scholarly journals SOX2-phosphorylation toggles a bistable differentiation-switch in squamous cell carcinoma

2021 ◽  
Author(s):  
Steven Hoang-Phou ◽  
Ana Sastre-Perona ◽  
Matteo Abbruzzese ◽  
Zhe Ying ◽  
Jasmin Siegle ◽  
...  

SummaryThe fate choice between stem cell self-renewal and differentiation is regulated by bistable transcriptional networks, which are balanced in homeostasis and imbalanced in tumors. Yet, how stem cells switch from self-renewal to differentiation remains a conundrum. Here, we discover a molecular mechanism that allows stem cell-like tumor propagating cells (TPCs) in squamous cell carcinomas (SCCs) to switch from a mutually exclusive SOX2-PITX1-TP63 self-renewal circuit to a KLF4 driven differentiation program, dependent on the relative occupancy of a novel Klf4-regulatory enhancer cluster (Klf4EC944) by SOX2 or KLF4, respectively. We find SOX2 occupies this site in TPCs to inhibit Klf4 transcription, but upon phosphorylation SOX2 becomes evicted from Klf4EC944, allowing residual KLF4 to occupy this site instead, boost the expression of KLF4 and its downstream targets, and differentiate self-renewing TPCs into post-mitotic SCC cells. This mechanism allows SOX2 to promote self-renewal and tumor formation, while preserving the differentiation potential in SCC cells. Our data suggest that stochastic cell fate decisions depend on the effective concentration of enzymatically regulated transcription factors. The surprising specificity by which SOX2-phosphorylation governs the bistable Klf4EC944 network-switch in SCCs reveals a conceptual framework for the identification of similar switches in other stem cell and cancer types and their potential development into cell type specific differentiation therapies for diseases in which tissue homeostasis has gone awry.

Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 3907-3907 ◽  
Author(s):  
Lev M Kats ◽  
Madison J Kelly ◽  
Gareth Gregory ◽  
Ricky W Johnstone ◽  
Stephin J Vervoort

Abstract Stem cell self-renewal and lineage specification are highly dynamic and tightly controlled processes that are essential for normal haematopoiesis and are dysregulated in cancer. The X-linked BCL6 Corepressor (BCOR) gene encodes a protein that is widely expressed across adult human tissues and is a component of a non-canonical Polycomb repressive complex 1 (PRC1). The BCOR gene is recurrently mutated in various malignant and non-malignant blood disorders, and we and others have recently provided experimental evidence that BCOR has cell-context dependent functions in regulating the proliferation, differentiation and survival of haematopoietic cells. To comprehensively examine the role of BCOR in haematopoiesis in vivo we used a conditional mouse model that mimics the truncating mutations observed in acute myeloid leukaemia (AML) and myelodysplastic syndrome (MDS). Using stem and progenitor populations isolated ex vivo we comprehensively analysed the role of BCOR in regulating gene expression, modifying chromatin and altering genome architecture. We demonstrate that BCOR has a pivotal role in down-regulating haematopoietic stem cell (HSC) associated transcriptional networks during the transition from multi-potent stem cells to lineage-committed myeloid progenitors. Inactivation of Bcor in HSCs results in expansion of myeloid progenitors and co-operates with oncogenic KrasG12D in the initiation of an aggressive and fully transplantable acute leukaemia. Mechanistically, Bcor regulates a subset of PRC1-target genes including key HSC super-enhancer-linked transcription factors that are normally down-regulated during myeloid differentiation. We used CRISPR/Cas9 to explore the function of Bcor target genes and identified those that are necessary for the proliferation of Bcor mutant leukaemic cells. This study provides a comprehensive mechanistic understanding of how BCOR regulates cell fate decisions and contributes to the development of leukaemia. Disclosures No relevant conflicts of interest to declare.


2013 ◽  
Vol 201 (3) ◽  
pp. 409-425 ◽  
Author(s):  
An Zeng ◽  
Yong-Qin Li ◽  
Chen Wang ◽  
Xiao-Shuai Han ◽  
Ge Li ◽  
...  

Adult stem cells (ASCs) capable of self-renewal and differentiation confer the potential of tissues to regenerate damaged parts. Epigenetic regulation is essential for driving cell fate decisions by rapidly and reversibly modulating gene expression programs. However, it remains unclear how epigenetic factors elicit ASC-driven regeneration. In this paper, we report that an RNA interference screen against 205 chromatin regulators identified 12 proteins essential for ASC function and regeneration in planarians. Surprisingly, the HP1-like protein SMED–HP1-1 (HP1-1) specifically marked self-renewing, pluripotent ASCs, and HP1-1 depletion abrogated self-renewal and promoted differentiation. Upon injury, HP1-1 expression increased and elicited increased ASC expression of Mcm5 through functional association with the FACT (facilitates chromatin transcription) complex, which consequently triggered proliferation of ASCs and initiated blastema formation. Our observations uncover an epigenetic network underlying ASC regulation in planarians and reveal that an HP1 protein is a key chromatin factor controlling stem cell function. These results provide important insights into how epigenetic mechanisms orchestrate stem cell responses during tissue regeneration.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 393-393
Author(s):  
Sonia Cellot ◽  
Kristin J Hope ◽  
Martin Sauvageau ◽  
Guy Sauvageau

Abstract Abstract 393 Epigenetic modifications influence cell fate decisions, such as stem cell self-renewal and differentiation, both in normal and leukemic stem cells (LSC). On this premise, an expression profile study of the emerging class of histone demethylating enzymes (HDE) in stem cell enriched populations was designed. Transcripts of all these enzymes (n=27) were detected in isolated HSC populations (frequency 1:2) from fetal liver and bone marrow. Similar results were obtained in HoxA9-Meis1 induced leukemia, with an inverse correlation between LSC content and the HDE Jarid1b expression level. As a functional follow-up, systematic knockdown of known or presumed HDE was undertaken in primary HSC using a RNA interference (RNAi) based approach. Five sh-RNA were designed and generated against every HDE and cloned into a retroviral vector, containing a GFP reporter gene. The screen was set as a loss-of-function assay, where sorted CD150+CD48-Lin- cells were co-cultured on retroviral producers for 5 days, in a 96-well setting, and a fraction of each well was then transplanted into 2 mice (day 0), along with competitor cells. The remaining cell fraction served to asses gene transfer by GFP epifluorescence measurements, and RNA isolated from sorted GFP+ cells was used to evaluate gene knockdown levels by Q-PCR analysis. Blood reconstitution was evaluated at regular intervals, tracking the contribution of the donor CD45.1+ transduced (GFP+) cells to recipient hematopoiesis over time. As baseline references, sh-RNA to luciferase (no effect) and the histone acetyl transferase myst3 (stem cell loss) were used, as well as hoxb4 over-expression (stem cell expansion). In the primary screen, RNAi against jarid1a, lsd1 and an arginine demethylating enzyme, resulted in a progressive decline in blood reconstitution levels from the transduced cells, ranging from a 50% to 80% reduction of the GFP content within the graft. On the other hand, knockdown of Jarid1b conferred an in vivo competitive advantage to the transduced cells. As a validation procedure, a 7 day culture step was introduced to the assay, hence increasing the signal to noise ratio, to better detect an increase in HSC activity, as recently reported. After a week in vitro (day 7), 1/8 equivalents of single well cultures were transplanted into 3 independent mice, and blood reconstitution levels serially assessed. Cells transduced with sh-RNA against jarid1b (3 constructs out of 6) contributed more significantly to host hematopoiesis than sh-RNA luciferase transduced cells (58±16% vs 26±3% GFP), or hoxb4 over-expressing cells (37±2% GFP), at comparable gene transfer rates, at the 14 week time point and beyond (3 independent experiments). In long-term recipients, differentiation potential of these cells was preserved, as evidenced by CD4+CD8+ thymic cells, B220+ splenic cells and CD11b+ bone marrow cells in the GFP positive contingent. Clonality studies on DNA isolated from these sorted populations are ongoing. There were no cases of leukemic transformation in all of the transplant recipients (n>30). In conclusion, this RNAi based study performed on primary hematopoietic cells links histone demethylases to modulations in cell fate. An opposite role seems to emerge among the H3K4me3/me2 HDE JARID1a and JARID1b regarding HSC activity, yet these proteins are highly homologous. Sequence alignments pinpoint to differences within the C-terminal portion, containing a PHD domain, and interestingly, dysregulation of this chromatin-binding PHD finger (NUP98-JARID1a) has been recently shown to play a direct role in leukemogenesis Disclosures: No relevant conflicts of interest to declare.


Cancers ◽  
2020 ◽  
Vol 12 (11) ◽  
pp. 3253
Author(s):  
Kamini Kaushal ◽  
Suresh Ramakrishna

Cancer stem cells (CSCs) have both the capacity for self-renewal and the potential to differentiate and contribute to multiple tumor properties, such as recurrence, metastasis, heterogeneity, multidrug resistance, and radiation resistance. Thus, CSCs are considered to be promising therapeutic targets for cancer therapy. The function of CSCs can be regulated by ubiquitination and deubiquitination of proteins related to the specific stemness of the cells executing various stem cell fate choices. To regulate the balance between ubiquitination and deubiquitination processes, the disassembly of ubiquitin chains from specific substrates by deubiquitinating enzymes (DUBs) is crucial. Several key developmental and signaling pathways have been shown to play essential roles in this regulation. Growing evidence suggests that overactive or abnormal signaling within and among these pathways may contribute to the survival of CSCs. These signaling pathways have been experimentally shown to mediate various stem cell properties, such as self-renewal, cell fate decisions, survival, proliferation, and differentiation. In this review, we focus on the DUBs involved in CSCs signaling pathways, which are vital in regulating their stem-cell fate determination.


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 4327-4327
Author(s):  
Nicola Vannini ◽  
Mukul Girotra ◽  
Olaia M. Naveiras ◽  
Vasco Campos ◽  
Evan Williams ◽  
...  

Abstract A tight control of hematopoietic stem cell (HSC) quiescence, self-renewal and differentiation is crucial for lifelong blood production. The mechanisms behind this control are still poorly understood. Here we show that mitochondrial activity determines HSC fate decisions. A low mitochondrial membrane potential (Δψm) predicts long-term multi-lineage blood reconstitution capability, as we show for freshly isolated and in vitro-cultured HSCs. However, as in vivo both quiescent and cycling HSCs have comparable Δψm distributions, a low Δψm is not per se related to quiescence but is also found in dividing cells. Indeed, using divisional tracking, we demonstrate that daughter HSCs with a low Δψm maintain stemness, whereas daughter cells with high Δψm have undergone differentiation. Strikingly, lowering the Δψm by chemical uncoupling of the electron transport chain leads to HSC self-renewal under culture conditions that normally induce rapid differentiation. Taken together, these data show that mitochondrial activity and fate choice are causally related in HSCs, and provides a novel method for identifying HSC potential after in vitro culture. Disclosures No relevant conflicts of interest to declare.


2017 ◽  
Author(s):  
Andrea Corsinotti ◽  
Frederick C. K. Wong ◽  
Tülin Tatar ◽  
Iwona Szczerbinska ◽  
Florian Halbritter ◽  
...  

AbstractDeletion of Sox2 from embryonic stem cells (ESCs) causes trophectodermal differentiation. While this can be prevented by enforced expression of the related SOXB1 proteins, SOX1 or SOX3, the roles of SOXB1 proteins in epiblast stem cell (EpiSC) pluripotency are unknown. Here we show that Sox2 can be deleted from EpiSCs with impunity. This is due to a shift in the balance of SoxB1 expression in EpiSCs, which have decreased Sox2 and increased Sox3 compared to ESCs. Consistent with functional redundancy, Sox3 can also be deleted from EpiSCs without eliminating self-renewal. However, deletion of both Sox2 and Sox3 prevents self-renewal. The overall SOXB1 levels in ESCs affect differentiation choices: neural differentiation of Sox2 heterozygous ESCs is compromised, while increased SOXB1 levels divert the ESC to EpiSC transition towards neural differentiation. Therefore, optimal SOXB1 levels are critical for each pluripotent state and for cell fate decisions during exit from naïve pluripotency.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 2369-2369
Author(s):  
Jonathan R Keller ◽  
Ming Ji ◽  
Huajie Li ◽  
Serguei V Kozlov ◽  
Lino Tessarollo ◽  
...  

Abstract Abstract 2369 Inhibitor of DNA binding protein-2 (Id2) is a member of the helix-loop-helix family of transcriptional regulators that is required for the normal development of natural killer (NK) cells, dendritic cells, B cells and erythrocytes. However, little is known about the expression and function of Id2 in hematopoietic stem and progenitor cells (HSPC). Recent evidence suggests that cell fate is resolved in HSPC by regulating the levels transcription factor expression. Therefore, to determine if Id2 functions in cell fate decisions, and in which progenitor populations these decisions are made, we generated an Id2-EYFP reporter mouse model and validated that EYFP accurately reflected Id2 expression. Using this model we mapped Id2 expression levels in purified HSPC and their differentiated progeny. Id2 is highly expressed in differentiated neutrophils, dendritic cells and NK cells, but is suppressed during erythroid development. Id2 expression is dynamically regulated during lymphoid development, with high levels of Id2 expression in lymphoid progenitors that are down regulated during the early stages of T cell (DN2-DN4) and B cell (Pre-B) differentiation. Id2 is then up-regulated and highly expressed in CD4+CD8+ and single positive CD4 and CD8 thymocytes, suggesting that Id2 may have novel functions in differentiated T cells and in neutrophils. Id2 is expressed in HSC and multi-potent progenitors (MPP), is decreased in common myeloid progenitors (CMP), and further decreased in myeloid/erythroid progenitors (MEP), but increased in granulocyte/macrophage progenitors (GMP). We also observed a range of Id2 expression within purified HSPC, suggesting that the levels of Id2 expression in purified HSC, MPP and CMP may correlate with potential of these progenitors for myeloid, erythroid and lymphoid development. To investigate this, CMP Id2-hi and CMP-Id2-low expressing cells were sorted and evaluated for differentiation potential in vitro. CMP-Id2-hi cells showed greatly increased myeloid developmental potential compared to the CMP-Id2-low cells as indicated by flow cytometry and growth in soft agar. Gene expression profiles confirmed these results and showed that the CMP-Id2-hi cell expression profile correlated with the expression profile of GMP, while the CMP-Id2-lo cells showed an expression profile that resembled MEP. Based on these observations we anticipate that MPP-Id2-hi and MPP-Id2-lo cells will show differences in myeloid and lymphoid potential. The expression of Id2 in primitive HSPC suggested that Id2 may be required for the maintenance and fate of HSC. Using the Id2−/− mouse model, we discovered that Id2−/− bone marrow cells have impaired ability to rescue mice after serial transplantation, suggesting that Id2 is required for HSC self-renewal. Competitive repopulation assays showed that Id2−/− HSC have diminished repopulation potential. Thus, these data suggest that Id2 is required for the self-renewal of HSC, and that changes in the levels of Id2 expression are associated with cell fate determination of HSPC. Disclosures: No relevant conflicts of interest to declare.


eLife ◽  
2017 ◽  
Vol 6 ◽  
Author(s):  
Andrea Corsinotti ◽  
Frederick CK Wong ◽  
Tülin Tatar ◽  
Iwona Szczerbinska ◽  
Florian Halbritter ◽  
...  

Deletion of Sox2 from mouse embryonic stem cells (ESCs) causes trophectodermal differentiation. While this can be prevented by enforced expression of the related SOXB1 proteins, SOX1 or SOX3, the roles of SOXB1 proteins in epiblast stem cell (EpiSC) pluripotency are unknown. Here, we show that Sox2 can be deleted from EpiSCs with impunity. This is due to a shift in the balance of SoxB1 expression in EpiSCs, which have decreased Sox2 and increased Sox3 compared to ESCs. Consistent with functional redundancy, Sox3 can also be deleted from EpiSCs without eliminating self-renewal. However, deletion of both Sox2 and Sox3 prevents self-renewal. The overall SOXB1 levels in ESCs affect differentiation choices: neural differentiation of Sox2 heterozygous ESCs is compromised, while increased SOXB1 levels divert the ESC to EpiSC transition towards neural differentiation. Therefore, optimal SOXB1 levels are critical for each pluripotent state and for cell fate decisions during exit from naïve pluripotency.


Cells ◽  
2018 ◽  
Vol 7 (12) ◽  
pp. 237 ◽  
Author(s):  
Christian Kosan ◽  
Florian Heidel ◽  
Maren Godmann ◽  
Holger Bierhoff

In complex organisms, stem cells are key for tissue maintenance and regeneration. Adult stem cells replenish continuously dividing tissues of the epithelial and connective types, whereas in non-growing muscle and nervous tissues, they are mainly activated upon injury or stress. In addition to replacing deteriorated cells, adult stem cells have to prevent their exhaustion by self-renewal. There is mounting evidence that both differentiation and self-renewal are impaired upon aging, leading to tissue degeneration and functional decline. Understanding the molecular pathways that become deregulate in old stem cells is crucial to counteract aging-associated tissue impairment. In this review, we focus on the epigenetic mechanisms governing the transition between quiescent and active states, as well as the decision between self-renewal and differentiation in three different stem cell types, i.e., spermatogonial stem cells, hematopoietic stem cells, and muscle stem cells. We discuss the epigenetic events that channel stem cell fate decisions, how this epigenetic regulation is altered with age, and how this can lead to tissue dysfunction and disease. Finally, we provide short prospects of strategies to preserve stem cell function and thus promote healthy aging.


Sign in / Sign up

Export Citation Format

Share Document