Abstract

BackgroundPerivascular adipose tissue (PVAT) accelerates plaque progression and increases cardiovascular risk. We tested the hypothesis that PVAT contributed to plaque vulnerability and investigated whether endoplasmic reticulum stress (ER stress) in PVAT played an important role in vulnerable plaque.MethodsWe transplanted thoracic aortic PVAT or subcutaneous adipose tissue as a control, from donor mice to carotid arteries of recipient apolipoprotein E deficient (apoE−/−) mice after removing carotid artery collar placed for 6 weeks. Two weeks after transplantation, ER stress inhibitor 4-phenyl butyric acid (4-PBA) was locally administrated to the transplanted PVAT and then animals were euthanized after 4 weeks. Immunohistochemistry was performed to quantify plaque composition and neovascularization. Mouse angiogenesis antibody array kit was used to test the angiogenic factors produced by transplanted adipose tissue. In vitro tube formation assay, scratch wound migration assay and mouse aortic ring assay were used to assess the angiogenic capacity of supernatant of transplanted PVAT.ResultsUltrastructural detection by transmission electron microscopy showed transplanted PVAT was a mixed population of white and brown adipocytes with abundant mitochondria. Transplanted PVAT increased the intraplaque macrophage infiltration, lipid core, intimal and vasa vasorum neovascularization and MMP2/9 expression in plaque while decreased smooth muscle cells and collagen in atherosclerotic plaque, which were restored by local 4-PBA-treatment. Antibody array analysis showed that 4-PBA reduced several angiogenic factors [Granulocyte Macrophage Colony Stimulating Factor (GM-CSF), MCP-1, IL-6] secreted by PVAT. Besides, conditioned medium from 4-PBA treated-PVAT inhibited tube formation and migration capacity of endothelial cells and ex vivo mouse aortic ring angiogenesis compared to conditioned medium from transplanted PVAT. mRNA expression and protein levels of GM-CSF were markedly elevated in adipocytes under ER stress which would be suppressed by 4-PBA. In addition, ER stress enhanced NF-κB binding to the promoter of the mouse GM-CSF gene in adipocytes confirmed by Chromatin immunoprecipitation analyses.ConclusionsOur findings demonstrate that ER stress in PVAT destabilizes atherosclerotic plaque, in part through increasing GM-CSF paracrine via transcription factor NF-κB.

Highlights

  • Perivascular adipose tissue (PVAT) accelerates plaque progression and increases cardiovascular risk

  • We found that transplanted PVAT promoted plaque vulnerability in the setting of high-fat diet (HFD) which could be ameliorated by 4-phenyl butyric acid (4-PBA) at least in part dependent on decreased Granulocyte Macrophage Colony Stimu‐ lating Factor (GM-CSF) released locally by transplanted PVAT

  • Effects of adipose tissue transplantation To determine whether adipose tissue could contribute to characteristics of atherosclerotic plaque, ­ApoE−/− mice underwent PVAT, subcutaneous adipose tissue (SQAT) transplant or sham operation to the left common carotid artery a site that is devoid of perivascular adipose tissue and typically does not develop spontaneous atherosclerosis

Read more

Summary

Introduction

Perivascular adipose tissue (PVAT) accelerates plaque progression and increases cardiovascular risk. Scientists revealed that chemokines production from PVAT played a role in the pathogenesis of atherosclerosis [7] These molecules might contribute to the alterations of the function and structure of vascular wall, including chronic inflammation, alterations of vascular tone, smooth muscle cell dysfunction, neo-angiogenesis and to the development of atherosclerosis. Clinical observations suggest that coronary perivascular adipose tissue is related to the presence of lipid core, macrophage infiltration and severity of atherosclerotic plaque which are the characteristics of high risk plaque [13, 14] These findings urged us to hypothesize that PVAT contributes to plaque vulnerability through paracrine effects on the vasculature from ‘outside to inside’

Methods
Results
Conclusion
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call