Adgrf5 contributes to patterning of the endothelial deep layer in retina

Standard

Adgrf5 contributes to patterning of the endothelial deep layer in retina. / Niaudet, C; Petkova, M; Jung, B; Lu, S; Laviña, B; Offermanns, S; Brakebusch, C; Betsholtz, C.

In: ANGIOGENESIS, Vol. 22, No. 4, 11.2019, p. 491-505.

Research output: SCORING: Contribution to journalSCORING: Journal articleResearchpeer-review

Harvard

Niaudet, C, Petkova, M, Jung, B, Lu, S, Laviña, B, Offermanns, S, Brakebusch, C & Betsholtz, C 2019, 'Adgrf5 contributes to patterning of the endothelial deep layer in retina', ANGIOGENESIS, vol. 22, no. 4, pp. 491-505. https://doi.org/10.1007/s10456-019-09674-0

APA

Niaudet, C., Petkova, M., Jung, B., Lu, S., Laviña, B., Offermanns, S., Brakebusch, C., & Betsholtz, C. (2019). Adgrf5 contributes to patterning of the endothelial deep layer in retina. ANGIOGENESIS, 22(4), 491-505. https://doi.org/10.1007/s10456-019-09674-0

Vancouver

Niaudet C, Petkova M, Jung B, Lu S, Laviña B, Offermanns S et al. Adgrf5 contributes to patterning of the endothelial deep layer in retina. ANGIOGENESIS. 2019 Nov;22(4):491-505. https://doi.org/10.1007/s10456-019-09674-0

Bibtex

@article{59d3d8f3a4b84e07be4101b52fb9a9ce,
title = "Adgrf5 contributes to patterning of the endothelial deep layer in retina",
abstract = "Neovascularization of the inner retinal space is a major cause of vision loss. In retinal angiomatous proliferation (RAP) syndrome, newly formed vessels originate from the retinal plexus and invade the inner retinal space. However, the molecular pathways preventing subretinal vascularization remain largely unknown. In most murine models of RAP, pathological neovascularization occurs concomitantly with the development of the retinal vasculature. Here, we demonstrate that disturbing the sequence of morphogenetic events that shape the three-layered retinal vascular network leads to subretinal vascularization. Sprouts emerging from the perivenous region after the first postnatal week extended toward the retinal space where they merged into the deep layer. The small GTPase Rac1 was required for the formation of these vascular extensions and the vascular inner plexus is formed coaxially to the overarching veins. The adhesion receptor Adgrf5 was highly expressed in the endothelium of the central nervous system, where it regulates blood-brain barrier formation. The vascular superficial plexus of Adgrf5 mutant mouse retinae exhibited an increased vascular density in the perivenous areas with increased projections toward the inner plexus where they subsequently created hyper-dense endothelial cells (EC) clusters. Disturbing the perivenous pool of EC thus significantly altered the inner plexus formation. These abnormalities culminated in transient vascular protrusions in the inner retinal space. Taken together, these results reveal a previously unobserved vascular morphogenetic defect in Adgrf5 knockout mice, implicating a role for ADGRF5 in the initiation of subretinal vascularization. Our findings also illustrate how vein-derived EC shape the inner retinal layer formation and could control the appearance of angiomatous malformations.",
keywords = "Animals, Endothelium, Vascular/metabolism, Mice, Mice, Knockout, Receptors, G-Protein-Coupled/genetics, Retina/metabolism, Retinal Neovascularization/metabolism",
author = "C Niaudet and M Petkova and B Jung and S Lu and B Lavi{\~n}a and S Offermanns and C Brakebusch and C Betsholtz",
year = "2019",
month = nov,
doi = "10.1007/s10456-019-09674-0",
language = "English",
volume = "22",
pages = "491--505",
journal = "ANGIOGENESIS",
issn = "0969-6970",
publisher = "Springer Netherlands",
number = "4",

}

RIS

TY - JOUR

T1 - Adgrf5 contributes to patterning of the endothelial deep layer in retina

AU - Niaudet, C

AU - Petkova, M

AU - Jung, B

AU - Lu, S

AU - Laviña, B

AU - Offermanns, S

AU - Brakebusch, C

AU - Betsholtz, C

PY - 2019/11

Y1 - 2019/11

N2 - Neovascularization of the inner retinal space is a major cause of vision loss. In retinal angiomatous proliferation (RAP) syndrome, newly formed vessels originate from the retinal plexus and invade the inner retinal space. However, the molecular pathways preventing subretinal vascularization remain largely unknown. In most murine models of RAP, pathological neovascularization occurs concomitantly with the development of the retinal vasculature. Here, we demonstrate that disturbing the sequence of morphogenetic events that shape the three-layered retinal vascular network leads to subretinal vascularization. Sprouts emerging from the perivenous region after the first postnatal week extended toward the retinal space where they merged into the deep layer. The small GTPase Rac1 was required for the formation of these vascular extensions and the vascular inner plexus is formed coaxially to the overarching veins. The adhesion receptor Adgrf5 was highly expressed in the endothelium of the central nervous system, where it regulates blood-brain barrier formation. The vascular superficial plexus of Adgrf5 mutant mouse retinae exhibited an increased vascular density in the perivenous areas with increased projections toward the inner plexus where they subsequently created hyper-dense endothelial cells (EC) clusters. Disturbing the perivenous pool of EC thus significantly altered the inner plexus formation. These abnormalities culminated in transient vascular protrusions in the inner retinal space. Taken together, these results reveal a previously unobserved vascular morphogenetic defect in Adgrf5 knockout mice, implicating a role for ADGRF5 in the initiation of subretinal vascularization. Our findings also illustrate how vein-derived EC shape the inner retinal layer formation and could control the appearance of angiomatous malformations.

AB - Neovascularization of the inner retinal space is a major cause of vision loss. In retinal angiomatous proliferation (RAP) syndrome, newly formed vessels originate from the retinal plexus and invade the inner retinal space. However, the molecular pathways preventing subretinal vascularization remain largely unknown. In most murine models of RAP, pathological neovascularization occurs concomitantly with the development of the retinal vasculature. Here, we demonstrate that disturbing the sequence of morphogenetic events that shape the three-layered retinal vascular network leads to subretinal vascularization. Sprouts emerging from the perivenous region after the first postnatal week extended toward the retinal space where they merged into the deep layer. The small GTPase Rac1 was required for the formation of these vascular extensions and the vascular inner plexus is formed coaxially to the overarching veins. The adhesion receptor Adgrf5 was highly expressed in the endothelium of the central nervous system, where it regulates blood-brain barrier formation. The vascular superficial plexus of Adgrf5 mutant mouse retinae exhibited an increased vascular density in the perivenous areas with increased projections toward the inner plexus where they subsequently created hyper-dense endothelial cells (EC) clusters. Disturbing the perivenous pool of EC thus significantly altered the inner plexus formation. These abnormalities culminated in transient vascular protrusions in the inner retinal space. Taken together, these results reveal a previously unobserved vascular morphogenetic defect in Adgrf5 knockout mice, implicating a role for ADGRF5 in the initiation of subretinal vascularization. Our findings also illustrate how vein-derived EC shape the inner retinal layer formation and could control the appearance of angiomatous malformations.

KW - Animals

KW - Endothelium, Vascular/metabolism

KW - Mice

KW - Mice, Knockout

KW - Receptors, G-Protein-Coupled/genetics

KW - Retina/metabolism

KW - Retinal Neovascularization/metabolism

U2 - 10.1007/s10456-019-09674-0

DO - 10.1007/s10456-019-09674-0

M3 - SCORING: Journal article

C2 - 31256320

VL - 22

SP - 491

EP - 505

JO - ANGIOGENESIS

JF - ANGIOGENESIS

SN - 0969-6970

IS - 4

ER -