Journal of Cystic Fibrosis 10 (2011) 212–216 www.elsevier.com/locate/jcf

Short Communication Splicing defects in the CFTR gene: Minigene analysis of two mutations, 1811+1GNC and 1898+3ANG

Gwendal Dujardin a, Diane Commandeur b, Catherine Le Jossic-Corcos a, ⁎ Claude Ferec a, Laurent Corcos a,

a INSERM U613, Faculty of Medicine, 22, avenue Camille Desmoulins, 29200 Brest, France b Hôpital d'Instruction des Armées, rue du colonel fonferrier, 29019 Brest, France

Received 10 September 2010; received in revised form 22 December 2010; accepted 26 December 2010

Abstract

Background: Cystic fibrosis is caused by mutations of the Cystic Fibrosis Transmembrane conductance Regulator gene (CFTR). Among the 1795 reported mutations, 221 (12.31%) are believed to affect pre-mRNA splicing. Nevertheless, not all splicing mutations have been demonstrated, by functional assays, to affect splicing in living cells. Methods: We have used a minigene-based approach, coupled to site-specific mutagenesis, to analyze the effects of presumptive pre-mRNA splicing mutations. Results: We show here that the 11 1811+1GNC and the intron 12 1898+3ANG mutations strongly affected CFTR pre-mRNA splicing. The encoded are predicted to be defective, which would thus participate in the disease phenotype of carrier individuals. Conclusions: These results further validate the minigene strategy for the study of presumptive splice mutations, and report unanticipated defects in splicing. Such assays should improve the analysis of genotype–phenotype correlations. © 2011 European Cystic Fibrosis Society. Published by Elsevier B.V. All rights reserved.

Keywords: Cystic fibrosis; ; Mutation analysis; Minigene assays

1. Introduction analysis from patient samples, analysis of mRNA isolated from immortalized lymphocytes or minigene assays [1]. In this study, we Cystic fibrosis (CF) is caused by mutations of the CFTR gene, have chosen to analyze, with a minigene approach, the effect of two which encodes a chloride transporter . CF is the most intronic splice site mutations, one in intron 11 (1811+1GNC) and frequent autosomal recessive genetic disease, with 1 in 3000 one in intron 12 (1898+3ANG) associated with phenotypes of affected carrier individuals. The delta F508 (F508del) mutation is rather distinct severities. the most frequent, as it accounts for roughly 70% of the mutant The 1811+1GNC mutation has been previously described by alleles. Disease severity depends, in part, on the nature of the Petreska et al. [2]. This mutation was either associated with the mutation that may be present on the other allele. Among the 1795 deltaF508 (18/204), with non-deltaF508 (82/204) or with reported mutations, 221 (12.31%) are believed to affect pre-mRNA normal (104/204) chromosomes. The young patient who carried splicing, but the demonstration of a functional effect has not been the deltaF508 mutation on the other chromosome showed systematically obtained. Several methods can be used to look for malnutrition due to malabsorption, bilateral bronchopneumonia potential pre-mRNA splicing defects, including direct mRNA with a compensatory emphysema, early manifested anemia, extremely high sweat C1- values (240 mmole L−1), pancreatic insufficiency and positive Pseudomonas aeruginosa coloniza- ⁎ Corresponding author. Tel.: +33 2 98 01 83 01; fax: +33 2 98 01 83 22. tion. In Brest, two young subjects, one girl and one boy, were E-mail address: [email protected] (L. Corcos). also diagnosed (in 1996) with both the 1811+1GNC mutation

1569-1993/$ - see front matter © 2011 European Cystic Fibrosis Society. Published by Elsevier B.V. All rights reserved. doi:10.1016/j.jcf.2010.12.008 G. Dujardin et al. / Journal of Cystic Fibrosis 10 (2011) 212–216 213 and the deltaF508 defect on the other allele, sweat Cl- values of inclusion of 11, with no detectable exclusion (Fig. 2a). By 120 and 110 mmole L−1 and pancreatic insufficiency. Hence in contrast, the mutant (1811+1GNC) minigene showed only little all three cases, the severity of the disease was quite strong. exon 11 inclusion with both a slower and a higher mobility The 1898+3ANG mutation was detected in one female bands in higher amounts. The cDNA products were sequenced subject at age 20, with a severe 3659delC frameshift mutation and, while the correctly sized band was identical with that from on the other allele. She showed a 96 mmol L−1 sweat Cl- value the WT minigene, the lower band showed complete exon 11 and pancreatic sufficiency. Hence, the severity of the phenotype skipping while the upper band showed partial intron 11 was relatively mild. retention (Fig. 2b). Usage of the donor splice site was shifted We used a minigene that had been previously validated to towards the 3′ end (as shown by the arrow in the lower part of provide a good matrix with which to assay splicing modifications the figure) of the CFTR sequence. Translation of the encoded [3–6]. The results presented herein demonstrate that the intron 11 sequences showed that the resulting proteins were likely to be 1811+1GNC mutation indeed affected CFTR pre-mRNA splicing non functional, as a stop codon appeared early in the reading with a complex pattern of anomalies, including strongly reduced frame (Fig. 2c). levels of correctly spliced transcript, exon exclusion and partial The second mutation (1898+3ANG) also drove aberrant intron retention. The intron 12 1898+3ANG mutation led to full CFTR mRNA production, with a full skipping of exon 12 skipping of exon 12. Protein structure prediction from the (Fig. 3a). A defective protein was also predicted from sequence corresponding transcripts suggested that both mutations would be analysis, although in this case, it was not truncated but internally expected to encode aberrant proteins most probably lacking deleted (Fig. 3b). chloride channel function. 4. Discussion 2. Materials and methods These analyses demonstrate that the minigene strategy could The minigene construct used has been described previously functionally detect the effects of these CFTR mutations, which [7,8]. In order to clone the fragment of interest, genomic DNA were either anticipated (1811+1GNC) or not (1898+3ANG) to from a healthy subject was extracted and used for PCR be splicing mutations. In addition, quite striking results were amplification using primers from sites within the 3′ portion of obtained. intron 10 or 11 and the 5′ portion of intron 11 or 12. The primers Concerning the 1811+1GNC mutation, which was located in were engineered to contain NdeI restriction sites (sequences on the invariant donor splice site (+1) and was thus supposed to request). The resulting fragment was first cloned into the PCR fully prevent splicing, we observed, surprisingly, alternative Topo2.1 plasmid (Invitrogen) and the sequence was verified splicing events with three distinct mRNAs including correctly using the Big Dye terminator cycle sequencing kit (Applied spliced mRNA, albeit in strongly diminished amounts (in a 20 Biosystems) and an ABI 3100 Genetic Analyzer (Applied to 1 ratio, as determined by densitometry analysis), when Biosystems). It was then removed with the NdeI restriction compared to the wild type control. We believe that this last enzyme, cloned into the minigene insertion vector and observation might be related to the high sensitivity of the sequenced again for orientation analysis. Two different approach. Indeed, the minigene strategy allows analyzing strategies were used to generate the minigenes carrying the mRNA expression from high copy numbers of transfected mutations of interest: a—the same strategy as for the WT plasmids, which translate into multiple mRNA copies detect- minigene but using the mutant genomic DNA as a PCR able by RT-PCR. Hence, it can be proposed that such a mutation template, or b—introducing the mutations by site-directed might not be incompatible with production of splicing at the mutagenesis (QuikChange II XL Site-Directed Mutagenesis, right position, provided that it is possible to detect low mRNA Stratagene). The 1811+1GNC and the 1898+3ANG mutations levels. The other two transcripts, which were more abundant were selected to design mutant minigene constructs, which were than the wild type transcript, should be interpreted as a then used to transfect HeLa cells transiently with the consequence of the low recognition efficiency of the splicing Lipofectamine 2000 reagent (Invitrogen). The cells were then machinery for the mutant site, and the use of alternative, lower incubated for 24 h and harvested to prepare total RNA. The affinity, splice sites. structure of the amplicons was analyzed by RT-PCR (OneStep At position 1898, we observed 2 different transcripts in the RT-PCR kit, Qiagen) and the products were gel purified absence of mutation. In fact, the activity of the splicing (NucleoSpin extract, Macherey-Nagel) and sequenced. The machinery can certainly be quite versatile, based on differences procedure outline is shown in Fig. 1. in the efficiency of splice site recognition. A very interesting model suggests that splice sites that may be relatively weak 3. Results would be less efficiently recognised than strong splice sites during elongation of transcription [11]. Hence, if the RNA polII The minigene strategy for pre-mRNA splicing analysis has progresses rapidly, only strong splice sites would be efficiently been validated previously [9]. Following plasmid in recognised, whereas if progression is average or slow, weaker HeLa cells, which have been commonly used for pre-mRNA sites would be correctly recognised, and the exon spliced in. It splicing analysis [10], the WT minigene centred on exon 11 can thus be postulated that in the wild type context, the showed a strong cDNA band corresponding to the normal proportion of the correctly spliced transcript from a normal 214 G. Dujardin et al. / Journal of Cystic Fibrosis 10 (2011) 212–216

Fig. 1. Preparation of CFTR hybrid minigenes. Strategy for engineering CFTR hybrid minigenes from either WT (left panel) or mutant (right panel) genomic DNA. The minigene structure has been described in [9]. The size of the flanking in the minigene is 1038 bp in 5′ and 1126 bp in 3′.

allele is in equilibrium with some level of exon 12 skipping. In mutant transcripts for the 1811+1 GNC mutation, and the higher the mutant, one possibility would be that splice site recognition intensity of the mutant transcript for the 1898+3 ANG mutation. would be hampered by proteins that could interact better, or In addition, the minigene approach does not allow analyzing even in full, with the mutant site, which was very proximal to the quantitative defects related to mRNA stability. It can the splice site. Such proteins are not likely to be SR proteins, as certainly be expected that the insertion of premature stop ESE finder did not predict a modulation in the binding ability of codons, as occurs for both of the studied mutations, could lead SR proteins according to the mutation (one site is predicted for to unstable mRNAs that might be subjected to nonsense mRNA SRp40 on the TGAAAGG sequence at the end of exon 12, a site decay (NMD). By contrast, an increased stability of the mature conserved between the wild type and the mutant). Rather, we mRNAs produced in the context of such genomic mutations that can hypothesize that negatively acting proteins, like hnRNP lead to the insertion of premature STOP codons is not expected. proteins, could be recruited to hamper normal splicing. This Nevertheless, what the approach reveals is only the ability of hypothesis will require further analysis. such pre-mRNAs to be aberrantly spliced because of the From a pathophysiological viewpoint, this study both confirms mutations. Consequently, the results of the RT-PCR approach previous expectations for the occurrence of a splicing defect in the should be interpreted as fair indicators of the ability of the case of the 1811+1GNC mutation, and uncovers a striking splicing aberrant transcripts to encode a CFTR-related polypeptide, were defect with the full exclusion of exon 12 in the case of 1898+3GNC. these stable enough in the patient cells, or no polypeptide at all As observed in the present study, variations in the levels of if these alternative transcripts are subjected to NMD. the RT-PCR products were obtained between the wild type and The results obtained with the two experimental strategies, the mutant minigenes. Some of these might be experimental, cloning mutant genomic DNA or site-directed mutagenesis on although consistently observed. We believe, rather, that the WT DNA, were identical. The site-directed mutagenesis differences in signal intensities may mainly reflect either the strategy allows the engineering of the mutant minigene in a competition between primer binding sites or the relative couple of days, much faster than the other method. In addition, efficiencies of the DNA polymerase over the amplicons that the site-directed mutagenesis approach makes it possible to are partially shared by all the transcripts produced from the analyze splicing mutations without DNA samples from patients, minigenes. This would explain both the lower intensity of the i.e. it can be used both retrospectively and prospectively. G. Dujardin et al. / Journal of Cystic Fibrosis 10 (2011) 212–216 215

Fig. 2. 1811+1 GNC mutation analysis. a, CFTR hybrid minigenes containing exon 11 and its flanking introns, carrying or not the 1811+1 GNC , were transfected into HeLa cells. RT-PCR products were resolved by agarose gel electrophoresis. b, Sequencing of the intron retention fragment (in the middle), compared to both EDB+1 (upper) and intron 11 WT (lower) sequences. The sequences are shown at the bottom part of the figure, and the used spliced sites are shown within squares. MUT: The 1811+1 GNC mutation. c, Comparison of the predicted translation product of both WT (upper sequence lane) and 1811+1 GNC (lower sequence name) mutant transcripts. No RT-PCR product was obtained in mock transfected cells.

This approach could be extended to other mutations and patterns found in patients or in immortalized cells from patients, polymorphisms believed to affect splicing. In addition, this provided that biological samples are available. methodology, while directly applicable to DNA sequence variations that occur in regions where consensus sequences Acknowledgements have been compiled, i.e. in the close vicinity of intron–exon junctions, should also be useful to analyze the effects of variant We thank MP Audrezet and K Giteau for their help with the sequences remote from these sites. For example, branch point selection of mutations from Brest genetic CFTR database. G mutations or modifications of the polypyrimidine tract in the 3′ Dujardin was supported by Vaincre la Mucoviscidose. D region of introns, or any other sequence predicted to affect the Commandeur was appointed by Brest Army Hospital. The binding of splicing regulatory proteins (like splicing enhancers authors thank Pr P Lehn for his comments on the manuscript. or silencers) could also be analyzed. To further validate the This work was supported by grants from Vaincre la Mucov- approach, these results could be confronted to the splicing iscidose, the INSERM, Brest University, the Medical Faculty of 216 G. Dujardin et al. / Journal of Cystic Fibrosis 10 (2011) 212–216

Fig. 3. 1898+3 ANG mutation analysis. a, CFTR hybrid minigenes containing exon 12 and its flanking introns, carrying or not the 1898+3 ANG point mutation, were transfected into HeLa cells. RT-PCR products were resolved by agarose gel electrophoresis (upper part). The localisation of the 1898+3 ANG point mutation is shown with an arrow (lower part). b, Comparison of the predicted translation product of both WT (upper sequence lane) and 1898+3 ANG (lower sequence name) mutant transcripts. No RT-PCR product was obtained in mock transfected cells.

Brest and the Institut Fédératif de Recherche 148-ScInBioS. We [5] Pagani F, Buratti E, Stuani C, Bendix R, Dork T, Baralle FE. A new type of are grateful to the Gaetan Saleun Association for its expert mutation causes a splicing defect in ATM. Nat Genet 2002 Apr;30(4): 426–9. administrative help. [6] Dujardin G, Buratti E, Charlet-Berguerand N, Martins de Araujo M, Mbopda A, Le Jossic-Corcos C, et al. CELF proteins regulate CFTR pre- mRNA splicing: essential role of the divergent domain of ETR-3. Nucleic References Acids Res 2010 Nov 1;38(20):7273–85. [7] Niksic M, Romano M, Buratti E, Pagani F, Baralle FE. Functional analysis [1] Mardon HJ, Sebastio G, Baralle FE. A role for exon sequences in of cis-acting elements regulating the alternative splicing of human CFTR alternative splicing of the human fibronectin gene. Nucleic Acids Res 1987 exon 9. Hum Mol Genet 1999 Dec;8(13):2339–49. Oct 12;15(19):7725–33. [8] D'Ambrogio A, Buratti E, Stuani C, Guarnaccia C, Romano M, Ayala [2] Petreska L, Koceva S, Efremov GD. A donor splice site mutation (1811 + YM, et al. Functional mapping of the interaction between TDP-43 and 1G–NC) in intron 11 of the CFTR gene identified in a patient of hnRNP A2 in vivo. Nucleic Acids Res 2009 Jul;37(12):4116–26. Macedonian origin. Hum Mutat 1996;7(4):375. [9] Amaral MD, Clarke LA, Ramalho AS, Beck S, Broackes-Carter F, [3] Vibe-Pedersen K, Kornblihtt AR, Baralle FE. Expression of a human Rowntree R, et al. Quantitative methods for the analysis of CFTR alpha-globin/fibronectin gene hybrid generates two mRNAs by alternative transcripts/splicing variants. J Cyst Fibros 2004 Aug;3(Suppl 2):17–23. splicing. EMBO J 1984 Nov;3(11):2511–6. [10] Norton PA. Alternative pre-mRNA splicing: factors involved in splice site [4] Pagani F, Buratti E, Stuani C, Romano M, Zuccato E, Niksic M, et al. selection. J Cell Sci 1994 Jan;107(Pt 1):1–7. Splicing factors induce cystic fibrosis transmembrane regulator exon 9 [11] Nogues G, Kadener S, Cramer P, de la Mata M, Fededa JP, Blaustein M, skipping through a nonevolutionary conserved intronic element. J Biol et al. Control of alternative pre-mRNA splicing by RNA Pol II elongation: Chem 2000 Jul 14;275(28):21041–7. faster is not always better. IUBMB Life 2003 Apr–May;55(4–5):235–41.