scholarly journals Aberrant splicing of the E-cadherin transcript is a novel mechanism of gene silencing in chronic lymphocytic leukemia cells

Blood ◽  
2009 ◽  
Vol 114 (19) ◽  
pp. 4179-4185 ◽  
Author(s):  
Sanjai Sharma ◽  
Alan Lichtenstein

Abstract Premature termination codon (PTC) mutations are due to insertion or deletion of nucleotides causing a frameshift and premature termination codon in RNA. These transcripts are degraded by the nonsense-mediated decay pathway and have a very short half-life. We used a microarray technique to screen for genes that up-regulate their RNA signal upon nonsense-mediated decay pathway blockade in chronic lymphocytic leukemia (CLL) specimens and identified an E-cadherin transcript with PTC. Sequencing revealed an aberrant E-cadherin transcript lacking exon 11, resulting in a frameshift and PTC. The aberrant E-cadherin transcript was also identified in normal B cells, but occurred at a much lower level compared with CLL cells. In CLL specimens, E-cadherin expression was depressed more than 50% in 62% cases (relative to normal B cells). By real-time polymerase chain reaction analysis, the relative amounts of wild-type transcript inversely correlated with amounts of aberrant transcript (P = .018). Ectopic expression of E-cadherin in CLL specimens containing high amounts of aberrant transcript resulted in down-regulation of the wnt–β-catenin pathway reporter, a pathway known to be up-regulated in CLL. Our data point to a novel mechanism of E-cadherin gene inactivation, with CLL cells displaying a higher proportion of aberrant nonfunctional transcripts and resulting up-regulation of the wnt–β-catenin pathway.

Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 2935-2935
Author(s):  
Sanjai Sharma ◽  
Alan Lichtenstein

Abstract The acquisition of a premature termination codon (PTC) is a mutation that results in premature termination of transcription and degradation of the RNA by the non-sense mediated decay (NMD) pathway. This results in loss of expression of the allele, which harbors the PTC and is frequently the “second hit” in the loss of function of tumor suppressor genes. The NMD pathway can be blocked by protein translation inhibitors like emetine resulting in accumulation of abnormal truncated transcripts with PTC. By combining emetine-induced paralysis of the NMD pathway and microarray technology, a large number of genes were screened for the presence of PTCs in chronic lymphocytic leukemia (CLL) cells. Isolated normal peripheral blood B lymphocytes from two volunteers and five CLL samples were initially analyzed by emetine treatment and microarray. Cells were treated with and without emetine and the RNA was hybridized to Affymetrix 133A chip. Data was analyzed by calculating the fold up-regulation of signal intensity with emetine treatment for all the 23,000 genes on the chip. To take into account any non-specific up-regulation of signal intensity with emetine treatment, the fold induction observed with CLL samples was divided by fold induction obtained from the normal B cell samples to get a Nonsense enrichment index (NEI). Genes with the highest NEI in more than one CLL sample were selected for further study. From this cohort, individual genes were then sequenced and PTC mutations successfully identified in at least three genes; E-cadherin, plakophillin-4 (member of the armadillo family of proteins, a component of desmosomes) and TLE-2 (groucho homolog, a transcriptional repressor that represses transcription by TCF/LEF family members). Because E-cadherin has been described as a potential tumor suppressor whose expression is lost in CLL, we focused on this gene in a bigger cohort of 10 CLL patients. CLL cells were treated with emetine and total RNA was isolated. RNA from emetine treated and untreated cells was analyzed by RT-PCR. Amplified DNA fragments were isolated and sequenced. All 10 patients had an E-cadherin PTC: in 8/10 patients an E-cadherin allele had an exon 11 deletion resulting in premature termination codon and a smaller fragment on the RT-PCR assay. In 2/10 patients, a deletion was identified in exon 13 and 14 both of which were partially deleted resulting in RNA with PTC. Thus in CLL cells from 10 of 10 patients of all clinical stages, an inactivating PTC mutation is present in one of the alleles along with a wild type E-cadherin allele. E-cadherin is a transmembrane glycoprotein that is a component of the adherens junction and plays a role in cell-cell interaction, signaling via the beta-catenin pathway and is a known tumor suppressor. In addition, the plakophillin-4 and TLE-2 genes are negative regulators of the Wnt/catenin pathway and potential loss-of-function PTCs in those genes would further dysregulate this cascade. These data further support the role of E-cadherin as a potential tumor suppressor in CLL and suggest that our novel screen can be used to identify additional tumor suppressors in this disease.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 2842-2842
Author(s):  
Sanjai Sharma ◽  
Alan Lichtenstein

Abstract Premature termination codons (PTCs) in tumor suppressor genes can contribute to oncogenesis. PTCs prematurely terminate transcription, leading to expression of a truncated, mRNA. The nonsense-mediated degradation (NMD) pathway degrades the aberrant transcripts resulting in loss of expression. To identify RNA transcripts with PTCs in chronic lymphocytic leukemia specimens (CLL), we employed a microarray technique to screen for genes which upregulate their RNA signal upon NMD blockade. In the list of genes with highest upregulation, we identified the E-cadherin gene, a known tumor suppressor gene. On further investigation, we found that the PTC was due to stabilization of an alternatively spliced E-cadherin transcript. This transcript completely lacks exon 11 (exon skipping), resulting in a frameshift and a PTC which is recognized by the NMD pathway and degraded. RT-PCR analysis demonstrated that the exon 11 skipping occurs in normal B cells as well but at a much lower frequency. A real time PCR based strategy to quantify the relative amount of spliced transcript confirmed a 10–30 fold increase in exon 11 skipping in CLL cells (n=29) as compared to normal B cells (n=4) and the degree of skipping increased with increased Rai stage. In addition, real time PCR also demonstrated a significant decrease in total wild type E-cadherin RNA expression in 58% of CLL specimens compared to normal B cells. As prior work demonstrates an upregulated wnt/β-catenin pathway in CLL and, and that E-cadherin can be a physiologic regulator of this pathway, we tested wnt pathway reporter expression in CLL specimens. High expression was identified and ectopic expression of E-cadherin in some samples with E-cadherin loss was sufficient to inhibit the wnt-reporter activity. This suggests that E-cadherin loss, possibly due to exon 11 alternative splicing and exon skipping plays a role in upregulated wnt signaling. As there are no mutations in exon 11 or its flanking intronic regions in CLL cells, our results suggest a novel mechanism of E-cadherin gene inactivation in which the trans-factors/splicing factors in malignant B lymphocytes induce an increased nonproductive splicing of a tumor suppressor gene.


2013 ◽  
Vol 6 (3) ◽  
pp. 755-767 ◽  
Author(s):  
J. N. Ahmed ◽  
R. G. Ali ◽  
N. Warr ◽  
H. M. Wilson ◽  
H. M. Bellchambers ◽  
...  

Author(s):  
Vincent Chu ◽  
Qing Feng ◽  
Yang Lim ◽  
Sichen Shao

The translation of mRNAs that contain a premature termination codon (PTC) generates truncated proteins that may have toxic dominant negative effects. Nonsense-mediated decay (NMD) is an mRNA surveillance pathway that degrades PTC-containing mRNAs to limit the production of truncated proteins. NMD activation requires a ribosome terminating translation at a PTC, but what happens to the polypeptides synthesized during the translation cycle needed to activate NMD is incompletely understood. Here, by establishing reporter systems that encode the same polypeptide sequence before a normal or premature termination codon, we show that termination of protein synthesis at a PTC is sufficient to selectively destabilize polypeptides in mammalian cells. Proteasome inhibition specifically rescues the levels of nascent polypeptides produced from PTC-containing mRNAs within an hour, but also disrupts mRNA homeostasis within a few hours. PTC-terminated polypeptide destabilization is also alleviated by depleting the central NMD factor UPF1 or SMG1, the kinase that phosphorylates UPF1 to activate NMD, but not by inhibiting SMG1 kinase activity. Our results suggest that polypeptide degradation is linked to PTC recognition in mammalian cells and clarify a framework to investigate these mechanisms.


Blood ◽  
1998 ◽  
Vol 91 (9) ◽  
pp. 3390-3396 ◽  
Author(s):  
M. Bernardetta Majolini ◽  
Mario M. D'Elios ◽  
Piero Galieni ◽  
Marianna Boncristiano ◽  
Francesco Lauria ◽  
...  

Src family kinases play a key role in mitogenesis. The exquisitely tissue-specific distribution of different Src family members suggests that a fine tuning of their expression might be a key prerequisite for cell homeostasis. We tested B cells from patients affected by B-cell chronic lymphocytic leukemia (B-CLL) for expression of Src family kinases. The T-cell–specific tyrosine kinase Lck was found to be expressed at significant levels in CLL B-cells. This finding could be accounted for either by ectopic expression of Lck in B-CLL or by specific expression of this kinase in normal B-1 cells, which are believed to be the normal counterpart of CLL B cells. To answer this question B cells from different sources, characterized by a different size of the B-1 subpopulation, were tested for Lck expression. The results show that Lck expression is a feature of CD5+, B-1 cells, suggesting a potential role for Lck in the self-renewal capacity of this B-cell subpopulation and supporting the notion that B-1 cells are the subset undergoing oncogenic transformation in B-CLL. Furthermore, we show that the CD5−, B-2 subpopulation, while normally lacking Lck expression, acquires the capacity to express Lck ectopically upon transformation by EBV.


2020 ◽  
Author(s):  
Haowei Jiang ◽  
Stephen Martin Pederson ◽  
Morgan Newman ◽  
Yang Dong ◽  
Michael Lardelli

AbstractPRESENILIN 2 (PSEN2) is one of the genes mutated in early onset familial Alzheimer’s disease (EOfAD). PSEN2 shares significant amino acid sequence identity with another EOfAD-related gene PRESENILIN 1 (PSEN1), and partial functional redundancy is seen between these two genes. However, the complete range of functions of PSEN1 and PSEN2 is not yet understood. In this study, we performed targeted mutagenesis of the zebrafish psen2 gene to generate a premature termination codon close downstream of the translation start with the intention of creating a null mutation. Homozygotes for this mutation, psen2S4Ter, are viable and fertile, and adults do not show any gross pigmentation defects, arguing against significant loss of γ-secretase activity. Also, assessment of the numbers of Dorsal Longitudinal Ascending (DoLA) interneurons that are responsive to psen2 but not psen1 activity during embryogenesis did not reveal decreased psen2 function. Transcripts containing the S4Ter mutation show no evidence of destabilization by nonsense-mediated decay. Forced expression in zebrafish embryos of fusions of psen2S4Ter 5’ mRNA sequences with sequence encoding enhanced green fluorescent protein (EGFP) indicated that the psen2S4Ter mutation permits utilization of cryptic, novel downstream translation start codons. These likely initiate translation of N-terminally truncated Psen2 proteins that obey the “reading frame preservation rule” of PRESENILIN EOfAD mutations. Transcriptome analysis of entire brains from a 6-month-old family of wild type, heterozygous and homozygous psen2S4Ter female siblings revealed profoundly dominant effects on gene expression likely indicating changes in ribosomal, mitochondrial, and anion transport functions.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 1232-1232 ◽  
Author(s):  
Arianna Bottoni ◽  
Rizzotto Lara ◽  
Lai Tzung-Huei ◽  
Rose Mantel ◽  
Lisa Simith ◽  
...  

Abstract The B-Cell Receptor (BCR) pathway is overactive and plays a crucial role in the survival, proliferation and trafficking of chronic lymphocytic leukemia (CLL), the most common leukemia in adults. Brutons agammaglobulenemia tyrosine kinase (BTK) is a key downstream component of the BCR pathway and targeting BTK with ibrutinib, an orally bioavailable irreversible inhibitor of BTK has had remarkable success in relapsed CLL including in patients with high risk genetic features. However, therapy-induced point mutations affecting BTK p.C481, which convert ibrutinib from an irreversible inhibitor to a less potent reversible inhibitor, have emerged in a subset of patients with high risk genomic factors. Alternative strategies are therefore needed to prevent and treat ibrutinib-resistant disease for these high risk patients. Micro-RNAs (miRNAs) are small non coding RNAs that are aberrantly expressed in many cancers including CLL. We have previously shown that epigenetic silencing, due to the overexpression of HDACs in CLL, was responsible for the low expression of specific miRNAs in CLL. In this study we identified a key set of microRNA (miR147b, miR-210, miR425, miR1253, miR4267 and miR4667-3p) that targeted BTK. Ectopic expression of each individual miRNA reduced BTK expression by 30-50% whereas a pool of these miRNA reduced BTK by 80%. When the expression of the BTK targeting microRNAs was compared in the CLL cells from 80 patients with a pool of healthy B-cells (5 donors) we found that CLL cells expressed lower levels of the BTK targeting microRNA compared to healthy B cells. Chromatin immunoprecipitation followed by deep sequencing as well as real time-PCR experiments identified that a gene repressive complex, consisting of HDAC1, HDAC2 and KDM1, became recruited to the promoters of these microRNA to silence their expression. Consequently, targeting the HDACs with the novel 2nd generation HDAC inhibitor Abexinostat induced the expression of BTK targeting miRNAs by 5-200 fold with reciprocal decreases in the levels of BTK protein. Similarly RNAi against HDAC1 induced the BTK targeting miRNA as well as decreased BTK in primary CLL cells as well as in Mec2, a CLL cell line. We then demonstrated that HDAC inhibition with Abexinostat synergized with ibrutinib to dually target BTK (where abexinostat decreased total BTK protein and ibrutinib inhibits kinase activity of remaining BTK to achieve a more complete inhibition of BTK signaling and synergistic cytotoxicity. Importantly, we also demonstrated that HDAC inhibitors retain the ability to induce the BTK targeting microRNA and reduce BTK protein in ibrutinib-resistant CLL cells that developed the mutant p.C481S BTK. Together, our results show that the over-expression of HDACs in CLL causes epigenetic silencing of BTK targeting miRNAs leading to a higher expression of BTK in these patients. HDAC inhibition restores the expression of these miRNAs which in turn target BTK effecting a quantitative reduction. Since Abex is well toleratd in phase 1 clinical trials for heme malignancies, it makes an attractive therapeutic strategy to target BTK as part of a combination therapy as well as in Ibrutinib resistant patients. Disclosures Byrd: Acerta Pharma BV: Research Funding.


Blood ◽  
1998 ◽  
Vol 91 (9) ◽  
pp. 3390-3396 ◽  
Author(s):  
M. Bernardetta Majolini ◽  
Mario M. D'Elios ◽  
Piero Galieni ◽  
Marianna Boncristiano ◽  
Francesco Lauria ◽  
...  

Abstract Src family kinases play a key role in mitogenesis. The exquisitely tissue-specific distribution of different Src family members suggests that a fine tuning of their expression might be a key prerequisite for cell homeostasis. We tested B cells from patients affected by B-cell chronic lymphocytic leukemia (B-CLL) for expression of Src family kinases. The T-cell–specific tyrosine kinase Lck was found to be expressed at significant levels in CLL B-cells. This finding could be accounted for either by ectopic expression of Lck in B-CLL or by specific expression of this kinase in normal B-1 cells, which are believed to be the normal counterpart of CLL B cells. To answer this question B cells from different sources, characterized by a different size of the B-1 subpopulation, were tested for Lck expression. The results show that Lck expression is a feature of CD5+, B-1 cells, suggesting a potential role for Lck in the self-renewal capacity of this B-cell subpopulation and supporting the notion that B-1 cells are the subset undergoing oncogenic transformation in B-CLL. Furthermore, we show that the CD5−, B-2 subpopulation, while normally lacking Lck expression, acquires the capacity to express Lck ectopically upon transformation by EBV.


2022 ◽  
Vol 23 (2) ◽  
pp. 656
Author(s):  
Marta Vallverdú-Prats ◽  
Ramon Brugada ◽  
Mireia Alcalde

Arrhythmogenic cardiomyopathy is a heritable heart disease associated with desmosomal mutations, especially premature termination codon (PTC) variants. It is known that PTC triggers the nonsense-mediated decay (NMD) mechanism. It is also accepted that PTC in the last exon escapes NMD; however, the mechanisms involving NMD escaping in 5′-PTC, such as reinitiation of translation, are less known. The main objective of the present study is to evaluate the likelihood that desmosomal genes carrying 5′-PTC will trigger reinitiation. HL1 cell lines were edited by CRISPR/Cas9 to generate isogenic clones carrying 5′-PTC for each of the five desmosomal genes. The genomic context of the ATG in-frame in the 5′ region of desmosomal genes was evaluated by in silico predictions. The expression levels of the edited genes were assessed by Western blot and real-time PCR. Our results indicate that the 5′-PTC in PKP2, DSG2 and DSC2 acts as a null allele with no expression, whereas in the DSP and JUP gene, N-truncated protein is expressed. In concordance with this, the genomic context of the 5′-region of DSP and JUP presents an ATG in-frame with an optimal context for the reinitiation of translation. Thus, 5′-PTC triggers NMD in the PKP2, DSG2* and DSC2 genes, whereas it may escape NMD through the reinitiation of the translation in DSP and JUP genes, with no major effects on ACM-related gene expression.


Sign in / Sign up

Export Citation Format

Share Document