Your browser doesn't support javascript.
loading
The Pericytic Phenotype of Adipose Tissue-Derived Stromal Cells Is Promoted by NOTCH2.
Terlizzi, Vincenzo; Kolibabka, Matthias; Burgess, Janette Kay; Hammes, Hans Peter; Harmsen, Martin Conrad.
Afiliação
  • Terlizzi V; Lab for Cardiovascular Regenerative Medicine (CAVAREM), Department of Pathology and Medical Biology, University of Groningen, Groningen, The Netherlands.
  • Kolibabka M; 5th Medical Department, Section of Endocrinology, Medical Faculty Mannheim, University of Heidelberg, Germany.
  • Burgess JK; Research Institute W.J.Kolff, University of Groningen, Groningen, The Netherlands.
  • Hammes HP; 5th Medical Department, Section of Endocrinology, Medical Faculty Mannheim, University of Heidelberg, Germany.
  • Harmsen MC; Research Institute W.J.Kolff, University of Groningen, Groningen, The Netherlands.
Stem Cells ; 36(2): 240-251, 2018 02.
Article em En | MEDLINE | ID: mdl-29067740
ABSTRACT
Long-term diabetes leads to macrovascular and microvascular complication. In diabetic retinopathy (DR), persistent hyperglycemia causes permanent loss of retinal pericytes and aberrant proliferation of microvascular endothelial cells (ECs). Adipose tissue-derived stromal cells (ASCs) may serve to functionally replace retinal pericytes and normalize retinal microvasculature during disease progression. We hypothesized that Notch signaling in ASC underlies regulation and stabilization of dysfunctional retinal microvascular networks such as in DR. ASC prominently and constitutively expressed NOTCH2. Genetic knockdown of NOTCH2 in ASC (SH-NOTCH2) disturbed the formation of vascular networks of human umbilical cord vein endothelial cells both on monolayers of ASC and in organotypical three-dimensional cocultures with ASC. On ASC SH-NOTCH2, cell surface platelet-derived growth factor receptor beta was downregulated which disrupted their migration toward the chemoattractant platelet-derived growth factor beta subunits (PDGF-BB) as well as to conditioned media from EC and bovine retinal EC. This chemoattractant is secreted by pro-angiogenic EC in newly formed microvascular networks to attract pericytes. Intravitreal injected ASC SH-NOTCH2 in oxygen-induced retinopathy mouse eyes did not engraft in the preexisting retinal microvasculature. However, the in vivo pro-angiogenic capacity of ASC SH-NOTCH2 did not differ from controls. In this respect, multifocal electroretinography displayed similar b-wave amplitudes in the avascular zones when either wild type ASC or SH-NOTCH2 ASC were injected. In conclusion, our results indicate that NOTCH2 is essential to support in vitro vasculogenesis via juxtacrine interactions. In contrast, ongoing in vivo angiogenesis is influenced by paracrine signaling of ASC, irrespective of Notch signaling. Stem Cells 2018;36240-251.
Assuntos
Palavras-chave

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Tecido Adiposo / Células Estromais / Pericitos / Receptor Notch2 Idioma: En Ano de publicação: 2018 Tipo de documento: Article

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Tecido Adiposo / Células Estromais / Pericitos / Receptor Notch2 Idioma: En Ano de publicação: 2018 Tipo de documento: Article