Báo cáo khoa học: Alternative splicing: role of pseudoexons in human disease and potential therapeutic strategies pot

15 467 0
Báo cáo khoa học: Alternative splicing: role of pseudoexons in human disease and potential therapeutic strategies pot

Đang tải... (xem toàn văn)

Tài liệu hạn chế xem trước, để xem đầy đủ mời bạn chọn Tải xuống

Thông tin tài liệu

MINIREVIEW Alternative splicing: role of pseudoexons in human disease and potential therapeutic strategies Ashish Dhir and Emanuele Buratti International Centre for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy Keywords alternative splicing; antisense oligonucleotides; mRNA; pseudoexons; splicing therapy Correspondence E Buratti, Padriciano 99, 34012 Trieste, Italy Fax: +39 040 226555 Tel: +39 040 3757316 E-mail: buratti@icgeb.org (Received 26 August 2009, revised 15 October 2009, accepted November 2009) doi:10.1111/j.1742-4658.2009.07520.x What makes a nucleotide sequence an exon (or an intron) is a question that still lacks a satisfactory answer Indeed, most eukaryotic genes are full of sequences that look like perfect exons, but which are nonetheless ignored by the splicing machinery (hence the name ‘pseudoexons’) The existence of these pseudoexons has been known since the earliest days of splicing research, but until recently the tendency has been to view them as an interesting, but rather rare, curiosity In recent years, however, the importance of pseudoexons in regulating splicing processes has been steadily revalued Even more importantly, clinically oriented screening studies that search for splicing mutations are beginning to uncover a situation where aberrant pseudoexon inclusion as a cause of human disease is more frequent than previously thought Here we aim to provide a review of the mechanisms that lead to pseudoexon activation in human genes and how the various cis- and trans-acting cellular factors regulate their inclusion Moreover, we list the potential therapeutic approaches that are being tested with the aim of inhibiting their inclusion in the final mRNA molecules Introduction Towards the end of the 1970s, in the beginning of pre-mRNA splicing research [1,2], defining exons and introns was essentially based on observing the final composition of the mature mRNA molecule In 1978, any sequence that was included in a mature mRNA became tagged as an ‘exon’, whereas all the intervening genomic sequences that were left out during the splicing process became defined as ‘introns’ [3] However, this way of thinking did not explain what makes an exon an exon or an intron an intron The discovery of the basic splice site consensus sequences during the same years [4,5], and later on of enhancer and repressor elements, has taken us a long way in the direction of discovering exon- and intron-definition complexes [6–8] Nowadays, the splicing signals that define exons ⁄ introns have been greatly aided by basic research, bioinformatic approaches and advanced sequencing tools [9,10] In this regard, we certainly know much more about splicing regulation than we did 20 years ago Considering that several reviews have been written recently on the subject, the reader is referred to them for further information on the latest discoveries [11–14] Most important, in this respect, have been the initial observations that in alternative splicing processes the same nucleotide sequence could be defined by the spliceosome as an intron or an exon in response to specific signals [15,16] It is now clear that these kinds of decision (What is an exon? What is an intron?) are of paramount importance in explaining genome complexity and evolutionary pathways [17–20] However, the sum of this new knowledge does not necessarily mean that we are near the goal of Abbreviations 3¢ss, 3¢ splice site; 5¢ss, 5¢ splice site; AON, antisense oligonucleotide; LINE, long interspersed elements; NMD, nonsense-mediated decay; PTB, polypyrimidine tract binding protein; SINE, short interspersed elements FEBS Journal 277 (2010) 841–855 ª 2010 ICGEB Trieste (Italy) Journal compilation ª 2010 FEBS 841 Pseudoexons in human disease A Dhir and E Buratti understanding most splicing decisions Indeed, even the latest attempts at ‘designing’ exons based on current state-of-the-art knowledge have basically demonstrated that there is still a long way to go before we can become as good as the spliceosome in deciding what is an exon and what is an intron [21] Where pseudoexon sequences come into the story? Central to the issue of deciding what is an exon and what is an intron is the question of their origin, a very much debated field to this day that basically deals with deciding the order of appearance of introns during evolution, whether first, early or late [22] Whatever the answer to this question will turn out to be, it is now clear that many of the ‘new’ exons in our genome originate from the insertion of transposable sequence elements belonging to the SINE and LINE classes in the eukaryotic genome [23–25] In particular, exonization of Alu elements (which are primate specific and represent the most abundant mobile elements in the human genome) through retrotranposition–mutation events is a prominent source of new exons in the eukaryotic transcriptome, as schematically depicted in Fig [26,27] However, even if we ignore this particular class of exonization event, every in silico analysis shows that ‘false exons’ are very abundant in the intronic sequences of most genes [with this term we refer to any nucleotide sequence between 50 and 200–300 nucleotides in length with apparently viable 5¢ and 3¢ splice sites (5¢ss and 3¢ss) at either end] Presently, there is evidence that inclusion of many of these sequences is actively inhibited due to the presence of intrinsic defects [28], the presence of silencer elements [29–31] or the formation of inhibiting RNA secondary structures [32] Even if a combination of all these elements succeeds in repressing the use of many of these pseudoexon sequences, we have to consider the possibility that there must be many exceptions to this rule First, it is probable that several of these pseudoexons may actually be recognized only in particular circumstances, such as a consequence of particular external stimuli [33,34] or present in a given tissue or developmental stage Proof of this possibility is the observation that ‘novel’ exons keep being identified even in well-known and studied genes, such as the DMD gene [35] Second, our failure to observe their use in normal conditions may also be due to the fact that their inclusion can intentionally lead to premature insertion of a termination codon in the mature mRNA and the consequent rapid degradation by nonsense-mediated decay (NMD) pathways [36] (Fig 1) Such an occurrence has been described in the rat a-tropomyosin gene with a putative pseudoexon sequence localized downstream of two mutually exclusive exons: an upstream exon that is included only in smooth muscle tissue and a downstream exon that is included in most cell types [37] Fig The left panel shows a schematic model of Alu element exonization The element (Alu) is inserted by retrotransposition and during the course of evolution mutations within this sequence create viable splicing sequences The middle panel shows the effect of the inclusion of a nonsense exon sequence (NE) in a transcript When this nonsense exon sequence is included, the resulting transcript is degraded by NMD (lower diagram) The right panel shows the classical pathway of pseudoexon (PE) inclusion in human disease In this case, a nucleotide sequence on the brink of becoming an exon becomes activated following a number of different mutational events 842 FEBS Journal 277 (2010) 841–855 ª 2010 ICGEB Trieste (Italy) Journal compilation ª 2010 FEBS A Dhir and E Buratti Experimental analysis has shown that, when this pseudoexon is included in the mRNA molecule together with the ubiquitously expressed downstream exon, the formation of a stop codon causes activation of the NMD pathway On the other hand, when inclusion of this pseudoexon occurs with the upstream smooth muscle tissue-specific exon, then it can still be removed through a resplicing pathway (and a normally processed mRNA molecule can be generated) For this reason, the term ‘nonsense’ exon is now preferred to define these kinds of sequence, which according to bioinformatic analyses may be more prevalent in human genes than previously thought [37] Nonetheless, from a human disease point of view, many pseudoexon intronic sequences seem poised on the brink of becoming exons (Fig 1) and a comprehensive list of more than 60 published pathological pseudoexon events is presented in Table Although briefly reviewed previously elsewhere [38], the recent advances in pseudoexon research warrant a second look at several pseudoexon-related issues, especially with regards to novel therapeutic approaches Cis-acting sequences in pseudoexon inclusion As previously mentioned, most pathological pseudoexon inclusion events originate from the creation of new splicing donor or acceptor splice sites within an intronic sequence, followed by the subsequent selection of weaker ‘opportunistic’ acceptor or donor site sequences (Fig 2A) A preliminary analysis of the strength of donor sites activated in pseudoexon inclusion events has highlighted their relatively high strength (according to in silico prediction programs) with respect to normally processed exons and to cryptic donor sites activated following normal donor site inactivation [39] In a slightly lower number of cases, pseudoexon activation has been observed following the creation of de novo acceptor sites (Table 1), whereas branch-point creation still represents a minority (probably owing to the fact that a new branch point needs to find both a viable acceptor and donor site nearby, rather than just one of them) In addition to de novo creation of strong donor, acceptor and branch site sequences, the other most frequent mechanisms that may lead to pseudoexon activation involves the creation ⁄ deletion of splicing regulatory sequences that will be discussed more in detail below (Fig 2B) Finally, in two individual cases, the rearrangement of genomic regions through gross deletions (Fig 2C) [40] or genomic inversions (Fig 2D) [41] has also been described to give rise to Pseudoexons in human disease pseudoexon inclusion events This has come about either by bringing together viable splice sites that would normally be too far away from each other on the gene sequence or by activating exons in what would normally have been the antisense genomic strand In a few genes, a particularly interesting method of pseudoexon activation event has also occurred following the inactivation of naturally occurring up stream 5¢ss (FAA, IDS, MUT) [42–45] or downstream 3¢ss (BRCA2, CFTR) [46,47] (Fig 2E) These findings suggest that the processivity of these mRNA transcripts probably represents an element capable of determining pseudoexon repression apart from being capable of influencing normal splicing levels [48] On a more general note, a still underappreciated aspect of pseudoexon recognition that concerns the effect of cis-acting sequences is represented by the potential influence of RNA secondary structure on splicing efficiency [49] Recently, it has been shown that donor site usage in the inclusion of two pseudoexon sequences in the ATM and CFTR genes is strongly dependent on their availability in the single-stranded region [50] Interestingly, the same conclusion was reached in a recent study by Schwartz et al [51] analysing the differences between exonized and nonexonized Alu elements In this work, it was found that one of the major discriminating factors between these two classes of Alu elements was represented by the potential availability of 5¢ss sequences in an unstructured conformation Trans-acting factors in pseudoexon inclusion Not many studies have focused on identifying the role played by trans-acting factors in pseudoexon inclusion However, because of its significance, this is an area of research that would probably benefit from increased attention by researchers in the future In the case of nonpathologically related pseudoexons carrying nonsense codons, the presence of splicing regulatory elements may well provide a clue with regards to the possible roles played by these sequences For example, in the case of the previously described tropomyosin pseudoexon [37], the specific binding of hnRNP H ⁄ F proteins has been described as a potential key modifier of this pseudoexon inclusion event [52] The fact that these proteins are particularly downregulated in cardiomyocytes may explain the cell-specific repression of the downstream ‘normal’ exon that is otherwise present in all cell types (Fig 3A) FEBS Journal 277 (2010) 841–855 ª 2010 ICGEB Trieste (Italy) Journal compilation ª 2010 FEBS 843 Pseudoexons in human disease A Dhir and E Buratti Table Pathological pseudoexon inclusion events in human disease NA, not available; SRE, splicing regulatory element Gene Size (bp) a-Gal A ATM ATM b-globin b-globin b-globin BRCA1 BRCA2 57 65 137 165 126 73 66 93 CD40L CEP290 CFTR CFTR CFTR CFTR 59 128 49 84 101 184 CFTR CHM COL4A3a COL4A5 COL4A5 CTDP1 CYBB CYBB DHPR ⁄ QDPR DMD 214 98 74 30 147 95 56 61 152 DMD 108 DMD 125 DMD 149 DMD 160 DMD 180 DMD DMD DMD DMD DMD DMD DMD DMD DMD FBN1 FGB FGG FVIII GALC GHER GHR GUSBa 58 67 89 90 95 147 149 172 ⁄ 202 46 ⁄ 132 93 50 75 191 34 69 102 68 844 98 Activating mutation Reference DBASS3 ⁄ DBASS5 reference SRE creation SRE deletion 5¢ss creation 5¢ss creation 5¢ss creation 5¢ss creation 3¢ss creation Downstream 3¢ss deletion 5¢ss creation 5¢ss creation 5¢ss creation 5¢ss creation SRE creation Downstream 3¢ss deletion 5¢ss creation 3¢ss creation 3¢ss creation 3¢ss creation SRE creation 5¢ss creation 5¢ss creation 5¢ss creation 5¢ss creation [78] [56] [79] [80] [81] [82] [83] [46] http: ⁄ ⁄ www.som.soton.ac.uk ⁄ research ⁄ geneticsdiv ⁄ dbass5 ⁄ viewsplicesite.aspx?id=317 http: ⁄ ⁄ www.som.soton.ac.uk ⁄ research ⁄ geneticsdiv ⁄ dbass5 ⁄ viewsplicesite.aspx?id=324 http://www.som.soton.ac.uk/research/geneticsdiv/dbass5/viewsplicesite.aspx?id=331 http://www.som.soton.ac.uk/research/geneticsdiv/dbass5/viewsplicesite.aspx?id=323 http://www.som.soton.ac.uk/research/geneticsdiv/dbass5/viewsplicesite.aspx?id=336 http://www.som.soton.ac.uk/research/geneticsdiv/dbass5/viewsplicesite.aspx?id=335 NA NA [84] [85] [86] [87] [88] [47] http://www.som.soton.ac.uk/research/geneticsdiv/dbass5/viewsplicesite.aspx?id=437 http://www.som.soton.ac.uk/research/geneticsdiv/dbass5/viewsplicesite.aspx?id=342 http://www.som.soton.ac.uk/research/geneticsdiv/dbass5/viewsplicesite.aspx?id=330 http://www.som.soton.ac.uk/research/geneticsdiv/dbass5/viewsplicesite.aspx?id=328 NA http://www.som.soton.ac.uk/research/geneticsdiv/dbass3/view.asp?item=splice&id=31 [89] [90] [91] [92] [92] [93] [94] [95] [96] http://www.som.soton.ac.uk/research/geneticsdiv/dbass5/viewsplicesite.aspx?id=322 NA NA http://www.som.soton.ac.uk/research/geneticsdiv/dbass3/view.asp?item=splice&id=240 NA http://www.som.soton.ac.uk/research/geneticsdiv/dbass5/viewsplicesite.aspx?id=333 NA http://www.som.soton.ac.uk/research/geneticsdiv/dbass5/viewsplicesite.aspx?id=306 http://www.som.soton.ac.uk/research/geneticsdiv/dbass5/viewsplicesite.aspx?id=334 28 kb gene inversion 28 kb gene inversion 28 kb gene inversion 28 kb gene inversion 28 kb gene inversion 28 kb gene inversion 5¢ss creation 5¢ss creation 5¢ss creation 5¢ss creation 3¢ss creation 5¢ss creation 3¢ss creation 5¢ss creation 3¢ss creation 5¢ss creation SRE creation 5¢ss creation 5¢ss creation ND 5¢ss creation SRE deletion 5¢ss creation [41] NA [41] NA [41] NA [41] NA [41] NA [41] NA [97] [98] [98] [98] [97] [99] [98] [100] [101] [102] [63] [103] [104] [105] [106] [57,107] [108] http://www.som.soton.ac.uk/research/geneticsdiv/dbass5/viewsplicesite.aspx?id=340 NA http://www.som.soton.ac.uk/research/geneticsdiv/dbass5/viewsplicesite.aspx?id=338 http://www.som.soton.ac.uk/research/geneticsdiv/dbass5/viewsplicesite.aspx?id=339 http://www.som.soton.ac.uk/research/geneticsdiv/dbass3/view.asp?item=splice&id=275 http://www.som.soton.ac.uk/research/geneticsdiv/dbass5/viewsplicesite.aspx?id=326 http://www.som.soton.ac.uk/research/geneticsdiv/dbass3/view.asp?item=splice&id=274 http://www.som.soton.ac.uk/research/geneticsdiv/dbass5/viewsplicesite.aspx?id=320 http://www.som.soton.ac.uk/research/geneticsdiv/dbass3/view.asp?item=splice&id=273 NA NA NA http://www.som.soton.ac.uk/research/geneticsdiv/dbass5/viewsplicesite.aspx?id=332 http://www.som.soton.ac.uk/research/geneticsdiv/dbass5/viewsplicesite.aspx?id=344 NA http://www.som.soton.ac.uk/research/geneticsdiv/dbass5/viewsplicesite.aspx?id=316 http://www.som.soton.ac.uk/research/geneticsdiv/dbass5/viewsplicesite.aspx?id=311 FEBS Journal 277 (2010) 841–855 ª 2010 ICGEB Trieste (Italy) Journal compilation ª 2010 FEBS A Dhir and E Buratti Pseudoexons in human disease Table (Continued.) Gene Activating Size (bp) mutation Reference DBASS3 ⁄ DBASS5 reference HADHB HSPG2 IDS IDS 56 ⁄ 106 130 78 103 [109] [110] [111] [42,43] NA http://www.som.soton.ac.uk/research/geneticsdiv/dbass5/viewsplicesite.aspx?id=346 http://www.som.soton.ac.uk/research/geneticsdiv/dbass5/viewsplicesite.aspx?id=329 http://www.som.soton.ac.uk/research/geneticsdiv/dbass5/viewsplicesite.aspx?id=282 INI1 ⁄ SNF5 ISCU JK 72 5¢ss creation 5¢ss creation 5¢ss creation Upstream 5¢ss deletion 5¢ss creation [112] http://www.som.soton.ac.uk/research/geneticsdiv/dbass5/viewsplicesite.aspx?id=410 [113–115] [40] NA http://www.som.soton.ac.uk/research/geneticsdiv/dbass5/viewsplicesite.aspx?id=341 MCBB MYO6 MUT 64 108 76 [116] [117] [45,68] NA NA http://www.som.soton.ac.uk/research/geneticsdiv/dbass5/viewsplicesite.aspx?id=434 http://www.som.soton.ac.uk/research/geneticsdiv/dbass5/viewsplicesite.aspx?id=394 NDUFS7 NF-1 NF-1 NF-1 NF-1 NF-1 NF-1 NF-1 NF-2 122 70 107 172 58 76 54 177 106 [118] [70] [70] [119] [120] [120] [121] [70,122,123] [124] http://www.som.soton.ac.uk/research/geneticsdiv/dbass5/viewsplicesite.aspx?id=357 NA NA http://www.som.soton.ac.uk/research/geneticsdiv/dbass3/view.asp?item=splice&id=277 NA NA http://www.som.soton.ac.uk/research/geneticsdiv/dbass5/viewsplicesite.aspx?id=411 http://www.som.soton.ac.uk/research/geneticsdiv/dbass5/viewsplicesite.aspx?id=318 NA NPC1 OA1 ⁄ GPR143 OATa OTC PCCA PCCB PHEX 194 165 3¢ss creation Internal kb deletion SRE deletion 5¢ss creation 5¢ss creation or upstream 5¢ss deletion 5¢ss creation 5¢ss creation 5¢ss creation 3¢ss creation 3¢ss creation 5¢ss creation 5¢ss creation 5¢ss creation Branch-point creation 5¢ss creation 3¢ss creation [125] [69] NA http://www.som.soton.ac.uk/research/geneticsdiv/dbass3/view.asp?item=splice&id=114 5¢ss creation 3¢ss creation SRE creation 5¢ss creation 5¢ss creation [126] [127] [68] [68] [128] http://www.som.soton.ac.uk/research/geneticsdiv/dbass5/viewsplicesite.aspx?id=433 NA NA http://www.som.soton.ac.uk/research/geneticsdiv/dbass5/viewsplicesite.aspx?id=436 http://www.som.soton.ac.uk/research/geneticsdiv/dbass5/viewsplicesite.aspx?id=321 5¢ss creation 3¢ss creation 5¢ss creation 5¢ss creation Branch-point optimization Py-tract optimization 3¢ss creation 5¢ss creation 5¢ss creation 5¢ss creation [129] [130] [130,131] [132] [133] NA NA NA NA NA [133] NA [134] [135] [136] [137] NA http://www.som.soton.ac.uk/research/geneticsdiv/dbass5/viewsplicesite.aspx?id=337 NA http://www.som.soton.ac.uk/research/geneticsdiv/dbass5/viewsplicesite.aspx?id=307 86 ⁄ 100 136 PKHD1 PMM2 PMM2 PRPF31 PTS a 142 135 84 72 50 ⁄ 100 ⁄ 170 116 66 123 175 45 PTS b 79 RB1 RYR1 SOD-1 TSC2 103 119 43 89 a Alu-derived pseudoexons b LINE-2-derived pseudoexons Interestingly, repression of the tropomyosin nonsense exon was also observed following PTB overexpression PTB is a well-known and powerful splicing modifier that plays a major role in alternative splicing regulation [8,53] Recently, this protein has been reported to also downregulate the inclusion efficiency of a pathological pseudoexon in NF-1 intron 31 independently of the activating mutation that creates a very strong splicing acceptor site [54] (Fig 3B) This finding suggests that silencer binding sites may be FEBS Journal 277 (2010) 841–855 ª 2010 ICGEB Trieste (Italy) Journal compilation ª 2010 FEBS 845 Pseudoexons in human disease A Dhir and E Buratti A B C D E Fig The mutational events that determine pathological pseudoexon inclusion The most frequent is represented by the creation of de novo functional splice sites or branch-point elements through a single or few point mutation (A) Other mechanisms include the creation or deletion of splicing regulatory elements (B), genomic rearrangements (C, D) and inactivation of upstream or downstream splice sites (E) actively used by evolutionary mechanisms to decrease the probability that random activating mutations may determine the constitutive inclusion of pseudoexon sequences In this respect, one interesting molecular complex is U1snRNP, a ribonucleoprotein complex normally associated with 5¢ss recognition in the normal splicing process [55] First, U1snRNP binding to an intronic splicing processing element has been found to inhibit pathological pseudoexon inclusion in intron 20 of the ATM gene (Fig 3C) Inactivation of this element through a four nucleotide deletion causes pseudoexon inclusion and occurrence of ataxia telangiectasia in a patient [56] In a second case, binding of hnRNP E1 and U1snRNP to a weak 5¢ss efficiently silences pseudoexon inclusion in the GHR gene [57], preventing the development of Laron syndrome (Fig 3D) Finally, it should also be noted that in a variety of pseudoexon inclusion events, the activating mutations potentially created new splicing enhancer sequences (Table 1) Although in very few of these cases was trans-acting factors binding to these elements identified, in silico and experimental analyses have shown that several of the newly created enhancer sequences strongly correlate with potential binding to the SR protein class of splicing regulators 846 Therapeutic strategies aimed at correcting pseudoexon inclusion in genetic diseases Therapeutic strategies based on antisense oligonucleotide (AON) chemistry, which uses base pairing to target specific sequences in RNAs, have been extensively employed to correct splicing disorders in human genes [58,59] Interestingly, apart from these therapeutic applications, short nuclear RNAs may also play a similar functional role to physiologically regulate exon inclusion, such as the case of snoRNA HBII-52 in the regulation of exon Vb inclusion in the serotonin receptor 2C [60] AONs are thought to modulate the splicing pattern by steric hindrance of the recruitment of the splicing factors to the targeted splicing competent cis-elements, thus forcing the machinery to use the natural sites Dominski and Kole [61] were the first to pioneer the antisense-mediated modulation of premRNA splicing In the earliest examples, AONs were aimed at activated cryptic splice sites in the b-globin and CFTR genes in order to restore normal splicing in b-thalassaemia and cystic fibrosis patients [61,62] Currently, however, AON strategies have been used successfully to restore normal splicing in several disease models FEBS Journal 277 (2010) 841–855 ª 2010 ICGEB Trieste (Italy) Journal compilation ª 2010 FEBS A Dhir and E Buratti Pseudoexons in human disease A B C D Fig A schematic diagram of the tropomyosin gene with exons and 3, which are mutually exclusive (exon is the predominant form in most cell types), and the nonsense exon (NE), which causes transcript degradation following its joining to exon (but not exon 2) The levels of hnRNP H ⁄ F proteins can regulate the extent of NE inclusion (B) shows that in the NF-1 intron, 30 pseudoexon inclusion levels are regulated by silencer elements in UCUU-rich motifs that bind the PTB (hnRNP I) splicing regulator In the ATM gene, a four nucleotide deletion (GUAA) in the intronic region between exons 20 and 21 causes the insertion of a 65 nucleotide long pseudoexon (C) Functional analysis has demonstrated that this deletion abolished binding of an U1snRNP molecule in this position and activated a 3¢ss lying 12 nucleotides upstream of this element In the last case, binding of hnRNP E1 and U1snRNP to a silencer motif near a weak 5¢ss efficiently silences pseudoexon inclusion in the GHR gene, preventing the development of Laron syndrome (D) Afibrinogenemia is caused by genetic abnormalities within any of the three genes that encode the fibrinogen molecule: FGA, FGB, FGG Recently, Davis et al [63] showed that a homozygous c.115–600A>G point mutation located deep within intron of FGB causes pseudoexon inclusion In this study, pseudoexon inclusion was corrected by targeting this mutation with an antisense phosphorodiamidate morpholino oligonucleotide In several forms of b-thalassaemia, two single nucleotide mutations (IVS2-705 and IVS2-654) in the b-globin gene have been reported to cause pathological pseudoexon insertion In 1993, Dominski and Kole [61] successfully tested 2¢-O-methylribose AONs to restore correct splicing Later, Sierakowska et al [64] also restored correct splicing and b-globin polypeptide production using a phosphorothioate 2¢-O-methyloligoribonucleotide targeted to the aberrant 3¢ss More recently, Gorman et al [65,66] engineered the U7 snRNA gene to correct pre-mRNA splicing by replacing the antihistone sequence with sequences targeting b-globin aberrant splice sites (Fig 4A) The congenital disorders of glycosylation are caused by defects in the PMM2 gene Recently, Vega et al [130] studied a c.640–15479C>T deep intronic mutation that creates a new aberrant 5¢ss in intron and caused pseudoexon activation Antisense morpholino oligonucleotides that targeted the aberrant 5¢ss and 3¢ss sites achieved 100% restoration of correctly spliced mRNA Pseudoexon-activating mutation 3849 + 10 kb C > T in intron 19 of the CFTR gene has been reported to frequently cause cystic fibrosis In their study, Friedman et al [62] reported that a cocktail of 2¢-O-methyl phosphorothioate oligoribonucleotides against different regions of this pseudoexon abolished pseudoexon inclusion and partially restored production of normal mRNA and CFTR processed protein (Fig 4B) Mutations in the DMD gene are known to cause Duchenne and Becker muscular dystrophies Recently, Gurvich et al [67] demonstrated that 2¢-O-methyl ribose phosphorothioate AONs restored normal splicing in primary myoblast cultures established from two individual patients carrying out-of-frame pseudoexon insertion mutations (Fig 4C) Methylmalonic acidaemia and propionic acidaemia are caused by different gene defects in the MUT, FEBS Journal 277 (2010) 841–855 ª 2010 ICGEB Trieste (Italy) Journal compilation ª 2010 FEBS 847 Pseudoexons in human disease A Dhir and E Buratti A B C Fig A schematic representation of three different 5¢ss activating mutations in various disease-causing genes that activate pseudoexon inclusion where therapeutic correction has been attempted with an antisense approach (A) represents the IVS2-705 T>G splicing mutation that activates a 126 nucleotide pseudoexon in intron of the b-globin gene In this case, 2¢-O-methyl ribose AONs and functionally modified U7 snRNA were employed to block the acceptor and donor splice sites In (B), the 3849+10kbC>T splicing mutation activates a 84 nucleotide pseudoexon in intron 19 of the CFTR gene Three 2¢-O-methyl phosphorothioate oligoribonucleotides were targeted against the splice sites and against the pseudoexonic premature stop codon sequence to rescue normal splicing Finally, (C) shows the c.6614+3310G>T splicing mutation that activates a 137 nucleotide pseudoexon in intron 45 of the DMD gene To restore normal splicing, 2¢-O-methyl ribose AONs were also targeted against the donor splice site and a predicted cluster of exonic splicing enhancer sequences within the pseudoexon PCCA and PCCB genes Ugarte et al [68] recently reported the identification of three novel deep intronic mutations in each of these genes that potentially lead to pseudoexon activation through diverse mechanisms Antisense therapeutics using antisense morpholino oligomers correctly restored almost complete normal splicing that was effectively translated Ocular albinism type involves mutations in the OA1 gene Vetrini et al [69] identified a deep intronic point mutation g.25288G>A that created a new acceptor splice site in intron of this gene and resulted in pseudoexon inclusion Treatment of a patient’s melanocytes with antisense morpholino AONs complementary to the mutant sequence rescued mRNA and protein expression levels Mutations in the NF-1 gene cause neurofibromatosis type Recently, Pros et al [70] identified six neurofibromatosis type patients carrying three different deep intronic mutations that create new 5¢ss leading to the activation of the pseudoexon in the mature mRNA In this study, antisense morpholino oligonucleotides were targeted against these newly created 5¢ss, effectively restoring normal NF-1 splicing All of these different therapeutic strategies are summarized in Table 848 Concluding remarks This review is part of a miniseries co-ordinated by Diana Baralle [71] to look at emerging topics in splicing research, such as the correct assessment of sequence variants as pathogenic mutations [72]; the development of novel splicing-based therapeutic agents to treat HIV-1 infections [73]; and new methods in the global analysis of alternative splicing profiles [74] We decided to examine the role of pseudoexons in recent research, as no specialized reviews have appeared in the past dealing with this particular kind of event From a basic science point of view, the possibility for researchers to look at the splicing process on a much more global scale than the single exon or the individual gene will clarify the issues examined in this review by helping to distinguish clearly between exons and pseudoexons [19,75,76] In turn, this will provide a better appreciation regarding how the splicing process has evolved to define ‘exons’, how it distinguishes them from similar potentially pathological sequences (pseudoexons) and what is the preferential way it has chosen to repress their recognition In this respect, pseudoexon research will also provide us with an unparalleled opportunity to understand evolutionary mechanisms FEBS Journal 277 (2010) 841–855 ª 2010 ICGEB Trieste (Italy) Journal compilation ª 2010 FEBS A Dhir and E Buratti Pseudoexons in human disease Table Therapeutic approaches PMO, phosphorodiamidate morpholino oligonucleotide Therapeutic approaches Gene and diseases caused by pseudoexon inclusion Mode of action Antisense PMOs FGB, afibrinogenaemia Engineered U7 snRNA HBB, b-thalassaemia Antisense morpholino oligonucleotides PMM2, congenital disorders of glycosylation; MUT, PCCA and PCCB, methylmalonic acidaemia and propionic acidemia; OA1, ocular albinism type 1; NF-1, neurofibromatosis type CFTR, cystic fibrosis; HBB, b-thalassaemia; DMD, Duchenne and Becker muscular dystrophies Antisense PMO specifically target the predicted ESE motif created by the mutation Use of modified U7 snRNA to target aberrant splice sites for long-term restoration of correct splicing Targeting aberrant splice sites activated due to mutations AONs block access of the splicing machinery to the pseudoexonic regions in the pre-mRNAs Targeting of antisense against aberrant splice sites, in-frame stop codon and predicted exonic splicing enhancers within pseudoexons Antisense 2¢-O-methyl ribose phosphorothioate oligonucleotides; 2¢-O- methylribo-oligonucleotides that cause some of these sequences to become exons and, of course, vice versa Considering that aberrant pseudoexon inclusion events are an increasing phenomenon linked with disease, just the simple characterization of these sequences may have some very practical consequences The studies reported in this review clearly highlight the feasibility of using AONs to correct these types of splicing defect (even in the absence of a complete or even partial understanding of the ‘basic science’ explaining their occurrence) From a therapeutic point of view, the major advantage of targeting pseudoexon inclusion events is provided by the supposition that AONs targeted against what would normally be intronic sequences would not be expected to remain bound to the mature mRNA (and thus interfere with later stages of RNA processing, such as export ⁄ translation) However, several factors will still need to be improved before human application becomes a reality These start from basic studies aimed at optimizing gene ⁄ exon specificity (that will necessarily have to be made on an individual gene-specific basis) to the development of appropriate carrier systems These systems will be absolutely necessary to achieve successful delivery, low toxicity and avoidance of undesired immune responses Furthermore, even after achieving all of these aims, there will still remain the need to optimize recurrent administration protocols (this is an often overlooked consideration, as none of these methods will cause a permanent correction of mRNA splicing defects), and determining their clearance ⁄ accumulation in human organs ⁄ tissues However, notwithstanding all of these difficulties, AON technology [59,77] has already entered the clinical trial stage for diseases such as Duchenne muscular dystrophy (http://www.clinicaltrials.gov) and this represents a bright hope for the not too distant future Acknowledgements This work was supported by Telethon Onlus Foundation (Italy) (grant no GGP06147) and by a European community grant (EURASNET-LSHG-CT-2005518238) We thank Professor F E Baralle for helpful discussion References Berget SM, Moore C & Sharp PA (1977) Spliced segments at the 5¢ terminus of adenovirus late mRNA Proc Natl Acad Sci USA 74, 3171–3175 Chow LT, Gelinas RE, Broker TR & Roberts RJ (1977) An amazing sequence arrangement at the 5¢ ends of adenovirus messenger RNA Cell 12, 1–8 Gilbert W (1978) Why genes in pieces? Nature 271, 501 Breathnach R, Benoist C, O’Hare K, Gannon F & Chambon P (1978) Ovalbumin gene: evidence for a leader sequence in mRNA and DNA sequences at the exon-intron boundaries Proc Natl Acad Sci USA 75, 4853–4857 Catterall JF, O’Malley BW, Robertson MA, Staden R, Tanaka Y & Brownlee GG (1978) Nucleotide sequence homology at 12 intron–exon junctions in the chick ovalbumin gene Nature 275, 510–513 Black DL (2003) Mechanisms of alternative pre-messenger RNA splicing Annu Rev Biochem 72, 291–336 Berget SM (1995) Exon recognition in vertebrate splicing J Biol Chem 270, 2411–2414 Sharma S, Kohlstaedt LA, Damianov A, Rio DC & Black DL (2008) Polypyrimidine tract binding protein controls the transition from exon definition to an intron defined spliceosome Nat Struct Mol Biol 15, 183–191 Zhang MQ (1998) Statistical features of human exons and their flanking regions Hum Mol Genet 7, 919–932 FEBS Journal 277 (2010) 841–855 ª 2010 ICGEB Trieste (Italy) Journal compilation ª 2010 FEBS 849 Pseudoexons in human disease A Dhir and E Buratti 10 Chasin LA (2007) Searching for splicing motifs Adv Exp Med Biol 623, 85–106 11 Wang Z & Burge CB (2008) Splicing regulation: from a parts list of regulatory elements to an integrated splicing code RNA 14, 802–813 12 Hertel KJ (2008) Combinatorial control of exon recognition J Biol Chem 283, 1211–1215 13 Cartegni L, Chew SL & Krainer AR (2002) Listening to silence and understanding nonsense: exonic mutations that affect splicing Nat Rev Genet 3, 285–298 14 Sperling J, Azubel M & Sperling R (2008) Structure and function of the pre-mRNA splicing machine Structure 16, 1605–1615 15 Amara SG, Jonas V, Rosenfeld MG, Ong ES & Evans RM (1982) Alternative RNA processing in calcitonin gene expression generates mRNAs encoding different polypeptide products Nature 298, 240–244 16 Kornblihtt AR, Umezawa K, Vibe-Pedersen K & Baralle FE (1985) Primary structure of human fibronectin: differential splicing may generate at least 10 polypeptides from a single gene EMBO J 4, 1755–1759 17 Stamm S, Ben-Ari S, Rafalska I, Tang Y, Zhang Z, Toiber D, Thanaraj TA & Soreq H (2005) Function of alternative splicing Gene 344, 1–20 18 Xing Y & Lee C (2006) Alternative splicing and RNA selection pressure–evolutionary consequences for eukaryotic genomes Nat Rev Genet 7, 499–509 19 Blencowe BJ (2006) Alternative splicing: new insights from global analyses Cell 126, 37–47 20 Tarrio R, Ayala FJ & Rodriguez-Trelles F (2008) Alternative splicing: a missing piece in the puzzle of intron gain Proc Natl Acad Sci USA 105, 7223– 7228 21 Zhang XH, Arias MA, Ke S & Chasin LA (2009) Splicing of designer exons reveals unexpected complexity in pre-mRNA splicing RNA 15, 367–376 22 Rodriguez-Trelles F, Tarrio R & Ayala FJ (2006) Origins and evolution of spliceosomal introns Annu Rev Genet 40, 47–76 23 Sorek R (2007) The birth of new exons: mechanisms and evolutionary consequences RNA 13, 1603–1608 24 Sela N, Mersch B, Gal-Mark N, Lev-Maor G, Hotz-Wagenblatt A & Ast G (2007) Comparative analysis of transposed element insertion within human and mouse genomes reveals Alu’s unique role in shaping the human transcriptome Genome Biol 8, R127 25 Meili D, Kralovicova J, Zagalak J, Bonafe L, Fiori L, Blau N, Thony B & Vorechovsky I (2009) Disease-causing mutations improving the branch site and polypyrimidine tract: pseudoexon activation of LINE-2 and antisense Alu lacking the poly(T)-tail Hum Mutat 30, 823–831 26 Gal-Mark N, Schwartz S & Ast G (2008) Alternative splicing of Alu exons–two arms are better than one Nucleic Acids Res 36, 2012–2023 850 27 Lev-Maor G, Ram O, Kim E, Sela N, Goren A, Levanon EY & Ast G (2008) Intronic Alus influence alternative splicing PLoS Genet 4, e1000204 28 Sun H & Chasin LA (2000) Multiple splicing defects in an intronic false exon Mol Cell Biol 20, 6414–6425 29 Sironi M, Menozzi G, Riva L, Cagliani R, Comi GP, Bresolin N, Giorda R & Pozzoli U (2004) Silencer elements as possible inhibitors of pseudoexon splicing Nucleic Acids Res 32, 1783–1791 30 Zhang XH & Chasin LA (2004) Computational definition of sequence motifs governing constitutive exon splicing Genes Dev 18, 1241–1250 31 Fairbrother WG & Chasin LA (2000) Human genomic sequences that inhibit splicing Mol Cell Biol 20, 6816–6825 32 Zhang XH, Leslie CS & Chasin LA (2005) Dichotomous splicing signals in exon flanks Genome Res 15, 768–779 33 Blaustein M, Pelisch F & Srebrow A (2007) Signals, pathways and splicing regulation Int J Biochem Cell Biol 39, 2031–2048 34 Biamonti G & Caceres JF (2009) Cellular stress and RNA splicing Trends Biochem Sci 34, 146–153 35 Tran VK, Zhang Z, Yagi M, Nishiyama A, Habara Y, Takeshima Y & Matsuo M (2005) A novel cryptic exon identified in the 3¢ region of intron of the human dystrophin gene J Hum Genet 50, 425–433 36 Maquat LE (2005) Nonsense-mediated mRNA decay in mammals J Cell Sci 118, 1773–1776 37 Grellscheid SN & Smith CW (2006) An apparent pseudo-exon acts both as an alternative exon that leads to nonsense-mediated decay and as a zero-length exon Mol Cell Biol 26, 2237–2246 38 Buratti E, Baralle M & Baralle FE (2006) Defective splicing, disease and therapy: searching for master checkpoints in exon definition Nucleic Acids Res 34, 3494–3510 39 Buratti E, Chivers M, Kralovicova J, Romano M, Baralle M, Krainer AR & Vorechovsky I (2007) Aberrant 5¢ splice sites in human disease genes: mutation pattern, nucleotide structure and comparison of computational tools that predict their utilization Nucleic Acids Res 35, 4250–4263 40 Lucien N, Chiaroni J, Cartron JP & Bailly P (2002) Partial deletion in the JK locus causing a Jk(null) phenotype Blood 99, 1079–1081 41 Madden HR, Fletcher S, Davis MR & Wilton SD (2009) Characterization of a complex Duchenne muscular dystrophy-causing dystrophin gene inversion and restoration of the reading frame by induced exon skipping Hum Mutat 30, 22–28 42 Lualdi S, Pittis MG, Regis S, Parini R, Allegri AE, Furlan F, Bembi B & Filocamo M (2006) Multiple cryptic splice sites can be activated by IDS point mutations generating misspliced transcripts J Mol Med 84, 692–700 FEBS Journal 277 (2010) 841–855 ª 2010 ICGEB Trieste (Italy) Journal compilation ª 2010 FEBS A Dhir and E Buratti 43 Alves S, Mangas M, Prata MJ, Ribeiro G, Lopes L, Ribeiro H, Pinto-Basto J, Lima MR & Lacerda L (2006) Molecular characterization of Portuguese patients with mucopolysaccharidosis type II shows evidence that the IDS gene is prone to splicing mutations J Inherit Metab Dis 29, 743–754 44 Savino M, Borriello A, D’Apolito M, Criscuolo M, Del Vecchio M, Bianco AM, Di Perna M, Calzone R, Nobili B, Zatterale A, et al (2003) Spectrum of FANCA mutations in Italian Fanconi anemia patients: identification of six novel alleles and phenotypic characterization of the S858R variant Hum Mutat 22, 338–339 45 Martinez MA, Rincon A, Desviat LR, Merinero B, Ugarte M & Perez B (2005) Genetic analysis of three genes causing isolated methylmalonic acidemia: identification of 21 novel allelic variants Mol Genet Metab 84, 317–325 46 Chen X, Truong TT, Weaver J, Bove BA, Cattie K, Armstrong BA, Daly MB & Godwin AK (2006) Intronic alterations in BRCA1 and BRCA2: effect on mRNA splicing fidelity and expression Hum Mutat 27, 427–435 47 Will K, Dork T, Stuhrmann M, Meitinger T, BerteleHarms R, Tummler B & Schmidtke J (1994) A novel exon in the cystic fibrosis transmembrane conductance regulator gene activated by the nonsense mutation E92X in airway epithelial cells of patients with cystic fibrosis J Clin Invest 93, 1852–1859 48 Kornblihtt AR (2006) Chromatin, transcript elongation and alternative splicing Nat Struct Mol Biol 13, 5–7 49 Buratti E & Baralle FE (2004) Influence of RNA secondary structure on the pre-mRNA splicing process Mol Cell Biol 24, 10505–10514 50 Buratti E, Dhir A, Lewandowska MA & Baralle FE (2007) RNA structure is a key regulatory element in pathological ATM and CFTR pseudoexon inclusion events Nucleic Acids Res 35, 4369–4383 51 Schwartz S, Gal-Mark N, Kfir N, Oren R, Kim E & Ast G (2009) Alu exonization events reveal features required for precise recognition of exons by the splicing machinery PLoS Comput Biol 5, e1000300 52 Coles JL, Hallegger M & Smith CW (2009) A nonsense exon in the Tpm1 gene is silenced by hnRNP H and F RNA 15, 33–43 53 Spellman R & Smith CW (2006) Novel modes of splicing repression by PTB Trends Biochem Sci 31, 73–76 54 Raponi M, Buratti E, Llorian M, Stuani C, Smith CW & Baralle D (2008) Polypyrimidine tract binding protein regulates alternative splicing of an aberrant pseudoexon in NF1 FEBS J 275, 6101–6108 55 Mount SM, Pettersson I, Hinterberger M, Karmas A & Steitz JA (1983) The U1 small nuclear RNA-protein complex selectively binds a 5¢ splice site in vitro Cell 33, 509–518 Pseudoexons in human disease 56 Pagani F, Buratti E, Stuani C, Bendix R, Dork T & Baralle FE (2002) A new type of mutation causes a splicing defect in ATM Nat Genet 30, 426–429 57 Akker SA, Misra S, Aslam S, Morgan EL, Smith PJ, Khoo B & Chew SL (2007) Pre-spliceosomal binding of U1 small nuclear ribonucleoprotein (RNP) and heterogenous nuclear RNP E1 is associated with suppression of a growth hormone receptor pseudoexon Mol Endocrinol 21, 2529–2540 58 Crooke ST (2004) Antisense strategies Curr Mol Med 4, 465–487 59 Aartsma-Rus A & van Ommen GJ (2007) Antisensemediated exon skipping: a versatile tool with therapeutic and research applications RNA 13, 1609–1624 60 Kishore S & Stamm S (2006) The snoRNA HBII-52 regulates alternative splicing of the serotonin receptor 2C Science 311, 230–232 61 Dominski Z & Kole R (1993) Restoration of correct splicing in thalassemic pre-mRNA by antisense oligonucleotides Proc Natl Acad Sci USA 90, 8673–8677 62 Friedman KJ, Kole J, Cohn JA, Knowles MR, Silverman LM & Kole R (1999) Correction of aberrant splicing of the cystic fibrosis transmembrane conductance regulator (CFTR) gene by antisense oligonucleotides J Biol Chem 274, 36193–36199 63 Davis RL, Homer VM, George PM & Brennan SO (2009) A deep intronic mutation in FGB creates a consensus exonic splicing enhancer motif that results in afibrinogenemia caused by aberrant mRNA splicing, which can be corrected in vitro with antisense oligonucleotide treatment Hum Mutat 30, 221–227 64 Sierakowska H, Sambade MJ, Agrawal S & Kole R (1996) Repair of thalassemic human beta-globin mRNA in mammalian cells by antisense oligonucleotides Proc Natl Acad Sci USA 93, 12840–12844 65 Gorman L, Suter D, Emerick V, Schumperli D & Kole R (1998) Stable alteration of pre-mRNA splicing patterns by modified U7 small nuclear RNAs Proc Natl Acad Sci USA 95, 4929–4934 66 Gorman L, Mercatante DR & Kole R (2000) Restoration of correct splicing of thalassemic beta-globin pre-mRNA by modified U1 snRNAs J Biol Chem 275, 35914–35919 67 Gurvich OL, Tuohy TM, Howard MT, Finkel RS, Medne L, Anderson CB, Weiss RB, Wilton SD & Flanigan KM (2008) DMD pseudoexon mutations: splicing efficiency, phenotype, and potential therapy Ann Neurol 63, 81–89 68 Ugarte M, Aguado C, Desviat LR, Sanchez-Alcudia R, Rincon A & Perez B (2007) Propionic and methylmalonic acidemia: antisense therapeutics for intronic variations causing aberrantly spliced messenger RNA Am J Hum Genet 81, 69 Vetrini F, Tammaro R, Bondanza S, Surace EM, Auricchio A, De Luca M, Ballabio A & Marigo V FEBS Journal 277 (2010) 841–855 ª 2010 ICGEB Trieste (Italy) Journal compilation ª 2010 FEBS 851 Pseudoexons in human disease 70 71 72 73 74 75 76 77 78 79 80 81 82 83 852 A Dhir and E Buratti (2006) Aberrant splicing in the ocular albinism type gene (OA1 ⁄ GPR143) is corrected in vitro by morpholino antisense oligonucleotides Hum Mutat 27, 420–426 ´ Pros E, Fernandez-Rodrı´ guez J, Canet B, Benito L, ´ ´ Sanchez A, Benavides A, Ramos FJ, Lopez-Ariztegui ´ MA, Capella G, Blanco I, et al (2009) Antisense therapeutics for neurofibromatosis type caused by deep intronic mutations Hum Mutat 30, 454–462 Baralle D (2009) Splicing minireviews: introduction FEBS J, doi:10.1111/j.1742-4658.2009.07518.x Raponi M & Baralle D (2009) Alternative Splicing: Translationally silent substitutions that affect splicing: the bad and the good FEBS J, doi:10.1111/j.17424658.2009.07519.x Tazi J, Bakkour N, Aboufirassi A & Branlant C (2009) Alternative Splicing: Alternative splicing in the regulation of HIV-1 multiplication – a target for therapeutic action FEBS J, doi:10.1111/j.1742-4658 2009.07522.x Halleger M, Llorian M & Smith C (2009) Alternative Splicing: Global insights into alternative splicing FEBS J, doi:10.1111/j.1742-4658.2009.07521.x Ben-Dov C, Hartmann B, Lundgren J & Valcarcel J (2008) Genome-wide analysis of alternative pre-mRNA splicing J Biol Chem 283, 1229–1233 Moore MJ & Silver PA (2008) Global analysis of mRNA splicing RNA 14, 197–203 Garcia-Blanco MA (2005) Making antisense of splicing Curr Opin Mol Ther 7, 476–482 Ishii S, Nakao S, Minamikawa-Tachino R, Desnick RJ & Fan JQ (2002) Alternative splicing in the alphagalactosidase A gene: increased exon inclusion results in the Fabry cardiac phenotype Am J Hum Genet 70, 994–1002 McConville CM, Stankovic T, Byrd PJ, McGuire GM, Yao QY, Lennox GG & Taylor MR (1996) Mutations associated with variant phenotypes in ataxia-telangiectasia Am J Hum Genet 59, 320–330 Treisman R, Orkin SH & Maniatis T (1983) Specific transcription and RNA splicing defects in five cloned beta-thalassaemia genes Nature 302, 591–596 Dobkin C, Pergolizzi RG, Bahre P & Bank A (1983) Abnormal splice in a mutant human beta-globin gene not at the site of a mutation Proc Natl Acad Sci USA 80, 1184–1188 Cheng TC, Orkin SH, Antonarakis SE, Potter MJ, Sexton JP, Markham AF, Giardina PJ, Li A & Kazazian HH Jr (1984) Beta-thalassemia in Chinese: use of in vivo RNA analysis and oligonucleotide hybridization in systematic characterization of molecular defects Proc Natl Acad Sci USA 81, 2821–2825 Balz V, Prisack HB, Bier H & Bojar H (2002) Analysis of BRCA1, TP53, and TSG101 germline mutations in German breast and ⁄ or ovarian cancer families Cancer Genet Cytogenet 138, 120–127 84 Lee WI, Torgerson TR, Schumacher MJ, Yel L, Zhu Q & Ochs HD (2005) Molecular analysis of a large cohort of patients with the hyper immunoglobulin M (IgM) syndrome Blood 105, 1881–1890 85 den Hollander AI, Koenekoop RK, Yzer S, Lopez I, Arends ML, Voesenek KE, Zonneveld MN, Strom TM, Meitinger T, Brunner HG, et al (2006) Mutations in the CEP290 (NPHP6) gene are a frequent cause of Leber congenital amaurosis Am J Hum Genet 79, 556–561 ´ ´ 86 Chillon M, Dork T, Casals T, Gimenez J, Fonknechten ă N, Will K, Ramos D, Nunes V & Estivill X (1995) A novel donor splice site in intron 11 of the CFTR gene, created by mutation 1811+1.6kbA–>G, produces a new exon: high frequency in Spanish cystic fibrosis chromosomes and association with severe phenotype Am J Hum Genet 56, 623–629 87 Highsmith WE, Burch LH, Zhou Z, Olsen JC, Boat TE, Spock A, Gorvoy JD, Quittel L, Friedman KJ, Silverman LM, et al (1994) A novel mutation in the cystic fibrosis gene in patients with pulmonary disease but normal sweat chloride concentrations N Engl J Med 331, 974–980 ` 88 Faa V, Incani F, Meloni A, Corda D, Masala M, Baffico AM, Seia M, Cao A & Rosatelli MC (2009) Characterization of a disease-associated mutation affecting a putative splicing regulatory element in intron 6b of the cystic fibrosis transmembrane conductance regulator (CFTR) gene J Biol Chem 284, 30024–30031 89 Monnier N, Gout JP, Pin I, Gauthier G & Lunardi J (2001) A novel 3600+11.5 kb C>G homozygous splicing mutation in a black African, consanguineous CF family J Med Genet 38, E4 90 van den Hurk JA, van de Pol DJ, Wissinger B, van Driel MA, Hoefsloot LH, de Wijs IJ, van den Born LI, Heckenlively JR, Brunner HG, Zrenner E, et al (2003) Novel types of mutation in the choroideremia (CHM) gene: a full-length L1 insertion and an intronic mutation activating a cryptic exon Hum Genet 113, 268–275 91 Knebelmann B, Forestier L, Drouot L, Quinones S, Chuet C, Benessy F, Saus J & Antignac C (1995) Splice-mediated insertion of an Alu sequence in the COL4A3 mRNA causing autosomal recessive Alport syndrome Hum Mol Genet 4, 675–679 92 King K, Flinter FA, Nihalani V & Green PM (2002) Unusual deep intronic mutations in the COL4A5 gene cause X linked Alport syndrome Hum Genet 111, 548–554 93 Varon R, Gooding R, Steglich C, Marns L, Tang H, Angelicheva D, Yong KK, Ambrugger P, Reinhold A, Morar B, et al (2003) Partial deficiency of the C-terminal-domain phosphatase of RNA polymerase II is associated with congenital cataracts facial dysmorphism neuropathy syndrome Nat Genet 35, 185–189 FEBS Journal 277 (2010) 841–855 ª 2010 ICGEB Trieste (Italy) Journal compilation ª 2010 FEBS A Dhir and E Buratti 94 Rump A, Rosen-Wolff A, Gahr M, Seidenberg J, Roos C, Walter L, Gunther V & Roesler J (2006) A splicesupporting intronic mutation in the last bp position of a cryptic exon within intron of the CYBB gene induces its incorporation into the mRNA causing chronic granulomatous disease (CGD) Gene 371, 174–181 95 Noack D, Heyworth PG, Newburger PE & Cross AR (2001) An unusual intronic mutation in the CYBB gene giving rise to chronic granulomatous disease Biochim Biophys Acta 1537, 125–131 96 Ikeda H, Matsubara Y, Mikami H, Kure S, Owada M, Gough T, Smooker PM, Dobbs M, Dahl HH, Cotton RG, et al (1997) Molecular analysis of dihydropteridine reductase deficiency: identification of two novel mutations in Japanese patients Hum Genet 100, 637–642 97 Tuffery-Giraud S, Saquet C, Chambert S & Claustres M (2003) Pseudoexon activation in the DMD gene as a novel mechanism for Becker muscular dystrophy Hum Mutat 21, 608–614 ´ ´ 98 Beroud C, Carrie A, Beldjord C, Deburgrave N, Llense ´ S, Carelle N, Peccate C, Cuisset JM, Pandit F, CarrePigeon F, et al (2004) Dystrophinopathy caused by mid-intronic substitutions activating cryptic exons in the DMD gene Neuromusc Disord 14, 10–18 ´ 99 Deburgrave N, Daoud F, Llense S, Barbot JC, Recan ´ D, Peccate C, Burghes AH, Beroud C, Garcia L, Kaplan JC, et al (2007) Protein- and mRNA-based phenotype–genotype correlations in DMD ⁄ BMD with point mutations and molecular basis for BMD with nonsense and frameshift mutations in the DMD gene Hum Mutat 28, 183–195 100 Ikezawa M, Nishino I, Goto Y, Miike T & Nonaka I (1999) Newly recognized exons induced by a splicing abnormality from an intronic mutation of the dystrophin gene resulting in Duchenne muscular dystrophy Mutations in brief no 213 Online Hum Mutat 13, 170 101 Yagi M, Takeshima Y, Wada H, Nakamura H & Matsuo M (2003) Two alternative exons can result from activation of the cryptic splice acceptor site deep within intron of the dystrophin gene in a patient with as yet asymptomatic dystrophinopathy Hum Genet 112, 164–170 102 Guo DC, Gupta P, Tran-Fadulu V, Guidry TV, Leduc MS, Schaefer FV & Milewicz DM (2008) An FBN1 pseudoexon mutation in a patient with Marfan syndrome: confirmation of cryptic mutations leading to disease J Hum Genet 53, 1007–1011 103 Spena S, Asselta R, Plate M, Castaman G, Duga S & Tenchini ML (2007) Pseudo-exon activation caused by a deep-intronic mutation in the fibrinogen gammachain gene as a novel mechanism for congenital afibrinogenaemia Br J Haematol 139, 128–132 Pseudoexons in human disease 104 Bagnall RD, Waseem NH, Green PM, Colvin B, Lee C & Giannelli F (1999) Creation of a novel donor splice site in intron of the factor VIII gene leads to activation of a 191 bp cryptic exon in two haemophilia A patients Br J Haematol 107, 766–771 105 De Gasperi R, Gama Sosa MA, Sartorato EL, Battistini S, MacFarlane H, Gusella JF, Krivit W & Kolodny EH (1996) Molecular heterogeneity of late-onset forms of globoid-cell leukodystrophy Am J Hum Genet 59, 1233–1242 106 Wang M, Dotzlaw H, Fuqua SA & Murphy LC (1997) A point mutation in the human estrogen receptor gene is associated with the expression of an abnormal estrogen receptor mRNA containing a 69 novel nucleotide insertion Breast Cancer Res Treat 44, 145–151 107 Metherell LA, Akker SA, Munroe PB, Rose SJ, Caulfield M, Savage MO, Chew SL & Clark AJ (2001) Pseudoexon activation as a novel mechanism for disease resulting in atypical growth-hormone insensitivity Am J Hum Genet 69, 641–646 108 Vervoort R, Gitzelmann R, Lissens W & Liebaers I (1998) A mutation (IVS8+0.6kbdelTC) creating a new donor splice site activates a cryptic exon in an Alu-element in intron of the human beta-glucuronidase gene Hum Genet 103, 686–693 109 Purevsuren J, Fukao T, Hasegawa Y, Fukuda S, Kobayashi H & Yamaguchi S (2008) Study of deep intronic sequence exonization in a Japanese neonate with a mitochondrial trifunctional protein deficiency Mol Genet Metab 95, 46–51 110 Stum M, Davoine CS, Vicart S, Guillot-Noel L, ă Topaloglu H, Carod-Artal FJ, Kayserili H, Hentati F, Merlini L, Urtizberea JA, et al (2006) Spectrum of HSPG2 (Perlecan) mutations in patients with Schwartz–Jampel syndrome Hum Mutat 27, 1082–1091 111 Rathmann M, Bunge S, Beck M, Kresse H, TylkiSzymanska A & Gal A (1996) Mucopolysaccharidosis type II (Hunter syndrome): mutation ‘‘hot spots’’ in the iduronate-2-sulfatase gene Am J Hum Genet 59, 1202–1209 ´ 112 Sevenet N, Lellouch-Tubiana A, Schofield D, Hoang-Xuan K, Gessler M, Birnbaum D, Jeanpierre C, Jouvet A & Delattre O (1999) Spectrum of hSNF5 ⁄ INI1 somatic mutations in human cancer and genotype–phenotype correlations Hum Mol Genet 8, 2359–2368 113 Olsson A, Lind L, Thornell LE & Holmberg M (2008) Myopathy with lactic acidosis is linked to chromosome 12q23.3-24.11 and caused by an intron mutation in the ISCU gene resulting in a splicing defect Hum Mol Genet 17, 1666–1672 114 Mochel F, Knight MA, Tong WH, Hernandez D, Ayyad K, Taivassalo T, Andersen PM, Singleton A, Rouault TA, Fischbeck KH, et al (2008) Splice mutation in the iron–sulfur cluster scaffold protein ISCU FEBS Journal 277 (2010) 841–855 ª 2010 ICGEB Trieste (Italy) Journal compilation ª 2010 FEBS 853 Pseudoexons in human disease 115 116 117 118 119 120 121 122 123 124 125 854 A Dhir and E Buratti causes myopathy with exercise intolerance Am J Hum Genet 82, 652–660 Kollberg G, Tulinius M, Melberg A, Darin N, Andersen O, Holmgren D, Oldfors A & Holme E (2009) Clinical manifestation and a new ISCU mutation in iron–sulphur cluster deficiency myopathy Brain 132, 2170–2179 Stucki M, Suormala T, Fowler B, Valle D & Baumgartner MR (2009) Cryptic exon activation by disruption of an exon splice enhancer: a novel mechanism causing 3-methylcrotonyl-CoA carboxylase deficiency J Biol Chem 284, 28953–28957 Hilgert N, Topsakal V, van Dinther J, Offeciers E, Van de Heyning P & Van Camp G (2008) A splice-site mutation and overexpression of MYO6 cause a similar phenotype in two families with autosomal dominant hearing loss Eur J Hum Genet 16, 593–602 Lebon S, Minai L, Chretien D, Corcos J, Serre V, Kadhom N, Steffann J, Pauchard JY, Munnich A, Bonnefont JP, et al (2007) A novel mutation of the NDUFS7 gene leads to activation of a cryptic exon and impaired assembly of mitochondrial complex I in a patient with Leigh syndrome Mol Genet Metab 92, 104–108 Raponi M, Upadhyaya M & Baralle D (2006) Functional splicing assay shows a pathogenic intronic mutation in neurofibromatosis type (NF1) due to intronic sequence exonization Hum Mutat 27, 294–295 Pros E, Gomez C, Martin T, Fabregas P, Serra E & Lazaro C (2008) Nature and mRNA effect of 282 different NF1 point mutations: focus on splicing alterations Hum Mutat 29, E173–E193 Wimmer K, Roca X, Beiglbock H, Callens T, Etzler J, ¨ Rao AR, Krainer AR, Fonatsch C & Messiaen L (2007) Extensive in silico analysis of NF1 splicing defects uncovers determinants for splicing outcome upon 5¢ splice-site disruption Hum Mutat 28, 599–612 Perrin G, Morris MA, Antonarakis SE, Boltshauser E & Hutter P (1996) Two novel mutations affecting mRNA splicing of the neurofibromatosis type (NF1) gene Hum Mutat 7, 172–175 Ars E, Serra E, Garcia J, Kruyer H, Gaona A, Lazaro C & Estivill X (2000) Mutations affecting mRNA splicing are the most common molecular defects in patients with neurofibromatosis type Hum Mol Genet 9, 237–247 De Klein A, Riegman PH, Bijlsma EK, Heldoorn A, Muijtjens M, den Bakker MA, Avezaat CJ & Zwarthoff EC (1998) A G–>A transition creates a branch point sequence and activation of a cryptic exon, resulting in the hereditary disorder neurofibromatosis Hum Mol Genet 7, 393–398 Rodriguez-Pascau L, Coll MJ, Vilageliu L & Grinberg D (2009) Antisense oligonucleotide treatment for a 126 127 128 129 130 131 132 133 134 pseudoexon-generating mutation in the NPC1 gene causing Niemann–Pick type C disease Hum Mutat, E993–E1001 Mitchell GA, Labuda D, Fontaine G, Saudubray JM, Bonnefont JP, Lyonnet S, Brody LC, Steel G, Obie C & Valle D (1991) Splice-mediated insertion of an Alu sequence inactivates ornithine delta-aminotransferase: a role for Alu elements in human mutation Proc Natl Acad Sci USA 88, 815–819 Ogino W, Takeshima Y, Nishiyama A, Okizuka Y, Yagi M, Tsuneishi S, Saiki K, Kugo M & Matsuo M (2007) Mutation analysis of the ornithine transcarbamylase (OTC) gene in five Japanese OTC deficiency patients revealed two known and three novel mutations including a deep intronic mutation Kobe J Med Sci 53, 229–240 Christie PT, Harding B, Nesbit MA, Whyte MP & Thakker RV (2001) X-linked hypophosphatemia attributable to pseudoexons of the PHEX gene J Clin Endocrinol Metab 86, 3840–3844 Michel-Calemard L, Dijoud F, Till M, Lambert JC, Vercherat M, Tardy V, Coubes C & Morel Y (2009) Pseudoexon activation in the PKHD1 gene: a French founder intronic mutation IVS46+653A>G causing severe autosomal recessive polycystic kidney disease Clin Genet 75, 203–206 Vega AI, Perez-Cerda C, Desviat LR, Matthijs G, Ugarte M & Perez B (2009) Functional analysis of three splicing mutations identified in the PMM2 gene: toward a new therapy for congenital disorder of glycosylation type Ia Hum Mutat 30, 795– 803 Schollen E, Keldermans L, Foulquier F, Briones P, ´ Chabas A, Sanchez-Valverde F, Adamowicz M, Pronicka E, Wevers R & Matthijs G (2007) Characterization of two unusual truncating PMM2 mutations in two CDG-Ia patients Mol Genet Metab 90, 408–413 Frio TR, McGee TL, Wade NM, Iseli C, Beckmann JS, Berson EL & Rivolta C (2009) A single-base substitution within an intronic repetitive element causes dominant retinitis pigmentosa with reduced penetrance Hum Mutat 30, 1340–1347 Meili D, Kralovicova J, Zagalak J, Bonafe L, Fiori L, Blau N, Thony B & Vorechovsky I (2009) Diseasecausing mutations improving the branch site and polypyrimidine tract: pseudoexon activation of LINE-2 and antisense Alu lacking the poly(T)-tail Hum Mutat 30, 823–831 ` Dehainault C, Michaux D, Pages-Berhouet S, Caux-Moncoutier V, Doz F, Desjardins L, Couturier J, Parent P, Stoppa-Lyonnet D, Gauthier-Villars M, et al (2007) A deep intronic mutation in the RB1 gene leads to intronic sequence exonisation Eur J Hum Genet 15, 473–477 FEBS Journal 277 (2010) 841–855 ª 2010 ICGEB Trieste (Italy) Journal compilation ª 2010 FEBS A Dhir and E Buratti 135 Monnier N, Ferreiro A, Marty I, Labarre-Vila A, Mezin P & Lunardi J (2003) A homozygous splicing mutation causing a depletion of skeletal muscle RYR1 is associated with multi-minicore disease congenital myopathy with ophthalmoplegia Hum Mol Genet 12, 1171–1178 136 Valdmanis PN, Belzil VV, Lee J, Dion PA, St-Onge J, Hince P, Funalot B, Couratier P, Clavelou P, Camu Pseudoexons in human disease W, et al (2009) A mutation that creates a pseudoexon in SOD1 causes familial ALS Ann Hum Genet 73, 652–657 137 Mayer K, Ballhausen W, Leistner W & Rott H (2000) Three novel types of splicing aberrations in the tuberous sclerosis TSC2 gene caused by mutations apart from splice consensus sequences Biochim Biophys Acta 1502, 495–507 FEBS Journal 277 (2010) 841–855 ª 2010 ICGEB Trieste (Italy) Journal compilation ª 2010 FEBS 855 ... (even in the absence of a complete or even partial understanding of the ‘basic science’ explaining their occurrence) From a therapeutic point of view, the major advantage of targeting pseudoexon inclusion... several of the newly created enhancer sequences strongly correlate with potential binding to the SR protein class of splicing regulators 846 Therapeutic strategies aimed at correcting pseudoexon inclusion... recognition in the normal splicing process [55] First, U1snRNP binding to an intronic splicing processing element has been found to inhibit pathological pseudoexon inclusion in intron 20 of the ATM

Ngày đăng: 06/03/2014, 09:22

Từ khóa liên quan

Tài liệu cùng người dùng

Tài liệu liên quan