STEM-19. NON-REDUNDANT, ISOFORM-SPECIFIC ROLES OF HDAC1 IN THE REGULATION OF THE GLIOMA STEM CELL PHENOTYPE

2021 ◽  
Vol 23 (Supplement_6) ◽  
pp. vi25-vi25
Author(s):  
Costanza Lo Cascio ◽  
James McNamara ◽  
Ernesto Luna Melendez ◽  
Erika Lewis ◽  
Matthew Dufault ◽  
...  

Abstract Glioblastoma (GBM) is characterized by an aberrant yet druggable epigenetic landscape. One major family of epigenetic regulators, the Histone Deacetylases (HDACs), are considered promising therapeutic targets for GBM due to their repressive influences on transcription. Although HDACs share redundant functions and common substrates, the unique isoform-specific roles of different HDACs in GBM remain unclear. There is a temporal and cell-type specific requirement of HDAC1 and 2 during normal brain development, with HDAC2 being indispensable in neural stem cells. Here, we specifically investigated the functional importance of HDAC1 in glioma stem cells, an HDAC isoform whose expression increases with brain tumor grade and is correlated with decreased survival. Using cell-based and biochemical assays, transcriptomic analyses and patient-derived xenograft models, we report that knockdown of HDAC1 alone has profound effects on the glioma stem cell (GSC) phenotype and survival in a p53-dependent manner. HDAC1 is the essential class I deacetylase in glioma stem cells, and its loss is not compensated for by its paralogue HDAC2 or other HDACs. Loss of HDAC1 expression significantly suppresses viability of GSCs harboring functional p53, and that HDAC2 expression is completely dispensable in GSCs. In addition, HDAC1 silencing but not HDAC2, stabilizes and acetylates p53 in GSCs, resulting in upregulation of key p53 target genes and induction of programmed cell death. Furthermore, ablation of HDAC1 function alone results in histone hyperacetylation and a collapse of the stemness state in GSCs. We demonstrate significant suppression in tumor growth upon targeting of HDAC1 and identify compensatory pathways that provide insights into combination therapies for GBM. Our study highlights the importance of HDAC1 in GBM and the need to develop isoform-specific HDAC inhibitor drugs.

2020 ◽  
Author(s):  
Adam Kosti ◽  
Rodrigo Barreiro ◽  
Gabriela Guardia ◽  
Shiva Ostadrahimi ◽  
Erzsebet Kokovay ◽  
...  

Abstract Background: Tumor suppressor microRNAs (miRNAs) have been explored as agents to target cancer stem cells. Most strategies use a single miRNA mimic and present many disadvantages, such as the amount of reagent required and the diluted effect on target genes. miRNAs work in a cooperative fashion to regulate distinct biological processes and pathways. This information can be used to design more efficient ways to target cancer stem cells via miRNA combinations. Methods: We transfected glioma stem cell and neuroblastoma lines with miRNA combinations and evaluate their impact on cancer relevant phenotypes and neuronal differentiation. RNA-seq analysis was conducted then to determine the expression alterations induced by the miRNA combination and map target genes and affected pathways. Results: We have shown that miR-124, miR-128, and miR-137 function synergistically to regulate neurogenesis. We used a combination of these three miRNAs to treat glioma stem cells. miR-124, miR-128 and miR-137 combined treatment was much more effective than single miRNAs in disrupting cell proliferation and survival and promoting differentiation and response to radiation. Transcriptomic analyses indicated that transcription regulation, angiogenesis, metabolism, and neuronal differentiation are among the main biological processes affected by transfection of this miRNA combination. Finally, in search of other combinations of other tumor suppressor/pro-neurogenic miRNAs, we identified a miRNA cluster based on target predictions and established miR-29, miR-101, and miR-218 combination as an alternative to be used in cancer therapy.Conclusions: We demonstrated the value of using combinations of neurogenic miRNAs to disrupt cancer phenotypes and glioma stem cell growth. Our genomic analyses in GSCs established that the synergistic effect of these three miRNA amplified the repression of oncogenic factors and the effect on cancer relevant pathways. These results suggest that future therapeutic approaches would benefit from utilizing miRNA combinations, especially when targeting cancer-initiating cell populations.


2019 ◽  
Vol 21 (Supplement_3) ◽  
pp. iii57-iii57
Author(s):  
M Dontenwill ◽  
M Mercier ◽  
G Gillmann ◽  
D Reita ◽  
I Lelong-Rebel ◽  
...  

Abstract BACKGROUND Glioblastoma (GBM) is the most aggressive primary brain tumor. Treatment failure and recurrence are explained in part by tumoral heterogeneity. Our previous results showed that the integrin α5β1 is implicated in GBM aggressiveness and represents a relevant therapeutic target. Recently, we observed intra- and inter-tumor heterogeneity of integrin α5β1 expression. Heterogeneity may be linked to different glioma stem cell populations. MATERIAL AND METHODS Ten glioma stem cell lines were grown as neurospheres in stem cell medium and their differentiation was induced by serum and/or ATRA. Two cell lines (NCH421k and NCH644) were selected and were modified by depletion (CrisprCas9) or transfection of the α5 integrin gene. Polyclonal lines and individual clones were analyzed phenotypically in vitro, before and after differentiation, and in vivo in orthotopic xenografts of 2x104 cells in nude mice. TCGA datasets were used to validate the heterogeneous expression of α5 integrin in GBM. RESULTS TCGA data validate that α5 integrin mRNA was only over-expressed in the mesenchymal subclass of GBM. Our results show that α5 integrin protein is not expressed in stem cell culture conditions. However, α5 integrin expression is induced after differentiation in only half of the cell lines supporting the notion of tumoral heterogeneity of glioma stem cells. Interestingly, single cell-derived clone evaluation showed that intra-tumoral stem cell heterogeneity also exists at the level of α5 protein expression. When glioma stem cells are programmed or transduced to express α5 integrin, differentiated cells became more aggressive. Notably, they acquired a fibronectin-dependent motility and a proliferative phenotype. Interestingly, integrin α5 remained expressed in secondary stem cells obtained after dedifferentiation. The in vivo assays suggested that glioma stem cells, programmed to express the integrin, were prone to form larger tumors. CONCLUSION Our data support the hypothesis that some glioma stem cells are programmed to express the α5 integrin subunit in their differentiated progeny to form a more aggressive tumor. They add new evidences that both cell populations may be considered for new therapeutic strategies against GBM.


2019 ◽  
Vol 21 (Supplement_3) ◽  
pp. iii53-iii54
Author(s):  
J Auzmendi-Iriarte ◽  
A Saenz-Antoñanzas ◽  
J Andermatten ◽  
A Elua-Pinin ◽  
E Aldaba ◽  
...  

Abstract BACKGROUND Glioblastoma’s origin and development is not only associated to genetic alterations, but also to epigenetic changes. Indeed, an altered expression or activity of epigenetic enzymes such as histone deacetylases (HDAC) has been associated to cancer stem cell activity, which has been widely described as a major feature for therapy resistance and tumor recurrence. In particular, inhibition of HDAC6 is an increasingly attractive pharmacological strategy, due to its association with low toxicity. Thus, the aim of the present study was to determine the impact of a new HDAC6-selective-inhibitor in glioblastoma and glioma stem cells. MATERIAL AND METHODS To test the effect of QTX compound in glioblastoma and glioma stem cell lines, cell viability after 72h of treatment was studied by MTT assay. After evaluation of IC50, QTX in vitro activity was analyzed, focusing on proliferation, apoptosis and stemness of U87-MG cell line and confirmed in a patient-derived glioma stem cell line. In vivo antitumor effect was evaluated using U87-MG cells xenografted in immunocompromised mice; after tumor formation, 5 mice were randomly selected as control group and another 5 for QTX treatment (intraperitoneal administration of 50 mg/kg; 5 days of dosing / 2 days off for 2 weeks). Mice weight was measured daily and tumor volume every two days. RESULTS We demonstrated that QTX reduces viability of all tested glioblastoma cells, even more greatly than normal astrocytes. Indeed, QTX diminishes proliferation and induces apoptosis in both conventional and patient-derived glioma cell lines. In particular, this effect was accompanied by a reduction of self-renewal properties of glioma stem cells. Interestingly, QTX in vitro activity was more effective comparing to the pan-inhibitor SAHA or the HDAC6-selective inhibitor Tubastatin A. Furthermore, QTX delayed tumor initiation and progression in vivo, without presenting significant side effects. CONCLUSION QTX compound presents a promising anti-tumor effect both in vitro and in vivo in glioblastoma, at least in part, inhibiting glioma stem cell activity.


2018 ◽  
Vol 20 (suppl_6) ◽  
pp. vi256-vi256 ◽  
Author(s):  
A B Madhankumar ◽  
Oliver Mrowczynski ◽  
Becky Slagle-Webb ◽  
Abraham Thomas ◽  
Brad Zacharia ◽  
...  

2020 ◽  
Vol 2 (Supplement_3) ◽  
pp. ii5-ii5
Author(s):  
Daisuke Ogawa ◽  
Takeshi Fujimori ◽  
Yasunori Toyota ◽  
Tetsuhiro Hatakeyama ◽  
Masanobu Okauchi ◽  
...  

Abstract (Pro)renin receptor((P)RR) is a part of the Wnt receptor complex. Wnt/β-catenin signaling pathway (Wnt signaling) plays important role in pathogenesis and self-renewal of glioblastoma (GBM), or differentiation of glioma stem cell. We previously reported that (P)RR activated Wnt signaling, (P)RR expression correlated with malignancy of glioma, and treatment with (P)RR siRNA reduced the proliferative capacity. This time, we have searched for over 2632 microRNAs by microRNA microarray that its expression is affected by (P)RR whether overexpressed or suppressed and examined their effects in GBM cell lines or its glioma stem cells.


2019 ◽  
Vol 21 (Supplement_6) ◽  
pp. vi236-vi237
Author(s):  
Costanza Lo Cascio ◽  
Ernesto Luna Melendez ◽  
James McNamara ◽  
Robert Schultz ◽  
An-Chi Tien ◽  
...  

Abstract The current standard-of-care treatment for GBM is ineffective and fails to significantly prolong survival. Following exposure to aggressive multimodal treatment, GBMs have been shown to frequently shift their biological features upon recurrence, acquiring a more resistant phenotype. However, the temporal dynamics and molecular mechanisms that facilitate GBM recurrence are poorly understood. The objective of our study was to assess the acute response to ionizing radiation in glioma stem cells (GSCs) from the Classical subtype of GBM in vitro and in vivo. We find that Classical GSCs rapidly undergo dramatic molecular and cellular changes in response to single and fractionated doses of ionizing radiation, resulting in a heterogeneous cell population. Ionizing radiation causes a transient decrease in the expression of key stemness genes (e.g., SOX2) followed by drastic morphological changes and a concomitant significant increase in the levels of key cell fate markers expressed in adult quiescent neural stem cells. Radiation-induced alteration of SOX2 levels in Classical GSCs is dependent on intact p53 signaling. GSCs previously exposed to radiation are more radio-resistant upon re-treatment compared to their naïve, untreated counterparts – suggesting that the aforementioned phenotypic shift to a quiescent neural stem cell phenotype promotes treatment resistance. Our results suggest that cell-intrinsic factors dictate how GSCs respond to radiation, and that Classical GSCs are neurodevelopmentally predisposed to shift towards an astrocytic/neural stem cell identity in response to cellular stress.


Cancers ◽  
2021 ◽  
Vol 13 (2) ◽  
pp. 289
Author(s):  
Adam Kosti ◽  
Rodrigo Barreiro ◽  
Gabriela D. A. Guardia ◽  
Shiva Ostadrahimi ◽  
Erzsebet Kokovay ◽  
...  

Tumor suppressor microRNAs (miRNAs) have been explored as agents to target cancer stem cells. Most strategies use a single miRNA mimic and present many disadvantages, such as the amount of reagent required and the diluted effect on target genes. miRNAs work in a cooperative fashion to regulate distinct biological processes and pathways. Therefore, we propose that miRNA combinations could provide more efficient ways to target cancer stem cells. We have previously shown that miR-124, miR-128, and miR-137 function synergistically to regulate neurogenesis. We used a combination of these three miRNAs to treat glioma stem cells and showed that this treatment was much more effective than single miRNAs in disrupting cell proliferation and survival and promoting differentiation and response to radiation. Transcriptomic analyses indicated that transcription regulation, angiogenesis, metabolism, and neuronal differentiation are among the main biological processes affected by transfection of this miRNA combination. In conclusion, we demonstrated the value of using combinations of neurogenic miRNAs to disrupt cancer phenotypes and glioma stem cell growth. The synergistic effect of these three miRNA amplified the repression of oncogenic factors and the effect on cancer relevant pathways. Future therapeutic approaches would benefit from utilizing miRNA combinations, especially when targeting cancer-initiating cell populations.


Author(s):  
Merve Erkisa ◽  
Nazlihan Aztopal ◽  
Elif Erturk ◽  
Engin Ulukaya ◽  
Veysel T. Yilmaz ◽  
...  

Background: Cancer stem cells (CSC) are subpopulation within the tumor that acts a part in the initiation, progression, recurrence, resistance to drugs and metastasis of cancer. It is well known that epigenetic changes lead to tumor formation in cancer stem cells and show drug resistance. Epigenetic modulators and /or their combination with different agents have been used in cancer therapy. Objective: In our study we scope out the effects of combination of a histone deacetylases inhibitor, valproic acid (VPA), and Cu(II) complex [Cu(barb-κN)(barb-κ2N,O)(phen-κN,N’)]·H2O] on cytotoxicity/apoptosis in a stem-cell enriched population (MCF-7s) obtained from parental breast cancer cell line (MCF-7). Methods: Viability of the cells was measured by the ATP assay. Apoptosis was elucidated via the assessment of caspase-cleaved cytokeratin 18 (M30 ELISA) and a group of flow cytometry analysis (caspase 3/7 activity, phosphatidylserine translocation by annexin V-FITC assay, DNA damage and oxidative stress) and 2ˈ,7ˈ–dichlorofluorescein diacetate staining. Results: The VPA combined with Cu(II) complex showed anti proliferative activity on MCF-7s cells in a dose- and time-dependently. Treatment with combination of 2.5 mM VPA and 3.12 μM Cu(II) complex induces oxidative stress in a time-dependent manner, as well as apoptosis that is evidenced by the increase in caspase 3/7 activity, positive annexin-V-FITC, and increase in M30 levels. Conclusion: The results suggest that the combination therapy induces apoptosis following increased oxidative stress, thereby making it a possible promising therapeutic strategy that further analysis is required.


Cells ◽  
2021 ◽  
Vol 10 (2) ◽  
pp. 225
Author(s):  
Claire Racaud-Sultan ◽  
Nathalie Vergnolle

In adult stem cells, Glycogen Synthase Kinase 3β (GSK3β) is at the crossroad of signaling pathways controlling survival, proliferation, adhesion and differentiation. The microenvironment plays a key role in the regulation of these cell functions and we have demonstrated that the GSK3β activity is strongly dependent on the engagement of integrins and protease-activated receptors (PARs). Downstream of the integrin α5β1 or PAR2 activation, a molecular complex is organized around the scaffolding proteins RACK1 and β-arrestin-2 respectively, containing the phosphatase PP2A responsible for GSK3β activation. As a consequence, a quiescent stem cell phenotype is established with high capacities to face apoptotic and metabolic stresses. A protective role of GSK3β has been found for hematopoietic and intestinal stem cells. Latters survived to de-adhesion through PAR2 activation, whereas formers were protected from cytotoxicity through α5β1 engagement. However, a prolonged activation of GSK3β promoted a defect in epithelial regeneration and a resistance to chemotherapy of leukemic cells, paving the way to chronic inflammatory diseases and to cancer resurgence, respectively. In both cases, a sexual dimorphism was measured in GSK3β-dependent cellular functions. GSK3β activity is a key marker for inflammatory and cancer diseases allowing adjusted therapy to sex, age and metabolic status of patients.


Sign in / Sign up

Export Citation Format

Share Document