Inhibition of DLEU1 Following siRNA Transfection in Burkett’s Lymphoma (BL): Implication for Tumor Repressor Role of DLEU1 in C-Myc-Activated BL Lymphomagenesis

Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 5335-5335
Author(s):  
Nancy Day ◽  
Janet Ayello ◽  
Ian Waxman ◽  
Evan Shereck ◽  
Catherine McGuinn ◽  
...  

Abstract Background: The progress of childhood BL and DLBCL has improved dramatically in the past three decades; however, patients with a 13q-deletion have a significantly poorer outcome (Cairo/Patte et al Blood, 2007 and Patte/Cairo et al Blood, 2007; Poirel/Cairo et al Leukemia 2008). DLEU1, a potential tumor suppressor gene, is located within the 13q-deletion. DLEU1 was reported to be a key gene in the Burkitt classifier genes (Dave/Staudt et al NEJM, 2006) and c-myc binds to the promoter region of DLEU1. DLEU1-network proteins include, among others, E3 ubiquitin-protein ligase (UBR1), Tubulin beta-2C (TUBB2C) and RASSF1A. We previously demonstrated that UBR1, TUBB2C, and RASSF1A, were differentially expressed in BL vs DLBCL patients and cell lines by global gene profiles and real time RT-PCR studies (Day/Cairo et al AACR 2008; Day/Cairo et al ICML 2008). We further demonstrated decreased expression of UBR1 (33.2±4.5% reduction compared to control (p<0.02)) and TUBB2C (30.0±3.5% reduction compared to control (p<0.001)) by DLEU1 gene siRNA knock down, while expression of RASSF1A was not changed (Day/Cairo, et al SIOP 2008). Taken together, these data suggest the hypothesis that DLEU1 interacting with UBR1 may interfere with microtubule function, and therefore act as a tumor repressor in c-myc-activated BL lymphomagenesis, by arrest of the cell cycle at G2/M and subsequent inducion of apoptosis. Objective: In this study, we investigated the role of DLEU1 in regulation of apoptosis in BL by inhibition of DLEU1 gene expression by a DLEU1 siRNA and evaluated it effects on the apoptotic rate in a BL cell line. Methods: The Ramos BL cell line was transiently transfected with a 25-nucleotide modified DLEU1 siRNA (5′-AUACUUGGCAUGAAUGAACUUAUGU-3′ and 3′-UAUGAACCGUACUUACUUGAAUACA-5′). Stealth RNAi whose GC content is similar to that of this DLEU1 siRNA was used as negative control. The transient transfection of DLEU1 siRNA (10 – 20 nM) was achieved using Lipofectamine RNAiMAX. The transfection efficiency of siRNA was evaluated using Alexa Fluor Red Fluorescent Oligo. DLEU1 contents were measured by qRT-PCR with ddCt relative quantitative determination. GAPDH was used as endogenous control. Statistical analysis was conducted by one-way analysis of variance (ANOVA) followed by Tukey-Kramer multiple comparisons test. To determine the early and late stages of apoptosis, we transfected Ramos BL cells with DLEU1 siRNA, and then incubated cells with Annexin V-FITC and Propidium Iodide for 15 minutes, respectively (BD Pharmingen), followed by FACS using BD LSRII with FACSDiva. Results: The DLEU1 siRNA decreased the expression of DLEU1 RNA (52±13%; p<0.0006). The transfection efficiency of siRNA was 85 – 90%. Comparing to untreated cells, DLEU1 siRNA treatment significantly reduced early apoptosis (16.90±0.37%; p<0.001) and late stage apoptosis (14.70±0.27%; p<0.0001). Conclusion: These results suggest that when DLEU1 gene expression is decreased in BL cells, there is a significant reduction in both early and late apoptosis. The results strongly support a relationship between DLEU1 gene and regulation of BL apoptotic mechanisms. In concert with previous investigations, this data suggests that DLEU1 may function as a tumor growth repressor via UBR1 and TUBB2C-regulated mechanism in the cellular apoptotic process. Since c-myc binds promoter region of DLEU1 and these two genes are a part of the c-myc signaling network, this further underscores the importance of DLEU1 and its network proteins may play in c-myc-activated BL lymphomagenesis.

Author(s):  
Daniel M. Sapozhnikov ◽  
Moshe Szyf

AbstractAlthough associations between DNA methylation and gene expression were established four decades ago, the causal role of DNA methylation in gene expression remains unresolved. Different strategies to address this question were developed; however, all are confounded and fail to disentangle cause and effect. We developed here a highly effective new method using only deltaCas9(dCas9):gRNA site-specific targeting to physically block DNA methylation at specific targets in the absence of a confounding flexibly-tethered enzymatic activity, enabling examination of the role of DNA methylation per se in living cells. We show that the extensive induction of gene expression achieved by TET/dCas9-based targeting vectors is confounded by DNA methylation-independent activities, inflating the role of DNA methylation in the promoter region. Using our new method, we show that in several inducible promoters, the main effect of DNA methylation is silencing basal promoter activity. Thus, the effect of demethylation of the promoter region in these genes is small, while induction of gene expression by different inducers is large and DNA methylation independent. In contrast, targeting demethylation to the pathologically silenced FMR1 gene targets robust induction of gene expression. We also found that standard CRISPR/Cas9 knockout generates a broad unmethylated region around the deletion, which might confound interpretation of CRISPR/Cas9 gene depletion studies. In summary, this new method could be used to reveal the true extent, nature, and diverse contribution to gene regulation of DNA methylation at different regions.


2014 ◽  
Vol 2014 ◽  
pp. 1-9 ◽  
Author(s):  
Li-Juan Fu ◽  
Yu-Bin Ding ◽  
Lan-Xiang Wu ◽  
Chun-Jie Wen ◽  
Qiang Qu ◽  
...  

DNA (cytosine-5-) methylation silencing of GSTP1 function occurs in prostate adenocarcinoma (PCa). Previous studies have shown that there is an inverse relationship between dietary lycopene intake and the risk of PCa. However, it is unknown whether lycopene reactivates the tumor suppressor gene glutathioneS-transferase-π(GSTP1) by demethylation of the hypermethylated CpGs that act to silence the GSTP1 promoter. Here, we demonstrated that lycopene treatment significantly decreased the methylation levels of the GSTP1 promoter and increased the mRNA and protein levels of GSTP1 in an androgen-independent PC-3 cell line. In contrast, lycopene treatment did not demethylate the GSTP1 promoter or increase GSTP1 expression in the androgen-dependent LNCaP cell line. DNA methyltransferase (DNMT) 3A protein levels were downregulated in PC-3 cells following lycopene treatment; however, DNMT1 and DNMT3B levels were unchanged. Furthermore, the long interspersed element (LINE-1) and short interspersed element ALU were not demethylated when treated by lycopene. In LNCaP cells, lycopene treatment did not affect any detected DNMT protein expression, and the methylation levels of LINE-1 and ALU were decreased. These results indicated that the protective effect of lycopene on the prostate is different between androgen-dependent and androgen-independent derived PCa cells. Further, in vivo studies should be conducted to confirm these promising results and to evaluate the potential role of lycopene in the protection of the prostate.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 5043-5043
Author(s):  
Karel Svojgr ◽  
Tatiana Burjanivova ◽  
Martina Vaskova ◽  
Tomas Kalina ◽  
Tomas Brdicka ◽  
...  

Abstract PAG, LAT, NTAL and LIME belong to category of transmembrane adaptor proteins (TRAPs). They do not possess an enzymatic or kinase function, but they are involved in mediation of signal transmission from surface receptors to cell nucleus. We propose that some of them are engaged in development or maintenance of leukaemia. We have previously demonstrated that expression status of TRAPs at mRNA level is specific in some subgroups of childhood ALL, particularly in B-cell precursor acute lymphoblastic leukaemia (BCP-ALL) with the TEL/AML1 fusion gene. Furthermore, we have described that variable expression of NTAL mRNA is related to the response to initial glucocorticosteroid pre-phase in the treatment of childhood T-ALL; patients with high NTAL levels show better treatment response compared to the low-NTAL cases. In the current study, we aimed to prove experimentally that different levels of NTAL protein influence response of leukaemic T cells to glucocorticosteroids. In the wild type Jurkat cells (human T-cell leukaemia cell line) the NTAL protein is undetectable. For the in-vitro experiments we created a derivative Jurkat cell line transfected with the NTAL construct. In the derivative cell line, the NTAL positivity at both mRNA and protein level was verified using RT-PCR and Western blotting. The derivative cell line in a cell culture behaves similar as wild type Jurkat cell line. Transfectants and wild type Jurkat cells were incubated with methylprednisolone, dexamethasone or with solvent alone (H2O) as a negative control. Using flow cytometry we determined a percentage of surviving cells after 24, 48 and 72 hours of treatment. Cells were stained with Annexin V and DAPI and living cells were defined as Annexin V, DAPI negative. At each time point, the number of living cells in the negative control was set to 100%. After 48 hours of dexamethasone treatment the number of surviving cells in the Jurkat wild-type was higher by 12% compared to the Jurkat cells expressing NTAL. After 72 hours this difference was even more prominent reaching 46%. The same effect of methylprednisolone treatment was less pronounced (non-significant difference at 48 hours and 5% difference at 72 hours). Based on our experimental data we propose that NTAL acts in T-cells as a putative tumour suppressor. A plausible mechanism of its function is that NTAL competitively inhibits another adaptor LAT, which is required for signal propagation to the nucleus in T-cells. NTAL and LAT can compete for its localisation in proper “lipid rafts” or for palmitoylation. Our data can be used for subsequent functional analysis of signaling pathways in leukaemic blasts as well as in physiological lymphoid cells.


2018 ◽  
Vol 84 (19) ◽  
Author(s):  
Isaac V. Greenhut ◽  
Beryl L. Slezak ◽  
Johan H. J. Leveau

ABSTRACT We show for soil bacterium Enterobacter soli LF7 that the possession of an indole-3-acetic acid catabolic (iac) gene cluster is causatively linked to the ability to utilize the plant hormone indole-3-acetic acid (IAA) as a carbon and energy source. Genome-wide transcriptional profiling by mRNA sequencing revealed that these iac genes, chromosomally arranged as iacHABICDEFG and coding for the transformation of IAA to catechol, were the most highly induced (>29-fold) among the relatively few (<1%) differentially expressed genes in response to IAA. Also highly induced and immediately downstream of the iac cluster were genes for a major facilitator superfamily protein (mfs) and enzymes of the β-ketoadipate pathway (pcaIJD-catBCA), which channels catechol into central metabolism. This entire iacHABICDEFG-mfs-pcaIJD-catBCA gene set was constitutively expressed in an iacR deletion mutant, confirming the role of iacR, annotated as coding for a MarR-type regulator and located upstream of iacH, as a repressor of iac gene expression. In E. soli LF7 carrying the DNA region upstream of iacH fused to a promoterless gfp gene, green fluorescence accumulated in response to IAA at concentrations as low as 1.6 μM. The iacH promoter region also responded to chlorinated IAA, but not other aromatics tested, indicating a narrow substrate specificity. In an iacR deletion mutant, gfp expression from the iacH promoter region was constitutive, consistent with the predicted role of iacR as a repressor. A deletion analysis revealed putative −35/−10 promoter sequences upstream of iacH, as well as a possible binding site for the IacR repressor. IMPORTANCE Bacterial iac genes code for the enzymatic conversion of the plant hormone indole-3-acetic acid (IAA) to catechol. Here, we demonstrate that the iac genes of soil bacterium Enterobacter soli LF7 enable growth on IAA by coarrangement and coexpression with a set of pca and cat genes that code for complete conversion of catechol to central metabolites. This work contributes in a number of novel and significant ways to our understanding of iac gene biology in bacteria from (non-)plant environments. More specifically, we show that LF7's response to IAA involves derepression of the MarR-type transcriptional regulator IacR, which is quite fast (less than 25 min upon IAA exposure), highly specific (only in response to IAA and chlorinated IAA, and with few genes other than iac, cat, and pca induced), relatively sensitive (low micromolar range), and seemingly tailored to exploit IAA as a source of carbon and energy.


2020 ◽  
Author(s):  
Guang Yang ◽  
Hua Xiao Wang ◽  
Bin Yan ◽  
Yan Chun Xu ◽  
Yi Ru Zheng ◽  
...  

Abstract Background Burkitt lymphoma is a kind of non-Hodgkin B-cell-derived malignancy, derived from germinal center B cells. FAM54A has been proved to be involved in various physiological and pathological processes of cancers, but the biological function of FAM54A in Burkitt lymphoma remains unclear. Thus, the aim of our research was to elucidate the roles of FAM54A in proliferation, apoptosis and cell cycle of Burkitt lymphoma.Methods Burkitt lymphoma cell line (Namalwa) was chosen to perform the following experiments. FAM54A-shRNA and negative control-shRNA lentivirus that were synthesized, respectively by Qiagen were used to transfect targeted cells in order to knockdown FAM54A or as negative control. Then, cell proliferation, cell cycle and cell apoptosis were detected by using MTS assay, propidium iodide staining and Annexin V-APC staining, respectively.Results Our results showed that high expression of FAM54A protein was found in Namalwa cell line. Furthermore, MTS analysis exhibited that knockdown of FAM54A obviously inhibited cell proliferation in Namalwa cells. What’s more, cell cycle analysis showed that knockdown of FAM54A induced Namalwa cell apoptosis and arrested cell cycle in G2/M phase.Conclusions These findings suggest that FAM54A is essential for Namalwa cell proliferation and may be a potential therapeutic target for the treatment of Burkitt lymphoma.


Sign in / Sign up

Export Citation Format

Share Document