scholarly journals Fast1 functions as a transcriptional switch for nodal signaling in Xenopus mesodermal development

2008 ◽  
Vol 319 (2) ◽  
pp. 524
Author(s):  
Aaron Steiner ◽  
Christine Reid ◽  
Sergey Yaklichkin ◽  
Qun Lu ◽  
Shouwen Wang ◽  
...  
2016 ◽  
Vol 414 (1) ◽  
pp. 34-44 ◽  
Author(s):  
Christine D. Reid ◽  
Aaron B. Steiner ◽  
Sergey Yaklichkin ◽  
Qun Lu ◽  
Shouwen Wang ◽  
...  

2009 ◽  
Vol 331 (2) ◽  
pp. 482
Author(s):  
Daniel S. Kessler ◽  
Aaron B. Steiner ◽  
Christine D. Reid ◽  
Sergey Yaklichkin ◽  
Qun Lu ◽  
...  

2019 ◽  
Vol 20 (8) ◽  
pp. 1916 ◽  
Author(s):  
Marc L. Sprouse ◽  
Thomas Welte ◽  
Debasish Boral ◽  
Haowen N. Liu ◽  
Wei Yin ◽  
...  

Intratumoral infiltration of myeloid-derived suppressor cells (MDSCs) is known to promote neoplastic growth by inhibiting the tumoricidal activity of T cells. However, direct interactions between patient-derived MDSCs and circulating tumors cells (CTCs) within the microenvironment of blood remain unexplored. Dissecting interplays between CTCs and circulatory MDSCs by heterotypic CTC/MDSC clustering is critical as a key mechanism to promote CTC survival and sustain the metastatic process. We characterized CTCs and polymorphonuclear-MDSCs (PMN-MDSCs) isolated in parallel from peripheral blood of metastatic melanoma and breast cancer patients by multi-parametric flow cytometry. Transplantation of both cell populations in the systemic circulation of mice revealed significantly enhanced dissemination and metastasis in mice co-injected with CTCs and PMN-MDSCs compared to mice injected with CTCs or MDSCs alone. Notably, CTC/PMN-MDSC clusters were detected in vitro and in vivo either in patients’ blood or by longitudinal monitoring of blood from animals. This was coupled with in vitro co-culturing of cell populations, demonstrating that CTCs formed physical clusters with PMN-MDSCs; and induced their pro-tumorigenic differentiation through paracrine Nodal signaling, augmenting the production of reactive oxygen species (ROS) by PMN-MDSCs. These findings were validated by detecting significantly higher Nodal and ROS levels in blood of cancer patients in the presence of naïve, heterotypic CTC/PMN-MDSC clusters. Augmented PMN-MDSC ROS upregulated Notch1 receptor expression in CTCs through the ROS-NRF2-ARE axis, thus priming CTCs to respond to ligand-mediated (Jagged1) Notch activation. Jagged1-expressing PMN-MDSCs contributed to enhanced Notch activation in CTCs by engagement of Notch1 receptor. The reciprocity of CTC/PMN-MDSC bi-directional paracrine interactions and signaling was functionally validated in inhibitor-based analyses, demonstrating that combined Nodal and ROS inhibition abrogated CTC/PMN-MDSC interactions and led to a reduction of CTC survival and proliferation. This study provides seminal evidence showing that PMN-MDSCs, additive to their immuno-suppressive roles, directly interact with CTCs and promote their dissemination and metastatic potency. Targeting CTC/PMN-MDSC heterotypic clusters and associated crosstalks can therefore represent a novel therapeutic avenue for limiting hematogenous spread of metastatic disease.


2006 ◽  
Vol 295 (2) ◽  
pp. 743-755 ◽  
Author(s):  
Anne Camus ◽  
Aitana Perea-Gomez ◽  
Anne Moreau ◽  
Jérôme Collignon

Sign in / Sign up

Export Citation Format

Share Document