Oncotarget

Research Papers:

Isolation and characterization of renal cancer stem cells from patient-derived xenografts

Meriem Hasmim, Stefania Bruno, Sandy Azzi, Cindy Gallerne, Julien Giron Michel, Giulia Chiabotto, Vincent Lecoz, Cristina Romei, Grazia Maria Spaggiari, Annalisa Pezzolo, Vito Pistoia, Eric Angevin, Sophie Gad, Sophie Ferlicot, Yosra Messai, Claudine Kieda, Denis Clay, Federica Sabatini, Bernard Escudier, Giovanni Camussi, Pierre Eid, Bruno Azzarone and Salem Chouaib _

PDF  |  HTML  |  How to cite

Oncotarget. 2016; 7:15507-15524. https://doi.org/10.18632/oncotarget.6266

Metrics: PDF 2738 views  |   HTML 3088 views  |   ?  


Abstract

Meriem Hasmim1,2,*, Stefania Bruno3,*, Sandy Azzi2, Cindy Gallerne2, Julien Giron Michel2, Giulia Chiabotto4, Vincent Lecoz2, Cristina Romei5, Grazia Maria Spaggiari5, Annalisa Pezzolo6, Vito Pistoia6, Eric Angevin1,7, Sophie Gad1,8, Sophie Ferlicot1,9, Yosra Messai1, Claudine Kieda10, Denis Clay11, Federica Sabatini12, Bernard Escudier1,7, Giovanni Camussi4, Pierre Eid2, Bruno Azzarone5, Salem Chouaib1

1INSERM U 1186, Equipe labellisée Ligue Contre le Cancer, Gustave Roussy Campus, Villejuif, France

2INSERM UMR 1014, Lavoisier Building, Paul Brousse Hospital, Villejuif, France

3Department of Molecular Biotechnology and Healthy Science, Molecular Biotechnology Center, University of Torino, Turin, Italy

4Department of Medical Science, University of Torino, Medical School, Torino, Italy

5DIMES, UNIGE, Genova, Italy

6Laboratory of Oncology Giannina Gaslini Institute, Genoa, Italy

7Medical Oncology Department, Gustave Roussy Campus, Villejuif, France

8Laboratoire de Génétique Oncologique EPHE, Ecole Pratique des Hautes Etudes, Paris, France

9Université Paris-Sud, Assistance Publique-Hôpitaux de Paris, Service d'Anatomo-Pathologie, Hôpital Bicêtre, Le Kremlin-Bicêtre, France

10Centre de Biophysique Moléculaire, CNRS UPR 4301, Orléans, France

11INSERM UMR 972, Paul Brousse Hospital, Villejuif, France

12Stem Cell and Cell Therapy Laboratory, Istituto G. Gaslini, Genoa, Italy

*These authors have contributed equally to this work

Correspondence to:

Salem Chouaib, e-mail: [email protected]

Bruno Azzarone, e-mail: [email protected]

Keywords: clear cell renal cell carcinoma, cancer stem cells, patient-derived xenografts, CD133, EpCAM

Received: July 24, 2015        Accepted: October 08, 2015        Published: November 02, 2015

ABSTRACT

As rapidly developing patient-derived xenografts (PDX) could represent potential sources of cancer stem cells (CSC), we selected and characterized non-cultured PDX cell suspensions from four different renal carcinomas (RCC). Only the cell suspensions from the serial xenografts (PDX-1 and PDX-2) of an undifferentiated RCC (RCC-41) adapted to the selective CSC medium. The cell suspension derived from the original tumor specimen (RCC-41-P-0) did not adapt to the selective medium and strongly expressed CSC-like markers (CD133 and CD105) together with the non-CSC tumor marker E-cadherin. In comparison, PDX-1 and PDX-2 cells exhibited evolution in their phenotype since PDX-1 cells were CD133high/CD105-/Ecadlow and PDX-2 cells were CD133low/CD105-/Ecad-. Both PDX subsets expressed additional stem cell markers (CD146/CD29/OCT4/NANOG/Nestin) but still contained non-CSC tumor cells. Therefore, using different cell sorting strategies, we characterized 3 different putative CSC subsets (RCC-41-PDX-1/CD132+, RCC-41-PDX-2/CD133-/EpCAMlow and RCC-41-PDX-2/CD133+/EpCAMbright). In addition, transcriptomic analysis showed that RCC-41-PDX-2/CD133 over-expressed the pluripotency gene ERBB4, while RCC-41-PDX-2/CD133+ over-expressed several tumor suppressor genes. These three CSC subsets displayed ALDH activity, formed serial spheroids and developed serial tumors in SCID mice, although RCC-41-PDX-1/CD132+ and RCC-41-PDX-2/CD133+ displayed less efficiently the above CSC properties. RCC-41-PDX-1/CD132+ tumors showed vessels of human origin with CSC displaying peri-vascular distribution. By contrast, RCC-41-PDX-2 originated tumors exhibiting only vessels of mouse origin without CSC peri-vascular distribution.

Altogether, our results indicate that PDX murine microenvironment promotes a continuous redesign of CSC phenotype, unmasking CSC subsets potentially present in a single RCC or generating ex novo different CSC-like subsets.


Creative Commons License All site content, except where otherwise noted, is licensed under a Creative Commons Attribution 4.0 License.
PII: 6266