G Model

YSCDB-2404; No. of Pages 10 ARTICLE IN PRESS

Seminars in Cell & Developmental Biology xxx (2017) xxx–xxx

Contents lists available at ScienceDirect

Seminars in Cell & Developmental Biology

j ournal homepage: www.elsevier.com/locate/semcdb

Review

Minor and disease

1 ∗

Bhupendra Verma , Maureen V. Akinyi, Antto J. Norppa, Mikko J. Frilander

Institute of Biotechnology, PL56 (Viikinkaari 5), 000014, University of Helsinki, Finland

a r t i c l e i n f o a b s t r a c t

Article history: The U12-dependent (minor) spliceosome excises a rare group of introns that are characterized by a



Received 4 July 2017

highly conserved 5 splice site and branch point sequence. Several new congenital or somatic diseases

Received in revised form

have recently been associated with mutations in components of the minor spliceosome. A common

21 September 2017

theme in these diseases is the detection of elevated levels of transcripts containing U12-type introns,

Accepted 27 September 2017

of which a subset is associated with other splicing defects. Here we review the present understanding

Available online xxx

of minor spliceosome diseases, particularly those associated with the specific components of the minor

spliceosome. We also present a model for interpreting the molecular-level consequences of the different Keywords: diseases.

U12-type introns

© 2017 The Authors. Published by Elsevier Ltd. This is an open access article under the CC BY license

Minor spliceosome (http://creativecommons.org/licenses/by/4.0/). Human diseases

Pre-mRNA splicing

Cryptic splice sites

Exon skipping

Intron retention

Contents

1. Introduction ...... 00

2. Minor spliceosome in human disease ...... 00

2.1. Diseases affecting the intron recognition step ...... 00

2.1.1. Isolated growth hormone deficiency ...... 00

2.1.2. Cerebellar ataxia ...... 00

2.1.3. Myelodysplastic syndrome ...... 00

2.2. Diseases affecting the formation of catalytically active spliceosome ...... 00

2.2.1. MOPD1/TALS...... 00

2.2.2. Roifman syndrome ...... 00

2.3. Diseases with mutations in the U12-type splice sites ...... 00

2.4. Other spliceosome mutations affecting minor spliceosome activity ...... 00

3. Minor spliceosome components as biomarkers ...... 00

4. Consequences of minor spliceosome-specific mutations ...... 00

4.1. The fate of the mRNA: intron retention vs cryptic splicing ...... 00

4.2. Fate of the mRNA and disease severity ...... 00

4.3. Defects with individual genes vs a global splicing defect ...... 00

5. Future perspective ...... 00

Acknowledgements...... 00

References ...... 00

Corresponding author.

E-mail address: [email protected] (M.J. Frilander).

1

Present address: Department of Biotechnology, All India Institute of Medical Sciences, New Delhi, India.

https://doi.org/10.1016/j.semcdb.2017.09.036

1084-9521/© 2017 The Authors. Published by Elsevier Ltd. This is an open access article under the CC BY license (http://creativecommons.org/licenses/by/4.0/).

Please cite this article in press as: B. Verma, et al., Minor spliceosome and disease, Semin Cell Dev Biol (2017), https://doi.org/10.1016/j.semcdb.2017.09.036

G Model

YSCDB-2404; No. of Pages 10 ARTICLE IN PRESS

2 B. Verma et al. / Seminars in Cell & Developmental Biology xxx (2017) xxx–xxx

1. Introduction U11/U12 di-snRNP protein component, Urp/ZRSR2 takes up the role



of 3 ss recognition with minor introns [19]. Following this initial

Pre-mRNA splicing is an essential step in the gene expression recognition, both splicing pathways proceed with association of a

pathway of all eukaryotes. During splicing, the non-coding intron tri-snRNP, either U4atac/U6atac.U5 or U4/U6.U5 (Fig. 1B; [20,21]).

sequences are recognized and removed from precursor mRNA (pre- Further rearrangements in RNA–RNA and RNA–protein interac-

mRNA) by the spliceosome, a large ribonucleoprotein complex. tions lead to the formation of a catalytically active spliceosome and

Defects in splicing of one or more mRNA species are a major cause catalytic excision of the intron [20,22,23].

of human diseases and present estimates suggest that up to 60%, Here, we review the role of the minor spliceosome in human

are associated with pre-mRNA splicing [1,2]. In addition to mono- diseases, with a specific focus on diseases caused by mutations

genic disorders, genetic variants altering splicing are also thought in the integral components of the minor spliceosome. Given that

to be an important contributor to complex diseases and cancer most protein components and U5 snRNA are shared with the major

[3,4]. The majority of disease-associated splicing defects are cis- spliceosome, there are also several diseases where mutations dis-

acting mutations within a single gene that disrupt either the splice rupting shared components can potentially affect the functions of

site consensus sequences or splicing regulatory elements located in both . Those have been discussed in detail elsewhere

introns or [5]. Notably, exonic mutations interpreted as mis- [24].

sense, nonsense or silent also commonly affect splicing [4,6,7]. The

outcome of cis-acting splicing mutations is often either a formation

of abnormal or non-functional protein species or accelerated decay 2. Minor spliceosome in human disease

of the affected individual mRNAs. In contrast, mutations in splicing

factors, spliceosome components or spliceosome assembly factors The direct targets for human diseases specific for the minor

often lead to widespread defects in the processing of large numbers spliceosome are the unique snRNA and protein components. This

of pre-mRNAs [8–10]. includes several components of the U11/U12 intron recognition

Most metazoan species contain two distinct pre-mRNA splicing complex and the U4atac and U6atac snRNAs in the minor tri-snRNP.

machineries known as the major (U2-dependent) and minor (U12- The U11/U12 di-snRNP contains, in addition to the U11 and U12

dependent) spliceosomes, which recognize and excise either the snRNAs, seven integral proteins (65K, 48K, 59K, 35K, 31K, 25K and

major (U2-type) or minor (U12-type) class of introns, respectively. 20K) that are unique to the minor spliceosome [25,26]. Addition-

In contrast to the major introns, U12-type introns are character- ally, the Urp/ZRSR2 protein associated with the U11/U12 di-snRNP

 

ized by divergent and highly conserved 5 splice site (5 ss) and has been reported to function in both minor and major spliceo-



branch point sequences (BPS) (Fig. 1A; [11]). These introns also somes, with an essential role for U12-type intron 3 ss recognition

lack the characteristic polypyrimidine tract (PPT) that is present in [19].

 

U2-type introns immediately upstream of the 3 splice site (3 ss). To date five human diseases with mutations in the spe-

Minor introns constitute only 0.35% of all human introns and have cific components of the minor spliceosome have been described

been reported to be present in 700–800 genes, each of which typ- (Table 1). Three of them affect the components of the U11/U12 di-

ically carry only a single U12-type intron and multiple U2-type snRNP, namely the U11/U12-65K protein (RNPC3; [27]), U12 snRNA

introns. U12-type introns are enriched in genes that represent a (RNU12; [28]) and Urp protein (ZRSR2; [29]); while two diseases are

rather restricted set of functional classes and pathways. Particu- attributed to mutations in the U4atac snRNA (RNU4ATAC; [30–32]).

larly they are present in genes related to ‘information processing Each of these diseases is hypomorphic, leading only to a partial loss

functions’, such as DNA replication and repair, transcription, RNA of minor spliceosome function because correctly spliced mRNAs

processing, and translation, but can also be found in genes related can be detected in the patient cells. We briefly introduce each

to cytoskeletal organization, vesicular transport, and voltage-gated disease and later discuss the impact of disease mutations on the

ion channel activity, as suggested originally by Burge et al., [12] assembly of the minor spliceosome and the fate of affected mRNAs.

and verified later [13,14]. Both the identities of genes carrying U12- Apart from mutations in the core minor spliceosome compo-

type introns and their positions within the genes are evolutionarily nents, we also describe the few reported human diseases caused

conserved [15]. by mutations at the splice sites of U12-type introns, though we

The overall organisational and functional features of both note that this appears to be underexplored territory given the small

spliceosomes are highly similar. Both are composed of five small number of cases reported so far. Finally, we mention the emerging

nuclear RNA (snRNA) molecules that associate with protein factors role for the minor spliceosome in cancer and autoimmune disorders

to give rise to small nuclear ribonucleoproteins (). Within as well as neurodegenerative diseases.

the minor spliceosome, four of the five snRNAs are unique. Specifi-

cally, U11, U12, U4atac, and U6atac replace the major spliceosome

counterparts U1, U2, U4 and U6 snRNAs, respectively. U5 snRNA 2.1. Diseases affecting the intron recognition step

is shared between the two spliceosomes. Of the 200–300 pro-

teins associated with spliceosomes, most are thought to be shared 2.1.1. Isolated growth hormone deficiency

between the two systems and only 7 proteins, associated with U11 Recessive mutations in the RNPC3 gene, encoding U11/U12-

and U12 snRNPs, are unique to the minor spliceosome [16,17]. 65K, one of the seven minor spliceosome-specific proteins, have

The highly similar spliceosome composition is reflected in been associated with isolated growth hormone deficiency (IGHD)

the conserved assembly pathway and catalytic mechanism. Both and associated pituitary hypoplasia. The 65K protein is part of a

spliceosomes are assembled sequentially starting from intron molecular bridge that connects the U11 and U12 snRNPs into a di-

recognition, followed by formation of a catalytically active spliceo- snRNP (Fig 1B; [33]). Initially, RNPC3 mutations were detected in a

some and joining of the exons flanking the excised intron. With single family only [27], but additional cases with overlapping muta-



minor introns, the 5 ss and BPS are co-operatively recognized by tions and similar phenotypes have been described subsequently

a pre-formed U11/U12 di-snRNP [18], contrary to the sequential [34]. IGHD is a condition characterized by a shortage or absence

recognition of these sequences by individual U1 and U2 snRNPs of growth hormone, with the absence of associated pituitary hor-

in major introns. Since the PPT is lacking in minor introns, the mone deficiencies. Genetically, IGHD is a diverse disease and can



U2AF1/2 heterodimer that recognizes the PPT and 3 ss of major result from either recessive or dominant mutations in various genes

introns does not associate with minor introns. Instead, an integral involved in pituitary development or function [35].

Please cite this article in press as: B. Verma, et al., Minor spliceosome and disease, Semin Cell Dev Biol (2017), https://doi.org/10.1016/j.semcdb.2017.09.036

G Model

YSCDB-2404; No. of Pages 10 ARTICLE IN PRESS

B. Verma et al. / Seminars in Cell & Developmental Biology xxx (2017) xxx–xxx 3

Fig. 1. (A) Consensus sequences of human U12-type and U-2 type introns. Adapted from [11]. (B) A simplified assembly pathway of the U12-dependent spliceosome

highlighting the intron recognition and catalytic stages of the assembly. The schematic structure of the U11/U12 intron recognition complex includes protein components

specific to the U12-dependent spliceosome (65K (RNPC3), 59K (PDCD7), 48K (SNRP48), 35K (SNRNP35), 31K (SNRNP31/ZCRB1), 25K (SNRP25K), 20K (SNRNP20/ZMAT5), Urp

(ZRSR2) and also the SF3b complex shared between the two spliceosomes. Additionally, human diseases specifically associated with either assembly step of the minor

spliceosome assembly are indicated on right.

All reported IGHD cases associated with RNPC3 mutations are terminal RNA recognition motif (RRM) of the protein, while the

compound heterozygous with a missense P474T mutation com- R205X maps to the proline-rich region between the two RRMs

bined with either R502X or R205X nonsense mutation [27,34]. At (Fig. 2A). Although the tissue affected in IGHD, the pituitary gland,

the protein level the P474T and R502X mutations map to the C- cannot be sampled from the patients, RT-PCR and RNA-seq analyses

Please cite this article in press as: B. Verma, et al., Minor spliceosome and disease, Semin Cell Dev Biol (2017), https://doi.org/10.1016/j.semcdb.2017.09.036

G Model

YSCDB-2404; No. of Pages 10 ARTICLE IN PRESS

4 B. Verma et al. / Seminars in Cell & Developmental Biology xxx (2017) xxx–xxx

Table 1

Human diseases showing defects in splicing of U12-type introns.

Defect in Locus Disease Abbreviation References

Integral components of RNPC3 Isolated Growth Hormone Deficiency IGHD [27]

minor snRNPs ZRSR2 Myelodysplastic Syndrome MDS [29]

RNU12 Early-onset Cerebellar ataxia EOCA [28]

RNU4ATAC Microcephalic osteodysplastic primordial dwarfism/Taybi-Linder Syndrome MOPD1/TALS [31,32]

RNU4ATAC Roifman syndrome RFMN [30]



U12-type 5 ss STK11 Peutz-Jegher’s syndrome PJS [49]

TRAPPC2 Spondyloepiphyseal dysplasia tarda SEDT [50]

Biogenesis/Assembly FUS Amyotrophic lateral sclerosis ALS [64]

SMN1 Spinal muscular atrophy SMA [10,57–61]

Fig. 2. Disease-associated mutations in the specific protein and snRNA components of the minor spliceosome.

A) Domain structure of the U11/U12-65K proteins with the locations of IGHD-associated mutations indicated.

B) Secondary structure of U12 snRNA showing the location of a point mutation associated with EOCA. Binding site for the U11/U12-65K protein is also indicated.

C) Secondary structure of the U4atac/U6atac di-snRNA complex showing mutations associated with MOPDI (red) and RFMN (cyan). A mutation shared between MOPD1/TALS

and RFNM patients is indicated in red text surrounded by a cyan rectangle. Binding sites for 15.5K, PRPF31, the PRPF4/PRPF3/PPIH complex proteins and the U11/U12-65K

protein are also shown.

D) Domain structure of the ZRSR2 protein showing mutations associated with MDS. Mutations are from the COSMIC database [76]. Mutations studied by Madan et al. [29]

are shown in blue.

was carried out on P474T/R502X patient lymphocytes [27]. Consis- as well as reduced stability of the U11/U12 di-snRNP complex.

tent with the function of the 65K protein in the intron recognition This result is consistent with the function of the 65K protein as a

complex, several types of aberrant splicing events associated with component of themolecular bridge between U11 and U12 snRNPs

a subset of U12-type introns were detected, including increased through interactions with the U11-59K protein and U12 snRNA

retention of U12-type introns, activation of nearby cryptic U2- [33]. The 65K-U12 snRNA interaction is mediated by the C-terminal

type splice sites and skipping events. Biochemical analysis RRM domain and, as predicted, the IGHD mutations were shown

of patient cells revealed a reduction in levels of the 65K protein, to either reduce (P474T) or eliminate (R502X) the binding of 65K

Please cite this article in press as: B. Verma, et al., Minor spliceosome and disease, Semin Cell Dev Biol (2017), https://doi.org/10.1016/j.semcdb.2017.09.036

G Model

YSCDB-2404; No. of Pages 10 ARTICLE IN PRESS

B. Verma et al. / Seminars in Cell & Developmental Biology xxx (2017) xxx–xxx 5

to the stem-loop III of U12 snRNA (Fig. 2B; [36]). The picture that domain (UHM) flanked on each side by a CCCH-type zinc finger, and

is emerging from these studies is that the IGHD mutations lead an RS domain near the C-terminus (Fig. 2D). ZRSR2 is an integral

to impaired interaction between the 65K protein and U12 snRNA, component of the U11/U12 di-snRNP and functions in the recogni-



which in turn causes destabilization or reduced formation of the tion of the U12-type 3 splice site, but it has also been reported to

U11/U12 di-snRNP. Additionally, it is likely that both RNPC3 non- function during the second catalytic step of U2-type intron splicing

sense mutations described for IGHD lead to allele-specific decay of [19,25]. MDS-associated mutations in ZRSR2 include a wide spec-

the 65K mRNA via nonsense mediated decay (NMD) pathway, as trum of nonsense, missense, frameshift and splice site mutations

shown recently for the R502X allele [36]. This is expected to signif- which are uniformly scattered across the gene (Fig. 2D). This distri-

icantly reduce the levels of 65K proteins encoded by the nonsense bution is in striking contrast to MDS cases with mutations in U2AF1,

alleles. SRSF2 and SF3B1 genes that show a narrower mutation spectrum,

However, we note that additional mechanisms may be at play, with particular preference for missense mutations that are located

as it is not currently known whether the 65K protein is purely a mostly within a small number of hotspot positions.

structural component of the U11/U12 di-snRNP, or whether it has Of the numerous somatic mutations reported in the ZRSR2

additional functions in the splicing process. A zebrafish study indi- (Fig. 2D), only few have been studied in detail [29]. In the Madan

cated that the 65K protein may have other functions later in the et al. [29] RNAseq study, bone marrow samples from eight ZRSR2-

spliceosome assembly [37]. Consistently, recent work reported an mutant MDS patients, each carrying a different mutation, were



interaction between 65K and the 3 terminal stem-loop of U6atac compared to both ZRSR2 wild-type MDS patients and healthy indi-

snRNA (Fig. 2C; [38]), which is similarly impaired by the IGHD viduals. The analysis revealed a global increase in U12-type intron

mutations [36]. retention, as well as activation of cryptic U2-type splice sites only in

MDS patients carrying the mutated ZRSR2 allele. Importantly, the

2.1.2. Cerebellar ataxia effect of ZRSR2 mutations and knockdown of ZRSR2 on U2-type

A mutation in the noncoding U12 snRNA gene (RNU12) has intron splicing were comparatively small. The affected U2-type

recently been associated with an early-onset cerebellar ataxia introns were mostly in genes containing U12-type introns. Similar

(EOCA) [28]. In general, ataxias are a diverse group of disorders conclusions have also been reported in plants [41]. These findings

characterized by defects in muscle coordination which, in the case are in stark contrast to the earlier biochemical experiments which

of cerebellar ataxias, results from abnormal development and/or suggested that an essential role for ZRSR2 was in splicing of both

degeneration of the cerebellum. The patients in this single study U2- and U12-type introns [19].

are homozygous for a point mutation (84C > T; see Fig. 2B) with

symptoms of hypotonia at infancy, delayed motor development, 2.2. Diseases affecting the formation of catalytically active

abnormal gait, speech and learning difficulties [28]. RNAseq and spliceosome

RT-PCR analyses of patient mononuclear cells showed increased

U12-type intron retention and upregulation of the U12 snRNA. 2.2.1. MOPD1/TALS

The 84C > T mutation is located at the base of U12 snRNA stem- Recessive mutations in the U4atac snRNA gene (RNU4atac)

loop III, and is predicted to weaken the stem-closing G-C base-pair associated with MOPD1/TALS, a rare autosomal recessive disease,

by converting it to a weaker G-U base-pair (Fig. 2B). Mechanistic represent the first human disease with mutations in a specific

understanding of how this mutation leads to a defect in minor component of the minor spliceosome [31,32,42]. U4atac snRNA

spliceosome function is currently lacking. The mutation does not is an essential component of the U4atac/U6atac.U5 tri-snRNP

directly overlap with any known functional elements, such as (minor tri-snRNP), which joins the nascent spliceosome after the

sequences involved in known RNA–RNA or RNA–protein interac- intron boundaries have been recognized by the U11/U12 di-snRNP

tions; however, the apical part of this long stem-loop acts as the (Fig. 1B). During the assembly of the minor tri-snRNP, U4atac forms

binding site for the 65K protein (Fig 2B; [33]) and the assembly site an extensively base-paired duplex with the U6atac snRNA (Figs. 1B

for the Sm protein ring, an integral part of all snRNPs, is located and 2C). This U4atac/U6atac di-snRNA complex forms a platform

only two nucleotides away. Furthermore, as the U12 snRNA not for the binding of tri-snRNP-specific proteins that are needed for

only recognizes the branch point sequence during intron recogni- the association of the U5 snRNP to the complex.

tion, but is also part of the catalytic core of the minor spliceosome, MOPD1/TALS in its severe form is characterized by intrauterine

several additional mechanistic scenarios are conceivable. Further and postnatal growth retardation, developmental defects in sev-

functional studies are needed to uncover the mechanistic basis of eral organs, including microcephaly, and death typically within 3

this disease. years after birth. However, milder forms of the disease have been

reported displaying more subtle growth retardation, less severe

2.1.3. Myelodysplastic syndrome developmental defects and/or survival to at least to 12 years of age

Myelodysplastic syndrome (MDS; myelodysplasia) is a term for or even adulthood [43,44]. Presently, individual single-nucleotide

a group of disorders characterized by inefficient hematopoiesis, point mutations at ten different positions within the U4atac snRNA

with a risk of progression to acute myeloid leukemia (AML). have been associated with MOPD1/TALS (Fig. 2C); in addition, one

In recent years, several whole-exome sequencing studies have patient with a duplication of U4atac nucleotides 16–100 has been

revealed frequent somatic mutations in genes encoding splicing reported [44]. All patients are either homozygous or compound

factors, including U2AF1, ZRSR2, SRSF2 and SF3B1 in patients with heterozygous for the disease mutations. In contrast, heterozy-

MDS reviewed by [9,39]. Although patients with a combination of gous parents carrying only a single disease-causing mutation are

different splicing factor mutations have been reported, typically phenotypically normal, suggesting that a loss of a single allele is

only one splicing factor gene in each patient has been mutated. Fur- well-tolerated at the cellular level.

thermore, the splicing factor mutations frequently co-occur with The majority of disease-causing mutations, particularly those

mutations in genes encoding epigenetic factors, cell signaling reg- leading to severe forms of the disease, are found in the U4atac



ulators and transcriptional regulators [39,40]. Here, we focus on the 5 stemloop, with only few pathogenic mutations located else-



Urp/ZRSR2 protein involved in U12-type 3 ss recognition. where in the U4atac snRNA (Fig. 2C). Earlier studies from the major

The ZRSR2 protein shares both primary structure and functional spliceosome have indicated that this region is recognized by a group



features with the splicing factor U2AF1, that facilitates 3 ss recogni- of proteins (Fig. 2C), particularly the 15.5K (Snu13) and PRPF31 pro-

tion with the major introns. Both contain a central U2AF-homology teins, that are both necessary for the association of U5 snRNP to

Please cite this article in press as: B. Verma, et al., Minor spliceosome and disease, Semin Cell Dev Biol (2017), https://doi.org/10.1016/j.semcdb.2017.09.036

G Model

YSCDB-2404; No. of Pages 10 ARTICLE IN PRESS

6 B. Verma et al. / Seminars in Cell & Developmental Biology xxx (2017) xxx–xxx

the tri-snRNP [45]. Consistently, the MOPD1/TALS mutations in this These three cases do not only demonstrate that a single point



region reduce the 15.5K binding, which in turn decreases the cellu- mutation in the highly conserved U12-type 5 ss sequence can

lar levels of U4atac/U6atac.U5 tri-snRNP [46]. Importantly, there is efficiently disable the U12-type intron recognition, but also that

a correlation between disease severity and the affinity of the 15.5K splicing of a U12-type intron is linked to other splicing events in



protein. Mutations on the tip of the 5 stemloop that reduce most the same transcript and that in some cases the outcome may be

the 15.5K binding, also lead to severe forms of the disease. In con- difficult to predict. PJS, in which a mutation at the +1 position

trast, the more distal mutations (such as U4atac positions 30, 53 and of an AT–AC-type U12-type intron is changed to G, is a particu-

55–see Fig. 2C) have little effect on the binding of the 15.5K pro- larly illuminating case [49]. At the sequence level, it appears to



tein and are consequently associated with milder symptoms, such be a simple U12-type 5 ss subtype change, but the experimentally

as longer life expectancy [43,46]. Additionally, at least one of the determined outcome is in fact complex, showing activation of sev-



mutations (124G > A) reduces the U4atac snRNA expression levels eral non-canonical 3 splice sites that illuminate the flexibility of



[46]. the U12-dependent spliceosome in 3 ss recognition. Even though

No transcriptome studies have yet been conducted for the outcome in each case is the same, i.e. introduction of a pre-

MOPD1/TALS, but qPCR analyses have shown increased U12-type mature STOP-codon and mRNA decay via the NMD pathway, this

intron retention in patient cells that can be rescued by expression particular mutation demonstrates that simple point mutations can

of wild type U4atac snRNA [31,32]. Significantly, an analysis with lead to unexpected consequences.

a reporter system indicates that >90% of the minor spliceosome

activity may have been compromised [31]. However, it is possible

that the reporter system used may have exacerbated the splic-

ing defect as the endogenous transcripts in patient cell lines and

2.4. Other spliceosome mutations affecting minor spliceosome

iPS cells carrying the mutation show much milder splicing defects

[31,32,46]. activity

In addition to diseases caused by mutations in the specific

2.2.2. Roifman syndrome

components of the minor spliceosome, there are several addi-

In addition to MOPD1/TALS, mutations in the RNU4atac locus

tional human diseases caused by congenital or somatic mutations

have been associated with a phenotypically different Roifman

in either the shared components of the two spliceosomes or the

syndrome (RFMN) [30]. Patients suffering from RFMN are char-

assembly factors necessary for both major and minor snRNPs.

acterized by poor pre- and post-natal growth, distinct facial

Examples of the former include Retinitis pigmentosa (RP) mutations

dystrophies, cognitive delay and immunological defects which,

in genes encoding the tri-snRNP specific proteins PRPF3, PRPF4,

with the exception of growth defects, are distinct to Roifman

PRPF6, PRPF8, PRPF31 and BRR2 [Reviewed in 8], and MDS muta-

patients [30]. Interestingly, all RFMN patients are compound het-

tions in PRPF8 [52], in SF3b1, a component of both U2 snRNP and

erozygotes, with one mutation shared with MOPD1/TALS patients

U11/U12 di-snRNP [26], and in SRSF2, a splicing activator poten-

with severe forms of the diseases (Fig. 2C). The other mutation is

tially affecting both spliceosomes [53]. Spinal muscular atrophy

predominantly located among the highly conserved positions of the

(SMA) represents the latter group and is caused by mutations in

RNU4atac stem II, except for a single case with a mutation within

the SMN1 gene, coding for an essential factor for the assembly of all

the Sm site.

Sm-class snRNPs, including components of both major and minor

No functional studies have been performed with the RFMN

spliceosomes [54].

mutations, but it is safe to assume that the mutations shared with

With RP, the shared protein composition between the minor

the MOPD1/TALS lead to a similar defect in minor tri-snRNP assem-

and major tri-snRNPs [17] and functional studies [55,56] predict

bly [46]. In contrast, the stem II mutations presumably lead to a

that the mutations should have an equal effect on both the major

milder functional defect in minor splicing, given the overall less

and minor tri-snRNPs. With SMA, however, several investigations

severe disease phenotype. While the molecular defects resulting

using different model systems have reported a preferential reduc-

from the stem II mutations remain to be determined, the investiga-

tion in the cellular levels of minor snRNPs [10,57–61]. In a subset of

tion from the major spliceosome assembly tentatively suggest that

the studies this has been reported to lead to an increased retention

the association of the PRPF4/PRPF3/PPIH complex (Fig. 2C) with the

U12-type introns, suggesting that defects in the splicing of minor

stem II may be compromised [30,47,48].

introns may contribute to the SMA symptoms. In a Drosophila model

In addition, RNAseq analyses of the patient cells revealed a

of SMA the effects on neuronal cells were attributed to a single

widespread retention of U12-type introns. Significantly, neither

U12-type intron containing gene, Stasimon, encoding a transmem-

increased retention of U2-type introns, nor other types of alter-

brane protein required for axonal growth and regulation of synaptic

native splicing changes were detected in the patient cells [30],

transmission of motor neurons [60]. However, some aspects of this

suggesting a very specific splicing defect affecting U12-type introns

only. study have been challenged subsequently [62].

Finally, two independent studies have linked minor spliceo-

some to amyotrophic lateral sclerosis (ALS). Ishihara et al. [63]

2.3. Diseases with mutations in the U12-type splice sites

reported that ALS-associated depletion of TDP-43 protein leads to

a co-depletion of U12 snRNA both in cultured cells and in patient

To our knowledge there are only two reported cases in lit-

samples, presumably due to defects in snRNP assembly. In another

erature that specifically attribute U12-type splice site mutations

study [64], ALS-associated mutations in the Fused in sarcoma (FUS)

with human disease. These include the autosomal dominant dis-

protein lead to reduced nuclear import of FUS and its accumulation

order Peutz-Jeghers syndrome (PJS), characterized by the presence

in the cytoplasm. FUS is a multifunctional RNA-binding protein that

of multiple intestinal polyps and a high risk for cancer [49], and

interacts with several splicing factors, including U11 snRNP and is

Spondyloepiphyseal dysplasia tarda (SEDT), an X-linked recessive



needed for the splicing of at least a subset of minor introns [64]. Sig-

disorder [50]. Both are caused by mutations in a U12-type 5 ss that

nificantly, the cytoplasmic accumulation of a particular FUS mutant

lead to incorrect splicing of the respective mRNAs. Additionally, the

leads to a concomitant mislocalization of U11 and U12 snRNPs in

Motor endplate disease in mouse reports a mutation of a U2-type



the cytoplasm, which in turn leads to defects in the splicing of

5 ss in a downstream intron, leading to skipping of the upstream

U12-type introns.

U12-type intron [51].

Please cite this article in press as: B. Verma, et al., Minor spliceosome and disease, Semin Cell Dev Biol (2017), https://doi.org/10.1016/j.semcdb.2017.09.036

G Model

YSCDB-2404; No. of Pages 10 ARTICLE IN PRESS

B. Verma et al. / Seminars in Cell & Developmental Biology xxx (2017) xxx–xxx 7

Fig. 3. Model for molecular consequences of the minor spliceosome diseases. Mutations in the minor spliceosome components can lead to nuclear retention of the partially

spliced mRNA, or intron retention and associated activation of cryptic U2-type splice sites or exon skipping events. These in turn lead to nuclear or cytoplasmic decay, or

formation of the abnormal proteins as indicated.

3. Minor spliceosome components as biomarkers molecular consequences of the mutations, but may also help to

interpret the subsequent disease phenotypes.

In addition to being a target for disease-causing mutations, sev-

eral components of the minor spliceosome have been reported as

4.1. The fate of the mRNA: intron retention vs cryptic splicing

cancer biomarkers. A recent study by Xu et al. [65] using serum

from patients with scleroderma and coincident cancer reported

According to this classification the mutations associated with

autoantibodies targeted to components of the U11/U12 di-snRNP,

both the MOPD1/TALS and RFMN affect the formation of the

particularly the U11/U12-65K protein, but also the 25K, 35K and

U4atac/U6atac.U5 tri-SNRP (see 2.2.1 and 2.2.2), but not the initial

59K proteins. Of these, the U11/U12-65K autoantibodies appear

intron recognition step. Therefore, in these diseases the spliceo-

to show a reliable association with an increased risk of cancer

some assembly is arrested at the pre-spliceosome stage where

occurring within 2 years of the onset of scleroderma [66]. Similar

the U11/U12 di-snRNP is bound to the intron but the subsequent

associations have been described for U11 snRNA in familial prostate

assembly steps are inhibited. Consequently, the bound U11/U12

cancers [67] and U11-59K protein in Acute Myeloid Leukemia [68].

di-snRNP can be expected to maintain exon-definition interactions

However, the role of minor spliceosome-specific autoantibodies in

with the surrounding (U2-type) introns via the RS domain contain-

the pathogenesis of scleroderma and/or coincident cancer, or the

ing U11-35K [69] and possibly Urp/ZRSR2 proteins. Furthermore,

expression level changes of other minor spliceosome components

the U11/U12 di-snRNP bound to the partially spliced mRNA can trap

in other diseases is presently unclear.

such transcripts in the nucleus [70], where they may be targeted

by the nuclear quality control mechanisms [71,72].

In contrast, the loss of U11/U12 binding and the recognition of

4. Consequences of minor spliceosome-specific mutations U12-type introns in IGHD and MDS (and possibly EOCA) leads to a

different outcome. Due to the loss of U11/U12 binding, transcripts

Presently, mutations in four specific components of the minor containing unspliced U12-type introns are free to be exported to the

spliceosome have been associated with human diseases showing cytoplasm. Concomitantly, the loss of local exon definition interac-

very different disease phenotypes. RT-PCR and RNAseq analyses tions may lead to increased activation of cryptic splice sites or exon

have demonstrated that each of the five human diseases show skipping events near the U12-type introns [73]. The outcome can

the expected molecular level phenotype, i.e. the increased levels be either truncated or altered protein, or mRNA decay via the NMD

of unspliced U12-type introns in the patient cells. With a subset of quality control pathway (Fig. 3).

diseases, additional splicing defects, particularly activation of cryp- Is there evidence supporting this model? RNAseq analyses from

tic U2-type splice sites and exon skipping events have also been both IGHD and MDS patient cells provide extensive evidence of acti-

described. Presently, there has not been clear consensus on how to vation of cryptic U2-type splice sites and increased levels of exon

interpret the molecular consequences of minor spliceosome muta- skipping near U12-type introns in addition to the expected reten-

tions. Here, we propose a simple classification of disease-causing tion of U12-type introns [27,29]. In contrast, besides the increased

mutations according to their impact on the recognition of the U12- intron retention of U12-type introns seen with both MOPD1/TALS

type introns. This provides a simple framework for predicting the and RFMN, there is no evidence of the associated cryptic U2-type

Please cite this article in press as: B. Verma, et al., Minor spliceosome and disease, Semin Cell Dev Biol (2017), https://doi.org/10.1016/j.semcdb.2017.09.036

G Model

YSCDB-2404; No. of Pages 10 ARTICLE IN PRESS

8 B. Verma et al. / Seminars in Cell & Developmental Biology xxx (2017) xxx–xxx

splicing events [30–32,46] as stated by Merico et al. [30] in their significance and function of the U12-dependent spliceosome. How-

detailed RNAseq analysis of RFMN patient cells. Together, present ever, many aspects of the disease process, including the basis of the

evidence supports the proposal that the consequences of the minor tissue specificity and the detailed mechanism of the disease process

spliceosome mutations are linked to the type of assembly defect are yet to be determined for most diseases, requiring animal models

introduced. However, with both MOPD1/TALS and RFMN it is pos- of each disease. These will also help to address the more fundamen-

sible that some arrested transcripts trapped in the nucleus may tal question of the existence of two separate spliceosomes.

either lose the bound U11/U12 di-snRNP or undergo the splicing

process post-transcriptionally (see Fig. 3). Acknowledgements

4.2. Fate of the mRNA and disease severity

This work was supported by the Academy of Finland (Grant

140087 to MJF and Grant 278798 to BV) and Sigrid Jusélius Foun-

It is somewhat puzzling to note that the severity of congen-

dation (MJF). AJN was supported by the Integrative Life Science

ital minor spliceosome diseases and the level of mRNA splicing

doctoral program at the University of Helsinki.

defect follow a counterintuitive association as described above.

The two diseases showing only relatively mild intron retention References

defects (MOPD1/TALS and RFMN) show pleiotropic, and often

severe, defects in many tissues [30–32]. In contrast, IGHD and EOCA [1] N. Lopez-Bigas, B. Audit, C. Ouzounis, G. Parra, R. Guigo, Are splicing

show large splicing defects that were detected presumably ubiq- mutations the most frequent cause of hereditary disease? FEBS Lett. 579 (9)

(2005) 1900–1903.

uitously − also from cells/tissues not associated with the disease

[2] K.H. Lim, L. Ferraris, M.E. Filloux, B.J. Raphael, W.G. Fairbrother, Using

[27,28]. Even though the present small dataset does not allow gen-

positional distribution to identify splicing elements and predict pre-mRNA

eralization, it is tempting to speculate that the difference may processing defects in human genes, Proc. Natl. Acad. Sci. U. S. A. 108 (27)

(2011) 11093–11098.

be linked to the fate of the unspliced mRNAs. Specifically, it is

[3] Y.I. Li, B. van de Geijn, A. Raj, D.A. Knowles, A.A. Petti, D. Golan, Y. Gilad, J.K.

possible that the unspliced U12-type introns or transcripts con-

Pritchard, RNA splicing is a primary link between genetic variation and

taining cryptic splicing events are better tolerated when exported disease, Science 352 (6285) (2016) 600–604.

[4] F. Supek, B. Minana,˜ J. Valcárcel, T. Gabaldón, B. Lehner, Synonymous

to cytoplasm where they can be targeted by the cytoplasmic qual-

mutations frequently act as driver mutations in human cancers, Cell 156 (6)

ity control mechanisms. Transcripts trapped in the nucleus may,

(2014) 1324–1335.

in contrast, accumulate in some tissues or lead to other detrimen- [5] G.-S. Wang, T.A. Cooper, Splicing in disease: disruption of the splicing code

and the decoding machinery, Nat. Rev. Genet. 8 (10) (2007) 749–761.

tal downstream effects. This hypothesis receives tentative support

[6] H.Y. Xiong, B. Alipanahi, L.J. Lee, H. Bretschneider, D. Merico, R.K.C. Yuen, Y.

from a zebrafish study where a specific 65K mutation arrests the

Hua, S. Gueroussov, H.S. Najafabadi, T.R. Hughes, Q. Morris, Y. Barash, A.R.

spliceosome assembly at a late stage and presumably traps such Krainer, N. Jojic, S.W. Scherer, B.J. Blencowe, B.J. Frey, The human splicing

code reveals new insights into the genetic determinants of disease, Science

mRNAs in the nucleus. Unlike the more restricted phenotype seen

347 (6218) (2015).

with IGHD 65K mutations, the zebrafish phenotype is severe, with

[7] R. Soemedi, K.J. Cygan, C.L. Rhine, J. Wang, C. Bulacan, J. Yang, P.

developmental defects in multiple organs [37]. Bayrak-Toydemir, J. McDonald, W.G. Fairbrother, Pathogenic variants that

alter protein code often disrupt splicing, Nat. Genet. 49 (6) (2017) 848–855.

[8] S.ˇ Ru˚ ziˇ cková,ˇ D. Stanek,ˇ Mutations in spliceosomal proteins and retina

4.3. Defects with individual genes vs a global splicing defect

degeneration, RNA Biol. 14 (5) (2017) 544–552.

[9] D. Inoue, R.K. Bradley, O. Abdel-Wahab, Spliceosomal gene mutations in

One of the outstanding questions is whether the pathologi- myelodysplasia: molecular links to clonal abnormalities of hematopoiesis,

Genes Dev. 30 (9) (2016) 989–1001.

cal consequences of the minor spliceosome diseases are caused

[10] Z. Zhang, F. Lotti, K. Dittmar, I. Younis, L. Wan, M. Kasim, G. Dreyfuss, SMN

by a global defect in the splicing of U12-type introns or whether

deficiency causes tissue-specific perturbations in the repertoire of snRNAs

the various effects seen in different tissues are caused by mis- and widespread defects in splicing, Cell 133 (4) (2008) 585–600.

[11] J.J. Turunen, E.H. Niemelä, B. Verma, M.J. Frilander, The significant other:

splicing of a small subset of genes containing U12-type introns.

splicing by the minor spliceosome, Wiley Interdiscip. Rev. RNA 4 (2013)

Gene expression analyses have suggested various candidate genes 61–76.

responsible for the observed disease phenotypes. These must, how- [12] C.B. Burge, R.A. Padgett, P.A. Sharp, Evolutionary fates and origins of U12-type

introns, Mol. Cell 2 (1998) 773–785.

ever, be interpreted with caution since in all cases except MDS, the

[13] Q. Wu, A.R. Krainer, AT-AC pre-mRNA splicing mechanism and conservation

gene expression analyses have been done using cells other than the

of minor introns in voltage-gated ion channel genes, Mol. Cell Biol. 19 (1999)

actual affected tissue. 3225–3236.

[14] G.W. Yeo, E.L. Van Nostrand, T.Y. Liang, Discovery and analysis of

Furthermore, it is not yet possible to conclude whether a par-

evolutionarily conserved intronic splicing regulatory elements, PLoS Genet. 3

ticular tissue or cell type would be more susceptible to defects in

(5) (2007) e85.

the U12-dependent spliceosome. Of the present congenital minor [15] M.K. Basu, W. Makalowski, I.B. Rogozin, E.V. Koonin, U12 intron positions are

spliceosome diseases, the only common target appears to be the more strongly conserved between animals and plants than U2 intron

positions, Biol. Direct. 3 (2008) 19.

nervous system. Specifically, microcephaly has been reported for

[16] C.L. Will, R. Lührmann, Splicing of a rare class of introns by the

three of the diseases (severe for MOPD1/TALS, mild for RFMN

U12-dependent spliceosome, Biol. Chem. 386 (8) (2005) 713–724.

and IGHD), which with MOPD1/TALS is associated with struc- [17] C. Schneider, C.L. Will, O.V. Makarova, E.M. Makarov, R. Lührmann, Human

U4/U6.U5 and U4atac/U6atac.U5 tri-snRNPs exhibit similar protein

tural abnormalities of brain. Additionally, RFMN and EOCA patients

compositions, Mol. Cell Biol. 22 (10) (2002) 3219–3229.

display cognitive delay and cerebellar ataxia, respectively. The

[18] M.J. Frilander, J.A. Steitz, Initial recognition of U12-dependent introns

observed tissue-specific effects may be linked to the associations of requires both U11/5’ splice-site and U12/branchpoint interactions, Genes

Dev. 13 (1999) 851–863.

the minor spliceosome activity with cell proliferation [37,74] or cel-

[19] H. Shen, X. Zheng, S. Luecke, M.R. Green, The U2AF35-related protein Urp

lular response to stress [75]. Consistently, recent work has reported

contacts the 3’ splice site to promote U12-type intron splicing and the second

that components of the U11/U12 di-snRNP, including the 65K, 48K step of U2-type intron splicing, Genes Dev. 24 (21) (2010) 2389–2394.

[20] W.-Y. Tarn, J.A. Steitz, Highly diverged U4 and U6 small nuclear RNAs required

and Urp/ZRSR2 proteins are downregulated during neuron terminal

for splicing rare AT-AC introns, Science 273 (1996) 1824–1832.

differentiation [70].

[21] W.-Y. Tarn, J.A. Steitz, A novel spliceosome containing U11, U12 and U5

snRNPs excises a minor class (AT-AC) intron in vitro, Cell 84 (1996) 801–811.

[22] M.J. Frilander, J.A. Steitz, Dynamic exchanges of RNA interactions leading to

5. Future perspective

catalytic core formation in the U12-dependent spliceosome, Mol. Cell 7 (1)

(2001) 217–226.

The recent discoveries of human diseases affecting the minor [23] G.C. Shukla, R.A. Padgett, Conservation of functional features of U6atac and

U12 snRNAs between vertebrates and higher plants, RNA 5 (1999) 525–538.

spliceosome have significantly advanced our understanding of the

Please cite this article in press as: B. Verma, et al., Minor spliceosome and disease, Semin Cell Dev Biol (2017), https://doi.org/10.1016/j.semcdb.2017.09.036

G Model

YSCDB-2404; No. of Pages 10 ARTICLE IN PRESS

B. Verma et al. / Seminars in Cell & Developmental Biology xxx (2017) xxx–xxx 9

[24] M.M. Scotti, M.S. Swanson, RNA mis-splicing in disease, Nat. Rev. Genet. 17 (1) [47] K. Nottrott, P. Hartmuth, H. Fabrizio, I. Urlaub, R. Vidovic, R. Ficner, Functional

(2016) 19–32. interaction of a novel 15.5kD [U4/U6 U5] tri-snRNP protein with the 5’

[25] C.L. Will, C. Schneider, M. Hossbach, H. Urlaub, R. Rauhut, S. Elbashir, T. Tuschl, stem-loop of U4 snRNA, EMBO J. 18 (21) (1999) 6119–6133.

R. Lührmann, The human 18S U11/U12 snRNP contains a set of novel proteins [48] J.W. Hardin, C. Warnasooriya, Y. Kondo, K. Nagai, D. Rueda, Assembly and

not found in the U2-dependent spliceosome, RNA 10 (6) (2004) 929–941. dynamics of the U4/U6 di-snRNP by single-molecule FRET, Nucleic Acids Res.

[26] C.L. Will, C. Schneider, R. Reed, R. Lührmann, Identification of both shared and 43 (22) (2015) 10963–10974.

distinct proteins in the major and minor spliceosomes, Science 284 (1999) [49] M.L. Hastings, N. Resta, D. Traum, A. Stella, G. Guanti, A.R. Krainer, An LKB1

2003–2005. AT-AC intron mutation causes Peutz-Jeghers syndrome via splicing at

[27] J. Argente, R. Flores, A. Gutiérrez-Arumí, B. Verma, G.A. Martos-Moreno, I. noncanonical cryptic splice sites, Nat. Struct. Mol. Biol. 12 (1) (2005) 54–59.

Cuscó, A. Oghabian, J.A. Chowen, M.J. Frilander, L.A. Pérez-Jurado, Defective [50] M.A. Shaw, N. Brunetti-Pierri, L. Kádasi, V. Kovácová, L. Van Maldergem, D. De

minor spliceosome mRNA processing results in isolated familial growth Brasi, M. Salerno, J. Gécz, Identification of three novel SEDL mutations,

hormone deficiency, EMBO Mol. Med. 6 (2014) 299–306. including mutation in the rare, non-canonical splice site of exon 4, Clin.

[28] M.F. Elsaid, N. Chalhoub, T. Ben-Omran, P. Kumar, H. Kamel, K. Ibrahim, Y. Genet. 64 (3) (2003) 235–242.

Mohamoud, E. Al-Dous, I. Al-Azwani, J.A. Malek, K. Suhre, M.E. Ross, A.A. [51] D.C. Kohrman, J.B. Harris, M.H. Meisler, Mutation detection in the med and

Aleem, Mutation in noncoding RNA RNU12 causes early onset cerebellar medJ alleles of the sodium channel scn8a: unusual splicing due to a minor

ataxia, Ann. Neurol. 81 (1) (2017) 68–78. class AT-AC intron, J. Biol. Chem. 271 (29) (1996) 17576–17581.

[29] V. Madan, D. Kanojia, J. Li, R. Okamoto, A. Sato-Otsubo, A. Kohlmann, M. [52] A. Kurtovic-Kozaric, B. Przychodzen, J. Singh, M.M. Konarska, M.J. Clemente,

Sanada, V. Grossmann, J. Sundaresan, Y. Shiraishi, S. Miyano, F. Thol, A. Z.K. Otrock, M. Nakashima, E.D. Hsi, K. Yoshida, Y. Shiraishi, K. Chiba, H.

Ganser, H. Yang, T. Haferlach, S. Ogawa, H.P. Koeffler, Aberrant splicing of Tanaka, S. Miyano, S. Ogawa, J. Boultwood, H. Makishima, J.P. Maciejewski,

U12-type introns is the hallmark of ZRSR2 mutant myelodysplastic syndrome, R.A. Padgett, PRPF8 defects cause missplicing in myeloid malignancies,

Nat. Commun. 6 (2015) 6042. Leukemia 29 (1) (2015) 126–136.

[30] D. Merico, M. Roifman, U. Braunschweig, R.K.C. Yuen, R. Alexandrova, A. Bates, [53] M.L. Hastings, A.R. Krainer, Functions of SR proteins in the U12-dependent

B. Reid, T. Nalpathamkalam, Z. Wang, B. Thiruvahindrapuram, P. Gray, A. AT-AC pre-mRNA splicing pathway, RNA 7 (3) (2001) 471–482.

Kakakios, J. Peake, S. Hogarth, D. Manson, R. Buncic, S.L. Pereira, J.-A. Herbrick, [54] D.K. Li, S. Tisdale, F. Lotti, L. Pellizzoni, SMN control of RNP assembly: from

B.J. Blencowe, C.M. Roifman, S.W. Scherer, Compound heterozygous post-transcriptional gene regulation to motor neuron disease, Semin. Cell

mutations in the noncoding RNU4ATAC cause Roifman Syndrome by Dev. Biol. 32 (2014) 22–29.

disrupting minor intron splicing, Nat. Commun. 6 (2015) 8718. [55] H.R. Luo, G.A. Moreau, N. Levin, M.J. Moore, The human Prp8 protein is a

[31] H. He, S. Liyanarachchi, K. Akagi, R. Nagy, J. Li, R.C. Dietrich, W. Li, N. Sebastian, component of both U2- and U12-dependent spliceosomes, RNA 5 (1999)

B. Wen, B. Xin, J. Singh, P. Yan, H. Alder, E. Haan, D. Wieczorek, B. Albrecht, E. 893–908.

Puffenberger, H. Wang, J.A. Westman, R.A. Padgett, D.E. Symer, A. de la [56] M.-C. Keightley, M.O. Crowhurst, J.E. Layton, T. Beilharz, S. Markmiller, S.

Chapelle, Mutations in U4atac snRNA, a component of the minor spliceosome, Varma, B.M. Hogan, T.A. de Jong-Curtain, J.K. Heath, G.J. Lieschke, In vivo

in the developmental disorder MOPD I, Science 332 (6026) (2011) 238–240. mutation of pre-mRNA processing factor 8 (Prpf8) affects transcript splicing,

[32] P. Edery, C. Marcaillou, M. Sahbatou, A. Labalme, J. Chastang, R. Touraine, E. cell survival and myeloid differentiation, FEBS Lett. 587 (14) (2013)

Tubacher, F. Senni, M.B. Bober, S. Nampoothiri, P.-S. Jouk, E. Steichen, S. 2150–2157.

Berland, A. Toutain, C.A. Wise, D. Sanlaville, F. Rousseau, F. Clerget-Darpoux, [57] M. Jangi, C. Fleet, P. Cullen, S.V. Gupta, S. Mekhoubad, E. Chiao, N. Allaire, C.F.

A.-L. Leutenegger, Association of TALS developmental disorder with defect in Bennett, F. Rigo, A.R. Krainer, J.A. Hurt, J.P. Carulli, J.F. Staropoli, SMN

minor splicing component U4atac snRNA, Science 332 (6026) (2011) 240–243. deficiency in severe models of spinal muscular atrophy causes widespread

[33] H. Benecke, R. Lührmann, C.L. Will, The U11/U12 snRNP 65K protein acts as a intron retention and DNA damage, Proc. Natl. Acad. Sci. U. S. A. 114 (12)

molecular bridge, binding the U12 snRNA and U11-59K protein, EMBO J. 24 (2017) E2347–E2356.

(17) (2005) 3057–3069. [58] F. Gabanella, M.E.R. Butchbach, L. Saieva, C. Carissimi, A.H.M. Burghes, L.

[34] M. Guceva, V. Polenakovicb, Y. Tasica, J. LeBoucc, J. Klammtd, A. Pfaeffled, Pellizzoni, Ribonucleoprotein assembly defects correlate with spinal

Severe isolated growth hormone deficiency and myopathy in two brothers muscular atrophy severity and preferentially affect a subset of spliceosomal

with RNPC3 mutation, Horm. Res. Paediatr. 84 (Suppl. 1) (2015) 447. snRNPs, PLoS One 2 (9) (2007) e921.

[35] K.S. Alatzoglou, M.T. Dattani, Genetic causes and treatment of isolated growth [59] N. Boulisfane, M. Choleza, F. Rage, H. Neel, J. Soret, R. Bordonné, Impaired

hormone deficiency—an update, Nat. Rev. Endocrinol. 6 (10) (2010) 562–576. minor tri-snRNP assembly generates differential splicing defects of U12-type

[36] A.J. Norppa, T. Kauppala, H.A. Heikkinen, B. Verma, H. Iwai, M.J. Frilander, introns in lymphoblasts derived from a type I SMA patient, Hum. Mol. Genet.

Mutations in the U11/U12-65 K protein associated with isolated growth 20 (4) (2011) 641–648.

hormone deficiency lead to structural destabilization and impaired binding of [60] F. Lotti, Wendy L. Imlach, L. Saieva, Erin S. Beck, Le T. Hao, Darrick K. Li, W.

U12 snRNA, RNA, in press. Jiao, George Z. Mentis, Christine E. Beattie, Brian D. McCabe, L. Pellizzoni, An

[37] S. Markmiller, N. Cloonan, R.M. Lardelli, K. Doggett, M.-C. Keightley, Y. Boglev, SMN-dependent U12 splicing event essential for motor circuit function, Cell

A.J. Trotter, A.Y. Ng, S.J. Wilkins, H. Verkade, E.A. Ober, H.A. Field, S.M. 151 (2) (2012) 440–454.

Grimmond, G.J. Lieschke, D.Y.R. Stainier, J.K. Heath, Minor class splicing [61] T.K. Doktor, Y. Hua, H.S. Andersen, S. Brøner, Y.H. Liu, A. Wieckowska, M.

shapes the zebrafish transcriptome during development, Proc. Natl. Acad. Sci. Dembic, G.H. Bruun, A.R. Krainer, B.S. Andresen, RNA-sequencing of a

U. S. A. 111 (2014) 3062–3067. mouse-model of spinal muscular atrophy reveals tissue-wide changes in

[38] J. Singh, K. Sikand, H. Conrad, C.L. Will, A.A. Komar, G.C. Shukla, U6atac snRNA splicing of U12-dependent introns, Nucleic Acids Res. 45 (2016) 395–416.

stem-loop interacts with U12 p65 RNA binding protein and is functionally [62] E.L. Garcia, Z. Lu, M.P. Meers, K. Praveen, A.G. Matera, Developmental arrest of

interchangeable with the U12 apical stem-loop III, Sci. Rep. 6 (2016) 31393. Drosophila survival motor neuron (Smn) mutants accounts for differences in

[39] B. Saez, M.J. Walter, T.A. Graubert, Splicing factor gene mutations in expression of minor intron-containing genes, RNA 19 (11) (2013) 1510–1516.

hematologic malignancies, Blood 129 (10) (2017) 1260–1269. [63] T. Ishihara, Y. Ariizumi, A. Shiga, T. Kato, C.-F. Tan, T. Sato, Y. Miki, M. Yokoo, T.

[40] S.A. Mian, A.E. Smith, A.G. Kulasekararaj, A. Kizilors, A.M. Mohamedali, N.C. Fujino, A. Koyama, A. Yokoseki, M. Nishizawa, A. Kakita, H. Takahashi, O.

Lea, K. Mitsopoulos, K. Ford, E. Nasser, T. Seidl, G.J. Mufti, Spliceosome Onodera, Decreased number of Gemini of coiled bodies and U12 snRNA level

mutations exhibit specific associations with epigenetic modifiers and in amyotrophic lateral sclerosis, Hum. Mol. Genet. 22 (20) (2013) 4136–4147.

proto-oncogenes mutated in myelodysplastic syndrome, Haematologica 98 [64] S. Reber, J. Stettler, G. Filosa, M. Colombo, D. Jutzi, S.C. Lenzken, C.

(7) (2013) 1058–1066. Schweingruber, R. Bruggmann, A. Bachi, S.M. Barabino, O. Mühlemann, M.D.

[41] C.M. Gault, F. Martin, W. Mei, F. Bai, J.B. Black, W.B. Barbazuk, A.M. Settles, Ruepp, Minor intron splicing is regulated by FUS and affected by

Aberrant splicing in maize rough endosperm3 reveals a conserved role for ALS-associated FUS mutants, EMBO J. 35 (14) (2016) 1504–1521.

U12 splicing in eukaryotic multicellular development, Proc. Natl. Acad. Sci. U. [65] G.J. Xu, A.A. Shah, M.Z. Li, Q. Xu, A. Rosen, L. Casciola-Rosen, S.J. Elledge,

S. A. 114 (11) (2017) E2195–E2204. Systematic autoantigen analysis identifies a distinct subtype of scleroderma

[42] H.K.J. Pessa, M.J. Frilander, Minor splicing, disrupted, Science 332 (6026) with coincident cancer, Proc. Natl. Acad. Sci. U. S. A. 113 (47) (2016)

(2011) 184–185. E7526–E7534.

[43] G.M.H. Abdel-Salam, M.S. Abdel-Hamid, M. Issa, A. Magdy, A. El-Kotoury, K. [66] G. Shah, A. Xu, L.K. Rosen, F.M. Hummers, S.J. Wigley, L. Elledge, Anti–RNPC-3

Amr, Expanding the phenotypic and mutational spectrum in microcephalic antibodies as a marker of cancer-associated scleroderma, Arthritis

osteodysplastic primordial dwarfism type I, Am. J. Med. Genet. 158A (6) Rheumatol. 69 (6) (2017) 1306–1312.

(2012) 1455–1461. [67] D. Fischer, T. Wahlfors, H. Mattila, H. Oja, T.L.J. Tammela, J. Schleutker, MiRNA

[44] A.B. Krøigård, A.P. Jackson, L.S. Bicknell, E. Baple, K. Brusgaard, L.K. Hansen, profiles in lymphoblastoid cell lines of finnish prostate cancer families, PLoS

L.B. Ousager, Two novel mutations in RNU4ATAC in two siblings with an One 10 (5) (2015) e0127427.

atypical mild phenotype of microcephalic osteodysplastic primordial [68] Y. Tian, Z. Huang, Z. Wang, C. Yin, L. Zhou, L. Zhang, K. Huang, H. Zhou, X. Jiang,

dwarfism type 1, Clin. Dysmorphol. 25 (2) (2016) 68–72. J. Li, L. Liao, M. Yang, F. Meng, Identification of novel molecular markers for

[45] O.V. Makarova, E.M. Makarov, S. Liu, H.-P. Vornlocher, R. Lührmann, Protein prognosis estimation of acute myeloid leukemia: over-expression of PDCD7,

61K, encoded by a gene (PRPF31) linked to autosomal dominant retinitis FIS1 and ang2 may indicate poor prognosis in pretreatment patients with

pigmentosa, is required for U4/U6·U5 tri-snRNP formation and pre-mRNA acute myeloid leukemia, PLoS One 9 (1) (2014) e84150.

splicing, EMBO J. 21 (5) (2002) 1148–1157. [69] E.H. Niemelä, J. Verbeeren, P. Singha, V. Nurmi, M.J. Frilander, Evolutionarily

[46] F. Jafarifar, R.C. Dietrich, J.M. Hiznay, R.A. Padgett, Biochemical defects in conserved exon definition interactions with U11 snRNP mediate alternative

minor spliceosome function in the developmental disorder MOPD I, RNA 20 splicing regulation on U11-48K and U11/U12-65K genes, RNA Biol. 12 (2015)

(7) (2014) 1078–1089. 1256–1264.

Please cite this article in press as: B. Verma, et al., Minor spliceosome and disease, Semin Cell Dev Biol (2017), https://doi.org/10.1016/j.semcdb.2017.09.036

G Model

YSCDB-2404; No. of Pages 10 ARTICLE IN PRESS

10 B. Verma et al. / Seminars in Cell & Developmental Biology xxx (2017) xxx–xxx

[70] J. Verbeeren, B. Verma, E.H. Niemelä, K. Yap, E.V. Makeyev, M.J. Frilander, [74] H. König, N. Matter, R. Bader, W. Thiele, F. Müller, Splicing segregation: the

Alternative exon definition events control the choice between nuclear minor spliceosome acts outside the nucleus and controls cell proliferation,

retention and cytoplasmic export of U11/U12-65K mRNA, PLoS Genet. 13 (5) Cell 131 (4) (2007) 718–729.

(2017) e1006824. [75] I. Younis, K. Dittmar, W. Wang, S.W. Foley, M.G. Berg, K.Y. Hu, Z. Wei, L. Wan,

[71] M.K. Doma, R. Parker, RNA quality control in eukaryotes, Cell 131 (4) (2007) G. Dreyfuss, Minor introns are embedded molecular switches regulated by

660–668. highly unstable U6atac snRNA, eLife 2 (2013) e00780.

[72] E.H. Niemelä, A. Oghabian, R.H.J. Staals, G.J.M. Pruijn, M.J. Frilander, Global [76] S.A. Forbes, D. Beare, H. Boutselakis, S. Bamford, N. Bindal, J. Tate, C.G. Cole, S.

analysis of the nuclear processing of unspliced U12-type introns by the Ward, E. Dawson, L. Ponting, R. Stefancsik, B. Harsha, C.Y. Kok, M. Jia, H. Jubb,

exosome, Nucleic Acids Res. 42 (2014) 7358–7369. Z. Sondka, S. Thompson, T. De, P.J. Campbell, COSMIC: somatic cancer genetics

[73] J.J. Turunen, C.L. Will, M. Grote, R. Lührmann, M.J. Frilander, The U11-48K at high-resolution, Nucleic Acids Res. 45 (D1) (2017) D777–D783.

protein contacts the 5’ splice site of U12-type introns and the U11-59K

protein, Mol. Cell Biol. 28 (10) (2008) 3548–3560.

Please cite this article in press as: B. Verma, et al., Minor spliceosome and disease, Semin Cell Dev Biol (2017), https://doi.org/10.1016/j.semcdb.2017.09.036