathogenesis of atherosclerosis. Of note, we found that oxLDL downregulates IGF1R levels in human-derived THP1 macrophage (Figure VII in the online-only Data Supplement), which is consistent with our previous reports that both IGF-1 and IGF1R expression were significantly lower in human atherosclerotic plaque intimal regions with macrophage infiltration, where oxLDL was highly detected.79,80

We used MΦ-IGF1R-KO mice, which have 1 allele of Lys2 ablated; the IGF1R-flox mice (control) have both alleles intact. Previous studies in LysCre/+ mice with regard to potential alterations in monocyte/macrophage biology caused by the hemizygous deficiency of Lys2 showed no evidence of a heterozygous phenotype.21 In addition, complete ablation of both alleles of Lys2 does not influence atherosclerosis in Apoe−/− mice.81 Lys2-cre–mediated gene excision occurs in monocytes and neutrophils.21–23 We confirmed deletion of exon 3 of the Igf1r gene in both macrophages and neutrophils isolated from the MΦ-IGF1R-KO mouse, but we were unable to detect IGF1R protein even in Igf1r-normal neutrophils (ie, IGF1R-flox neutrophils), indicating extremely low expression levels. Thus, although contribution of neutrophils to the phenotype of MΦ-IGF1R-KO mice cannot be excluded, it appears much more likely that macrophages, which express significant levels of IGF1R, play the predominant role.

IGF1R and InsR are structurally similar and form a heteromeric hybrid receptor consisting of α+β subunits of IGF1R and α+β subunits of InsR. The hybrid receptor binds IGF-1 with high affinity but does not bind insulin at physiological ranges. To the best of our knowledge, this is the first study evaluating the expression ratio between IGF1R and InsR in macrophages, showing predominant expression of InsR. Thus, IGF1R-flox macrophages (expressing both IGF1R and InsR) responded to a physiological dose of insulin (Figure I in the online-only Data Supplement). Intriguingly, IGF1R deficiency in macrophages, although it should free up InsR hemidimers to form holoreceptors, did not increase insulin signaling. This finding is relevant because macrophage InsR deficiency has been shown to modulate atherosclerosis development (although results have been contradictory30,31). Because we found that MΦ-IGF1R-KO did not alter insulin signaling in macrophages, it is unlikely that changes in insulin action on macrophages plays a significant role in the phenotype of MΦ-IGF1R-KO mice.

Macrophages become activated as they infiltrate into a target tissue and are exposed to stimuli, expressing a highly proinflammatory phenotype or a less inflammatory but phagocytic and antigen-presenting phenotype. The former status was called classic activation or M1 activation; the latter was referred to as alternative activation or M2 activation. Recent investigations indicate, however, that macrophage activation status likely encompasses a broad spectrum in which M1 and M2 activation are likely 2 extremes.82–84 Recognizing that macrophage activation represents a continuum, we tested M1 and M2 activation, as well as Mox activation, which was described as a unique activation status found in plaque macrophages,39 to provide insights into the effects of IGF1R deficiency. Our results indicate that IGF1R deficiency resulted in macrophages acquiring a highly inflammatory (ie, M1) phenotype (Figure 3), whereas M2 marker gene expression levels were not altered (Figure 4F–4H). However, MΦ-IGF1R-KO macrophages are not simply skewed to a more inflammatory phenotype; these cells were also shifted to a phenotype induced by oxLDL, characterized by upregulation of the antioxidant genes Hmox1 and Txnrd1, described as Mox activation39 (Figure 5A and 5B). Reports of the effects of oxLDL and its specific lipid moieties on macrophage activation are variable and include enhancement of the inflammatory phenotype,36,38,85 induction of M2 activation,86 or a unique activation status.39 In summary, our results suggest that IGF1R deficiency skews macrophages to a unique activation state that can be characterized by enhanced proinflammatory response and elevated antioxidant system.

The effect of IGF1R deficiency on macrophage activation status prompted us to examine how IGF1R deficiency influences uptake of modified LDL, cholesterol efflux, and efferocytosis. It has been shown that M1-activated macrophages demonstrate reduced foam cell formation in response to oxLDL6,87 and that Mox-activated macrophages have attenuated phagocytosis and efferocytosis.39 It would thus have been reasonable to speculate that IGF1R deficiency, which enhances the proinflammatory phenotype and Mox marker expression levels, could lead to reduced foam cell formation. However, our results showed no evidence of altered acetylated LDL or oxLDL uptake by IGF1R-deficient macrophages (Figure 6A and 6B). We also assessed expression levels of genes that are functional in efferocytosis, but there was no apparent alteration caused by IGF1R deficiency (Figure V in the online-only Data Supplement). Thus, despite enhanced inflammatory responses and redox gene expression, IGF1R-deficient macrophages do not appear to have impairment in modified lipid uptake or efferocytosis. However, further assessment of lipid handling by IGF1R-deficient macrophages indicated that cholesterol efflux is impaired by IGF1R deficiency (Figure 6C and 6D), potentially promoting foam cell formation. Cholesterol efflux is mediated by the lipid transporters ABCA1, ABCG1, and SRB1.88 IGF1R deficiency caused lower expression levels of ABCG1 (Figure 7), which should, at least in part, account for lowered cholesterol efflux. Intriguingly, the LXR-dependent regulation of ABCG1 expression was compromised in MΦ-IGF1R-KO macrophages. LXR is a major regulator of ABCG1 expression.89,90 In fact, LXR drives ABCG1 expression on lipid loading by modified LDL (which causes accumulation of oxysterols).89,90 Thus, impaired LXR regulation of ABCG1 expression could be an important mechanism underlying lowered cholesterol efflux in MΦ-IGF1R-KO macrophages.

MΦ-IGF1R-KO increased atherosclerosis burden as assessed by Oil Red O staining of en face aortas and by histological analysis of aortic root cross sections (Figure 1). These mice had an increase in lesional macrophages and in recruitment of monocytes to atherosclerosis plaques (Figure 8). Consistent with the increased recruitment of monocytes, chemokine expression levels were upregulated in MΦ-IGF1R-KO macrophages (ie, MCP-1 and fractalkine; Figures 3–5). Moreover, MΦ-IGF1R-KO increased features of plaque vulnerability, as evidenced by histological features of intraplaque hemorrhage and medial elastin breaks in lesions from brachiocephalic artery (Figure IV in the online-only Data Supplement and Table). These findings were in accordance with the changes in plaque composition, notably the increased population of macrophages, the reduced population of smooth muscle cells particularly within the plaque cap, and decreased collagen (Figure 2). To obtain insights into the underlying mechanisms, we assessed MMP levels in peritoneal macrophages from MΦ-IGF1R-KO mice and showed significant upregulation of MMPs (Figure 2D), which have previously been shown to be relevant to atherogenesis and plaque vulnerability.91,92 In addition, we found significant upregulation of MMP-1, -2, -8, and -9 in lysates of ascending aorta from MΦ-IGF1R-KO animals, consistent with data obtained with peritoneal macrophages. A major source of collagen matrix synthesis and deposition in atherosclerotic plaques is smooth muscle cells, and thus, one can speculate that the enhanced inflammatory milieu induced by MΦ-IGF1R-KO altered smooth muscle cell homeostasis. In fact, it is noteworthy that MΦ-IGF1R-KO robustly enhanced macrophage production of proinflammatory cytokines such as IL-1α, TNFα, and IL-6 (Figure 3), and it has been reported that cytokines such as TNFα reduce IGF-1 and increase IGF binding protein-3 in vascular smooth muscle cells, leading to a reduction in bioactive IGF-1.93 IGF-1 positively regulates collagen synthesis by smooth muscle cells.94 Indeed, we have recently shown that increased IGF-1 signaling in vascular smooth muscle cells increases features of plaque stability, as determined by increased fibrous cap area, α-smooth muscle actin–positive smooth muscle cells, and collagen content, without affecting plaque burden,6 potentially mediated by IGF-1 induction of smooth muscle differentiation6 and collagen synthesis.94 Thus, it seems reasonable to speculate that increased production of inflammatory cytokines in MΦ-IGF1R-KO mice disrupts normal IGF-1 signaling in smooth muscle cells and reduces collagen deposition in plaques of MΦ-IGF1R-KO mice. Taken together with our previous results, the present study provides insights into potential interactions between macrophages and smooth muscle cells; that is, the enhanced inflammatory milieu by macrophages suppresses IGF-1 signaling in smooth muscle cells, leading to a reduction of plaque stability.

Our results are consistent with the growing body of experimental evidence that IGF1 has antiatherogenic effects. Infusion of IGF-1 in Apoe−/− mice reduces atherosclerotic burden,5 and overexpression of IGF-1 in vascular smooth muscle cells increases plaque collagen content and smooth muscle cell levels and reduces necrotic core size.6 IGF-1 has been shown to have antioxidant effects on the endothelium via upregulation of glutathione peroxidase levels.95 Our current findings demonstrate that IGF-1 signaling has a major effect on macrophage biology that is critical for atherogenesis. However, a limitation of experimental studies to date has been that they have been performed in murine models and that studies in larger animals phylogenetically closer to humans are lacking. Such studies will be important for the development of IGF-1–based therapeutic strategies.

Conclusions

We have shown that IGF1R deficiency in macrophages of Apoe−/− mice increases atherosclerotic burden and changes plaque composition to one of lowered smooth muscle cell and collagen content. Our data suggest that the loss of IGF-1 signaling skews macrophage activation to a proinflammatory status and promotes lipid accumulation in macrophages by lowering lipid efflux. There is increasing evidence linking low IGF-1 to multiple cardiovascular risk factors, including metabolic syndrome and aging.57,96 In fact, it has been reported that decreased IGF-1 bioavailability is an adverse prognostic factor for coronary heart disease (reviewed elsewhere97,98). Our findings herein suggesting that IGF-1 regulates macrophage inflammatory responses and lipid metabolism may provide the basis for a novel therapeutic approach for the treatment of atherosclerotic vascular disease development and progression.

Acknowledgments

We thank Chelsea Deroche, PhD (Biostatistics and Research Design Unit in the Office of Medical Research and Health Management and Informatics, University of Missouri School of Medicine), for her expert advice on statistical assessments.

Sources of Funding

This work was supported by National Institutes of Health grants R01-HL070241 (Dr Delafontaine), R01-HL080682 (Dr Delafontaine), R21-HL113705 (Dr Sukhanov), R01-HL059976 (Dr Korthuis), P01-HL095486 (Dr Korthuis), and R01-AA022108 (Dr Korthuis), as well as American Heart Association Grant-in-Aid 13GRNT17230069 (Dr Sukhanov).

Disclosures

None.

Footnotes

  • The online-only Data Supplement is available with this article at http://circ.ahajournals.org/lookup/suppl/doi:10.1161/CIRCULATIONAHA.116.021805/-/DC1.

  • Received July 13, 2015.
  • Accepted April 27, 2016.
  • © 2016 American Heart Association, Inc.

References

  1. 1.↵
    1. Juul A,
    2. Scheike T,
    3. Davidsen M,
    4. Gyllenborg J,
    5. Jørgensen T
    . Low serum insulin-like growth factor I is associated with increased risk of ischemic heart disease: a population-based case-control study. Circulation. 2002;106:939–944.
    OpenUrlAbstract/FREE Full Text
  2. 2.↵
    1. Laughlin GA,
    2. Barrett-Connor E,
    3. Criqui MH,
    4. Kritz-Silverstein D
    . The prospective association of serum insulin-like growth factor I (IGF-I) and IGF-binding protein-1 levels with all cause and cardiovascular disease mortality in older adults: the Rancho Bernardo Study. J Clin Endocrinol Metab. 2004;89:114–120. doi: 10.1210/jc.2003-030967.
    OpenUrlCrossRefPubMed
  3. 3.↵
    1. Ruidavets JB,
    2. Luc G,
    3. Machez E,
    4. Genoux AL,
    5. Kee F,
    6. Arveiler D,
    7. Morange P,
    8. Woodside JV,
    9. Amouyel P,
    10. Evans A,
    11. Ducimetière P,
    12. Bingham A,
    13. Ferrières J,
    14. Perret B
    . Effects of insulin-like growth factor 1 in preventing acute coronary syndromes: the PRIME study. Atherosclerosis. 2011;218:464–469. doi: 10.1016/j.atherosclerosis.2011.05.034.
    OpenUrlCrossRefPubMed
  4. 4.↵
    1. Shai SY,
    2. Sukhanov S,
    3. Higashi Y,
    4. Vaughn C,
    5. Rosen CJ,
    6. Delafontaine P
    . Low circulating insulin-like growth factor I increases atherosclerosis in ApoE-deficient mice. Am J Physiol Heart Circ Physiol. 2011;300:H1898–H1906. doi: 10.1152/ajpheart.01081.2010.
    OpenUrlAbstract/FREE Full Text
  5. 5.↵
    1. Sukhanov S,
    2. Higashi Y,
    3. Shai SY,
    4. Vaughn C,
    5. Mohler J,
    6. Li Y,
    7. Song YH,
    8. Titterington J,
    9. Delafontaine P
    . IGF-1 reduces inflammatory responses, suppresses oxidative stress, and decreases atherosclerosis progression in ApoE-deficient mice. Arterioscler Thromb Vasc Biol. 2007;27:2684–2690. doi: 10.1161/ATVBAHA.107.156257.
    OpenUrlAbstract/FREE Full Text
  6. 6.↵
    1. Shai SY,
    2. Sukhanov S,
    3. Higashi Y,
    4. Vaughn C,
    5. Kelly J,
    6. Delafontaine P
    . Smooth muscle cell-specific insulin-like growth factor-1 overexpression in Apoe-/- mice does not alter atherosclerotic plaque burden but increases features of plaque stability. Arterioscler Thromb Vasc Biol. 2010;30:1916–1924. doi: 10.1161/ATVBAHA.110.210831.
    OpenUrlAbstract/FREE Full Text
  7. 7.↵
    1. von der Thüsen JH,
    2. Borensztajn KS,
    3. Moimas S,
    4. van Heiningen S,
    5. Teeling P,
    6. van Berkel TJ,
    7. Biessen EA
    . IGF-1 has plaque-stabilizing effects in atherosclerosis by altering vascular smooth muscle cell phenotype. Am J Pathol. 2011;178:924–934. doi: 10.1016/j.ajpath.2010.10.007.
    OpenUrlCrossRefPubMed
  8. 8.↵
    1. Renier G,
    2. Clément I,
    3. Desfaits AC,
    4. Lambert A
    . Direct stimulatory effect of insulin-like growth factor-I on monocyte and macrophage tumor necrosis factor-alpha production. Endocrinology. 1996;137:4611–4618. doi: 10.1210/endo.137.11.8895324.
    OpenUrlCrossRefPubMed
  9. 9.↵
    1. Hochberg Z,
    2. Hertz P,
    3. Maor G,
    4. Oiknine J,
    5. Aviram M
    . Growth hormone and insulin-like growth factor-I increase macrophage uptake and degradation of low density lipoprotein. Endocrinology. 1992;131:430–435. doi: 10.1210/endo.131.1.1612024.
    OpenUrlCrossRefPubMed
  10. 10.↵
    1. Succurro E,
    2. Andreozzi F,
    3. Sciacqua A,
    4. Sciaqua A,
    5. Hribal ML,
    6. Perticone F,
    7. Sesti G
    . Reciprocal association of plasma IGF-1 and interleukin-6 levels with cardiometabolic risk factors in nondiabetic subjects. Diabetes Care. 2008;31:1886–1888. doi: 10.2337/dc08-0553.
    OpenUrlAbstract/FREE Full Text
  11. 11.↵
    1. Jeschke MG,
    2. Barrow RE,
    3. Herndon DN
    . Insulinlike growth factor I plus insulinlike growth factor binding protein 3 attenuates the proinflammatory acute phase response in severely burned children. Ann Surg. 2000;231:246–252.
    OpenUrlCrossRefPubMed
  12. 12.↵
    1. Spies M,
    2. Wolf SE,
    3. Barrow RE,
    4. Jeschke MG,
    5. Herndon DN
    . Modulation of types I and II acute phase reactants with insulin-like growth factor-1/binding protein-3 complex in severely burned children. Crit Care Med. 2002;30:83–88.
    OpenUrlCrossRefPubMed
  13. 13.↵
    1. Roubenoff R,
    2. Parise H,
    3. Payette HA,
    4. Abad LW,
    5. D’Agostino R,
    6. Jacques PF,
    7. Wilson PW,
    8. Dinarello CA,
    9. Harris TB
    . Cytokines, insulin-like growth factor 1, sarcopenia, and mortality in very old community-dwelling men and women: the Framingham Heart Study. Am J Med. 2003;115:429–435.
    OpenUrlCrossRefPubMed
  14. 14.↵
    1. Cappola AR,
    2. Xue QL,
    3. Ferrucci L,
    4. Guralnik JM,
    5. Volpato S,
    6. Fried LP
    . Insulin-like growth factor I and interleukin-6 contribute synergistically to disability and mortality in older women. J Clin Endocrinol Metab. 2003;88:2019–2025. doi: 10.1210/jc.2002-021694.
    OpenUrlCrossRefPubMed
  15. 15.↵
    1. Gough PJ,
    2. Gomez IG,
    3. Wille PT,
    4. Raines EW
    . Macrophage expression of active MMP-9 induces acute plaque disruption in apoE-deficient mice. J Clin Invest. 2006;116:59–69. doi: 10.1172/JCI25074.
    OpenUrlCrossRefPubMed
  16. 16.↵
    1. Tacke F,
    2. Alvarez D,
    3. Kaplan TJ,
    4. Jakubzick C,
    5. Spanbroek R,
    6. Llodra J,
    7. Garin A,
    8. Liu J,
    9. Mack M,
    10. van Rooijen N,
    11. Lira SA,
    12. Habenicht AJ,
    13. Randolph GJ
    . Monocyte subsets differentially employ CCR2, CCR5, and CX3CR1 to accumulate within atherosclerotic plaques. J Clin Invest. 2007;117:185–194. doi: 10.1172/JCI28549.
    OpenUrlCrossRefPubMed
  17. 17.↵
    1. Wang WZ,
    2. Jones AW,
    3. Wang M,
    4. Durante W,
    5. Korthuis RJ
    . Preconditioning with soluble guanylate cyclase activation prevents postischemic inflammation and reduces nitrate tolerance in heme oxygenase-1 knockout mice. Am J Physiol Heart Circ Physiol. 2013;305:H521–H532. doi: 10.1152/ajpheart.00810.2012.
    OpenUrlAbstract/FREE Full Text
  18. 18.↵
    1. Zhang X,
    2. Goncalves R,
    3. Mosser DM
    . The isolation and characterization of murine macrophages. Curr Protoc Immunol. 2008;chap 14:unit 14.1. doi: 10.1002/0471142735.im1401s83.
    OpenUrl
  19. 19.↵
    1. Higashi Y,
    2. Sukhanov S,
    3. Parthasarathy S,
    4. Delafontaine P
    . The ubiquitin ligase Nedd4 mediates oxidized low-density lipoprotein-induced downregulation of insulin-like growth factor-1 receptor. Am J Physiol Heart Circ Physiol. 2008;295:H1684–H1689. doi: 10.1152/ajpheart.00548.2008.
    OpenUrlAbstract/FREE Full Text
  20. 20.↵
    1. Sukhanov S,
    2. Higashi Y,
    3. Shai SY,
    4. Itabe H,
    5. Ono K,
    6. Parthasarathy S,
    7. Delafontaine P
    . Novel effect of oxidized low-density lipoprotein: cellular ATP depletion via downregulation of glyceraldehyde-3-phosphate dehydrogenase. Circ Res. 2006;99:191–200. doi: 10.1161/01.RES.0000232319.02303.8c.
    OpenUrlAbstract/FREE Full Text
  21. 21.↵
    1. Clausen BE,
    2. Burkhardt C,
    3. Reith W,
    4. Renkawitz R,
    5. Förster I
    . Conditional gene targeting in macrophages and granulocytes using LysMcre mice. Transgenic Res. 1999;8:265–277.
    OpenUrlCrossRefPubMed
  22. 22.↵
    1. Ribas V,
    2. Drew BG,
    3. Le JA,
    4. Soleymani T,
    5. Daraei P,
    6. Sitz D,
    7. Mohammad L,
    8. Henstridge DC,
    9. Febbraio MA,
    10. Hewitt SC,
    11. Korach KS,
    12. Bensinger SJ,
    13. Hevener AL
    . Myeloid-specific estrogen receptor alpha deficiency impairs metabolic homeostasis and accelerates atherosclerotic lesion development. Proc Natl Acad Sci U S A. 2011;108:16457–16462. doi: 10.1073/pnas.1104533108.
    OpenUrlAbstract/FREE Full Text
  23. 23.↵
    1. Park SH,
    2. Sui Y,
    3. Gizard F,
    4. Xu J,
    5. Rios-Pilier J,
    6. Helsley RN,
    7. Han SS,
    8. Zhou C
    . Myeloid-specific IκB kinase β deficiency decreases atherosclerosis in low-density lipoprotein receptor-deficient mice. Arterioscler Thromb Vasc Biol. 2012;32:2869–2876. doi: 10.1161/ATVBAHA.112.254573.
    OpenUrlAbstract/FREE Full Text
  24. 24.↵
    1. Moxham CP,
    2. Duronio V,
    3. Jacobs S
    . Insulin-like growth factor I receptor beta-subunit heterogeneity: evidence for hybrid tetramers composed of insulin-like growth factor I and insulin receptor heterodimers. J Biol Chem. 1989;264:13238–13244.
    OpenUrlAbstract/FREE Full Text
  25. 25.↵
    1. Treadway JL,
    2. Morrison BD,
    3. Goldfine ID,
    4. Pessin JE
    . Assembly of insulin/insulin-like growth factor-1 hybrid receptors in vitro. J Biol Chem. 1989;264:21450–21453.
    OpenUrlAbstract/FREE Full Text
  26. 26.↵
    1. Soos MA,
    2. Siddle K
    . Immunological relationships between receptors for insulin and insulin-like growth factor I: evidence for structural heterogeneity of insulin-like growth factor I receptors involving hybrids with insulin receptors. Biochem J. 1989;263:553–563.
    OpenUrlAbstract/FREE Full Text
  27. 27.↵
    1. Soos MA,
    2. Field CE,
    3. Siddle K
    . Purified hybrid insulin/insulin-like growth factor-I receptors bind insulin-like growth factor-I, but not insulin, with high affinity. Biochem J. 1993;290 (Pt 2):419–426.
    OpenUrl
  28. 28.↵
    1. Frattali AL,
    2. Pessin JE
    . Relationship between alpha subunit ligand occupancy and beta subunit autophosphorylation in insulin/insulin-like growth factor-1 hybrid receptors. J Biol Chem. 1993;268:7393–7400.
    OpenUrlAbstract/FREE Full Text
  29. 29.↵
    1. Abbas A,
    2. Imrie H,
    3. Viswambharan H,
    4. Sukumar P,
    5. Rajwani A,
    6. Cubbon RM,
    7. Gage M,
    8. Smith J,
    9. Galloway S,
    10. Yuldeshava N,
    11. Kahn M,
    12. Xuan S,
    13. Grant PJ,
    14. Channon KM,
    15. Beech DJ,
    16. Wheatcroft SB,
    17. Kearney MT
    . The insulin-like growth factor-1 receptor is a negative regulator of nitric oxide bioavailability and insulin sensitivity in the endothelium. Diabetes. 2011;60:2169–2178. doi: 10.2337/db11-0197.
    OpenUrlAbstract/FREE Full Text
  30. 30.↵
    1. Han S,
    2. Liang CP,
    3. DeVries-Seimon T,
    4. Ranalletta M,
    5. Welch CL,
    6. Collins-Fletcher K,
    7. Accili D,
    8. Tabas I,
    9. Tall AR
    . Macrophage insulin receptor deficiency increases ER stress-induced apoptosis and necrotic core formation in advanced atherosclerotic lesions. Cell Metab. 2006;3:257–266. doi: 10.1016/j.cmet.2006.02.008.
    OpenUrlCrossRefPubMed
  31. 31.↵
    1. Baumgartl J,
    2. Baudler S,
    3. Scherner M,
    4. Babaev V,
    5. Makowski L,
    6. Suttles J,
    7. McDuffie M,
    8. Tobe K,
    9. Kadowaki T,
    10. Fazio S,
    11. Kahn CR,
    12. Hotamisligil GS,
    13. Krone W,
    14. Linton M,
    15. Brüning JC
    . Myeloid lineage cell-restricted insulin resistance protects apolipoproteinE-deficient mice against atherosclerosis. Cell Metab. 2006;3:247–256. doi: 10.1016/j.cmet.2006.02.010.
    OpenUrlCrossRefPubMed
  32. 32.↵
    1. Murphy AJ,
    2. Akhtari M,
    3. Tolani S,
    4. Pagler T,
    5. Bijl N,
    6. Kuo CL,
    7. Wang M,
    8. Sanson M,
    9. Abramowicz S,
    10. Welch C,
    11. Bochem AE,
    12. Kuivenhoven JA,
    13. Yvan-Charvet L,
    14. Tall AR
    . ApoE regulates hematopoietic stem cell proliferation, monocytosis, and monocyte accumulation in atherosclerotic lesions in mice. J Clin Invest. 2011;121:4138–4149. doi: 10.1172/JCI57559.
    OpenUrlCrossRefPubMed
  33. 33.↵
    1. Swirski FK,
    2. Libby P,
    3. Aikawa E,
    4. Alcaide P,
    5. Luscinskas FW,
    6. Weissleder R,
    7. Pittet MJ
    . Ly-6Chi monocytes dominate hypercholesterolemia-associated monocytosis and give rise to macrophages in atheromata. J Clin Invest. 2007;117:195–205. doi: 10.1172/JCI29950.
    OpenUrlCrossRefPubMed
  34. 34.↵
    1. Carstairs KC
    . The identification of platelets and platelet antigens in histological sections. J Pathol Bacteriol. 1965;90:225–231.
    OpenUrlCrossRefPubMed
  35. 35.↵
    1. Kozaki K,
    2. Kaminski WE,
    3. Tang J,
    4. Hollenbach S,
    5. Lindahl P,
    6. Sullivan C,
    7. Yu JC,
    8. Abe K,
    9. Martin PJ,
    10. Ross R,
    11. Betsholtz C,
    12. Giese NA,
    13. Raines EW
    . Blockade of platelet-derived growth factor or its receptors transiently delays but does not prevent fibrous cap formation in ApoE null mice. Am J Pathol. 2002;161:1395–1407. doi: 10.1016/S0002-9440(10)64415-X.
    OpenUrlCrossRefPubMed
  36. 36.↵
    1. van Tits LJ,
    2. Stienstra R,
    3. van Lent PL,
    4. Netea MG,
    5. Joosten LA,
    6. Stalenhoef AF
    . Oxidized LDL enhances pro-inflammatory responses of alternatively activated M2 macrophages: a crucial role for Krüppel-like factor 2. Atherosclerosis. 2011;214:345–349. doi: 10.1016/j.atherosclerosis.2010.11.018.
    OpenUrlCrossRefPubMed
  37. 37.↵
    1. Park YM,
    2. Drazba JA,
    3. Vasanji A,
    4. Egelhoff T,
    5. Febbraio M,
    6. Silverstein RL
    . Oxidized LDL/CD36 interaction induces loss of cell polarity and inhibits macrophage locomotion. Mol Biol Cell. 2012;23:3057–3068. doi: 10.1091/mbc.E11-12-1051.
    OpenUrlAbstract/FREE Full Text
  38. 38.↵
    1. Wiesner P,
    2. Choi SH,
    3. Almazan F,
    4. Benner C,
    5. Huang W,
    6. Diehl CJ,
    7. Gonen A,
    8. Butler S,
    9. Witztum JL,
    10. Glass CK,
    11. Miller YI
    . Low doses of lipopolysaccharide and minimally oxidized low-density lipoprotein cooperatively activate macrophages via nuclear factor kappa B and activator protein-1: possible mechanism for acceleration of atherosclerosis by subclinical endotoxemia. Circ Res. 2010;107:56–65. doi: 10.1161/CIRCRESAHA.110.218420.
    OpenUrlAbstract/FREE Full Text
  39. 39.↵
    1. Kadl A,
    2. Meher AK,
    3. Sharma PR,
    4. Lee MY,
    5. Doran AC,
    6. Johnstone SR,
    7. Elliott MR,
    8. Gruber F,
    9. Han J,
    10. Chen W,
    11. Kensler T,
    12. Ravichandran KS,
    13. Isakson BE,
    14. Wamhoff BR,
    15. Leitinger N
    . Identification of a novel macrophage phenotype that develops in response to atherogenic phospholipids via Nrf2. Circ Res. 2010;107:737–746. doi: 10.1161/CIRCRESAHA.109.215715.
    OpenUrlAbstract/FREE Full Text
  40. 40.↵
    1. Zelcer N,
    2. Tontonoz P
    . Liver X receptors as integrators of metabolic and inflammatory signaling. J Clin Invest. 2006;116:607–614. doi: 10.1172/JCI27883.
    OpenUrlCrossRefPubMed
  41. 41.↵
    1. Robbins CS,
    2. Hilgendorf I,
    3. Weber GF,
    4. Theurl I,
    5. Iwamoto Y,
    6. Figueiredo JL,
    7. Gorbatov R,
    8. Sukhova GK,
    9. Gerhardt LM,
    10. Smyth D,
    11. Zavitz CC,
    12. Shikatani EA,
    13. Parsons M,
    14. van Rooijen N,
    15. Lin HY,
    16. Husain M,
    17. Libby P,
    18. Nahrendorf M,
    19. Weissleder R,
    20. Swirski FK
    . Local proliferation dominates lesional macrophage accumulation in atherosclerosis. Nat Med. 2013;19:1166–1172. doi: 10.1038/nm.3258.
    OpenUrlCrossRefPubMed
  42. 42.↵
    1. Randolph GJ
    . Mechanisms that regulate macrophage burden in atherosclerosis. Circ Res. 2014;114:1757–1771. doi: 10.1161/CIRCRESAHA.114.301174.
    OpenUrlAbstract/FREE Full Text
  43. 43.↵
    1. Tacke F,
    2. Ginhoux F,
    3. Jakubzick C,
    4. van Rooijen N,
    5. Merad M,
    6. Randolph GJ
    . Immature monocytes acquire antigens from other cells in the bone marrow and present them to T cells after maturing in the periphery. J Exp Med. 2006;203:583–597. doi: 10.1084/jem.20052119.
    OpenUrlAbstract/FREE Full Text
  44. 44.↵
    1. Brizova H,
    2. Kalinova M,
    3. Krskova L,
    4. Mrhalova M,
    5. Kodet R
    . A novel quantitative PCR of proliferation markers (Ki-67, topoisomerase IIalpha, and TPX2): an immunohistochemical correlation, testing, and optimizing for mantle cell lymphoma. Virchows Arch. 2010;456:671–679. doi: 10.1007/s00428-010-0922-8.
    OpenUrlCrossRefPubMed
  45. 45.↵
    1. Ozen M,
    2. Ittmann M
    . Increased expression and activity of CDC25C phosphatase and an alternatively spliced variant in prostate cancer. Clin Cancer Res. 2005;11:4701–4706. doi: 10.1158/1078-0432.CCR-04-2551.
    OpenUrlAbstract/FREE Full Text
  46. 46.↵
    1. Kelman Z
    . PCNA: structure, functions and interactions. Oncogene. 1997;14:629–640. doi: 10.1038/sj.onc.1200886.
    OpenUrlCrossRefPubMed
  47. 47.↵
    1. Foryst-Ludwig A,
    2. Hartge M,
    3. Clemenz M,
    4. Sprang C,
    5. Hess K,
    6. Marx N,
    7. Unger T,
    8. Kintscher U
    . PPARgamma activation attenuates T-lymphocyte-dependent inflammation of adipose tissue and development of insulin resistance in obese mice. Cardiovasc Diabetol. 2010;9:64. doi: 10.1186/1475-2840-9-64.
    OpenUrlCrossRefPubMed
  48. 48.↵
    1. Kuhn E,
    2. Bourgeois C,
    3. Keo V,
    4. Viengchareun S,
    5. Muscat A,
    6. Meduri G,
    7. Le Menuet D,
    8. Fève B,
    9. Lombès M
    . Paradoxical resistance to high-fat diet-induced obesity and altered macrophage polarization in mineralocorticoid receptor-overexpressing mice. Am J Physiol Endocrinol Metab. 2014;306:E75–E90. doi: 10.1152/ajpendo.00323.2013.
    OpenUrlAbstract/FREE Full Text
  49. 49.↵
    1. Higashi Y,
    2. Peng T,
    3. Du J,
    4. Sukhanov S,
    5. Li Y,
    6. Itabe H,
    7. Parthasarathy S,
    8. Delafontaine P
    . A redox-sensitive pathway mediates oxidized LDL-induced downregulation of insulin-like growth factor-1 receptor. J Lipid Res. 2005;46:1266–1277. doi: 10.1194/jlr.M400478-JLR200.
    OpenUrlAbstract/FREE Full Text
  50. 50.↵
    1. Fournier T,
    2. Riches DW,
    3. Winston BW,
    4. Rose DM,
    5. Young SK,
    6. Noble PW,
    7. Lake FR,
    8. Henson PM
    . Divergence in macrophage insulin-like growth factor-I (IGF-I) synthesis induced by TNF-alpha and prostaglandin E2. J Immunol. 1995;155:2123–2133.
    OpenUrlAbstract
  51. 51.↵
    1. Pelosi L,
    2. Giacinti C,
    3. Nardis C,
    4. Borsellino G,
    5. Rizzuto E,
    6. Nicoletti C,
    7. Wannenes F,
    8. Battistini L,
    9. Rosenthal N,
    10. Molinaro M,
    11. Musarò A
    . Local expression of IGF-1 accelerates muscle regeneration by rapidly modulating inflammatory cytokines and chemokines. FASEB J. 2007;21:1393–1402. doi: 10.1096/fj.06-7690com.
    OpenUrlAbstract/FREE Full Text
  52. 52.↵
    1. Sandhu MS,
    2. Heald AH,
    3. Gibson JM,
    4. Cruickshank JK,
    5. Dunger DB,
    6. Wareham NJ
    . Circulating concentrations of insulin-like growth factor-I and development of glucose intolerance: a prospective observational study. Lancet. 2002;359:1740–1745. doi: 10.1016/S0140-6736(02)08655-5.
    OpenUrlCrossRefPubMed
  53. 53.↵
    1. Succurro E,
    2. Andreozzi F,
    3. Marini MA,
    4. Lauro R,
    5. Hribal ML,
    6. Perticone F,
    7. Sesti G
    . Low plasma insulin-like growth factor-1 levels are associated with reduced insulin sensitivity and increased insulin secretion in nondiabetic subjects. Nutr Metab Cardiovasc Dis. 2009;19:713–719. doi: 10.1016/j.numecd.2008.12.011.
    OpenUrlCrossRefPubMed
  54. 54.↵
    1. Succurro E,
    2. Arturi F,
    3. Grembiale A,
    4. Iorio F,
    5. Laino I,
    6. Andreozzi F,
    7. Sciacqua A,
    8. Hribal ML,
    9. Perticone F,
    10. Sesti G
    . Positive association between plasma IGF1 and high-density lipoprotein cholesterol levels in adult nondiabetic subjects. Eur J Endocrinol. 2010;163:75–80. doi: 10.1530/EJE-10-0113.
    OpenUrlAbstract/FREE Full Text
  55. 55.↵
    1. Sesti G,
    2. Sciacqua A,
    3. Cardellini M,
    4. Marini MA,
    5. Maio R,
    6. Vatrano M,
    7. Succurro E,
    8. Lauro R,
    9. Federici M,
    10. Perticone F
    . Plasma concentration of IGF-I is independently associated with insulin sensitivity in subjects with different degrees of glucose tolerance. Diabetes Care. 2005;28:120–125.
    OpenUrlAbstract/FREE Full Text
  56. 56.↵
    1. Perticone F,
    2. Sciacqua A,
    3. Perticone M,
    4. Laino I,
    5. Miceli S,
    6. Care’ I,
    7. Galiano Leone G,
    8. Andreozzi F,
    9. Maio R,
    10. Sesti G
    . Low-plasma insulin-like growth factor-I levels are associated with impaired endothelium-dependent vasodilatation in a cohort of untreated, hypertensive Caucasian subjects. J Clin Endocrinol Metab. 2008;93:2806–2810. doi: 10.1210/jc.2008-0646.
    OpenUrlCrossRefPubMed
  57. 57.↵
    1. Aguirre GA,
    2. De Ita JR,
    3. de la Garza RG,
    4. Castilla-Cortazar I
    . Insulin-like growth factor-1 deficiency and metabolic syndrome. J Transl Med. 2016;14:3. doi: 10.1186/s12967-015-0762-z.
    OpenUrlCrossRefPubMed
  58. 58.↵
    1. Colao A
    . The GH-IGF-I axis and the cardiovascular system: clinical implications. Clin Endocrinol (Oxf). 2008;69:347–358. doi: 10.1111/j.1365-2265.2008.03292.x.
    OpenUrlCrossRefPubMed
  59. 59.↵
    1. Palmeiro CR,
    2. Anand R,
    3. Dardi IK,
    4. Balasubramaniyam N,
    5. Schwarcz MD,
    6. Weiss IA
    . Growth hormone and the cardiovascular system. Cardiol Rev. 2012;20:197–207. doi: 10.1097/CRD.0b013e318248a3e1.
    OpenUrlCrossRefPubMed
  60. 60.↵
    1. Andreassen M,
    2. Raymond I,
    3. Kistorp C,
    4. Hildebrandt P,
    5. Faber J,
    6. Kristensen LØ
    . IGF1 as predictor of all cause mortality and cardiovascular disease in an elderly population. Eur J Endocrinol. 2009;160:25–31. doi: 10.1530/EJE-08-0452.
    OpenUrlAbstract/FREE Full Text
  61. 61.↵
    1. Boquist S,
    2. Ruotolo G,
    3. Skoglund-Andersson C,
    4. Tang R,
    5. Björkegren J,
    6. Bond MG,
    7. de Faire U,
    8. Brismar K,
    9. Hamsten A
    . Correlation of serum IGF-I and IGFBP-1 and -3 to cardiovascular risk indicators and early carotid atherosclerosis in healthy middle-aged men. Clin Endocrinol (Oxf). 2008;68:51–58. doi: 10.1111/j.1365-2265.2007.02998.x.
    OpenUrlCrossRefPubMed
  62. 62.↵
    1. Bøtker HE,
    2. Skjaerbaek C,
    3. Eriksen UH,
    4. Schmitz O,
    5. Orskov H
    . Insulin-like growth factor-I, insulin, and angina pectoris secondary to coronary atherosclerosis, vasospasm, and syndrome X. Am J Cardiol. 1997;79:961–963.
    OpenUrlCrossRefPubMed
  63. 63.↵
    1. Fischer F,
    2. Schulte H,
    3. Mohan S,
    4. Tataru MC,
    5. Köhler E,
    6. Assmann G,
    7. von Eckardstein A
    . Associations of insulin-like growth factors, insulin-like growth factor binding proteins and acid-labile subunit with coronary heart disease. Clin Endocrinol (Oxf). 2004;61:595–602. doi: 10.1111/j.1365-2265.2004.02136.x.
    OpenUrlCrossRefPubMed
  64. 64.↵
    1. Kawachi S,
    2. Takeda N,
    3. Sasaki A,
    4. Kokubo Y,
    5. Takami K,
    6. Sarui H,
    7. Hayashi M,
    8. Yamakita N,
    9. Yasuda K
    . Circulating insulin-like growth factor-1 and insulin-like growth factor binding protein-3 are associated with early carotid atherosclerosis. Arterioscler Thromb Vasc Biol. 2005;25:617–621. doi: 10.1161/01.ATV.0000154486.03017.35.
    OpenUrlAbstract/FREE Full Text
  65. 65.↵
    1. Ruotolo G,
    2. Båvenholm P,
    3. Brismar K,
    4. Eféndic S,
    5. Ericsson CG,
    6. de Faire U,
    7. Nilsson J,
    8. Hamsten A
    . Serum insulin-like growth factor-I level is independently associated with coronary artery disease progression in young male survivors of myocardial infarction: beneficial effects of bezafibrate treatment. J Am Coll Cardiol. 2000;35:647–654.
    OpenUrlCrossRefPubMed
  66. 66.↵
    1. Janssen JA,
    2. Stolk RP,
    3. Pols HA,
    4. Grobbee DE,
    5. de Jong FH,
    6. Lamberts SW
    . Serum free IGF-I, total IGF-I, IGFBP-1 and IGFBP-3 levels in an elderly population: relation to age and sex steroid levels. Clin Endocrinol (Oxf). 1998;48:471–478.
    OpenUrlCrossRefPubMed
  67. 67.↵
    1. Friedrich N,
    2. Haring R,
    3. Nauck M,
    4. Lüdemann J,
    5. Rosskopf D,
    6. Spilcke-Liss E,
    7. Felix SB,
    8. Dörr M,
    9. Brabant G,
    10. Völzke H,
    11. Wallaschofski H
    . Mortality and serum insulin-like growth factor (IGF)-I and IGF binding protein 3 concentrations. J Clin Endocrinol Metab. 2009;94:1732–1739. doi: 10.1210/jc.2008-2138.
    OpenUrlCrossRefPubMed
  68. 68.↵
    1. Conti E,
    2. Andreotti F,
    3. Sestito A,
    4. Riccardi P,
    5. Menini E,
    6. Crea F,
    7. Maseri A,
    8. Lanza GA
    . Reduced levels of insulin-like growth factor-1 in patients with angina pectoris, positive exercise stress test, and angiographically normal epicardial coronary arteries. Am J Cardiol. 2002;89:973–975.
    OpenUrlCrossRefPubMed
  69. 69.↵
    1. Goodman-Gruen D,
    2. Barrett-Connor E,
    3. Rosen C
    . IGF-1 and ischemic heart disease in older people. J Am Geriatr Soc. 2000;48:860–861.
    OpenUrlCrossRefPubMed
  70. 70.↵
    1. Martin RM,
    2. Gunnell D,
    3. Whitley E,
    4. Nicolaides A,
    5. Griffin M,
    6. Georgiou N,
    7. Davey Smith G,
    8. Ebrahim S,
    9. Holly JM
    . Associations of insulin-like growth factor (IGF)-I, IGF-II, IGF binding protein (IGFBP)-2 and IGFBP-3 with ultrasound measures of atherosclerosis and plaque stability in an older adult population. J Clin Endocrinol Metab. 2008;93:1331–1338. doi: 10.1210/jc.2007-2295.
    OpenUrlCrossRefPubMed
  71. 71.↵
    1. Spallarossa P,
    2. Brunelli C,
    3. Minuto F,
    4. Caruso D,
    5. Battistini M,
    6. Caponnetto S,
    7. Cordera R
    . Insulin-like growth factor-I and angiographically documented coronary artery disease. Am J Cardiol. 1996;77:200–202.
    OpenUrlCrossRefPubMed
  72. 72.↵
    1. van den Beld AW,
    2. Bots ML,
    3. Janssen JA,
    4. Pols HA,
    5. Lamberts SW,
    6. Grobbee DE
    . Endogenous hormones and carotid atherosclerosis in elderly men. Am J Epidemiol. 2003;157:25–31.
    OpenUrlAbstract/FREE Full Text
  73. 73.↵
    1. Brugts MP,
    2. Ranke MB,
    3. Hofland LJ,
    4. van der Wansem K,
    5. Weber K,
    6. Frystyk J,
    7. Lamberts SW,
    8. Janssen JA
    . Normal values of circulating insulin-like growth factor-I bioactivity in the healthy population: comparison with five widely used IGF-I immunoassays. J Clin Endocrinol Metab. 2008;93:2539–2545. doi: 10.1210/jc.2007-2454.
    OpenUrlCrossRefPubMed
  74. 74.↵
    1. Brugts MP,
    2. van den Beld AW,
    3. Hofland LJ,
    4. van der Wansem K,
    5. van Koetsveld PM,
    6. Frystyk J,
    7. Lamberts SW,
    8. Janssen JA
    . Low circulating insulin-like growth factor I bioactivity in elderly men is associated with increased mortality. J Clin Endocrinol Metab. 2008;93:2515–2522. doi: 10.1210/jc.2007-1633.
    OpenUrlCrossRefPubMed
  75. 75.↵
    1. Bleumink GS,
    2. Schut AF,
    3. Sturkenboom MC,
    4. Janssen JA,
    5. Witteman JC,
    6. van Duijn CM,
    7. Hofman A,
    8. Stricker BH
    . A promoter polymorphism of the insulin-like growth factor-I gene is associated with left ventricular hypertrophy. Heart. 2005;91:239–240. doi: 10.1136/hrt.2003.019778.
    OpenUrlFREE Full Text
  76. 76.↵
    1. Schut AF,
    2. Janssen JA,
    3. Deinum J,
    4. Vergeer JM,
    5. Hofman A,
    6. Lamberts SW,
    7. Oostra BA,
    8. Pols HA,
    9. Witteman JC,
    10. van Duijn CM
    . Polymorphism in the promoter region of the insulin-like growth factor I gene is related to carotid intima-media thickness and aortic pulse wave velocity in subjects with hypertension. Stroke. 2003;34:1623–1627. doi: 10.1161/01.STR.0000076013.00240.B0.
    OpenUrlAbstract/FREE Full Text
  77. 77.↵
    1. Sesti G,
    2. Mannino GC,
    3. Andreozzi F,
    4. Greco A,
    5. Perticone M,
    6. Sciacqua A,
    7. Marini MA,
    8. Perticone F
    . A polymorphism at IGF1 locus is associated with carotid intima media thickness and endothelium-dependent vasodilatation. Atherosclerosis. 2014;232:25–30. doi: 10.1016/j.atherosclerosis.2013.10.024.
    OpenUrlCrossRefPubMed
  78. 78.↵
    1. Imrie H,
    2. Abbas A,
    3. Viswambharan H,
    4. Rajwani A,
    5. Cubbon RM,
    6. Gage M,
    7. Kahn M,
    8. Ezzat VA,
    9. Duncan ER,
    10. Grant PJ,
    11. Ajjan R,
    12. Wheatcroft SB,
    13. Kearney MT
    . Vascular insulin-like growth factor-I resistance and diet-induced obesity. Endocrinology. 2009;150:4575–4582. doi: 10.1210/en.2008-1641.
    OpenUrlCrossRefPubMed
  79. 79.↵
    1. Okura Y,
    2. Brink M,
    3. Itabe H,
    4. Scheidegger KJ,
    5. Kalangos A,
    6. Delafontaine P
    . Oxidized low-density lipoprotein is associated with apoptosis of vascular smooth muscle cells in human atherosclerotic plaques. Circulation. 2000;102:2680–2686.
    OpenUrlAbstract/FREE Full Text
  80. 80.↵
    1. Okura Y,
    2. Brink M,
    3. Zahid AA,
    4. Anwar A,
    5. Delafontaine P
    . Decreased expression of insulin-like growth factor-1 and apoptosis of vascular smooth muscle cells in human atherosclerotic plaque. J Mol Cell Cardiol. 2001;33:1777–1789. doi: 10.1006/jmcc.2001.1441.
    OpenUrlCrossRefPubMed
  81. 81.↵
    1. Rotzius P,
    2. Soehnlein O,
    3. Kenne E,
    4. Lindbom L,
    5. Nystrom K,
    6. Thams S,
    7. Eriksson EE
    . ApoE(-/-)/lysozyme M(EGFP/EGFP) mice as a versatile model to study monocyte and neutrophil trafficking in atherosclerosis. Atherosclerosis. 2009;202:111–118. doi: 10.1016/j.atherosclerosis.2008.04.009.
    OpenUrlCrossRefPubMed
  82. 82.↵
    1. Xue J,
    2. Schmidt SV,
    3. Sander J,
    4. Draffehn A,
    5. Krebs W,
    6. Quester I,
    7. De Nardo D,
    8. Gohel TD,
    9. Emde M,
    10. Schmidleithner L,
    11. Ganesan H,
    12. Nino-Castro A,
    13. Mallmann MR,
    14. Labzin L,
    15. Theis H,
    16. Kraut M,
    17. Beyer M,
    18. Latz E,
    19. Freeman TC,
    20. Ulas T,
    21. Schultze JL
    . Transcriptome-based network analysis reveals a spectrum model of human macrophage activation. Immunity. 2014;40:274–288. doi: 10.1016/j.immuni.2014.01.006.
    OpenUrlCrossRefPubMed
  83. 83.↵
    1. Martinez FO,
    2. Gordon S
    . The M1 and M2 paradigm of macrophage activation: time for reassessment. F1000Prime Rep. 2014;6:13. doi: 10.12703/P6-13.
    OpenUrl
  84. 84.↵
    1. Mosser DM,
    2. Edwards JP
    . Exploring the full spectrum of macrophage activation. Nat Rev Immunol. 2008;8:958–969. doi: 10.1038/nri2448.
    OpenUrlCrossRefPubMed
  85. 85.↵
    1. Qin X,
    2. Qiu C,
    3. Zhao L
    . Lysophosphatidylcholine perpetuates macrophage polarization toward classically activated phenotype in inflammation. Cell Immunol. 2014;289:185–190. doi: 10.1016/j.cellimm.2014.04.010.
    OpenUrlCrossRefPubMed
  86. 86.↵
    1. Rios FJ,
    2. Koga MM,
    3. Pecenin M,
    4. Ferracini M,
    5. Gidlund M,
    6. Jancar S
    . Oxidized LDL induces alternative macrophage phenotype through activation of CD36 and PAFR. Mediators Inflamm. 2013;2013:198193. doi: 10.1155/2013/198193.
    OpenUrlPubMed
  87. 87.↵
    1. Chu EM,
    2. Tai DC,
    3. Beer JL,
    4. Hill JS
    . Macrophage heterogeneity and cholesterol homeostasis: classically-activated macrophages are associated with reduced cholesterol accumulation following treatment with oxidized LDL. Biochim Biophys Acta. 2013;1831:378–386. doi: 10.1016/j.bbalip.2012.10.009.
    OpenUrl
  88. 88.↵
    1. Phillips MC
    . Molecular mechanisms of cellular cholesterol efflux. J Biol Chem. 2014;289:24020–24029. doi: 10.1074/jbc.R114.583658.
    OpenUrlAbstract/FREE Full Text
  89. 89.↵
    1. Lee SD,
    2. Tontonoz P
    . Liver X receptors at the intersection of lipid metabolism and atherogenesis. Atherosclerosis. 2015;242:29–36. doi: 10.1016/j.atherosclerosis.2015.06.042.
    OpenUrlCrossRefPubMed
  90. 90.↵
    1. Tall AR
    . Cholesterol efflux pathways and other potential mechanisms involved in the athero-protective effect of high density lipoproteins. J Intern Med. 2008;263:256–273. doi: 10.1111/j.1365-2796.2007.01898.x.
    OpenUrlCrossRefPubMed
  91. 91.↵
    1. Newby AC
    . Matrix metalloproteinase inhibition therapy for vascular diseases. Vascul Pharmacol. 2012;56:232–244. doi: 10.1016/j.vph.2012.01.007.
    OpenUrlCrossRefPubMed
  92. 92.↵
    1. Newby AC
    . Metalloproteinases promote plaque rupture and myocardial infarction: a persuasive concept waiting for clinical translation. Matrix Biol. 2015;44-46:157–166. doi: 10.1016/j.matbio.2015.01.015.
    OpenUrl
  93. 93.↵
    1. Anwar A,
    2. Zahid AA,
    3. Scheidegger KJ,
    4. Brink M,
    5. Delafontaine P
    . Tumor necrosis factor-alpha regulates insulin-like growth factor-1 and insulin-like growth factor binding protein-3 expression in vascular smooth muscle. Circulation. 2002;105:1220–1225.
    OpenUrlAbstract/FREE Full Text
  94. 94.↵
    1. Blackstock CD,
    2. Higashi Y,
    3. Sukhanov S,
    4. Shai SY,
    5. Stefanovic B,
    6. Tabony AM,
    7. Yoshida T,
    8. Delafontaine P
    . Insulin-like growth factor-1 increases synthesis of collagen type I via induction of the mRNA-binding protein LARP6 expression and binding to the 5’ stem-loop of COL1a1 and COL1a2 mRNA. J Biol Chem. 2014;289:7264–7274. doi: 10.1074/jbc.M113.518951.
    OpenUrlAbstract/FREE Full Text
  95. 95.↵
    1. Higashi Y,
    2. Pandey A,
    3. Goodwin B,
    4. Delafontaine P
    . Insulin-like growth factor-1 regulates glutathione peroxidase expression and activity in vascular endothelial cells: implications for atheroprotective actions of insulin-like growth factor-1. Biochim Biophys Acta. 2013;1832:391–399. doi: 10.1016/j.bbadis.2012.12.005.
    OpenUrl
  96. 96.↵
    1. Wang JC,
    2. Bennett M
    . Aging and atherosclerosis: mechanisms, functional consequences, and potential therapeutics for cellular senescence. Circ Res. 2012;111:245–259. doi: 10.1161/CIRCRESAHA.111.261388.
    OpenUrlAbstract/FREE Full Text
  97. 97.↵
    1. Ungvari Z,
    2. Csiszar A
    . The emerging role of IGF-1 deficiency in cardiovascular aging: recent advances. J Gerontol A Biol Sci Med Sci. 2012;67:599–610. doi: 10.1093/gerona/gls072.
    OpenUrlPubMed
  98. 98.↵
    1. Higashi Y,
    2. Sukhanov S,
    3. Anwar A,
    4. Shai SY,
    5. Delafontaine P
    . Aging, atherosclerosis, and IGF-1. J Gerontol A Biol Sci Med Sci. 2012;67:626–639. doi: 10.1093/gerona/gls102.
    OpenUrlPubMed

CLINICAL PERSPECTIVE

Atherosclerosis is an inflammatory disease, and acute coronary events result largely from erosion or rupture of unstable plaques with increased inflammatory cells and a relative reduction in vascular smooth muscle cells. Macrophages play a major role in atherogenesis by scavenging and accumulating lipids to become lipid-laden foam cells. Furthermore, proinflammatory macrophages induce smooth muscle cell death by secreting cytokines and degrade extracellular matrix by producing enzymes such as matrix metalloproteinases, weakening the tensile strength of plaques and predisposing them to rupture. In the past decade, insulin-like growth factor-1 (IGF-1) has demonstrated antiatherogenic effects in experimental models, but the mechanisms are poorly elucidated. In this study, we investigated macrophage–IGF-1 receptor deficiency in a murine model of atherosclerosis. We found that IGF-1 receptor deficiency increased monocyte/macrophage recruitment to lesions, skewed macrophage activation to a proinflammatory status, promoted lipid accumulation in macrophages by lowering lipid efflux, and upregulated matrix metalloproteinase production, resulting in an increase in atherosclerotic burden and a decrease in features of plaque stability. Our findings are consistent with epidemiological studies suggesting that low circulating IGF-1 is a predictor of ischemic heart disease and mortality. There is also evidence linking low IGF-1 to multiple cardiovascular risk factors, including metabolic syndrome. Our findings suggest that IGF-1 regulation of macrophage inflammatory responses and lipid metabolism may be the basis for novel approaches to reduce atherosclerotic lesion progression and to promote plaque stability.

View Abstract
Back to top
Previous ArticleNext Article

This Issue

Circulation
June 7, 2016, Volume 133, Issue 23
  • Table of Contents
Previous ArticleNext Article

Jump to

  • Article
    • Abstract
    • Introduction
    • Methods
    • Results
    • Discussion
    • Acknowledgments
    • Sources of Funding
    • Disclosures
    • Footnotes
    • References
  • Figures & Tables
  • Supplemental Materials
  • Info & Metrics
  • eLetters

Article Tools

  • Print
  • Citation Tools
    Insulin-Like Growth Factor-1 Receptor Deficiency in Macrophages Accelerates Atherosclerosis and Induces an Unstable Plaque Phenotype in Apolipoprotein E–Deficient MiceCLINICAL PERSPECTIVE
    Yusuke Higashi, Sergiy Sukhanov, Shaw-Yung Shai, Svitlana Danchuk, Richard Tang, Patricia Snarski, Zhaohui Li, Patricia Lobelle-Rich, Meifang Wang, Derek Wang, Hong Yu, Ronald Korthuis and Patrice Delafontaine
    Circulation. 2016;133:2263-2278, originally published May 6, 2016
    https://doi.org/10.1161/CIRCULATIONAHA.116.021805

    Citation Manager Formats

    • BibTeX
    • Bookends
    • EasyBib
    • EndNote (tagged)
    • EndNote 8 (xml)
    • Medlars
    • Mendeley
    • Papers
    • RefWorks Tagged
    • Ref Manager
    • RIS
    • Zotero
  •  Download Powerpoint
  • Article Alerts
    Log in to Email Alerts with your email address.
  • Save to my folders

Share this Article

  • Email

    Thank you for your interest in spreading the word on Circulation.

    NOTE: We only request your email address so that the person you are recommending the page to knows that you wanted them to see it, and that it is not junk mail. We do not capture any email address.

    Enter multiple addresses on separate lines or separate them with commas.
    Insulin-Like Growth Factor-1 Receptor Deficiency in Macrophages Accelerates Atherosclerosis and Induces an Unstable Plaque Phenotype in Apolipoprotein E–Deficient MiceCLINICAL PERSPECTIVE
    (Your Name) has sent you a message from Circulation
    (Your Name) thought you would like to see the Circulation web site.
  • Share on Social Media
    Insulin-Like Growth Factor-1 Receptor Deficiency in Macrophages Accelerates Atherosclerosis and Induces an Unstable Plaque Phenotype in Apolipoprotein E–Deficient MiceCLINICAL PERSPECTIVE
    Yusuke Higashi, Sergiy Sukhanov, Shaw-Yung Shai, Svitlana Danchuk, Richard Tang, Patricia Snarski, Zhaohui Li, Patricia Lobelle-Rich, Meifang Wang, Derek Wang, Hong Yu, Ronald Korthuis and Patrice Delafontaine
    Circulation. 2016;133:2263-2278, originally published May 6, 2016
    https://doi.org/10.1161/CIRCULATIONAHA.116.021805
    del.icio.us logo Digg logo Reddit logo Twitter logo CiteULike logo Facebook logo Google logo Mendeley logo

Related Articles

Cited By...

Subjects

  • Basic, Translational, and Clinical Research
    • Inflammation
    • Vascular Biology
    • Growth Factors/Cytokines
  • Vascular Disease
    • Vascular Disease
    • Atherosclerosis

Circulation

  • About Circulation
  • Instructions for Authors
  • Circulation CME
  • Statements and Guidelines
  • Meeting Abstracts
  • Permissions
  • Journal Policies
  • Email Alerts
  • Open Access Information
  • AHA Journals RSS
  • AHA Newsroom

Editorial Office Address:
200 Fifth Avenue, Suite 1020
Waltham, MA 02451
email: circ@circulationjournal.org
 

Information for:
  • Advertisers
  • Subscribers
  • Subscriber Help
  • Institutions / Librarians
  • Institutional Subscriptions FAQ
  • International Users
American Heart Association Learn and Live
National Center
7272 Greenville Ave.
Dallas, TX 75231

Customer Service

  • 1-800-AHA-USA-1
  • 1-800-242-8721
  • Local Info
  • Contact Us

About Us

Our mission is to build healthier lives, free of cardiovascular diseases and stroke. That single purpose drives all we do. The need for our work is beyond question. Find Out More about the American Heart Association

  • Careers
  • SHOP
  • Latest Heart and Stroke News
  • AHA/ASA Media Newsroom

Our Sites

  • American Heart Association
  • American Stroke Association
  • For Professionals
  • More Sites

Take Action

  • Advocate
  • Donate
  • Planned Giving
  • Volunteer

Online Communities

  • AFib Support
  • Garden Community
  • Patient Support Network
  • Professional Online Network

Follow Us:

  • Follow Circulation on Twitter
  • Visit Circulation on Facebook
  • Follow Circulation on Google Plus
  • Follow Circulation on Instagram
  • Follow Circulation on Pinterest
  • Follow Circulation on YouTube
  • Rss Feeds
  • Privacy Policy
  • Copyright
  • Ethics Policy
  • Conflict of Interest Policy
  • Linking Policy
  • Diversity
  • Careers

©2018 American Heart Association, Inc. All rights reserved. Unauthorized use prohibited. The American Heart Association is a qualified 501(c)(3) tax-exempt organization.
*Red Dress™ DHHS, Go Red™ AHA; National Wear Red Day ® is a registered trademark.

  • PUTTING PATIENTS FIRST National Health Council Standards of Excellence Certification Program
  • BBB Accredited Charity
  • Comodo Secured