Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 3 de 3
Filter
1.
BMC Mol Biol ; 18(1): 19, 2017 07 20.
Article in English | MEDLINE | ID: mdl-28728573

ABSTRACT

BACKGROUND: RBM10 is an RNA binding protein involved in message stabilization and alternative splicing regulation. The objective of the research described herein was to identify novel targets of RBM10-regulated splicing. To accomplish this, we downregulated RBM10 in human cell lines, using small interfering RNAs, then monitored alternative splicing, using a reverse transcription-PCR screening platform. RESULTS: RBM10 knockdown (KD) provoked alterations in splicing events in 10-20% of the pre-mRNAs, most of which had not been previously identified as RBM10 targets. Hierarchical clustering of the genes affected by RBM10 KD revealed good conservation of alternative exon inclusion or exclusion across cell lines. Pathway annotation showed RAS signaling to be most affected by RBM10 KD. Of particular interest was the finding that splicing of SMN pre-mRNA, encoding the survival of motor neuron (SMN) protein, was influenced by RBM10 KD. Inhibition of RBM10 resulted in preferential expression of the full-length, exon 7 retaining, SMN transcript in four cancer cell lines and one normal skin fibroblast cell line. SMN protein is expressed from two genes, SMN1 and SMN2, but the SMN1 gene is homozygously disrupted in people with spinal muscular atrophy; as a consequence, all of the SMN that is expressed in people with this disease is from the SMN2 gene. Expression analyses using primary fibroblasts from control, carrier and spinal muscle atrophy donors demonstrated that RBM10 KD resulted in preferential expression of the full-length, exon 7 retaining, SMN2 transcript. At the protein level, upregulation of the full-length SMN2 was also observed. Re-expression of RBM10, in a stable RBM10 KD cancer cell line, correlated with a reversion of the KD effect, demonstrating specificity. CONCLUSION: Our work has not only expanded the number of pre-mRNA targets for RBM10, but identified RBM10 as a novel regulator of SMN2 alternative inclusion.


Subject(s)
RNA Precursors/genetics , RNA Splicing , RNA-Binding Proteins/metabolism , Alternative Splicing , Cell Line , Cluster Analysis , Computational Biology/methods , Exons , Fibroblasts , Gene Expression Profiling , Humans , Reproducibility of Results , Signal Transduction , Survival of Motor Neuron 2 Protein/genetics , ras Proteins/metabolism
2.
In Vitro Cell Dev Biol Anim ; 52(3): 327-336, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26659391

ABSTRACT

We previously examined the expression of Rbm5 during myoblast differentiation and found significantly more protein in the early stages of skeletal myoblast differentiation than during the later stages. We decided to determine if this elevated level was necessary for differentiation. Our hypothesis was that if high levels of Rbm5 protein expression were necessary for the initiation of skeletal myoblast differentiation, then inhibition of expression would prevent differentiation. Our long-term objective is to inhibit Rbm5 expression and examine the effect on H9c2 differentiation. Towards this end, stable knockdown clones and transient knockdown populations were generated. Expression analyses in H9c2 myoblasts demonstrated significant Rbm5 messenger RNA (mRNA) inhibition but, surprisingly, no effect on RBM5 protein levels. Expression of the Rbm5 paralogue Rbm10 was examined in order to (a) ensure no off-target knockdown effect, and (b) investigate any possible compensatory effects. RBM10 protein levels were found to be elevated, in both the clonal and transiently transfected populations. These results suggest that myoblast RBM5 expression is regulated by a process that includes RNA sequestration and/or controlled translation, and that (a) RBM5 function is compensated for by RBM10, and/or (b) RBM5 regulates RBM10 expression. We have developed a model to describe our findings, and suggest further experiments for testing its validity. Since upregulation of Rbm10 might compensate for downregulated Rbm5, and consequently might mask any potential knockdown effect, it could lead to incorrect conclusions regarding the importance of Rbm5 for differentiation. It is therefore imperative to determine how both RBM5 and RBM10 protein expression is regulated.


Subject(s)
Cell Cycle Proteins/biosynthesis , Cell Cycle Proteins/genetics , DNA-Binding Proteins/biosynthesis , DNA-Binding Proteins/genetics , Gene Expression Regulation/genetics , Myoblasts/cytology , Nuclear Proteins/biosynthesis , RNA-Binding Proteins/biosynthesis , RNA-Binding Proteins/genetics , Transcriptional Activation/genetics , Tumor Suppressor Proteins/biosynthesis , Tumor Suppressor Proteins/genetics , Animals , Cell Cycle Proteins/metabolism , Cell Differentiation/genetics , Cell Line , DNA-Binding Proteins/metabolism , Gene Expression Profiling , RNA Interference , RNA, Messenger/genetics , RNA, Small Interfering/genetics , RNA-Binding Proteins/metabolism , Rats , Tumor Suppressor Proteins/metabolism
3.
BMC Res Notes ; 8: 46, 2015 Feb 19.
Article in English | MEDLINE | ID: mdl-25889998

ABSTRACT

BACKGROUND: RBM10 is an RNA binding protein involved in the regulation of transcription, alternative splicing and message stabilization. Mutations in RBM10, which maps to the X chromosome, are associated with TARP syndrome, lung and pancreatic cancers. Two predominant isoforms of RBM10 exist, RBM10v1 and RBM10v2. Both variants have alternate isoforms that differ by one valine residue, at amino acid 354 (RBM10v1) or 277 (RBM10v2). It was recently observed that a novel point mutation at amino acid 354 of RBM10v1, replacing valine with glutamic acid, correlated with preferential expression of an exon 11 inclusion variant of the proliferation regulatory protein NUMB, which is upregulated in lung cancer. FINDINGS: We demonstrate, using the GLC20 male-derived small cell lung cancer cell line - confirmed to have only one X chromosome - that the two (+/-) valine isoforms of RBM10v1 and RBM10v2 result from alternative splicing. Protein modeling of the RNA Recognition Motif (RRM) within which the alteration occurs, shows that the presence of valine inhibits the formation of one of the two α-helices associated with RRM tertiary structure, whereas the absence of valine supports the α-helical configuration. We then show 2-fold elevated expression of the transcripts encoding the minus valine RBM10v1 isoform in GLC20 cells, compared to those encoding the plus valine isoform. This expression correlates with preferential expression of the lung cancer-associated NUMB exon 11 inclusion variant. CONCLUSIONS: Our observations suggest that the ability of RBM10v1 to regulate alternative splicing depends, at least in part, on a structural alteration within the second RRM domain, which influences whether RBM10v1 functions to support or repress splicing. A model is presented.


Subject(s)
Alternative Splicing , Gene Expression Regulation, Neoplastic , Lung Neoplasms/genetics , Membrane Proteins/genetics , Nerve Tissue Proteins/genetics , RNA, Messenger/genetics , RNA-Binding Proteins/genetics , Small Cell Lung Carcinoma/genetics , Cell Line, Tumor , Exons , Glutamic Acid/genetics , Glutamic Acid/metabolism , Humans , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Male , Membrane Proteins/metabolism , Molecular Sequence Data , Nerve Tissue Proteins/metabolism , Nucleotide Motifs , Point Mutation , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Structure, Secondary , Protein Structure, Tertiary , RNA, Messenger/metabolism , RNA-Binding Proteins/metabolism , Signal Transduction , Small Cell Lung Carcinoma/metabolism , Small Cell Lung Carcinoma/pathology , Valine/genetics , Valine/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL