scholarly journals A subpopulation of mouse esophageal basal cells has properties of stem cells with the capacity for self-renewal and lineage specification

Author(s):  
Jiri Kalabis ◽  
Kenji Oyama ◽  
Takaomi Okawa ◽  
Hiroshi Nakagawa ◽  
Carmen Z. Michaylira ◽  
...  
2019 ◽  
Vol 5 (3) ◽  
pp. eaau7887 ◽  
Author(s):  
Xiangxiang Wei ◽  
Jieyu Guo ◽  
Qinhan Li ◽  
Qianqian Jia ◽  
Qing Jing ◽  
...  

The transcription factor BTB and CNC homology 1 (Bach1) is expressed in the embryos of mice, but whether Bach1 regulates the self-renewal and early differentiation of human embryonic stem cells (hESCs) is unknown. We report that the deubiquitinase ubiquitin-specific processing protease 7 (Usp7) is a direct target of Bach1, that Bach1 interacts with Nanog, Sox2, and Oct4, and that Bach1 facilitates their deubiquitination and stabilization via the recruitment of Usp7, thereby maintaining stem cell identity and self-renewal. Bach1 also interacts with polycomb repressive complex 2 (PRC2) and represses mesendodermal gene expression by recruiting PRC2 to the genes’ promoters. The loss of Bach1 in hESCs promotes differentiation toward the mesendodermal germ layers by reducing the occupancy of EZH2 and H3K27me3 in mesendodermal gene promoters and by activating the Wnt/β-catenin and Nodal/Smad2/3 signaling pathways. Our study shows that Bach1 is a key determinant of pluripotency, self-renewal, and lineage specification in hESCs.


2008 ◽  
Vol 134 (4) ◽  
pp. A-707-A-708
Author(s):  
Jiri Kalabis ◽  
Kenji Oyama ◽  
Takaomi Okawa ◽  
Hiroshi Nakagawa ◽  
Jose Figueiredo ◽  
...  

2008 ◽  
Vol 17 (R1) ◽  
pp. R67-R75 ◽  
Author(s):  
G. W. Yeo ◽  
N. Coufal ◽  
S. Aigner ◽  
B. Winner ◽  
J. A. Scolnick ◽  
...  

2012 ◽  
Vol 86 (2) ◽  
Author(s):  
Trimble L.B. Spitzer ◽  
Angela Rojas ◽  
Zara Zelenko ◽  
Lusine Aghajanova ◽  
David W. Erikson ◽  
...  

2018 ◽  
Author(s):  
Shiekh Tanveer Ahmad ◽  
Alexandra D. Rogers ◽  
Myra J. Chen ◽  
Rajiv Dixit ◽  
Lata Adnani ◽  
...  

ABSTRACTCapicua (Cic) is a transcriptional repressor mutated in the brain cancer oligodendroglioma. Despite its cancer link, little is known of Cic’s function in the brain. Here, we investigated the relationship between Cic expression and cell type specification in the brain. Cic is strongly expressed in astrocytic and neuronal lineage cells but is more weakly expressed in stem cells and oligodendroglial lineage cells. Using a new conditionalCicknockout mouse, we show that forebrain-specificCicdeletion increases proliferation and self-renewal of neural stem cells. Furthermore,Cicloss biases neural stem cells toward glial lineage selection, expanding the pool of oligodendrocyte precursor cells (OPCs). These proliferation and lineage selection effects in the developing brain are dependent on de-repression of Ets transcription factors. In patient-derived oligodendroglioma cells, CIC re-expression or ETV5 blockade decreases lineage bias, proliferation, self-renewal and tumorigenicity. Our results identify Cic is an important regulator of cell fate in neurodevelopment and oligodendroglioma, and suggest that its loss contributes to oligodendroglioma by promoting proliferation and an OPC-like identity via Ets overactivity.


2021 ◽  
Vol 4 (Supplement_1) ◽  
pp. 2-3
Author(s):  
J Douchin ◽  
V Giroux

Abstract Background Esophageal cancer is a particularly deadly cancer with a 5-year survival rate of only 14% in Canada. Treatment resistance ascribed for at least 30% of the death. The acquisition of resistance to radio- and chemotherapy is mostly attributed to the presence of cancer stem cells (CSCs) and their persistence following classical treatments. CSCs are a subpopulation of tumor cells with high self-renewal and multipotent capacity which amongst others contribute to tumor heterogeneity. Our previous work identified Krt15+ esophageal cells as a rare and long-lived subpopulation of basal cells with higher self-renewal and multipotent capacities than other basal cells. Furthermore, preliminary observations suggest that Krt15+ cells could act as the cell-of-origin for ESCC, the most prevalent type of esophageal cancer worldwide. Though, we still ignore the role of Krt15+ cells in later stages of esophageal cancer such as treatment resistance and if therefore, they could act as CSC. Aims Determine if Krt15+ cells act as CSCs in ESCC patients and if they could contribute to treatment resistance. Methods To do so, we used Krt15-CrePR1;R26mT/mG mice treated with the carcinogen 4 Nitroquinoline-1-oxide (4NQO) in their drinking water for 16 weeks to induce ESCC. Twelve weeks following the beginning of 4NQO treatment, we induced Cre recombination with RU486, a PR1 agonist, leading to GFP expression specifically in Krt15+ cells. Following 4NQO treatment, mice were put back on normal water for 8 to 12 weeks allowing tumors to grow. At euthanasia, esophageal tumor cells were FACS sorted to isolate Krt15+ (GFP+) and Krt15- (GFP-) cells, which were then grown as tumoroids. Results We first validated that 4NQO successfully induced the formation of esophageal lesions in our model, which comprises Krt15+ and Krt15- tumor cells. Tumoroids were then successfully derived from these FACS-sorted cell populations. We demonstrated the increase of CSC-like cells within Krt15+ tumoroids compared to Krt15- tumoroids by measuring the presence of CD44highCD24high cells, two well-known CSC markers, by flow cytometry. Interestingly, Krt15+ and Krt15- tumoroids are histologically distinct. As observed for normal cells, Krt15+ tumoroids appeared as more multipotent and heterogenous than Krt15- tumoroids. Furthermore, Krt15+ tumoroids display higher hyperplasia than Krt15- tumoroids suggesting that Krt15+ tumor cells are functionally distinct from Krt15- tumor cells. Conclusions Krt15+ tumoroids display higher CSC content and hyperplastic capacity suggesting their potential role in esophageal cancer. With this project, we aim to highlight the role of Krt15+ cells in treatment resistance and put forward new targets to overcome this deadly issue in ESCC patients. Funding Agencies CAGCanada research chair TIER 2


2021 ◽  
Vol 4 (Supplement_1) ◽  
pp. 1-2
Author(s):  
M Rolland ◽  
A Gonneaud ◽  
D Jean ◽  
V Giroux

Abstract Background The esophagus is lined by a stratified epithelium in which basal cells can proliferate and undergo differentiation while migrating towards the lumen. In the basal layer, we also find Krt15+ stem cells that are multipotent, self-renewing and that have regenerative capacity. However, mechanisms that specifically control their functions remain unknown. Interestingly, RNA sequencing and gene set enrichment analysis (GSEA) revealed an enrichment of a gene set associated with DNA repair in Krt15+ cells in comparison to Krt15- cells. We also observed that Msh2 (MutS homolog 2), a gene associated with the DNA mismatch repair (MMR) mechanism, is the most significantly upregulated gene in Krt15+ stem cells. Aims To determine the impact of Msh2 loss on self-renewal of esophageal organoids under normal and stress conditions. Methods Esophageal epithelial cells were isolated from a wild type mouse and grown as organoids, a 3D culture model that supports stem cell growth and morphologically reproduces the tissue of origin. To determine Msh2 role in esophageal epithelium, this gene was deleted through a CRISPR/Cas9 approach in mouse esophageal organoids. Invalidation was confirmed by Western Blot and immunofluorescence. Impact of Msh2 loss on self-renewal was measured under normal condition and following radiation. Results At baseline, loss of Msh2 decreases the organoid formation rate of esophageal organoids. Furthermore, following high-dose radiation, Msh2 deficient cells form less organoids than control cells. These results suggest that self-renewal capacity is reduced when Msh2 is depleted. Interestingly, following radiation, organoids depleted for Msh2 show higher residual levels of p-H2AX, a DNA damage marker, and p-ATM, a key kinase in DNA damage response, suggesting that their capacity to cope with DNA damages is reduced. Conclusions Our results suggest that Msh2 contributes to maintaining genomic integrity in esophageal cells and that contributes to maintaining self-renewal capacity of basal cells and possibly esophageal stem cells. Funding Agencies Canada Research Chair.


Sign in / Sign up

Export Citation Format

Share Document