2014
DOI: 10.1016/j.cell.2014.09.051
|View full text |Cite
|
Sign up to set email alerts
|

Exosome Transfer from Stromal to Breast Cancer Cells Regulates Therapy Resistance Pathways

Abstract: SUMMARY Stromal communication with cancer cells can influence treatment response. We show that stromal and breast cancer (BrCa) cells utilize paracrine and juxtacrine signaling to drive chemotherapy and radiation resistance. Upon heterotypic interaction, exosomes are transferred from stromal to BrCa cells. RNA within exosomes, which are largely non-coding transcripts and transposable elements, stimulates the pattern recognition receptor RIG-I to activate STAT1-dependent anti-viral signaling. In parallel, strom… Show more

Help me understand this report

Search citation statements

Order By: Relevance

Paper Sections

Select...
2
2
1

Citation Types

16
592
6
3

Year Published

2015
2015
2021
2021

Publication Types

Select...
9

Relationship

0
9

Authors

Journals

citations
Cited by 668 publications
(617 citation statements)
references
References 53 publications
16
592
6
3
Order By: Relevance
“…exoRNA RN7SL1 was then transferred to breast cancer cells, activated RIG‐I signaling including STAT1 activation and ISG induction, which was proposed to promote aggressive development of cancer. In another work, the same group has demonstrated that RIG‐I activation by exoRNA can amplify NOTCH3 signaling, which is responsible for the expansion of tumor‐initiating cells and therapy resistance in ISG‐R breast cancer cells, which are mostly basal/triple‐negative breast cancer (TNBC) cells (Boelens et al , 2014). However, in our work done in different types of breast cancer cells, expression profiles of the down‐stream genes indicated that NOTCH3 signaling was not amplified when interferon signaling was activated.…”
Section: Discussionmentioning
confidence: 99%
“…exoRNA RN7SL1 was then transferred to breast cancer cells, activated RIG‐I signaling including STAT1 activation and ISG induction, which was proposed to promote aggressive development of cancer. In another work, the same group has demonstrated that RIG‐I activation by exoRNA can amplify NOTCH3 signaling, which is responsible for the expansion of tumor‐initiating cells and therapy resistance in ISG‐R breast cancer cells, which are mostly basal/triple‐negative breast cancer (TNBC) cells (Boelens et al , 2014). However, in our work done in different types of breast cancer cells, expression profiles of the down‐stream genes indicated that NOTCH3 signaling was not amplified when interferon signaling was activated.…”
Section: Discussionmentioning
confidence: 99%
“…This finding suggests that 5′pppRNA exosomes transport stress-related signals to neighboring cells, priming them for 5′ppp-RNA detection. Interestingly, breast cancer stroma cells under stress deliver endogenous 5′pppRNAs that are recognized by RIG-I in tumor cells (54). Thus, a conserved intercellular pathway exists in which stress signals in the form of small RNAs are transported between cells via exosomes.…”
Section: Discussionmentioning
confidence: 99%
“…Besides TLRs, a recent study mentioned above showed that exosomes from stromal cells contained 5-Triphosphate RNAs, which could activate RIG-I in breast cancer cells and promote resistance to radiation therapy. 37 Decrease of NK cell cytotoxicity Natural killer (NK) cells are a typical cytotoxic lymphocyte in innate immunity and take a variety of strategies to kill cancer cells directly. 38 It has been reported that exosomes derived from anticancer drug-treated human HCC cells were rich in heat shock proteins, which could act as endogenous danger signals to stimulate NK cell-elicited antitumor responses in vitro.…”
Section: Polarization Of Tumor-promoting Macrophagesmentioning
confidence: 99%