2019
DOI: 10.1038/s41467-019-12349-5
|View full text |Cite
|
Sign up to set email alerts
|

R-spondin 3 promotes stem cell recovery and epithelial regeneration in the colon

Abstract: The colonic epithelial turnover is driven by crypt-base stem cells that express the R-spondin receptor Lgr5. Signals that regulate epithelial regeneration upon stem cell injury are largely unknown. Here, we explore the dynamics of Wnt signaling in the colon. We identify two populations of cells with active Wnt signaling: highly proliferative Lgr5+/Axin2+ cells, as well as secretory Lgr5−/Axin2+ cells. Upon Lgr5+ cell depletion, these cells are recruited to contribute to crypt regeneration. Chemical injury indu… Show more

Help me understand this report

Search citation statements

Order By: Relevance

Paper Sections

Select...
4
1

Citation Types

4
68
1
1

Year Published

2020
2020
2023
2023

Publication Types

Select...
5
3
1

Relationship

0
9

Authors

Journals

citations
Cited by 98 publications
(74 citation statements)
references
References 39 publications
4
68
1
1
Order By: Relevance
“…This is a unique finding when compared to recent single cell studies in the adult human colon, and compared to findings in the mouse. In the adult human colon, a source of WNT and RSPO external to the muscular mucosae has been identified as WNT2B/RSPO3+ fibroblasts (Smillie et al, 2019), whereas in mice the predominant source of RSPO3 are PDGRFA+ cells in the small intestine (Greicius et al, 2018), or MYH11+ myofibroblasts in the colon (Harnack et al, 2019). In addition, the identification of the Foxl1 + telocyte has represented a major advance in elucidating the cells and sources of many niche factors in the murine intestinal stem cell niche (Aoki et al, 2016; Shoshkes-Carmel et al, 2018).…”
Section: Discussionmentioning
confidence: 99%
“…This is a unique finding when compared to recent single cell studies in the adult human colon, and compared to findings in the mouse. In the adult human colon, a source of WNT and RSPO external to the muscular mucosae has been identified as WNT2B/RSPO3+ fibroblasts (Smillie et al, 2019), whereas in mice the predominant source of RSPO3 are PDGRFA+ cells in the small intestine (Greicius et al, 2018), or MYH11+ myofibroblasts in the colon (Harnack et al, 2019). In addition, the identification of the Foxl1 + telocyte has represented a major advance in elucidating the cells and sources of many niche factors in the murine intestinal stem cell niche (Aoki et al, 2016; Shoshkes-Carmel et al, 2018).…”
Section: Discussionmentioning
confidence: 99%
“…Mesenchymal stem cells (MSCs) have a significant therapeutic potential for tissue damage by improving the growth of intestinal crypts in vitro by activating the Wnt/β-catenin signalling pathway [47]. Similarly, stromal myofibroblasts through secreted Rspo3 proteins can stabilise the effects of Wnt ligands and further reprogram differentiated cells to support crypt regeneration upon damage [48]. A recent study showed that fibroblasts could secrete extracellular vesicles with Wnt and EGF activity, thereby rescuing Wnt-or EGF-deficient organoid growth [49].…”
Section: Stromal Cells Inflammatory Cells and Cytokine Mediators Invmentioning
confidence: 99%
“…Interestingly, all markers that have a favorable prognostic significance are SC markers which predominantly exhibited a basal expression pattern in early GCs, and the proportion of marker-positive cancer cells increased in submucosal invasion. Therefore, it seems less likely that SC marker expression has any functional implication in the [29,30,37]. We hypothesized that the niche factors released from MM upregulate ISC markers, such as EPHB2 or LGR5, in the basal areas of GCs.…”
Section: Discussionmentioning
confidence: 99%
“…human stomach (Supplementary Figure S8b), whereas in the mouse stomach and colon only RSPO3 was shown to be expressed in the MM and to play essential roles for stem cell reprogramming [29,37].…”
Section: Discussionmentioning
confidence: 99%