Role of lipid phosphate phosphatase 3 in human aortic endothelial cell function

Standard

Role of lipid phosphate phosphatase 3 in human aortic endothelial cell function. / Touat-Hamici, Zahia; Weidmann, Henri; Blum, Yuna; Proust, Carole; Durand, Hervé; Iannacci, Francesca; Codoni, Veronica; Gaignard, Pauline; Thérond, Patrice; Civelek, Mete; Karabina, Sonia A; Lusis, Aldons J; Cambien, François; Ninio, Ewa.

in: CARDIOVASC RES, Jahrgang 112, Nr. 3, 12.2016, S. 702-713.

Publikationen: SCORING: Beitrag in Fachzeitschrift/ZeitungSCORING: ZeitschriftenaufsatzForschungBegutachtung

Harvard

Touat-Hamici, Z, Weidmann, H, Blum, Y, Proust, C, Durand, H, Iannacci, F, Codoni, V, Gaignard, P, Thérond, P, Civelek, M, Karabina, SA, Lusis, AJ, Cambien, F & Ninio, E 2016, 'Role of lipid phosphate phosphatase 3 in human aortic endothelial cell function', CARDIOVASC RES, Jg. 112, Nr. 3, S. 702-713. https://doi.org/10.1093/cvr/cvw217

APA

Touat-Hamici, Z., Weidmann, H., Blum, Y., Proust, C., Durand, H., Iannacci, F., Codoni, V., Gaignard, P., Thérond, P., Civelek, M., Karabina, S. A., Lusis, A. J., Cambien, F., & Ninio, E. (2016). Role of lipid phosphate phosphatase 3 in human aortic endothelial cell function. CARDIOVASC RES, 112(3), 702-713. https://doi.org/10.1093/cvr/cvw217

Vancouver

Touat-Hamici Z, Weidmann H, Blum Y, Proust C, Durand H, Iannacci F et al. Role of lipid phosphate phosphatase 3 in human aortic endothelial cell function. CARDIOVASC RES. 2016 Dez;112(3):702-713. https://doi.org/10.1093/cvr/cvw217

Bibtex

@article{d3de39ca0d454750932ade72b35ba606,
title = "Role of lipid phosphate phosphatase 3 in human aortic endothelial cell function",
abstract = "AIMS: Lipid phosphate phosphatase 3; type 2 phosphatidic acid phosphatase β (LPP3; PPAP2B) is a transmembrane protein dephosphorylating and thereby terminating signalling of lipid substrates including lysophosphatidic acid (LPA) and sphingosine-1-phosphate (S1P). Human LPP3 possesses a cell adhesion motif that allows interaction with integrins. A polymorphism (rs17114036) in PPAP2B is associated with coronary artery disease, which prompted us to investigate the possible role of LPP3 in human endothelial dysfunction, a condition promoting atherosclerosis.METHODS AND RESULTS: To study the role of LPP3 in endothelial cells we used human primary aortic endothelial cells (HAECs) in which LPP3 was silenced or overexpressed using either wild type or mutated cDNA constructs. LPP3 silencing in HAECs enhanced secretion of inflammatory cytokines, leucocyte adhesion, cell survival, and migration and impaired angiogenesis, whereas wild-type LPP3 overexpression reversed these effects and induced apoptosis. We also demonstrated that LPP3 expression was negatively correlated with vascular endothelial growth factor expression. Mutations in either the catalytic or the arginine-glycine-aspartate (RGD) domains impaired endothelial cell function and pharmacological inhibition of S1P or LPA restored it. LPA was not secreted in HAECs under silencing or overexpressing LPP3. However, the intra- and extra-cellular levels of S1P tended to be correlated with LPP3 expression, indicating that S1P is probably degraded by LPP3.CONCLUSIONS: We demonstrated that LPP3 is a negative regulator of inflammatory cytokines, leucocyte adhesion, cell survival, and migration in HAECs, suggesting a protective role of LPP3 against endothelial dysfunction in humans. Both the catalytic and the RGD functional domains were involved and S1P, but not LPA, might be the endogenous substrate of LPP3.",
author = "Zahia Touat-Hamici and Henri Weidmann and Yuna Blum and Carole Proust and Herv{\'e} Durand and Francesca Iannacci and Veronica Codoni and Pauline Gaignard and Patrice Th{\'e}rond and Mete Civelek and Karabina, {Sonia A} and Lusis, {Aldons J} and Fran{\c c}ois Cambien and Ewa Ninio",
note = "Published on behalf of the European Society of Cardiology. All rights reserved. {\textcopyright} The Author 2016. For Permissions, please email: journals.permissions@oup.com.",
year = "2016",
month = dec,
doi = "10.1093/cvr/cvw217",
language = "English",
volume = "112",
pages = "702--713",
journal = "CARDIOVASC RES",
issn = "0008-6363",
publisher = "Oxford University Press",
number = "3",

}

RIS

TY - JOUR

T1 - Role of lipid phosphate phosphatase 3 in human aortic endothelial cell function

AU - Touat-Hamici, Zahia

AU - Weidmann, Henri

AU - Blum, Yuna

AU - Proust, Carole

AU - Durand, Hervé

AU - Iannacci, Francesca

AU - Codoni, Veronica

AU - Gaignard, Pauline

AU - Thérond, Patrice

AU - Civelek, Mete

AU - Karabina, Sonia A

AU - Lusis, Aldons J

AU - Cambien, François

AU - Ninio, Ewa

N1 - Published on behalf of the European Society of Cardiology. All rights reserved. © The Author 2016. For Permissions, please email: journals.permissions@oup.com.

PY - 2016/12

Y1 - 2016/12

N2 - AIMS: Lipid phosphate phosphatase 3; type 2 phosphatidic acid phosphatase β (LPP3; PPAP2B) is a transmembrane protein dephosphorylating and thereby terminating signalling of lipid substrates including lysophosphatidic acid (LPA) and sphingosine-1-phosphate (S1P). Human LPP3 possesses a cell adhesion motif that allows interaction with integrins. A polymorphism (rs17114036) in PPAP2B is associated with coronary artery disease, which prompted us to investigate the possible role of LPP3 in human endothelial dysfunction, a condition promoting atherosclerosis.METHODS AND RESULTS: To study the role of LPP3 in endothelial cells we used human primary aortic endothelial cells (HAECs) in which LPP3 was silenced or overexpressed using either wild type or mutated cDNA constructs. LPP3 silencing in HAECs enhanced secretion of inflammatory cytokines, leucocyte adhesion, cell survival, and migration and impaired angiogenesis, whereas wild-type LPP3 overexpression reversed these effects and induced apoptosis. We also demonstrated that LPP3 expression was negatively correlated with vascular endothelial growth factor expression. Mutations in either the catalytic or the arginine-glycine-aspartate (RGD) domains impaired endothelial cell function and pharmacological inhibition of S1P or LPA restored it. LPA was not secreted in HAECs under silencing or overexpressing LPP3. However, the intra- and extra-cellular levels of S1P tended to be correlated with LPP3 expression, indicating that S1P is probably degraded by LPP3.CONCLUSIONS: We demonstrated that LPP3 is a negative regulator of inflammatory cytokines, leucocyte adhesion, cell survival, and migration in HAECs, suggesting a protective role of LPP3 against endothelial dysfunction in humans. Both the catalytic and the RGD functional domains were involved and S1P, but not LPA, might be the endogenous substrate of LPP3.

AB - AIMS: Lipid phosphate phosphatase 3; type 2 phosphatidic acid phosphatase β (LPP3; PPAP2B) is a transmembrane protein dephosphorylating and thereby terminating signalling of lipid substrates including lysophosphatidic acid (LPA) and sphingosine-1-phosphate (S1P). Human LPP3 possesses a cell adhesion motif that allows interaction with integrins. A polymorphism (rs17114036) in PPAP2B is associated with coronary artery disease, which prompted us to investigate the possible role of LPP3 in human endothelial dysfunction, a condition promoting atherosclerosis.METHODS AND RESULTS: To study the role of LPP3 in endothelial cells we used human primary aortic endothelial cells (HAECs) in which LPP3 was silenced or overexpressed using either wild type or mutated cDNA constructs. LPP3 silencing in HAECs enhanced secretion of inflammatory cytokines, leucocyte adhesion, cell survival, and migration and impaired angiogenesis, whereas wild-type LPP3 overexpression reversed these effects and induced apoptosis. We also demonstrated that LPP3 expression was negatively correlated with vascular endothelial growth factor expression. Mutations in either the catalytic or the arginine-glycine-aspartate (RGD) domains impaired endothelial cell function and pharmacological inhibition of S1P or LPA restored it. LPA was not secreted in HAECs under silencing or overexpressing LPP3. However, the intra- and extra-cellular levels of S1P tended to be correlated with LPP3 expression, indicating that S1P is probably degraded by LPP3.CONCLUSIONS: We demonstrated that LPP3 is a negative regulator of inflammatory cytokines, leucocyte adhesion, cell survival, and migration in HAECs, suggesting a protective role of LPP3 against endothelial dysfunction in humans. Both the catalytic and the RGD functional domains were involved and S1P, but not LPA, might be the endogenous substrate of LPP3.

U2 - 10.1093/cvr/cvw217

DO - 10.1093/cvr/cvw217

M3 - SCORING: Journal article

C2 - 27694435

VL - 112

SP - 702

EP - 713

JO - CARDIOVASC RES

JF - CARDIOVASC RES

SN - 0008-6363

IS - 3

ER -