Differentiated smooth muscle cells generate a subpopulation of resident vascular progenitor cells in the adventitia regulated by Klf4

MW Majesky, H Horita, A Ostriker, S Lu… - Circulation …, 2017 - Am Heart Assoc
MW Majesky, H Horita, A Ostriker, S Lu, JN Regan, A Bagchi, XR Dong, J Poczobutt…
Circulation research, 2017Am Heart Assoc
Rationale: The vascular adventitia is a complex layer of the vessel wall consisting of vasa
vasorum microvessels, nerves, fibroblasts, immune cells, and resident progenitor cells.
Adventitial progenitors express the stem cell markers, Sca1 and CD34 (adventitial sca1-
positive progenitor cells [AdvSca1]), have the potential to differentiate in vitro into multiple
lineages, and potentially contribute to intimal lesions in vivo. Objective: Although emerging
data support the existence of AdvSca1 cells, the goal of this study was to determine their …
Rationale:
The vascular adventitia is a complex layer of the vessel wall consisting of vasa vasorum microvessels, nerves, fibroblasts, immune cells, and resident progenitor cells. Adventitial progenitors express the stem cell markers, Sca1 and CD34 (adventitial sca1-positive progenitor cells [AdvSca1]), have the potential to differentiate in vitro into multiple lineages, and potentially contribute to intimal lesions in vivo.
Objective:
Although emerging data support the existence of AdvSca1 cells, the goal of this study was to determine their origin, degree of multipotency and heterogeneity, and contribution to vessel remodeling.
Methods and Results:
Using 2 in vivo fate-mapping approaches combined with a smooth muscle cell (SMC) epigenetic lineage mark, we report that a subpopulation of AdvSca1 cells is generated in situ from differentiated SMCs. Our data establish that the vascular adventitia contains phenotypically distinct subpopulations of progenitor cells expressing SMC, myeloid, and hematopoietic progenitor-like properties and that differentiated SMCs are a source to varying degrees of each subpopulation. SMC-derived AdvSca1 cells exhibit a multipotent phenotype capable of differentiating in vivo into mature SMCs, resident macrophages, and endothelial-like cells. After vascular injury, SMC-derived AdvSca1 cells expand in number and are major contributors to adventitial remodeling. Induction of the transcription factor Klf4 in differentiated SMCs is essential for SMC reprogramming in vivo, whereas in vitro approaches demonstrate that Klf4 is essential for the maintenance of the AdvSca1 progenitor phenotype.
Conclusions:
We propose that generation of resident vascular progenitor cells from differentiated SMCs is a normal physiological process that contributes to the vascular stem cell pool and plays important roles in arterial homeostasis and disease.
Am Heart Assoc