Your browser doesn't support javascript.
loading
Apelin-driven endothelial cell migration sustains intestinal progenitor cells and tumor growth.
Bernier-Latmani, Jeremiah; Cisarovsky, Christophe; Mahfoud, Samantha; Ragusa, Simone; Dupanloup, Isabelle; Barras, David; Renevey, François; Nassiri, Sina; Anderle, Pascale; Squadrito, Mario Leonardo; Siegert, Stefanie; Davanture, Suzel; González-Loyola, Alejandra; Fournier, Nadine; Luther, Sanjiv A; Benedito, Rui; Valet, Philippe; Zhou, Bin; De Palma, Michele; Delorenzi, Mauro; Sempoux, Christine; Petrova, Tatiana V.
  • Bernier-Latmani J; Department of Oncology, Ludwig Center for Cancer Research Lausanne and University of Lausanne, Lausanne, Switzerland.
  • Cisarovsky C; Department of Oncology, Ludwig Center for Cancer Research Lausanne and University of Lausanne, Lausanne, Switzerland.
  • Mahfoud S; Department of Oncology, Ludwig Center for Cancer Research Lausanne and University of Lausanne, Lausanne, Switzerland.
  • Ragusa S; Department of Oncology, Ludwig Center for Cancer Research Lausanne and University of Lausanne, Lausanne, Switzerland.
  • Dupanloup I; Bioinformatics Core Facility, SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland.
  • Barras D; Department of Oncology, Ludwig Center for Cancer Research Lausanne and University of Lausanne, Lausanne, Switzerland.
  • Renevey F; Bioinformatics Core Facility, SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland.
  • Nassiri S; Department of Biochemistry, University of Lausanne, Lausanne, Switzerland.
  • Anderle P; Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, EPFL, Lausanne, Switzerland.
  • Squadrito ML; Bioinformatics Core Facility, SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland.
  • Siegert S; Bioinformatics Core Facility, SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland.
  • Davanture S; Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, EPFL, Lausanne, Switzerland.
  • González-Loyola A; Department of Biochemistry, University of Lausanne, Lausanne, Switzerland.
  • Fournier N; Department of Oncology, Ludwig Center for Cancer Research Lausanne and University of Lausanne, Lausanne, Switzerland.
  • Luther SA; Department of Oncology, Ludwig Center for Cancer Research Lausanne and University of Lausanne, Lausanne, Switzerland.
  • Benedito R; Bioinformatics Core Facility, SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland.
  • Valet P; Department of Biochemistry, University of Lausanne, Lausanne, Switzerland.
  • Zhou B; Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain.
  • De Palma M; Institut RESTORE, UMR 1301-INSERM, 5070-CNRS, Université Paul Sabatier, Université de Toulouse, Toulouse, France.
  • Delorenzi M; State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, China.
  • Sempoux C; Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, EPFL, Lausanne, Switzerland.
  • Petrova TV; Department of Oncology, Ludwig Center for Cancer Research Lausanne and University of Lausanne, Lausanne, Switzerland.
Nat Cardiovasc Res ; 1(5): 476-490, 2022 May.
Article en En | MEDLINE | ID: mdl-35602406
ABSTRACT
Stem and progenitor cells residing in the intestinal crypts drive the majority of colorectal cancers (CRCs), yet vascular contribution to this niche remains largely unexplored. VEGFA is a key driver of physiological and tumor angiogenesis. Accordingly, current anti-angiogenic cancer therapies target the VEGFA pathway. Here we report that in CRC expansion of the stem/progenitor pool in intestinal crypts requires VEGFA-independent growth and remodeling of blood vessels. Epithelial transformation induced expression of the endothelial peptide apelin, directs migration of distant venous endothelial cells towards progenitor niche vessels ensuring optimal perfusion. In the absence of apelin, loss of injury-inducible PROX1+ epithelial progenitors inhibited both incipient and advanced intestinal tumor growth. Our results establish fundamental principles for the reciprocal communication between vasculature and the intestinal progenitor niche and provide a mechanism for resistance to VEGFA-targeting drugs in CRCs.