scholarly journals CBIO-44RESTING MEMBRANE POTENTIAL CONTROLS STEM CELL STATE AND DIFFERENTIATION OF GLIOBLASTOMA CANCER STEM CELLS

2015 ◽  
Vol 17 (suppl 5) ◽  
pp. v64.1-v64
Author(s):  
Brian Williams ◽  
Jinkyu Jung ◽  
Deric Park
Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 466-466 ◽  
Author(s):  
Eric R. Lechman ◽  
Kristin J. Hope ◽  
Fernando J. Suarez Saiz ◽  
Katsuto Takenaka ◽  
Carlo M. Croce ◽  
...  

Abstract MicroRNAs (miRNAs) are a new class of non-coding small RNAs that negatively regulate the expression of protein-encoding genes. Mature miRNAs are excised sequentially from primary miRNA (pri-miRNA) foldback precursor transcripts, and regulate gene expression at the post-transcriptional level. miRNAs functionally suppress gene expression by either inhibition of protein synthesis or by direct cleavage of the target mRNA. miRNA expression is tissue and developmental stage restricted, suggesting important roles in tissue specification and/or cell lineage determination. miRNAs are implicated in the regulation of diverse processes including cell growth control, apoptosis, fat metabolism and insulin secretion, and may be involved in the maintenance of the embryonic stem cell state. Several recent lines of evidence suggest a role for miRNAs in hematological malignancies. Many characterized miRNAs are located at fragile sites, minimal loss of heterozygosity regions, minimal regions of amplification or common breakpoint regions in human cancers. For example, chromosomal translocation t(8;17) in an aggressive B-cell leukemia results in fusion of miR-142 precursor and a truncated MYC gene. Furthermore, both miR-15 and miR-16 are located within a 30 kb deletion in CLL, and in most cases of this cancer both genes are deleted or underexpressed. In addition, mice transplanted with hematopoietic stem cells (HSC) overexpressing both c-Myc and the miR-17–92 polycistron developed cancers earlier with a more aggressive nature when compared to lymphomas generated by c-myc alone. To address the role of miRNAs in the regulation and maintenance of the hematopoietic stem cell state and leukemogenesis, we sorted 6 primary AML patient samples into 4 populations based on the expression of CD34/CD38 and performed miRNA array analysis. We identified a subset of miRNAs whose expression profile could discriminate the CD34+/CD38- fractions from more mature populations. In particular, BIC/miR-155 was found to be over-expressed in leukemic stem cells (LSC). Validation by qRT-PCR revealed this expression pattern in 5 of the 6 sorted AML samples. Furthermore, within umbilical cord blood (CB) cells, BIC/miR-155 is more highly expressed in the primitive CD34+38- fraction as compared to mature sub-fractions as assessed by Affymetrix microarray. miRNA array analysis also revealed elevated levels of miR-155 in bulk primary AMLs as compared to normal BM. Intriguingly, BIC/miR-155 was first identified as a common retroviral insertion site in avian leucosis virus induced B cell lymphomas, and BIC/miR-155 overexpression has been observed in all subtypes of Hodgkin’s lymphoma. To test the hypothesis that miR-155 is important in LSC/HSC function, we designed lentiviral vectors for RNAi mediated knockdown of BIC/miR-155. Knockdown of BIC/miR-155 within a novel CD34+ leukemic cell line resulted in a loss of CD34 expression and reduced proliferative potential. Additionally, knockdown within CB led to alterations in colony forming capacity. Additionally, we have recently generated lentiviral vectors for the enforced overexpression of BIC/miR-155. In vivo studies to investigate the effects of BIC/miR-155 over-expression and knockdown are ongoing and will be discussed.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 2444-2444
Author(s):  
Il-Hoan Oh ◽  
Kim Tae-Min ◽  
Jae-Seung Shim

Abstract Multiple transcription factors (TFs) that regulate the self-renewal/stem cell state of hematopoietic stem cells (HSCs) have been identified, but understanding the molecular interplay of these TFs for their functional coordination remains a challenging issue. In this study, we investigated the functional integration and transcriptional coordination of STAT3 and HoxB4, which are TFs known to have similar effects on the self-renewal of HSCs. We found that while STAT3 (STAT3-C) or HoxB4 similarly enhanced the in vitro self-renewal and in vivo repopulating activities of HSCs, simultaneous transduction of both STAT3-C and HoxB4 did not have any additive enhancing effects. In contrast, the overexpression of HoxB4 caused a ligand-independent Tyr-phosphorylation in STAT3, and the inhibition of the STAT3 activity in HoxB4-overexpressing bone marrow cells significantly abrogated the enhancing effects of HoxB4 on both the bone marrow repopulation and maintenance of the undifferentiated state, revealing a molecular integration of these two TFs for HSC self-renewal. Expression microarray analysis revealed a significant overlap of the transcriptomes regulated by STAT3 and HoxB4 in undifferentiated hematopoietic cells. Moreover, a gene set enrichment analysis (GSEA) for TFs that can recapitulate the transcriptional changes induced by HoxB4 or STAT3 showed significant overlap in the candidate TFs. Interestingly, among these identified TFs were the puripotency-related genes, Oct-4 and Nanog. These results indicate the functional integration of tissue-specific TFs for HSC self-renewal and provide insights into the functional convergence of various TFs towards a conserved transcription program for the stem cell state. Disclosures: No relevant conflicts of interest to declare.


Author(s):  
Satish Kumar Tiwari ◽  
Sudip Mandal

Over the years, Drosophila has served as a wonderful genetically tractable model system to unravel various facets of tissue-resident stem cells in their microenvironment. Studies in different stem and progenitor cell types of Drosophila have led to the discovery of cell-intrinsic and extrinsic factors crucial for stem cell state and fate. Though initially touted as the ATP generating machines for carrying various cellular processes, it is now increasingly becoming clear that mitochondrial processes alone can override the cellular program of stem cells. The last few years have witnessed a surge in our understanding of mitochondria’s contribution to governing different stem cell properties in their subtissular niches in Drosophila. Through this review, we intend to sum up and highlight the outcome of these in vivo studies that implicate mitochondria as a central regulator of stem cell fate decisions; to find the commonalities and uniqueness associated with these regulatory mechanisms.


Development ◽  
2013 ◽  
Vol 140 (19) ◽  
pp. 3965-3976 ◽  
Author(s):  
Yingchun Li ◽  
Matteo Moretto-Zita ◽  
Francesca Soncin ◽  
Anna Wakeland ◽  
Lynlee Wolfe ◽  
...  

Author(s):  
Gehad Youssef ◽  
Luke Gammon ◽  
Leah Ambler ◽  
Bethan Wicker ◽  
Swatisha Patel ◽  
...  

AbstractCancer stem cells undergo epithelial-mesenchymal transition (EMT) to drive metastatic dissemination in experimental cancer models. However, tumour cells undergoing EMT have not been observed disseminating into the tissue surrounding human tumour specimens, leaving the relevance to human cancer uncertain. Here, we identify an EMT stem cell state that retains EpCAM and CD24 after undergoing EMT and exhibits enhanced plasticity. This afforded the opportunity to investigate whether retention of EpCAM and CD24 alongside upregulation of the EMT marker Vimentin can identify disseminating EMT stem cells in human oral cancer specimens. Examining disseminating tumour cells in the stromal region of 3500 imaging fields from 24 human oral cancer specimens, evenly divided into metastatic and non-metastatic specimens, we see a significant enrichment of EpCAM, CD24 and Vimentin co-stained cells in metastatic specimens. Through training an artificial neural network on the EpCAM, CD24 and Vimentin co-staining, we predict metastasis with high accuracy (F1 0.91; AUC 0.87). We have observed, for the first time, disseminating EMT stem cells in patient histological specimens and demonstrated their utility for predicting metastatic disease.


2019 ◽  
Vol 14 (5) ◽  
pp. 428-436 ◽  
Author(s):  
Gabriele D. Bigoni-Ordóñez ◽  
Daniel Czarnowski ◽  
Tyler Parsons ◽  
Gerard J. Madlambayan ◽  
Luis G. Villa-Diaz

Cancer is a highly prevalent and potentially terminal disease that affects millions of individuals worldwide. Here, we review the literature exploring the intricacies of stem cells bearing tumorigenic characteristics and collect evidence demonstrating the importance of integrin α6 (ITGA6, also known as CD49f) in cancer stem cell (CSC) activity. ITGA6 is commonly used to identify CSC populations in various tissues and plays an important role sustaining the self-renewal of CSCs by interconnecting them with the tumorigenic microenvironment.


Author(s):  
Nese Unver

: Cancer stem cells represent a rare subpopulation of cancer cells carrying self-renewal and differentiation features in the multi-step tumorigenesis, tumor recurrence and metastasis. Pro-inflammatory stress is highly associated with cancer stemness via induction of cytokines, tumor-promoting immune cells and cancer stemness-related signaling pathways. This review summarizes the major pro-inflammatory factors affecting cancer stem cell characteristics and the critical immunotherapeutic strategies to eliminate cancer stem cells.


Cancers ◽  
2021 ◽  
Vol 13 (3) ◽  
pp. 469
Author(s):  
Juan Carlos López-Gil ◽  
Laura Martin-Hijano ◽  
Patrick C. Hermann ◽  
Bruno Sainz

Cancer stem cells (CSCs) are defined as a subpopulation of “stem”-like cells within the tumor with unique characteristics that allow them to maintain tumor growth, escape standard anti-tumor therapies and drive subsequent repopulation of the tumor. This is the result of their intrinsic “stem”-like features and the strong driving influence of the CSC niche, a subcompartment within the tumor microenvironment that includes a diverse group of cells focused on maintaining and supporting the CSC. CXCL12 is a chemokine that plays a crucial role in hematopoietic stem cell support and has been extensively reported to be involved in several cancer-related processes. In this review, we will provide the latest evidence about the interactions between CSC niche-derived CXCL12 and its receptors—CXCR4 and CXCR7—present on CSC populations across different tumor entities. The interactions facilitated by CXCL12/CXCR4/CXCR7 axes seem to be strongly linked to CSC “stem”-like features, tumor progression, and metastasis promotion. Altogether, this suggests a role for CXCL12 and its receptors in the maintenance of CSCs and the components of their niche. Moreover, we will also provide an update of the therapeutic options being currently tested to disrupt the CXCL12 axes in order to target, directly or indirectly, the CSC subpopulation.


Sign in / Sign up

Export Citation Format

Share Document