scholarly journals GEN-09 Pursuing the function of microRNA targeting (Pro)renin receptor against glioma

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_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 51 (11) ◽  
pp. 1-20 ◽  
Author(s):  
Jun-Cheng Guo ◽  
Yi-Jun Yang ◽  
Jin-Fang Zheng ◽  
Jian-Quan Zhang ◽  
Min Guo ◽  
...  

AbstractHepatocellular carcinoma (HCC) is a major cause of cancer-related deaths, but its molecular mechanisms are not yet well characterized. Long noncoding RNAs (lncRNAs) play crucial roles in tumorigenesis, including that of HCC. However, the role of homeobox A11 antisense (HOXA11-AS) in determining HCC stem cell characteristics remains to be explained; hence, this study aimed to investigate the effects of HOXA11-AS on HCC stem cell characteristics. Initially, the expression patterns of HOXA11-AS and HOXA11 in HCC tissues, cells, and stem cells were determined. HCC stem cells, successfully sorted from Hep3B and Huh7 cells, were transfected with short hairpin or overexpression plasmids for HOXA11-AS or HOXA11 overexpression and depletion, with an aim to study the influences of these mediators on the self-renewal, proliferation, migration, and tumorigenicity of HCC stem cells in vivo. Additionally, the potential relationship and the regulatory mechanisms that link HOXA11-AS, HOXA11, and the Wnt signaling pathway were explored through treatment with Dickkopf-1 (a Wnt signaling pathway inhibitor). HCC stem cells showed high expression of HOXA11-AS and low expression of HOXA11. Both HOXA11-AS silencing and HOXA11 overexpression suppressed the self-renewal, proliferation, migration, and tumorigenicity of HCC stem cells in vivo, as evidenced by the decreased expression of cancer stem cell surface markers (CD133 and CD44) and stemness-related transcription factors (Nanog, Sox2, and Oct4). Moreover, silencing HOXA11-AS inactivated the Wnt signaling pathway by decreasing the methylation level of the HOXA11 promoter, thereby inhibiting HCC stem cell characteristics. Collectively, this study suggested that HOXA11-AS silencing exerts an antitumor effect, suppressing HCC development via Wnt signaling pathway inactivation by decreasing the methylation level of the HOXA11 promoter.


2012 ◽  
Vol 30 (15_suppl) ◽  
pp. e13543-e13543
Author(s):  
Monal Mehta ◽  
Atif J. Khan ◽  
Hatem E. Sabaawy ◽  
Bruce George Haffty

e13543 Background: Glioblastoma (GBM) is the most frequent and deadly brain cancer. Despite tolerance doses of radiation, control of tumor growth within the brain remains a formidable failure. Since the identification of brain cancer stem cells (BCSCs), efforts are underway to target the pathways regulating these cells. The role of Bmi-1 (B-cell specific MMLV insertion site-1), a polycomb member of chromatin-remodeling complex, in BCSCs self-renewal was elucidated. Here we utilize shRNA targeting or pharmacological inhibition of Bmi-1 in GBM cell lines and primary cells as a radiosensitizer to examine the effects of combination therapy on cell death and BCSCs differentiation. Methods: Cells were pre-treated with a Bmi-1 inhibitor before being irradiated. Serial neurosphere assay, a measure of self-renewal potential, was employed to study the effects of radiation, Bmi-1 inhibition, or the combination on BCSCs. The efficacy of this combination on cell death was assessed with MTT and clonogenic assays. Next, the abilities of the inhibitor and radiation to induce differentiation in GBM cell lines and primary cells were quantified. Further, by utilizing a novel zebrafish orthotropic xenograft model, small molecules targeting Bmi-1 and other BCSC pathways can be identified, and used to predict response to combination therapies. Results: Targeting of Bmi-1 in combination with radiation, specifically as a radiosensitizer, induced significant cell death in GBM cells, and was five-fold more effective than radiation only. Importantly, the neurosphere forming ability of BCSCs was severely compromised when the cells were treated with the combination, indicating a potent effect on the stem cell constituency. These effects may be due to loss of BCSC self-renewal potential, increased differentiation, and/or apoptosis as cells treated with the combination exhibited decreased expression of neural stem cell markers and abnormal phenotypes compared to single treatment. Conclusions: Targeting of Bmi-1 may eliminate the subpopulation of radioresistant BCSCs. Bmi-1 inhibition when combined with radiotherapy might provide an effective therapy for GBM patients specifically through its effect on BCSCs by affecting their survival, proliferation, and stem cell features.


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.


Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 1608-1608
Author(s):  
Xiaomin Zheng ◽  
Tim Beissert ◽  
Brigitte Ruster ◽  
Dieter Hoelzer ◽  
Reinhard Henschler ◽  
...  

Abstract The pathogenesis of acute myeloid leukemia (AML) is strictly related to a block of terminal differentiation combined with an increased proliferation of hematopoietic progenitors (HP) in the bone marrow (BM). Furthermore an aberrant self-renewal of leukemic stem cells seems to be obligatory for the establishment of the leukemic clone in the BM. The block of differentiation is due to a deregulated function of differentiation-relevant transcription factors, whereas the proliferation is induced by aberrantly activated signaling pathways related to growth factor-dependent receptor kinases such as Flt3 or c-Kit (CD117). The aberrant self renewal can be attributed to specific pathways such as the Wnt-signaling, which play an important role also in the maintenance of the normal hematopoietic stem cell (HSC). The acute promyelocytic leukemia (APL) is characterized by the t(15;17) which leads to the expression of the PML/RAR fusion protein in the leukemic cells. PML/RAR mediates the APL phenotype given by a differentiation block at the promyelocytic level and an increased self renewal of the APL stem cells by the activation of the Wnt-signaling. The differentiation block, but not the aberrant self renewal, can be overcome by treatment with ATRA resulting ina high percentage of complete remissions. Nevertheless, if ATRA is used as a monotherapy a relapse is inevitable. APL blasts are frequently positive for constitutive activating Flt3 mutations and are constantly c-Kit-positive. Given the fact that c-Kit is a stem cell marker, the expression of c-Kit has to be considered aberrant and not related to the differentiation stage of the promyelocytes in APL. Therefore we investigated first the relationship between PML/RAR and the aberrant expression of c-Kit and then the role of aberrantly activated c-Kit for the pathogenesis of APL by studying its influence on the biology of early HSC. Here we report that i.) in contrast to the t(8;21)-associated AML-1/ETO, PML/RAR activated the c-Kit promotor in HP; ii.) the inhibition of the endogenous c-Kit kinase activity by imatinib or by AZD2171 abrogated the aberrant “replating efficiency“ of PML/RAR-positive HSC; iii.) activated c-Kit (c-Kit-D814H) accelerated cell cycle progression of Sca1+/lin- HSC; iv.) activated c-Kit blocked the differentiation of Sca1+/lin- HSC and increased their “replating efficiency“; v.) “colony forming unit-spleen“ (CFU-S) as well as “competitive repopulation“ assays“ (CRA) revealed that c-Kit-D814H strongly increased the “self renewal“-potential of Sca1+/lin- HSC; vi) c-Kit-D814H augmented the migration of Sca1+/lin- HSC into a 3D stromal spheroid model based on M2-10B4 cells, but did not have any influence on their adhesion (flow chamber on TNFalpha-stimulated HUVEC cells) as well as on their chemotaxis (SDF-1 gradient in transwell assays); vii.) c-Kit-D814H further increased the aberrant replating efficiency of PML/RAR- as well as of AML-1/ETO-positive HSC. Taken together our results strongly indicate not only that c-Kit importantly contributes to the leukemogenesis of APL, but that PML/RAR has a dual effect on c-Kit - the amplification of its expression at the promotor level as well as driving its expression in cells which normally do not have this proliferation stimulus. Thus it seems that there is a sort of positive feedback between PML/RAR and c-Kit which establishes c-Kit as a valuable therapeutic target for the treatment of APL-patients.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 5409-5409 ◽  
Author(s):  
Mariana Tereza Lira Benício ◽  
Priscila S. Scheucher ◽  
Aglair Bergamo Garcia ◽  
Roberto P. Falcao ◽  
Eduardo M. Rego

Abstract The clinical significance of LSCs has recently been confirmed by the finding that stem cell-like gene expression signatures can predict the clinical outcome of acute myeloid leukemia (AML) patients, what suggests that the targeted elimination of LSCs would be an efficient therapeutic approach in AML. LSCs are rather infrequent in patients’ bone marrow, and hence are difficult to be identified and isolated. As an attempt to overcome such limitations, it has been previously shown that cell lines might be considered as attractive models aimed to better characterize LSC biological properties and their chemotherapy resistance mechanisms. Many attempts have been made to identify an immunophenotype that allow to discriminate between LSCs and normal hematopoietic stem cells (HSCs), but there is still controversy regarding the surface markers proposed so far. As a functional rather than immunological approach, the assessment of intracellular enzyme activities associated with the protection of primitive cells from oxidative insult during hematopoietic development has been proven as successful strategies, such as the use of Aldefluor reagent in the identification of AML LSCs. By taking advantage of the Aldefluor reagent in the field of stem cells research, we sought to identify LSC among the following AML cell lines, based on their high expression levels of the enzyme aldehyde dehydrogenase (ALDH): Kasumi-1, THP-1, MV-4;11, NB4 and OCI-AML3. Cells were stained with the Aldefluor reagent, following manufacturer’s specifications, and cell subsets presenting high ALDH activity were identified in Kasumi-1 (13%), THP-1 (25%) and OCI-AML3 (12%). To assess if the Aldefluor staining was correlated to the expression of genes involved in stem cell maintenance or myeloid commitment, these cell lines were further sorted in two subpopulations, according to ALDH activity (ALDHhigh and ALDHlow/int), and the respective expression levels of the following genes were evaluated by RQ-PCR: CEBPA, BMI-1, NOTCH-1, C-MYC, HOXA9, E2F1, NANOG and OCT3/4. The delta-delta Ct method was used to determine the fold change in target genes between ALDHhigh and ALDHlow/int subpopulations. CEBPA, BMI-1 and NOTCH-1 genes were upregulated (9, 2.3 and 2 fold, respectively) in Kasumi-1 ALDHhigh cells when compared to ALDHlow/int, whereas C-MYC and E2F1 were downregulated (0.3 and 0.5 fold, respectively). Little is known about the physiological role of CEBPA in adult HSC biology, though it has been described as a central determinant in the switch from fetal to adult HSCs. Bmi-1 has been shown to be essential for the generation of self-renewing adult HSCs. Despite the roles of Notch signaling on myeloid development and AML are poorly understood, it can promote self-renewal, and induce growth arrest and apoptosis in hematopoietic stem cells. Some molecular mechanisms responsible for self-renewal, like Bmi-1 and Notch signaling pathways, were found to be shared by both HSC and LSC, suggesting that a higher expression of both genes in ALDHhigh cells would be in accordance with stemness properties. In line with these results, lower levels of C-MYC and E2F1 expression in ALDHhigh cells would suggest that they are somehow more quiescent than their ALDHlow/int counterparts. No significant differences were observed on the target genes expression levels among the other cell lines studied. In conclusion, our results reveal an important correlation between ALDH activity and the expression of genes associated with stemness properties in identifying stem cells in Kasumi-1 cell line, suggesting that like KG-1 cell line, which is also CD34-enriched, Kasumi-1 would be considered as a valuable model to better understand AML stem cells properties. Disclosures: No relevant conflicts of interest to declare.


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 ◽  
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.


Sign in / Sign up

Export Citation Format

Share Document