Abstract

AimsVein grafts are frequently used to bypass coronary artery occlusions. Unfortunately, vein graft disease (VGD) causes impaired patency rates. ALK1 mediates signaling by TGF-β via TGFβR2 or BMP9/10 via BMPR2, which is an important pathway in fibrotic, inflammatory, and angiogenic processes in vascular diseases. The role of the TGF-β pathway in VGD is previously reported, however, the contribution of ALK1 signaling is not known. Therefore, we investigated ALK1 signaling in VGD in a mouse model for vein graft disease using either genetic or pharmacological inhibition of the Alk1 signaling.Methods and ResultsMale ALK1 heterozygous (ALK1+/−), control C57BL/6, as well as hypercholesterolemic ApoE3*Leiden mice, underwent vein graft surgery. Histologic analyses of ALK1+/− vein grafts demonstrated increased outward remodeling and macrophage accumulation after 28 days. In hypercholesterolemic ApoE3*Leiden mice receiving weekly ALK1-Fc injections, ultrasound imaging showed 3-fold increased outward remodeling compared to controls treated with control-Fc, which was confirmed histologically. Moreover, ALK1-Fc treatment reduced collagen and smooth muscle cell accumulation, increased macrophages by 1.5-fold, and resulted in more plaque dissections. No difference was observed in intraplaque neovessel density. Flow cytometric analysis showed increased systemic levels of Ly6CHigh monocytes in ALK1-Fc treated mice, supported by in vitro increased MCP-1 and IL-6 production of LPS-stimulated and ALK1-Fc-treated murine monocytes and macrophages.ConclusionReduced ALK1 signaling in VGD promotes outward remodeling, increases macrophage influx, and promotes an unstable plaque phenotype.Translational PerspectiveVein graft disease (VGD) severely hampers patency rates of vein grafts, necessitating research of key disease-driving pathways like TGF-β. The three-dimensional nature of VGD together with the multitude of disease driving factors ask for a comprehensive approach. Here, we combined in vivo ultrasound imaging, histological analyses, and conventional in vitro analyses, identifying the ambiguous role of reduced ALK1 signaling in vein graft disease. Reduced ALK1 signaling promotes outward remodeling, increases macrophage influx, and promotes an unstable plaque phenotype in murine vein grafts. Characterization of in vivo vascular remodeling over time is imperative to monitor VGD development and identify new therapies.

Highlights

  • The vena saphena magna is the commonly used vein for bypass surgery in coronary or peripheral arterial circulation in patients with cardiovascular disease

  • A significant increase in plaque macrophages was found in the ALK1+/− mice in comparison to the ALK1+/+ animals (p = 0.03) (Figures 1F,I)

  • Since ALK1 heterozygosity and ALK1-Fc treatment had a profound effect on macrophages in vivo, we studied the effects of ALK1 on monocytes and macrophages in more detail, in particular, zooming in on pro-inflammatory cytokines

Read more

Summary

Introduction

The vena saphena magna is the commonly used vein for bypass surgery in coronary or peripheral arterial circulation in patients with cardiovascular disease. The vein graft requires arterialization to adapt to the pressure of the arterial circulation. Critical processes for arterialization and successful long-term patency of the grafts are moderate intimal hyperplasia (IH) combined with outward remodeling [2]. The beneficial process of proportional outward remodeling is often accompanied by moderate inflammation and angiogenesis [1, 3, 4]. Excessive forms and overshooting of these mechanisms may induce disproportionate IH, causing inward, frequently partially occluding, vascular remodeling, and subsequent blood flow reduction. These processes together are indicated as vein graft disease (VGD).

Methods
Results
Discussion
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