Your browser doesn't support javascript.
loading
Breast cancer cells promote a notch-dependent mesenchymal phenotype in endothelial cells participating to a pro-tumoral niche.
Ghiabi, Pegah; Jiang, Jie; Pasquier, Jennifer; Maleki, Mahtab; Abu-Kaoud, Nadine; Halabi, Najeeb; Guerrouahen, Bella S; Rafii, Shahin; Rafii, Arash.
Afiliação
  • Ghiabi P; Stem Cell and Microenvironment Laboratory, Weill Cornell Medical College in Qatar, Education City, Qatar Foundation, P.O. Box 24144, Doha, Qatar. Peg2006@qatar-med.cornell.edu.
  • Jiang J; Department of Genetic Medicine, Weill Cornell Medical College, New york city, NY, USA. Jij2007@med.cornell.edu.
  • Pasquier J; Stem Cell and Microenvironment Laboratory, Weill Cornell Medical College in Qatar, Education City, Qatar Foundation, P.O. Box 24144, Doha, Qatar. Jep2026@qatar-med.cornell.edu.
  • Maleki M; Department of Genetic Medicine, Weill Cornell Medical College, New york city, NY, USA. Jep2026@qatar-med.cornell.edu.
  • Abu-Kaoud N; Stem Cell and Microenvironment Laboratory, Weill Cornell Medical College in Qatar, Education City, Qatar Foundation, P.O. Box 24144, Doha, Qatar. Mam2116@qatar-med.cornell.edu.
  • Halabi N; Stem Cell and Microenvironment Laboratory, Weill Cornell Medical College in Qatar, Education City, Qatar Foundation, P.O. Box 24144, Doha, Qatar. Naa2037@qatar-med.cornell.edu.
  • Guerrouahen BS; Stem Cell and Microenvironment Laboratory, Weill Cornell Medical College in Qatar, Education City, Qatar Foundation, P.O. Box 24144, Doha, Qatar. Nah2024@qatar-med.cornell.edu.
  • Rafii S; Department of Genetic Medicine, Weill Cornell Medical College, New york city, NY, USA. Nah2024@qatar-med.cornell.edu.
  • Rafii A; Stem Cell and Microenvironment Laboratory, Weill Cornell Medical College in Qatar, Education City, Qatar Foundation, P.O. Box 24144, Doha, Qatar. Bsg2004@qatar-med.cornell.edu.
J Transl Med ; 13: 27, 2015 Jan 27.
Article em En | MEDLINE | ID: mdl-25623554
ABSTRACT

BACKGROUND:

Endothelial cells (ECs) are responsible for creating a tumor vascular niche as well as producing angiocrine factors. ECs demonstrate functional and phenotypic heterogeneity when located under different microenvironments. Here, we describe a tumor-stimulated mesenchymal phenotype in ECs and investigate its impact on tumor growth, stemness, and invasiveness.

METHODS:

Xenograft tumor assay in NOD/SCID mice and confocal imaging were conducted to show the acquisition of mesenchymal phenotype in tumor-associated ECs in vivo. Immunocytochemistry, qPCR and flow cytometry techniques showed the appearance of mesenchymal traits in ECs after contact with breast tumor cell lines MDA-MB231 or MCF-7. Cell proliferation, cell migration, and sphere formation assays were applied to display the functional advantages of mesenchymal ECs in tumor growth, invasiveness, and enrichment of tumor initiating cells. qPCR and western blotting were used to investigate the mechanisms underlying EC mesenchymal transition.

RESULTS:

Our results showed that co-injection of ECs and tumor cells in NOD/SCID mice significantly enhanced tumor growth in vivo with tumor-associated ECs expressing mesenchymal markers while maintaining their intrinsic endothelial trait. We also showed that a mesenchymal phenotype is possibly detectable in human neoplastic breast biopsies as well as ECs pre-exposed to tumor cells (ECs(Mes)) in vitro. The ECs(Mes) acquired prolonged survival, increased migratory behavior and enhanced angiogenic properties. In return, ECs(Mes) were capable of enhancing tumor survival and invasiveness. The mesenchymal phenotypes in ECs(Mes) were the result of a contact-dependent transient phenomenon and reversed upon removal of the neoplastic contexture. We showed a synergistic role for TGFß and notch pathways in this phenotypic change, as simultaneous inhibition of notch and TGFß down-regulated Smad1/5 phosphorylation and Jag1(KD) tumor cells were unable to initiate the process.

CONCLUSIONS:

Overall, our data proposed a crosstalk mechanism between tumor and microenvironment where tumor-stimulated mesenchymal modulation of ECs enhanced the constitution of a transient mesenchymal/endothelial niche leading to significant increase in tumor proliferation, stemness, and invasiveness. The possible involvement of notch and TGFß pathways in the initiation of mesenchymal phenotype may propose new stromal targets.
Assuntos

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: Neoplasias da Mama / Receptores Notch / Microambiente Tumoral / Células Endoteliais da Veia Umbilical Humana / Mesoderma Tipo de estudo: Prognostic_studies Limite: Animals / Female / Humans Idioma: En Ano de publicação: 2015 Tipo de documento: Article

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: Neoplasias da Mama / Receptores Notch / Microambiente Tumoral / Células Endoteliais da Veia Umbilical Humana / Mesoderma Tipo de estudo: Prognostic_studies Limite: Animals / Female / Humans Idioma: En Ano de publicação: 2015 Tipo de documento: Article