Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
1.
Int J Mol Sci ; 22(10)2021 May 12.
Article in English | MEDLINE | ID: mdl-34065983

ABSTRACT

Dysregulation of messenger RNA (mRNA) processing-in particular mRNA splicing-is a hallmark of cancer. Compared to normal cells, cancer cells frequently present aberrant mRNA splicing, which promotes cancer progression and treatment resistance. This hallmark provides opportunities for developing new targeted cancer treatments. Splicing of precursor mRNA into mature mRNA is executed by a dynamic complex of proteins and small RNAs called the spliceosome. Spliceosomes are part of the supraspliceosome, a macromolecular structure where all co-transcriptional mRNA processing activities in the cell nucleus are coordinated. Here we review the biology of the mRNA splicing machinery in the context of other mRNA processing activities in the supraspliceosome and present current knowledge of its dysregulation in lung cancer. In addition, we review investigations to discover therapeutic targets in the spliceosome and give an overview of inhibitors and modulators of the mRNA splicing process identified so far. Together, this provides insight into the value of targeting the spliceosome as a possible new treatment for lung cancer.


Subject(s)
Lung Neoplasms/genetics , RNA Splicing , Spliceosomes/metabolism , Alternative Splicing , Cell Nucleus/metabolism , Disease Progression , Drug Resistance, Neoplasm , Gene Expression Regulation, Neoplastic , Humans , Lung Neoplasms/metabolism
2.
Int J Mol Sci ; 21(12)2020 Jun 12.
Article in English | MEDLINE | ID: mdl-32545483

ABSTRACT

The core spliceosomal Sm proteins were recently proposed as cancer-selective lethal targets in non-small cell lung cancer (NSCLC). In contrast, the loss of the commonly mutated cancer target SF3B1 appeared to be toxic to non-malignant cells as well. In the current study, the transcriptomes of A549 NSCLC cells, in which SF3B1 or SNRPD3 was silenced, were compared using RNA sequencing. The skipping of exon 4 of the proteasomal subunit beta type-3 (PSMB3) mRNA, resulting in a shorter PSMB3-S variant, occurred only after silencing SNRPD3. This observation was extended to the other six Sm genes. Remarkably, the alternative splicing of PSMB3 mRNA upon Sm gene silencing was not observed in non-malignant IMR-90 lung fibroblasts. Furthermore, PSMB3 was found to be overexpressed in NSCLC clinical samples and PSMB3 expression correlated with Sm gene expression. Moreover, a high PSMB3 expression corresponds to worse survival in patients with lung adenocarcinomas. Finally, silencing the canonical full-length PSMB3-L, but not the shorter PSMB3-S variant, was cytotoxic and was accompanied by a decrease in proteasomal activity. Together, silencing Sm genes, but not SF3B1, causes a cytotoxic alternative splicing switch in the PSMB3 mRNA in NSCLC cells only.


Subject(s)
Carcinoma, Non-Small-Cell Lung/genetics , Gene Expression Profiling/methods , Lung Neoplasms/genetics , Phosphoproteins/genetics , Proteasome Endopeptidase Complex/genetics , RNA Splicing Factors/genetics , snRNP Core Proteins/genetics , A549 Cells , Alternative Splicing , Down-Regulation , Exons , Gene Expression Regulation, Neoplastic , Gene Silencing , Humans , Nonsense Mediated mRNA Decay , Sequence Analysis, RNA
3.
Mol Cancer Ther ; 19(2): 552-563, 2020 02.
Article in English | MEDLINE | ID: mdl-31619462

ABSTRACT

Loss of function of BRCA1-associated protein 1 (BAP1) is observed in about 50% of malignant pleural mesothelioma (MPM) cases. The aim of this study was to investigate whether this aspect could be exploited for targeted therapy. A genetically engineered model was established expressing either functional or nonfunctional BAP1, and whole-genome siRNA synthetic lethality screens were performed assessing differentially impaired survival between the two cell lines. The whole-genome siRNA screen unexpectedly revealed 11 hits (FDR < 0.05) that were more cytotoxic to BAP1-proficient cells. Two actionable targets, ribonucleotide reductase (RNR) catalytic subunit M1 (RRM1) and RNR regulatory subunit M2 (RRM2), were validated. In line with the screen results, primary mesothelioma (BAP1 +/-) overexpressing BAP1 C91A (catalytically dead mutant) was more resistant to RNR inhibition, while BAP1 knockdown in the BAP1-proficient cell lines rescued the cells from their vulnerability to RNR depletion. Gemcitabine and hydroxyurea were more cytotoxic in BAP1-proficient cell line-derived spheroids compared with BAP1 deficient. Upregulation of RRM2 upon gemcitabine and hydroxyurea treatment was more profound in BAP1 mut/del cell lines. Increased lethality mediated by RNR inhibition was observed in NCI-H2452 cells reconstituted with BAP1-WT but not with BAP1 C91A. Upregulation of RRM2 in NCI-H2452-BAP1 WT spheroids was modest compared with control or C91A mutant. Together, we found that BAP1 is involved in the regulation of RNR levels during replication stress. Our observations reveal a potential clinical application where BAP1 status could serve as predictive or stratification biomarker for RNR inhibition-based therapy in MPM.


Subject(s)
Mesothelioma/drug therapy , Mesothelioma/genetics , Pleural Neoplasms/genetics , Ribonucleoside Diphosphate Reductase/antagonists & inhibitors , Tumor Suppressor Proteins/genetics , Ubiquitin Thiolesterase/genetics , Antimetabolites, Antineoplastic/pharmacology , Cell Line, Tumor , Deoxycytidine/analogs & derivatives , Deoxycytidine/pharmacology , Drug Resistance, Neoplasm , Enzyme Inhibitors/pharmacology , Gene Knockdown Techniques , Genomics , Humans , Hydroxyurea/pharmacology , Mesothelioma/enzymology , Pleural Neoplasms/drug therapy , Pleural Neoplasms/enzymology , Ribonucleoside Diphosphate Reductase/genetics , Ribonucleoside Diphosphate Reductase/metabolism , Transfection , Tumor Suppressor Proteins/metabolism , Ubiquitin Thiolesterase/metabolism , Gemcitabine
4.
Oncogene ; 38(21): 4142-4153, 2019 05.
Article in English | MEDLINE | ID: mdl-30705407

ABSTRACT

Novel therapeutic strategies for non-small-cell lung cancer (NSCLC) are urgently needed. RNA splicing, orchestrated by the spliceosome, is deregulated in many forms of cancer, including NSCLC. Here, we performed high-throughput screening with a small interfering RNA library targeting all annotated human spliceosome proteins to identify cancer-selective lethal targets in the RNA splicing machinery. Silencing of several spliceosome proteins reduced cell viability in a panel of NSCLC cell lines, but not in non-malignant lung fibroblasts and epithelial cells. Interestingly, the cancer-selective lethal target set comprised all seven Sm proteins that, together with small nuclear RNA, form the core structure of most spliceosome subunits. Interfering with Sm protein expression induced apoptosis in NSCLC cells, but not in non-malignant cells. In silico analysis revealed that Sm proteins are frequently upregulated in NSCLC. For several Sm proteins, increased expression showed a positive correlation with disease severity. Together, our results suggest that the Sm proteins represent particularly useful novel targets for selective treatment of NSCLC.


Subject(s)
RNA Interference/physiology , RNA, Small Interfering/genetics , Spliceosomes/genetics , A549 Cells , Apoptosis/genetics , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line , Cell Line, Tumor , Epithelial Cells/pathology , Fibroblasts/pathology , High-Throughput Screening Assays/methods , Humans , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Membrane Proteins/genetics , RNA Splicing/genetics , Up-Regulation/genetics
5.
EBioMedicine ; 39: 215-225, 2019 Jan.
Article in English | MEDLINE | ID: mdl-30581150

ABSTRACT

INTRODUCTION: Therapeutic options for diffuse malignant peritoneal mesothelioma (DMPM) are limited to surgery and locoregional chemotherapy. Despite improvements in survival rates, patients eventually succumb to disease progression. We investigated splicing deregulation both as molecular prognostic factor and potential novel target in DMPM, while we tested modulators of SF3b complex for antitumor activity. METHODS: Tissue-microarrays of 64 DMPM specimens were subjected to immunohistochemical assessment of SF3B1 expression and correlation to clinical outcome. Two primary cell cultures were used for gene expression profiling and in vitro screening of SF3b modulators. Drug-induced splicing alterations affecting downstream cellular pathways were detected through RNA sequencing. Ultimately, we established bioluminescent orthotopic mouse models to test the efficacy of splicing modulation in vivo. RESULTS: Spliceosomal genes are differentially upregulated in DMPM cells compared to normal tissues and high expression of SF3B1 correlated with poor clinical outcome in univariate and multivariate analysis. SF3b modulators (Pladienolide-B, E7107, Meayamycin-B) showed potent cytotoxic activity in vitro with IC50 values in the low nanomolar range. Differential splicing analysis of Pladienolide-B-treated cells revealed abundant alterations of transcripts involved in cell cycle, apoptosis and other oncogenic pathways. This was validated by RT-PCR and functional assays. E7107 demonstrated remarkable in vivo antitumor efficacy, with significant improvement of survival rates compared to vehicle-treated controls. CONCLUSIONS: SF3B1 emerged as a novel potential prognostic factor in DMPM. Splicing modulators markedly impair cancer cell viability, resulting also in potent antitumor activity in vivo. Our data designate splicing as a promising therapeutic target in DMPM.


Subject(s)
Antineoplastic Agents/administration & dosage , Lung Neoplasms/drug therapy , Mesothelioma/drug therapy , Peritoneal Neoplasms/drug therapy , Phosphoproteins/metabolism , RNA Splicing Factors/metabolism , RNA Splicing/drug effects , Tissue Array Analysis/methods , Aged , Animals , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Epoxy Compounds/administration & dosage , Epoxy Compounds/pharmacology , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Gene Regulatory Networks/drug effects , Humans , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Macrolides/administration & dosage , Macrolides/pharmacology , Male , Mesothelioma/genetics , Mesothelioma/metabolism , Mesothelioma, Malignant , Mice , Middle Aged , Morpholines/administration & dosage , Morpholines/pharmacology , Peritoneal Neoplasms/genetics , Peritoneal Neoplasms/metabolism , Phosphoproteins/genetics , Pyrans/administration & dosage , Pyrans/pharmacology , RNA Splicing Factors/genetics , Xenograft Model Antitumor Assays
6.
Mol Oncol ; 11(5): 534-551, 2017 05.
Article in English | MEDLINE | ID: mdl-28296343

ABSTRACT

Reinstating wild-type tumor suppressor p53 activity could be a valuable option for the treatment of cancer. To contribute to development of new treatment options for non-small cell lung cancer (NSCLC), we performed genome-wide siRNA screens for determinants of p53 activity in NSCLC cells. We identified many genes not previously known to be involved in regulating p53 activity. Silencing p53 pathway inhibitor genes was associated with loss of cell viability. The largest functional gene cluster influencing p53 activity was mRNA splicing. Prominent p53 activation was observed upon silencing of specific spliceosome components, rather than by general inhibition of the spliceosome. Ten genes were validated as inhibitors of p53 activity in multiple NSCLC cell lines: genes encoding the Ras pathway activator SOS1, the zinc finger protein TSHZ3, the mitochondrial membrane protein COX16, and the spliceosome components SNRPD3, SF3A3, SF3B1, SF3B6, XAB2, CWC22, and HNRNPL. Silencing these genes generally increased p53 levels, with distinct effects on CDKN1A expression, induction of cell cycle arrest and cell death. Silencing spliceosome components was associated with alternative splicing of MDM4 mRNA, which could contribute to activation of p53. In addition, silencing splice factors was particularly effective in killing NSCLC cells, albeit in a p53-independent manner. Interestingly, silencing SNRPD3 and SF3A3 exerted much stronger cytotoxicity to NSCLC cells than to lung fibroblasts, suggesting that these genes could represent useful therapeutic targets.


Subject(s)
Alternative Splicing , Carcinoma, Non-Small-Cell Lung/genetics , Lung Neoplasms/genetics , RNA, Small Interfering/physiology , Tumor Suppressor Protein p53/metabolism , A549 Cells , Alternative Splicing/drug effects , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Apoptosis , Carcinoma, Non-Small-Cell Lung/drug therapy , Cell Cycle Checkpoints , Cell Cycle Proteins , Cell Survival , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Fibroblasts/drug effects , Gene Silencing , Genome-Wide Association Study , Humans , Lung Neoplasms/drug therapy , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Spliceosomes/genetics , Spliceosomes/metabolism , Tumor Suppressor Protein p53/genetics
7.
Int J Biol Sci ; 11(12): 1363-75, 2015.
Article in English | MEDLINE | ID: mdl-26681916

ABSTRACT

Genetic and epigenetic alterations have been identified as to contribute directly or indirectly to the generation of transitional cell carcinoma of the urinary bladder (TCC-UB). We have previously found that amplification of chromosome 6p22 is significantly associated with the muscle-invasive rather than superficial TCC-UB. Here, we demonstrated that Sox4, one of the candidate oncogenes located within the chromosome 6p22 amplicon, confers bladder cancer stem cell (CSC) properties. Down-regulation of Sox4 led to the inhibition of cell migration, colony formation as well as mesenchymal-to-epithelial transition (MET). Interestingly, knockdown of Sox4 also reduced the sphere formation, enriched cell population with high levels of aldehyde dehydrogenase (ALDH (high)) and tumor formation potential. Using gene expression profiling, we further identified novel Sox4 target genes. Last, immunohistochemistry analysis of human bladder tumor tissue microarrays (TMAs) indicated that high Sox4 expression was correlated with advanced cancer stages and poor survival rate. In summary, our data show that Sox4 is an important regulator of the bladder CSC properties and it may serve as a biomarker of the aggressive phenotype in bladder cancer.


Subject(s)
Carcinoma, Transitional Cell/genetics , Neoplastic Stem Cells/pathology , SOXC Transcription Factors/genetics , Urinary Bladder Neoplasms/genetics , Biomarkers, Tumor/genetics , Carcinoma, Transitional Cell/pathology , Cell Line, Tumor , Chromosomes, Human, Pair 6 , Cohort Studies , Epithelial-Mesenchymal Transition , Humans , Prognosis , Urinary Bladder Neoplasms/pathology
8.
Oncotarget ; 5(23): 12166-76, 2014 Dec 15.
Article in English | MEDLINE | ID: mdl-25361000

ABSTRACT

Triple-negative breast cancer (TNBC) accounts for approximately 15-20% of all breast cancer (BC) cases and contributes disproportionately to BC mortality. TAZ, a key transducer of the Hippo pathway, has recently been demonstrated to confer breast cancer stem cell (CSC) traits. However, TAZ target genes and the underlying transcriptional regulatory pathways responsible for the CSC phenomenon remain unknown. Here, we demonstrate that the oncogenic activity of TAZ is essential for propagation of the malignant phenotype. We further show that constitutively active TAZ tumor-derived cells exhibit unique tumor-initiating properties, including increased self-renewal and metastatic seeding potential, acquired chemotherapy resistance and the ability to efficiently regenerate tumor formation in vivo. Combined digital RNA expression analysis and computational network approaches identify several signaling pathways that distinguish breast cancer tumor-initiating cells (T-ICs) from bulk tumor cells. We demonstrate the utility of this approach by repositioning the small molecule tyrosine kinase inhibitor, Dasatinib, which selectively targets T-ICs and inhibits TNBC growth in vivo.


Subject(s)
Cell Transformation, Neoplastic/genetics , Intracellular Signaling Peptides and Proteins/genetics , Signal Transduction/genetics , Transcriptome/genetics , Triple Negative Breast Neoplasms/genetics , Animals , Blotting, Western , Cell Line, Tumor , Dasatinib , Flow Cytometry , Heterografts , Humans , Mice , Mice, SCID , Neoplastic Stem Cells , Protein Kinase Inhibitors/pharmacology , Pyrimidines/pharmacology , Thiazoles/pharmacology , Trans-Activators , Transcription Factors , Transcriptional Coactivator with PDZ-Binding Motif Proteins , Transduction, Genetic , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...