English
 
Help Privacy Policy Disclaimer
  Advanced SearchBrowse

Item

ITEM ACTIONSEXPORT

Released

Journal Article

A Subpopulation of Stromal Cells Controls Cancer Cell Homing to the Bone Marrow

MPS-Authors
/persons/resource/persons132138

Rossnagl,  Stephanie
Nakchbandi, Inaam / Translational Medicine, Max Planck Institute of Biochemistry, Max Planck Society;

/persons/resource/persons209081

Ghura,  Hiba
Nakchbandi, Inaam / Translational Medicine, Max Planck Institute of Biochemistry, Max Planck Society;

/persons/resource/persons215299

Groth,  Christopher
Nakchbandi, Inaam / Translational Medicine, Max Planck Institute of Biochemistry, Max Planck Society;

/persons/resource/persons145290

Altrock,  Eva
Nakchbandi, Inaam / Translational Medicine, Max Planck Institute of Biochemistry, Max Planck Society;

/persons/resource/persons78436

Nakchbandi,  Inaam
Nakchbandi, Inaam / Translational Medicine, Max Planck Institute of Biochemistry, Max Planck Society;

External Resource
No external resources are shared
Fulltext (restricted access)
There are currently no full texts shared for your IP range.
Fulltext (public)
There are no public fulltexts stored in PuRe
Supplementary Material (public)
There is no public supplementary material available
Citation

Rossnagl, S., Ghura, H., Groth, C., Altrock, E., Jakob, F., Schott, S., et al. (2018). A Subpopulation of Stromal Cells Controls Cancer Cell Homing to the Bone Marrow. Cancer Research, 78(1), 129-142. doi:10.1158/0008-5472.CAN-16-3507.


Cite as: https://hdl.handle.net/21.11116/0000-0000-6DFA-D
Abstract
Breast and prostate cancer cells home to the bone marrow, where they presumably hijack the hematopoietic stem cell niche. We characterize here the elusive premetastatic niche by examining the role of mesenchymal stromal cells (MSC) in cancer cell homing. Decreasing the number of MSC pharmacologically enhanced cancer cell homing to the bone marrow in mice. In contrast, increasing the number of these MSCs by various interventions including G-CSF administration diminished cancer cell homing. The MSC subpopulation that correlated best with cancer cells expressed stem, endothelial, and pericytic cell markers, suggesting these cells represent an undifferentiated component of the niche with vascular commitment. In humans, a MSC subpopulation carrying markers for endothelial and pericytic cells was lower in the presence of cytokeratin(+) cells in bone marrow. Taken together, our data show that a subpopulation of MSC with both endothelial and pericytic cell surface markers suppresses the homing of cancer cells to the bone marrow. Similar to the presence of cytokeratin(+) cells in the bone marrow, this MSC subpopulation could prove useful in determining the risk of metastatic disease, and its manipulation might offer a new possibility for diminishing bone metastasis formation. (C) 2017 AACR.