You are on page 1of 19

Advanced Review

SMN in spinal muscular atrophy


and snRNP biogenesis
Tristan H. Coady† and Christian L. Lorson∗

Ribonucleoprotein (RNP) complexes function in nearly every facet of cellular


activity. The spliceosome is an essential RNP that accurately identifies introns
and catalytically removes the intervening sequences, providing exquisite control
of spatial, temporal, and developmental gene expressions. U-snRNPs are the
building blocks for the spliceosome. A significant amount of insight into the
molecular assembly of these essential particles has recently come from a seemingly
unexpected area of research: neurodegeneration. Survival motor neuron (SMN)
performs an essential role in the maturation of snRNPs, while the homozygous
loss of SMN1 results in the development of spinal muscular atrophy (SMA), a
devastating neurodegenerative disease. In this review, the function of SMN is
examined within the context of snRNP biogenesis and evidence is examined which
suggests that the SMN functional defects in snRNP biogenesis may account for the
motor neuron pathology observed in SMA.  2011 John Wiley & Sons, Ltd. WIREs RNA
2011 2 546–564 DOI: 10.1002/wrna.76

SURVIVAL MOTOR NEURON regulation, however, they are not active sites of pre-
mRNA processing.11 SMN is also diffusely present in
S urvival motor neuron (SMN) is a 38 kDa
protein, encoded by nine exons (Figure 1), that is
ubiquitously expressed throughout development and
the cytoplasm where it performs its most thoroughly
described biochemical function: snRNP assembly and
maturation.12,13 SMNs role in snRNP biogenesis and
has been identified as a protein-binding partner for its relation to SMA will be the focus of this review.
nearly 50 cellular and viral proteins.1,2 While the
SMN expression can be detected in all tissues, levels
are not equivalent.3 SMN has been implicated in a SMN AND SPINAL MUSCULAR
variety of cellular activities, including transcription,
ATROPHY
RNA stability, apoptosis, pre-mRNA splicing, stress
responses, axonal RNA trafficking, and snRNP Spinal muscular atrophy (SMA) is a leading genetic
biogenesis.4 In the early analysis of the SMN protein, it cause of infantile death worldwide and is the second
was identified in punctate nuclear structures originally most common autosomal recessive disorder.14,15 SMA
referred to as Gemini-of-coiled-bodies or gems5,6 is a neurodegenerative disease that occurs in approx-
(Figure 2). These nuclear structures are distinct from imately 1:6000 live births and has a carrier frequency
splicing speckles and other nuclear structures such between 1:30 and 1:40.16,17 The overwhelming major-
as PML bodies, however, gems—as the Gemini- ity of the clinical manifestations in SMA are due to
derived moniker suggests—are frequently immediately the progressive loss of α motor neurons within the
adjacent to or overlapping Cajal bodies.5–10 Gems are anterior horns of the spinal cord. Loss of the motor
enriched in a variety of cellular factors, including neurons and disruption of the motor unit results in
those involved in RNA processing and transcription the atrophy of proximal voluntary muscles, eventually
progressing to the point where the trunk muscles are

Current address: Department of Biological Sciences, Columbia compromised. Respiratory complications are a major
University, New York, NY, USA cause of mortality. There are four clinical forms of
∗ Correspondence to: lorsonc@missouri.edu the disease, based upon the age of disease onset,
Department of Veterinary Pathobiology, Bond Life Sciences Center, the severity of the symptoms, and the achievement
University of Missouri, Columbia, MO, USA of defined physical milestones.18–21 At the molecular
DOI: 10.1002/wrna.76 level, all SMA patients lack SMN1; however, the copy

546  2011 Jo h n Wiley & So n s, L td. Vo lu me 2, Ju ly/Au gu s t 2011


WIREs RNA SMN in spinal muscular atrophy and snRNP biogenesis

Cytoplasmic
localization/
Nucleic acid Tudor domain Y/G stability
binding Sm-fold Poly-proline box

AA 28 52 92 159 210 242 279 294

Exon 1 2a 2b 3 4 5 6 7

Oligomerization Sm core, Profilin Oligomerization


Coilin
Gemin2 fibrillarin
GAR-1

FIGURE 1 | Survival motor neuron (SMN) protein subdomains. The amino acid number corresponding to the exon peptides and the size of the
SMN protein is indicated above the red exon boxes. The SMN is 294 amino acids and runs at 38 kDa on Western blots. The relative sizes of the exons
are diagrammed with the exon number below. Identified protein subdomains and associated functions are indicated. Below is an abbreviated list of
proteins that interact with the SMN along with the approximate region of interaction.

can never stand without assistance. Typically, the dis-


ease presents after 2 years of age and patients have
a significantly longer life span compared to Type I
patients.21,23,24 Type III SMA or Kugelberg–Welander
disease, is often referred to as the juvenile form of SMA
because symptoms typically develop later in life and
all patients can walk unassisted at some point during
their lives.24 Life span is normal for Type III individu-
als. Type IV SMA, often referred to as the adult form,
is the least severe form and onset typically occurs in
patients older than 30 years.2 Although these defini-
tions are useful, SMA is an extremely heterogeneous
disease. There are many cases that do not fall cleanly
into one category; conversely, there is additional strat-
ification within these classifications, used for clinical
management as well as clinical trials development.
FIGURE 2 | Intracellular localization of the survival motor neuron
Full-length SMN is actually produced by two
(SMN). An anti-SMN monoclonal antibody (4B7) was used to
specifically detect SMN (green) in the HeLa cells, while DAPI stained the
nearly identical genes, SMN1 and SMN2.1 These
nucleus. The SMN is present within discrete nuclear foci, often referred genes are located on the long arm of chromo-
to as gems. These gems have been used as a surrogate marker for the some 5 and are present within two relatively large
overall SMN concentration as the gem numbers frequently correspond genomic repeats consisting of approximately 500 kb
to the total intracellular SMN levels. (Figure 4). Several additional genes are present within
the repeats, although they are not involved in the
number of a nearly identical copy gene called SMN2 SMA development.1,25 The repeats lie in a head-to-
is a critical disease modifier as less severe forms of head orientation and are regulated by nearly identical
the disease are associated with increased SMN2 gene promoter elements. SMN1 expresses high levels of
copies (Figure 3). Type I SMA or Werdnig–Hoffmann full-length SMN, while SMN2 produces low levels of
disease, is the most severe form and is by far the most full-length SMN transcript and an abundance of an
common, comprising nearly 60% of all newly diag- alternatively spliced isoform that lacks the final coding
nosed cases. While the life expectancy has improved exon, Exon 7. Although RT-PCR can detect the pres-
for SMA I patients in the developed world, it is due in ence of other alternatively spliced isoforms lacking
large part to supportive care. In the absence of aggres- Exons 5, 7 or both 5 and 7,26 these are only produced
sive intervention, life expectancy is less than 2 years.22 at very low levels and it is clear that the critical differ-
Type II SMA is less severe, where patients can sit, but ence between SMN1 and SMN2 is in the processing

Vo lu me 2, Ju ly/Au gu s t 2011  2011 Jo h n Wiley & So n s, L td. 547


Advanced Review wires.wiley.com/rna

Unaffected Embryonic lethal Type I SMA

SMN2 SMN1 SMN2 SMN1 SMN2 SMN1

SMN2 SMN1 SMN2 SMN1 SMN2 SMN1

Type II SMA Type III SMA Unaffected carrier

SMN2 SMN1 SMN2 SMN2 SMN2 SMN2

SMN2 SMN2 SMN2 SMN2 SMN2 SMN2 SMN2

FIGURE 3 | SMN2 is a critical modifier of disease severity. The genomic organization of a typical unaffected individual contains two SMN1 and
two SMN2 genes, one on each chromosome (5q). No individuals have been identified that lacked both SMN1 and SMN2, presumably because this
would result in early embryonic lethality. As the SMN2 gene copy number increases in the SMN1-null spinal muscular atrophy (SMA) patients, the
disease severity tends to decrease. This is likely due to the low, residual level of full-length SMN produced by each SMN2, approximately 10% per
SMN2 copy. High SMN2 numbers have been observed in SMN1-null individuals who are phenotypically normal, although in genetic terms are defined
as having SMA.

5q11.2-13.3 its total absence.30–32 This is why no SMA patients


500 kb 500 kb
have been identified, who are null for both SMN1 and
GTF2H2T NAIPψ SMN2 SERF1C SERF1T SMN1 NAIP GTF2H2T SMN2. While humans are the only species with SMN1
and SMN2 genes,33 work performed in mice is consis-
tent with the notion that low levels, not the complete
Sp1 Ets
+1 absence, of SMN results in SMA.32,34 The knockout of
the murine SMN gene, which is functionally equivalent
to SMN1, results in pre-implantation lethality, while
Caa
g G introduction of the human SMN2 in the SMN-null
1 2a 2b 3 4 5 6 7 8 background confers viability.32 This work is con-
a gg A sistent with the majority of SMA cases in which
T
increasing SMN2 copy numbers are associated with
FIGURE 4 | Genomic organization of the survival motor neuron less severe forms of disease14 (Figure 3). It is impor-
(SMN) locus. SMN1 and SMN2 are present on a single region of the tant to stress that the truncated product produced by
same chromosome: 5q11.2-13.3. An evolutionarily recent genetic SMN2, often referred to as the SMN7 protein, is not
duplication of approximately 500 kb resulted in the duplication and a dominant-negative factor and does retain some resid-
inversion of the SMN and several flanking genes, including SERF1,
ual activity compared to the full-length SMN.35–37
NAIP, and GTF2H2. Initially, genes other than SMN were implicated in
the spinal muscular atrophy (SMA) development because larger
deletions frequently excised SMN and additional sequences. However, SMN Pre-mRNA Splicing
the detection of intragenic mutations within the SMN conclusively
While SMN1 and SMN2 encode identical amino acid
demonstrated that SMN was the only SMA-determining gene. SMN1
and SMN2 are nearly identical and the regulatory mechanisms
sequences, the majority of SMN2-derived transcripts
governing their promoters are essentially unchanged (Sp1, Ets sites, and are alternatively spliced and lack Exon 7, encoding the
many more). Methylation and histone regulation appear critical for the SMN7 protein1,26 (Figure 5). This protein is unable
SMN expression. Five silent, nonpolymorphic nucleotide differences are to form complexes with most of the typical SMN-
indicated at the 3 end of SMN1 (SMN1-derived sequences are on the associated protein and RNAs and thus is unstable and
top; SMN2 sequences are at the bottom). The critical difference is within is rapidly degraded.38,39 A number of recent reviews
Exon 7 (a C–T transition). In the full-length transcript, Exon 8 is entirely have focused upon the dynamic interplay between
noncoding, while in the SMN7 transcript, four additional amino acids the complex regulatory factors governing SMN Exon
are derived from the 5 end of Exon 8. 7 splicing; therefore, only a brief description of this
regulatory aspect will be included.40–43
of SMN Exon 7.27,28 Interestingly, only the loss of Pre-mRNA splicing of SMN Exon 7 is regulated
SMN1 results in SMA development whereas the loss of by a variety of cis- and trans-acting factors.2,42,44,45
SMN2 has no clinical consequence.29 It is important to Some elements are essential for Exon 7’s inclusion
stress that complete loss of SMN is a lethal condition for either SMN1 or SMN2, such as the hTraβ1-
and that SMA is caused by low levels of SMN—not binding site located within the central region of the 54

548  2011 Jo h n Wiley & So n s, L td. Vo lu me 2, Ju ly/Au gu s t 2011


WIREs RNA SMN in spinal muscular atrophy and snRNP biogenesis

SMN1
hnRNP-G
U2 SRp30c
snRNP
hnRNP-Q
U2AF35 SF2/ASF hTra2-β1 70K
U2AF65 U1 snRNP
Exon 6 CAGACAA Exon 8

SE1 SE2 SE3 Element 2

100% 100%
SMN exon 7 region

U2
SMN2 snRNP
U2AF35
U2AF65 70K
U1 snRNP
Sam68

hnRNP-A1
PTB/FUSE hnRNP-A1 hnRNP-A1 hnRNP-A1
Exon 6 TAGACAA Exon 8
G
EXTINCT ISS-N1

10% 90% 10%

FIGURE 5 | Pre-mRNA splicing regulation of survival motor neuron (SMN) genes. SMN1 exclusively produces the full-length form of the protein.
A number of cis - and trans -regulatory elements facilitate SMN Exon 7 inclusion. Positively acting factors are shown in the top panel, including various
SR, SR-like, and heterogeneous nuclear ribonucleoprotein (hnRNP) proteins (green), as well as putative splice enhancers (SE1–3) and Element 2. The
short sequence motif ‘CAGACAA’ represents a high-affinity SF2/ASF-binding motif that is bound by SF2 only in the SMN1 context. The first ‘C’
position (+6) is altered in SMN2 to a ‘T’. The general splicing machinery is indicated (blue). The bottom panel represents the regulatory mechanisms
governing SMN2 pre-mRNA splicing. The SF2/ASF-binding motif has been severely impaired by the introduction of a ‘T’ at the +6 position:
‘TAGACAA’. Additionally, this site is now bound efficiently by hnRNP-A1 and is within an additionally regulatory element referred to as EXTINCT.
Additional hnRNP-A1-binding sites have been identified within the flanking intron, including the ISS-N1 regions. Collectively, these factors prevent
the identification of the already weak splice sites flanking Exon 7 and promote high levels of exon skipping, ∼90%. A small, but important level
(10%) of SMN2 transcripts include Exon 7 and therefore encodes a fully functional SMN protein.

nucleotide exon.46 Other elements, however, appear RNP-A1 (hnRNP-A1).52 Within SMN2 pre-mRNA,
to function only within the context of one gene or hnRNP-A1 functions as a repressor of Exon 7’s
the other.47–52 Remarkably, there are only a handful inclusion, thereby promoting the exon-skipping event.
of nucleotides that differ between SMN1 and SMN2 Finally, an additional regulatory element referred to
and none change the protein coding sequence. One of as EXTINCT, likely mediated by the secondary struc-
these nonpolymorphic nucleotide differences resides tures within the 5 end of Exon 7, forms in the presence
within the SMN Exon 7 (+6) and results in a C–T of the SMN2-specific context.51 Collectively, the loss
transition. While this appears to be a relatively subtle of a positively acting factor and the novel presence
alteration, it clearly has a dramatic impact upon the of negatively acting factors results in the dramatic
retention of SMN Exon 7. The extreme phenotype reduction in full-length SMN expression.
of this nucleotide transition can likely be explained Recently, SMN2 has been viewed as an essential
based upon the disruption of several overlapping target for therapeutic development because it is
regulatory elements (Figure 5). First, SF2/ASF binds retained in all SMA patients and it has the capacity
this region in the SMN1 context and facilitates inclu- to encode a fully functional SMN protein.2,42,53,54
sion of SMN Exon 7.47 In contrast, the SMN2 RNA Therefore, strategies that modulate SMN2 Exon 7’s
is poorly recognized by SF2/ASF and therefore, a pos- inclusion and promote full-length expressions have
itively acting factor is lost. Second, the C–T transition been at the forefront of SMA translational research.
creates a novel binding site, heterogeneous nuclear A variety of modified oligonucleotides, antisense

Vo lu me 2, Ju ly/Au gu s t 2011  2011 Jo h n Wiley & So n s, L td. 549


Advanced Review wires.wiley.com/rna

sequences, and trans-splicing RNAs have been contains a Tudor domain which mimics a truncated
developed that efficiently increase Exon 7’s inclusion Smith protein fold with five β-pleated sheets and
in SMA patient fibroblasts and more recently in SMA appears to nucleate the formation of newly formed
animal models.49,55–71 As with many central nervous Smith core proteins.130–134 Exon 5 contains a polypro-
system disorders, these translational efforts now face line motif similar to the VASP protein family and is
the challenge of moving beyond cell-based assays or responsible for mediating interactions with the pro-
small animals and will need to address critical issues filin protein.98,130–136 Profilin binds ATP-actin at a 1:1
such as delivery and the therapeutic window. ratio to sequester it in an inactive globular form. SMN
protein contains an evolutionarily conserved Y/G box
which mediates self-association through Exon 6.87,137
SMN Protein Exon 2 also contributes to self-association through a
SMN is ubiquitously expressed, consistent with its separate oligomerization domain.129,138 Exon 7 con-
essential role in survival. The full-length SMN protein fers stability and contains a proposed cytoplasmic
is a 294 amino acid protein with a molecular weight localization motif ‘QNQKE’.38,93,139–143 This motif is
of 38 kDa and is expressed at high levels in the brain, sufficient to dramatically alter a heterologous nuclear
spinal cord, kidneys, and liver. Moderate expression protein and it is clear that this sequence can target
is found in cardiac and skeletal muscle. When the proteins to the cytoplasm. Refined characterization
spinal cord from an SMA type I patient was exam- of the stability and localization conferred by the
ined for SMN protein there was a 100-fold reduction SMN tail region determined that a minimal (4–6
compared to age-matched standards.72 amino acids), nonspecific sequence was sufficient to
at least partially compensate for much of the native
SMN Biochemical Properties sequence.139–142 The C-terminally truncated SMN7
Through a series of biochemical fractionation experi- mRNA stops translation at the next available termi-
ments, a tightly associated complex has been identified nation codon within Exon 8. The similarities between
that is present in all cells and tissues examined to the two isoforms are an equivalent mRNA half-life
date: the SMN/Gemin complex12,13,73–78 (Table 1). and the nuclear export of the SMN7 and full-
This multiprotein complex is nucleated by the SMN length mRNAs.144 The failure of the SMN7 protein
protein and contains a discrete set of interacting pro- to subsume those functions attributed to full-length
teins called Gemin2–Gemin8, and an additional factor SMN protein devolves primarily from its inability to
unrip.74,79–85 A detailed interaction map of these fac- oligomerize, an essential precondition for all SMN-
tors has been developed and while not all factors make associated functions. The inability to be incorporated
direct contact with the SMN, the complex is remark- into the appropriate RNP complexes presumably ren-
ably stable.86 Interestingly, while the ability of SMN ders the protein less stable.
to self-associate is linked to disease severity and it
has been presumed that SMN exists in a multimerized
state, the precise stoichiometry of SMN and Gemins SMN in snRNP Assembly
is yet to be determined.38,78,87 This macromolecular Eukaryotic messenger pre-mRNA splicing is an essen-
complex exhibits localization patterns similar to the tial step in nearly all gene expressions, impacting the
SMN protein: enriched in nuclear gems and diffusely diversity of the transcriptome, and allowing for tem-
abundant throughout the cytoplasm.5–10 The primary poral, spatial, and developmental control of various
exception to this rule would be unrip which appears to gene products. The general splicing machinery largely
be restricted to the cytoplasm.85 Additionally, in cells consists of small nuclear RNA (snRNA)/protein com-
of neuronal origin including cultured neurons or pri- plexes comprised of snRNAs, Smith core proteins, and
mary motor neuron cultures, additional SMN/RNP additional conserved protein factors. These RNPs will
complexes are present that are independent of Sm perform the catalytic reactions necessary to identify
proteins.88–93 Non-snRNP complexes are discussed in the intervening intron sequences and the ligation of
a subsequent section. the exon sequences. While SMN was initially believed
Functional and biochemical domains of SMN to have a direct role in pre-mRNA splicing,123,145 it
have been identified based upon cellular assays as well is now clear that SMN is intimately involved in the
as insight derived from intragenic SMN point muta- formation and maturation of snRNP particles and
tions from SMA patients4,125,126 (Figure 1). The SMN therefore performs an essential, yet indirect, role in
protein has an amino-terminal nucleic acid-binding pre-mRNA splicing.4,12,13,76
domain in the Exon 2a 2b region which overlaps The formation of a snRNP particle starts with
with the Gemin2 interaction regions.127–129 Exon 3 the polymerase II-mediated transcription of one of the

550  2011 Jo h n Wiley & So n s, L td. Vo lu me 2, Ju ly/Au gu s t 2011


WIREs RNA SMN in spinal muscular atrophy and snRNP biogenesis

TABLE 1 Previously Identified Survival Motor Neuron (SMN) Interacting Proteins Are Shown
SMN Interacting Factor Function Direct/Indirect Motifs References
Core SMN/Gemin components
SMN snRNP assembly Direct Tudor, Y/G box 74
Gemin2/SIP1 snRNP assembly Direct – 74
Gemin3/DDX20/DP103 snRNP assembly Direct DEXDc, ELICc 81
Gemin4/GIP1 snRNP assembly Indirect Leucine zipper 82
Gemin5/p175 snRNP assembly Direct WD40 83
Gemin6 snRNP assembly Indirect – 84
Gemin7/SIP3 snRNP assembly Indirect RG-rich 79
Gemin8 snRNP assembly Direct – 80
Unrip snRNP assembly Indirect WD40 85
snRNP assembly/maturation
Sm proteins/Sm B/B ,D1–3,E,F,G snRNP assembly Direct/indirect Sm-fold, RG-rich 74,94
Sm-like proteins/Lsm 10,11 snRNP assembly Direct/indirect Sm-fold, RG-rich 94–96
Snurportin and importin β snRNP nuclear import Direct/indirect HEAT repeat 96
TGSI (trimethylguanosine synthase1) Hypermethylation of m7 G cap Direct – 97
Actin dynamics
Profilin Actin transport Direct – 98
Plastin 3/ T-plastin Actin bundling Indirect EFh, CH 99
RNPs/RNA metabolism
hnRNP-Q RNA metabolism Direct RRM, RGG 88,100
hnRNP-R RNA metabolism Direct RRM, RGG 88,100
hnRNP-U RNA metabolism Direct SAP, SPRY, RGG 6
FMRP RNA metabolism Direct KH 101
KSRP/FBP2/ZBP2/MARTA1 RNA metabolism Direct KH, RGG 102
U1A Pre-mRNA splicing ND RRM 74
Galectin 1 and 3/LGALS1 Pre-mRNA splicing Indirect GLECT 103
RNA helicase A Transcription Direct DSRM, DEXD, HELIC, RGG 104
RNA polymerase II Transcription Direct – 104
Rpp20 tRNA/rRNA metabolism Direct – 105
Nucleolin rRNA metabolism Indirect RRM, RGG box 106
ISG20 ssRNA degradation ND EXOIII 107
TIAR Stress granules RRM 108,109
TDP43 Pre-mRNA splicing/transcription ND RRM 109
NFAR1/2 RNA metabolism Direct DZF, DSRM 110
Additional factors
p80 Coilin Cajal body recruitment Direct RGG 111
mSin3A Transcription ND PAH 112
EWS Transcription Direct RGG 113
Papilloma virus E2 Transcription Direct DNA binding 114
EBNA2 Transcription/viral replication Direct Acidic activation domain 115
MVM NS1/NS2 Transcription/viral replication Direct Nucleic acid binding 116,117
p53 Apoptosis Direct DNA binding 118
Osteoclast-stimulating factor Signal transduction Direct SH3, ANK 119

Vo lu me 2, Ju ly/Au gu s t 2011  2011 Jo h n Wiley & So n s, L td. 551


Advanced Review wires.wiley.com/rna

TABLE 1 Continued
SMN Interacting Factor Function Direct/Indirect Motifs References
USP9X (ubiquitin-specific protease 9) Deubiquitylating enzyme ND – 120
PPP4 (protein phosphatase 4) Ser/Thr protein phosphatase Indirect PP2Ac 121
FGF2 (fibroblast growth factor 2) Growth factor Direct FGF 122
HSC70 Protein chaperone ND – 75
ZPR1 Splicing/transcription Direct Zinc finger domain 123
BAT3 Apoptosis ND UBQ domain 124
UNC119 Photoreceptor synaptic protein ND GMP-PDE 124
RIF1 Transcriptional repressor ND – 124
GDF9 Growth factor ND TGFB 124
COPS6 Ubiquitin cascade ND JAB/MPN 124

The function of the cellular protein is included when known. However, the functional connections to the interacting proteins have not been determined in all
cases.

snRNAs (Figure 6). For the majority of pre-mRNA methylosome.12,94,95,160–162 The interaction between
splicing events, the snRNAs of interest are the the Sm proteins and PRMT5 and an additional factor,
uridine-rich U1, U2, U4, U5, and U6.146 Within the PRMT7, results in the symmetrical dimethylation of
nucleus, a 7-methylguanosine (m7 G) cap is added to arginine residues in SmB, SmD1, and SmD3, thereby
the nascent transcripts,147,148 which in turn, leads increasing the binding affinity between SMN and the
to the formation of a cap complex consisting of Sm proteins.163,164 Prior to binding to the snRNA, the
snRNA, PHAX, XPO1/CRM1, and cap-binding com- Sm proteins exist in four identifiable complexes: (1) a
plex (CBC).149–156 Cap formation and the processing hexameric complex consisting of Sm E, F, G dimers;
of the 3 end of the RNA are the molecular triggers (2) Sm D3 and B; (3) a Sm B complex; and (4) a Sm
for nuclear export.11 Collectively, these factors facil- D1, D2, and hexameric complex consisting of Sm E,
itate the nuclear export of the m7 G-capped snRNA F, and G dimers.158
into the cytoplasm through a heterodimeric CBC con- At this stage, the SMN/Gemin complex interacts
sisting of cap-binding protein 20 and 80 (CBP20 with the Sm proteins and mediates the formation of
and CBP80).151,152,155,157 Once in the cytoplasm,
a ring-like structure around the highly conserved Sm
the export complex dissociates in a GTP-dependent
site on the snRNA.13,75,76,164–166 The region of SMN
step.155
encoded by Exon 3 includes a Tudor domain which
Within the cytoplasm, a series of proteins
serves as a binding site for the Sm proteins.130–134
referred to as Smith core antigens or Sm protein
Remarkably, while the SMN Tudor region as well as
forms an ordered protein structure resembling a hep-
Gemins6 and 7 do not contain any primary amino
tameric ring.158 The Sm proteins common to all
snRNPs are encoded by seven genes: Sm B/B , D1, acid sequence similarities to the Sm proteins, each
D2, D3, E, F, and G.146 B and B are derived of these proteins are capable of forming structural
from the same gene but are generated through an motifs that closely resemble the Sm or Sm-like
alternative splicing mechanism.159 While Sm proteins protein folds.133,167–169 The heterodimeric structure of
can thermodynamically self-assemble in vitro, the Gemin6 and Gemin7 forms a five-stranded antiparallel
unassembled Sm proteins within the cytoplasm cannot β sheet flanked by an amino-terminal helix, and
merely assemble upon the recently imported snRNA. while not conclusively demonstrated, may facilitate
To initiate the heptameric formation of the Sm pro- the loading of individual Sm proteins into the complex
teins, the first step involves the interaction of the Sm and the eventual closure of the ring formation.169,170
proteins with the chloride conductance regulatory pro- The Sm site within the snRNA serves as a nucle-
tein (pICln).12,94,95,160–162 As these are RNA-binding ation site for the binding and formation of the hep-
proteins, this step is at least partly designed to pre- tameric Sm ring.167 In this ATP-dependent reaction,
vent spurious RNA binding by the Sm proteins. At there are interactions between the Sm proteins, SMN,
this stage, the Sm proteins retain a relatively low and the Gemin proteins.13,75,76,164–166 The Gemin5
affinity for snRNAs. pICln, however, is a compo- protein appears to provide specific guidance to the Sm
nent of a macromolecular complex referred to as proteins as it is responsible for specifically identifying
the protein arginine methyltransferase 5 (PRMT5) or target RNAs that contain the Sm-binding site.171,172

552  2011 Jo h n Wiley & So n s, L td. Vo lu me 2, Ju ly/Au gu s t 2011


WIREs RNA SMN in spinal muscular atrophy and snRNP biogenesis

Sm
proteins Cajal body
Gem
RNAPII
5′ 3′
PRMT7
UsnRNA
PRMT5 m3G Sm

Sm
methylation SMN/Gemin
complex
Sm

SMN/Gemin
complex

Snurportin

Sm
m7G Sm
Importin-β
7G Sm
m

FIGURE 6 | Overview of the survival motor neuron (SMN) functions in the assembly of mature spliceosome particles. Small nuclear RNAs
(snRNAs) are transcribed via polymerase II-dependent expression. The presence of the m7 G cap initiates binding by the nuclear export complex
consisting of PHAX, XPO1/CRM1, RAN, and both subunits of the cap-binding complex (CBC). Upon entering the cytoplasm, the nuclear export
complex dissociates. Within the cytoplasm, several complexes consisting of partially assembled, snRNA-free Sm proteins reside in the cytoplasm:
SmD1/SmD2; SmB/B /SmD3; and SmE/SmF/SmG. Prior to binding the SMN complex, PRMT5 and PRMT7 specifically methylate the C-terminal tails of
SmB, SmD1, and SmD3, thereby enhancing the affinity for the SMN complex. The SMN/Gemin/unrip complex binds the methylated Sm complex,
allowing the specific recruitment of snRNAs. The Sm complex is guided onto the Sm-binding site on the snRNA by Gemin5 at which point the
heptameric ring structure is formed. The SMN/Gemin/small nuclear ribonucleoprotein (snRNP) complex interacts with trimethylguanosine synthetase
1 (TGS1) which results in the hypermythylation of the m7 G cap and the formation of the m3 G cap. The nuclear import complex consisting of
snurportin and importin-β interacts with the m3 G cap and the SMN complex and transports the entire complex into the nucleus. Entry into the
nucleus results in the release of the import complex, the SMN complex, and the snRNP’s initial transitions through the Cajal body.

Once the Sm core has assembled, additional core complex then interacts with snurportin-1 and the
maturation steps are required prior to nuclear import. complex transitions the nuclear membrane.96
The snRNA 5 m7 G cap undergoes a series of tran- Within the nucleus, the SMN/Gemin/Sm core
sitions, first of which involves the hypermethylation complex is bound by RanGTP, triggering the release
by trimethylguanosine synthetase 1 (TGS1), result- of the U-snRNP.96,175 Snurportin-1 is quickly shuttled
ing in the formation of a 2,2,7-trimethyl-guanosine back into the cytoplasm as it becomes bound by
(TMG or m3 G) cap.173 This complex is now primed XPO1/CRM1. The U-snRNP is now able to transition
for nuclear import. The TMG cap and the assembled between the nucleoplasm and several discrete
Sm core serve as two separate nuclear localization subnuclear structures, the first of which are Cajal
signals each utilizing the importin β nuclear import bodies.176–178 While the precise role of the Cajal body
receptor.174 While nuclear localization signals are in the function or redirection of the U-snRNP remains
present within several members of the SMN/Gemin unclear, these regions accumulate the newly imported
complex, the direct involvement of these elements U-snRNPs. Importantly, however, these are not sites
has not been fully characterized. The SMN/Gemin/Sm of active transcription or RNA processing. Rather,

Vo lu me 2, Ju ly/Au gu s t 2011  2011 Jo h n Wiley & So n s, L td. 553


Advanced Review wires.wiley.com/rna

U-snRNPs are subsequently transported to additional would ensue, a vast array of genes could be expected to
subdomains that are the sites intimately associated be impacted. Recent work has demonstrated patho-
with pre-mRNA splicing, including perichromatin logically low levels of SMN results in general gene
fibrils and interchromatin granule clusters.11,178–182 expression defects.191 In one study that examined a
variety of tissues near end-stage in a severe model of
SMA, expression anomalies were detected in nearly
Connecting SMA to snRNP-Associated all tissues and across broad gene families. One poten-
Defects: Major Versus Minor Pathways tial caveat is that these animals typically exhibit a
One of the vexing questions in SMA focuses upon life span of 13–15 days and tissues were examined at
the SMN-associated function in disease development. 11 days; therefore, some of these differences may be
Two primary, although not necessarily, mutually attributable to the debilitated state of the animals.191
exclusive views of the SMN role(s) which, when Additionally, recent work by Baumer et al. revealed
lost, contribute most to SMA pathology have recently that widespread modifications in gene expression and
emerged4 : snRNP biogenesis and axonal mRNA splicing are associated with late-symptomatic stages
trafficking. The snRNP assembly activity of the of SMA and are not detectable earlier in development,
SMN/Gemin complex is clearly involved in a general suggesting that global gene expression alterations
cellular function required for survival of any cell or could be due to secondary effects rather than a SMN-
tissue type. So how could this general function account specific deficiency.192 While these results are difficult
for the motor neuron-specific pathology observed in to reconcile with motor neuron-specific defects, it is
most SMA cases? Are specific transcripts altered from worth noting that in very severe forms of SMA, both
motor neuron-specific genes? Recent work has begun human and murine, (Smn−/− ; SMN2+/+ and Smn−/− ;
to shed light on one potential molecular mechanism SMN2+/+ ; SMN7 mice), additional developmen-
that may explain the tissue specificity: the minor tal defects have been detected within the brain and
spliceosome.183–185 The overwhelming majority of cardiac tissue.193–196 Therefore, in extremely low lev-
introns are excised using a relatively abundant form of els of SMN, it may be that a threshold is breached
the spliceosomal snRNP population referred to as the that goes beyond that necessary to cause diseases in
‘major’ pathway or the U2-dependent spliceosome.183 a motor neuron-restricted manner, and results in a
In this RNP complex, U1, U2, U4/U6, and U5 snRNPs more far-reaching disease.
mediate the identification and excision of intron Similarly, snRNP assembly levels were shown
sequences. In contrast, an alternative spliceosomal to be broadly reduced in several animal models with
complex exists that is responsible for the excision additional subtle differences detected when specific
of introns in ∼600–700 genes, or less than 1% of snRNP profiles were examined.190 In spinal cord and
introns within the transcriptome.183–185 This pathway, brain extracts from severe SMA pups, many of the
referred to as the ‘minor’ pathway or U12-dependent snRNPs were reduced, however, there was a further
pathway, consists of U11, U12, U4atac/U6atac, and reduction in the U11 and U12 snRNPs.190 This sug-
U5 snRNPs. U5 snRNP is unique in that it is common gested that while there was a global reduction in
among both pathways. Unlike U2-dependent introns snRNP activity, there was an even greater preferential
which typically contain the canonical GT/AG splice reduction in the minor snRNP population.
sites, the U12-dependent introns contain a 5 AT splice Interestingly, U12 introns are not randomly
site and an AC 3 splice site.183,186 Additionally, for scattered and evenly distributed throughout the
many of the AT/AC introns, the typical branch site genome.189,197 In fact, an early analysis of the AT/AC
is less conserved than the comparable motif in U2- introns identified a cluster of voltage-gated ion chan-
dependent introns and it is believed that the 3 splice nel genes that were enriched for U12 introns.189,197
site and the branch site may function as a single entity As, these genes are associated with axonal function
rather than two separate regulatory domains. Con- and mutation, in many of these genes give rise to neu-
sistent with this, it appears that greater regulatory romuscular and neurodegenerative disorders, splicing
properties are associated with the 5 splice site in U12 defects in these genes could provide a mechanistic
introns.187–189 model explaining how splicing defects might result
In SMA animal models, SMN levels and snRNP in a neuronal pathology. Not only did these genes
assembly/activity during development are lower than contain a U12 intron, there were many instances in
that seen in control animals.3,190 Analysis of animals which multiple minor introns were contained within
with different forms of disease revealed an inverse a single gene.197 A scenario could be envisioned in
correlation between disease severity and snRNP which SMN is required for general snRNP assem-
activity.190 This work suggested that as SMN levels bly, however, as SMN levels decrease, the pool of

554  2011 Jo h n Wiley & So n s, L td. Vo lu me 2, Ju ly/Au gu s t 2011


WIREs RNA SMN in spinal muscular atrophy and snRNP biogenesis

existing minor spliceosome snRNPs would be the first (Figure 7). These factors have been shown to facil-
impacted because the concentration of these factors is itate the active transport of a subset of mRNAs,
lower than the major spliceosome snRNPs. Genes with including β-actin, KSRP/ZBP2/MARTA1,102 FBP,201
a single minor intron could be adversely impacted, hnRNP-R, and hnRNP-Q88,100,200 to the axon growth
but genes harboring multiple minor introns—such as cone allowing for localized translation.93,200,202–206
the voltage-gated ion channel genes—could be pref- This is accomplished through a direct association with
erentially misregulated. To date, splicing defects in the 3 UTR of β-actin mRNA known as the zip code
specific U12-dependent genes in SMA contexts have sequence. In cultured motor neurons derived from a
not been identified; however, this line of investigation SMA mouse model, β-actin mRNA and protein was
would provide important insight into the molecular significantly reduced in axonal growth cones.88,200,207
basis of SMA as well as provide additional targets for This molecular defect correlated with smaller growth
therapeutic development. cone size and shorter axons. Additionally, the knock-
down of SMN in a zebrafish model resulted in specific
motor neuron pathway finding defects and presynaptic
SMN and Axonal RNP Complexes neuromuscular junction (NMJ) defects.208,209 How-
While the primary focus of this review is on the role ever, studies in several mouse models have shown
of SMN in snRNP maturation, it is also instructive to that the majority of NMJs appear to develop nor-
examine the SMNs role in the formation of additional mally, albeit slower, suggesting that defects in axon
RNPs as it has been described in a variety of other outgrowth and related synaptogenesis pathways are
functional RNP complexes. Another RNP-associated unlikely to be the primary development defect in
role for SMN has proposed that may account for dis- SMA in vivo.210,211 Interestingly, the reintroduction
ease development: axonal mRNA transport.198 SMN of SMN-free snRNP particles was shown to over-
has been identified with a complex found within come the SMN-associated defects in zebrafish motor
axons that contains non Sm-related proteins, including neurons.212
hnRNPs Q and R (hnRNP-Q/R) as well as Z-DNA- Further evidence for SMN in an axonal-specific
binding protein 1/2 (ZBP1, ZBP2)88–90,92,93,136,199,200 function has been reported by the identification of

Well developed
hnRNP-Q/R NMJ
ZPB
Cell body

SMN
β-actin mRNA

Unaffected motor neuron

Poorly developed
hnRNP-Q/R NMJ
ZPB
Cell body

β-actin mRNA

SMA motor neuron

FIGURE 7 | Proposed role for the survival motor neuron (SMN) in neurons involves a novel ribonucleoprotein (RNP) complex. Within axons, a pool
of SMN exists that is not complexed with Sm proteins and the entire Gemin complex. Additional SMN–RNP forms that contain hnRNP-Q and -R (blue)
are ZPB (green) and β-actin mRNA. The active transport of this complex results in the accumulation of β-actin mRNA and protein at the distal end of
the axon and at the neuromuscular junction (NMJ). In the spinal muscular atrophy (SMA; bottom panel), low levels of the SMN result in reduced
levels of β-actin accumulation and poorly developed NMJs.

Vo lu me 2, Ju ly/Au gu s t 2011  2011 Jo h n Wiley & So n s, L td. 555


Advanced Review wires.wiley.com/rna

a SMA modifying gene called Plastin 3/T-plastin patients and their use in determining SMN-associated
(PLS3).99 PLS3 is involved in actin dynamics and biochemical activities has been invaluable. Ideally, a
aids in the stabilization of filamentous actin, thereby mutation would be identified that allowed the dis-
increasing the pool of F-actin.213,214 In discordant sib- crimination between snRNP assembly activity and
lings, increased expression of PLS3 correlated with the β-actin/axonal-specific activity. The transgenic
decreased disease severity in females.99 Defects in introduction of this type of SMN missense mutations
cultured motor neurons from SMA mice or from into SMA mice would provide conclusive evidence
SMN-depleted zebrafish could also be overcome by for an SMA pathogenic role for either functional
the transient expression of PLS3.99 While additional pathway. To date, however, attempts such as with
work such as SMA transgenic PLS3 mice will need to
the SMNA111G mutation have proven problematic
be developed, this work supports the notion that actin
as expression levels in the various assays can vary
dynamics plays a key role in SMA development. Addi-
and snRNP assembly assays in zebrafish have proven
tionally, administration of a synthetic peptide that
challenging.217 Currently, it is not clear which func-
inhibits Rho-kinase (ROCK) significantly corrected
the NMJ defects and extended the survival of a milder tional pathway is responsible for SMA, however, it is
model of SMA.215,216 Through this pathway, RhoA also interesting that each mechanistic line of inquiry
has been shown to perform important roles in the has provided compelling supportive biochemical and
regulation of actin dynamics. Collectively, this work functional evidence in disease-relevant contexts, and
illustrates the potential importance of actin transport it may emerge that there is a role for both. As trans-
and actin dynamics in the development of SMA. lational studies continue to push forward in the SMA
field, it will be essential to continue to push forward
with the basic biology underpinning this devastat-
CONCLUSION ing disease as identification of the true functional
SMN is clearly a multifunctional protein that serves as pathway that when defective leads to motor neu-
a nucleation point for a variety of RNPs. A number of ron attrition will help guide in the development and
intragenic mutations have been identified within SMA implementation of optimized therapeutics for SMA.

REFERENCES
1. Lefebvre S, Burglen L, Reboullet S, Clermont O, Burlet 7. Carvalho T, Almeida F, Calapez A, Lafarga M, Berc-
P, Viollet L, Benichou B, Cruaud C, Millasseau P, iano MT, Carmo-Fonseca M. The spinal muscular
Zeviani M, et al. Identification and characterization atrophy disease gene product, SMN: a link between
of a spinal muscular atrophy-determining gene. Cell snRNP biogenesis and the Cajal (coiled) body. J Cell
1995, 80:155–165. Biol 1999, 147:715–728.
2. Wirth B, Brichta L, Hahnen E. Spinal muscular atro- 8. Matera AG, Frey MR. Coiled bodies and gems: janus
phy: from gene to therapy. Semin Pediatr Neurol 2006, or gemini? Am J Hum Genet 1998, 63:317–321.
13:121–131.
9. Young PJ, Le TT, Dunckley M, Nguyen TM, Burghes
3. Gabanella F, Carissimi C, Usiello A, Pellizzoni L. The AH, Morris GE. Nuclear gems and Cajal (coiled) bod-
activity of the spinal muscular atrophy protein is regu-
ies in fetal tissues: nucleolar distribution of the spinal
lated during development and cellular differentiation.
muscular atrophy protein SMN. Exp Cell Res 2001,
Hum Mol Genet 2005, 14:3629–3642.
265:252–261.
4. Burghes AH, Beattie CE. Spinal muscular atrophy:
10. Young PJ, Le TT, Thi Man N, Burghes AH, Morris
why do low levels of survival motor neuron protein
make motor neurons sick? Nat Rev Neurosci 2009, GE. The relationship between SMN, the spinal mus-
10:597–609. cular atrophy protein, and nuclear coiled bodies in
differentiated tissues and cultured cells. Exp Cell Res
5. Coovert DD, Le TT, McAndrew PE, Strasswimmer J, 2000, 256:365–374.
Crawford TO, Mendell JR, Coulson SE, Androphy
EJ, Prior TW, Burghes AH, et al. The survival motor 11. Matera AG, Terns RM, Terns MP. Non-coding RNAs:
neuron protein in spinal muscular atrophy. Hum Mol lessons from the small nuclear and small nucleolar
Genet 1997, 6:1205–1214. RNAs. Nat Rev Mol Cell Biol 2007, 8:209–220.
6. Liu Q, Dreyfuss G. A novel nuclear structure contain- 12. Paushkin S, Gubitz AK, Massenet S, Dreyfuss G. The
ing the survival of motor neurons protein. EMBO J SMN complex, an assemblyosome of ribonucleopro-
1996, 15:3555–3565. teins. Curr Opin Cell Biol 2002, 14:305–312.

556  2011 Jo h n Wiley & So n s, L td. Vo lu me 2, Ju ly/Au gu s t 2011


WIREs RNA SMN in spinal muscular atrophy and snRNP biogenesis

13. Pellizzoni L. Chaperoning ribonucleoprotein biogen- 27. Lorson CL, Hahnen E, Androphy EJ, Wirth B. A sin-
esis in health and disease. EMBO Rep 2007, 8: gle nucleotide in the SMN gene regulates splicing and
340–345. is responsible for spinal muscular atrophy. Proc Natl
14. Feldkotter M, Schwarzer V, Wirth R, Wienker TF, Acad Sci U S A 1999, 96:6307–6311.
Wirth B. Quantitative analyses of SMN1 and SMN2 28. Monani UR, Lorson CL, Parsons DW, Prior TW,
based on real-time lightCycler PCR: fast and highly Androphy EJ, Burghes AH, McPherson JD. A single
reliable carrier testing and prediction of severity of nucleotide difference that alters splicing patterns dis-
spinal muscular atrophy. Am J Hum Genet 2002, tinguishes the SMA gene SMN1 from the copy gene
70:358–368. SMN2. Hum Mol Genet 1999, 8:1177–1183.
15. Pearn J. Classification of spinal muscular atrophies. 29. Hahnen E, Schonling J, Rudnik-Schoneborn S, Zer-
Lancet 1980, 1:919–922. res K, Wirth B. Hybrid survival motor neuron genes
16. Gitlin JM, Fischbeck K, Crawford TO, Cwik V, Fleis- in patients with autosomal recessive spinal muscu-
chman A, Gonye K, Heine D, Hobby K, Kaufmann P, lar atrophy: new insights into molecular mechanisms
Keiles S, et al. Carrier testing for spinal muscular atro- responsible for the disease. Am J Hum Genet 1996,
phy. Genet Med 2010, 12:621–622. 59:1057–1065.
17. Prior TW. Spinal muscular atrophy: newborn and car- 30. Monani UR. Spinal muscular atrophy: a deficiency in
rier screening. Obstet Gynecol Clin North Am 2010, a ubiquitous protein; a motor neuron-specific disease.
37:23–36. Neuron 2005, 48:885–896.
18. Kabra M, Arora S, Maria A, Aggarwal R. Preserved 31. Park GH, Kariya S, Monani UR. Spinal muscular atro-
umbilical cord facilitates antenatal diagnosis of spinal phy: new and emerging insights from model mice. Curr
muscular atrophy. Indian Pediatr 2003, 40:415–418. Neurol Neurosci Rep 2010, 10:108–117.
19. Kaufmann P, Iannaccone ST. Clinical trials in spinal 32. Schrank B, Gotz R, Gunnersen JM, Ure JM, Toyka
muscular atrophy. Phys Med Rehabil Clin N Am 2008, KV, Smith AG, Sendtner M. Inactivation of the sur-
19:653–660, xii. vival motor neuron gene, a candidate gene for human
20. Oskoui M, Kaufmann P. Spinal muscular atrophy. spinal muscular atrophy, leads to massive cell death in
Neurotherapeutics 2008, 5:499–506. early mouse embryos. Proc Natl Acad Sci U S A 1997,
21. Swoboda KJ, Kissel JT, Crawford TO, Bromberg MB, 94:9920–9925.
Acsadi G, D’Anjou G, Krosschell KJ, Reyna SP, 33. Rochette CF, Gilbert N, Simard LR. SMN gene dupli-
Schroth MK, Scott CB, et al. Perspectives on clini- cation and the emergence of the SMN2 gene occurred
cal trials in spinal muscular atrophy. J Child Neurol in distinct hominids: SMN2 is unique to Homo sapi-
2007, 22:957–966. ens. Hum Genet 2001, 108:255–266.
22. Oskoui M, Levy G, Garland CJ, Gray JM, O’Hagen J, 34. Schaal TD, Maniatis T. Selection and characteriza-
De Vivo DC, Kaufmann P. The changing natural his- tion of pre-mRNA splicing enhancers: identification
tory of spinal muscular atrophy type 1 Neurology of novel SR protein-specific enhancer sequences. Mol
2007, 69:1931–1936. Cell Biol 1999, 19:1705–1719.
23. Swoboda KJ, Prior TW, Scott CB, McNaught TP, 35. Hsieh-Li HM, Chang JG, Jong YJ, Wu MH, Wang
Wride MC, Reyna SP, Bromberg MB. Natural his- NM, Tsai CH, Li H. A mouse model for spinal mus-
tory of denervation in SMA: relation to age, SMN2 cular atrophy. Nat Genet 2000, 24:66–70.
copy number, and function. Ann Neurol 2005,
57:704–712. 36. Monani UR, Coovert DD, Burghes AH. Animal mod-
els of spinal muscular atrophy. Hum Mol Genet 2000,
24. Zerres K, Rudnik-Schoneborn S. Natural history in
9:2451–2457.
proximal spinal muscular atrophy: clinical analysis
of 445 patients and suggestions for a modifica- 37. Monani UR, Sendtner M, Coovert DD, Parsons DW,
tion of existing classifications. Arch Neurol 1995, Andreassi C, Le TT, Jablonka S, Schrank B, Rossol W,
52:518–523. Prior TW, et al. The human centromeric survival
motor neuron gene (SMN2) rescues embryonic lethal-
25. Roy N, Mahadevan MS, McLean M, Shutler G,
ity in SMN(−/−) mice and results in a mouse with
Yaraghi Z, Farahani R, Baird S, Besner-Johnston A,
spinal muscular atrophy. Hum Mol Genet 2000,
Lefebvre C, Kang X, et al. The gene for neuronal
9:333–339.
apoptosis inhibitory protein is partially deleted in
individuals with spinal muscular atrophy. Cell 1995, 38. Burnett BG, Munoz E, Tandon A, Kwon DY, Sum-
80:167–178. ner CJ, Fischbeck KH. Regulation of SMN protein
26. Novelli G, Calza L, Amicucci P, Giardino L, Pozza M, stability. Mol Cell Biol 2009, 29:1107–1115.
Silani V, Pizzuti A, Gennarelli M, Piombo G, Capon F, 39. Lorson CL, Androphy EJ. An exonic enhancer is
et al. Expression study of survival motor neuron gene required for inclusion of an essential exon in the
in human fetal tissues. Biochem Mol Med 1997, SMA-determining gene SMN. Hum Mol Genet 2000,
61:102–106. 9:259–265.

Vo lu me 2, Ju ly/Au gu s t 2011  2011 Jo h n Wiley & So n s, L td. 557


Advanced Review wires.wiley.com/rna

40. Farrar MA, Johnston HM, Grattan-Smith P, Turner expression by delivering bifunctional RNAs via a viral
A, Kiernan MC. Spinal muscular atrophy: molecular vector. Mol Ther 2006, 14:54–62.
mechanisms. Curr Mol Med 2009, 9:851–862. 56. Baughan TD, Dickson A, Osman EY, Lorson CL.
41. Horne C, Young PJ. Is RNA manipulation a viable Delivery of bifunctional RNAs that target an intronic
therapy for spinal muscular atrophy? J Neurol Sci repressor and increase SMN levels in an animal model
2009, 287:27–31. of spinal muscular atrophy. Hum Mol Genet 2009,
42. Lorson CL, Rindt H, Shababi M. Spinal muscular 18:1600–1611 (Epub ahead of print; February 19,
atrophy: mechanisms and therapeutic strategies. Hum 2009).
Mol Genet 2010, 19:R111–R118. 57. Coady TH, Baughan TD, Shababi M, Passini MA,
43. Stavarachi M, Apostol P, Toma M, Cimponeriu D, Lorson CL. Development of a single vector system that
Gavrila L. Spinal muscular atrophy disease: a litera- enhances trans-splicing of SMN2 transcripts. PLoS
ture review for therapeutic strategies. J Med Life 2010, One 2008, 3:e3468.
3:3–9. 58. Coady TH, Lorson CL. Trans-splicing-mediated im-
44. Bebee TW, Gladman JT, Chandler DS. Splicing regu- provement in a severe mouse model of spinal muscular
lation of the survival motor neuron genes and implica- atrophy. J Neurosci 2010, 30:126–130.
tions for treatment of spinal muscular atrophy. Front 59. Coady TH, Shababi M, Tullis GE, Lorson CL. Res-
Biosci 2010, 15:1191–1204. toration of SMN function: delivery of a trans-splicing
RNA re-directs SMN2 pre-mRNA splicing. Mol Ther
45. Singh RN. Evolving concepts on human SMN pre-
2007, 15:1471–1478.
mRNA splicing. RNA Biol 2007, 4:7–10 (Epub ahead
of print; June 4, 2007). 60. Dickson A, Osman E, Lorson C. A negatively-acting
bifunctional RNA increases survival motor neu-
46. Hofmann Y, Lorson CL, Stamm S, Androphy EJ,
ron in vitro and in vivo. Hum Gene Ther 2008,
Wirth B. Htra2-β 1 stimulates an exonic splicing
19:1307–1315.
enhancer and can restore full-length SMN expression
to survival motor neuron 2 (SMN2). Proc Natl Acad 61. Hua Y, Sahashi K, Hung G, Rigo F, Passini MA, Ben-
Sci U S A 2000, 97:9618–9623. nett CF, Krainer AR. Antisense correction of SMN2
splicing in the CNS rescues necrosis in a type III SMA
47. Cartegni L, Krainer AR. Disruption of an SF2/ASF-
mouse model. Genes Dev 2010, 24:1634–1644.
dependent exonic splicing enhancer in SMN2 causes
spinal muscular atrophy in the absence of SMN1. Nat 62. Hua Y, Vickers TA, Baker BF, Bennett CF, Krainer
Genet 2002, 30:377–384. AR. Enhancement of SMN2 exon 7 inclusion by anti-
sense oligonucleotides targeting the exon. PLoS Biol
48. Gladman JT, Chandler DS. Intron 7 conserved
2007, 5:e73.
sequence elements regulate the splicing of the SMN
genes. Hum Mol Genet 2010, 19:4239–4252 (Epub 63. Hua Y, Vickers TA, Okunola HL, Bennett CF,
ahead of print; August 12, 2010). Krainer AR. Antisense masking of an hnRNP A1/A2
intronic splicing silencer corrects SMN2 splicing in
49. Singh NK, Singh NN, Androphy EJ, Singh RN. Splic-
transgenic mice. Am J Hum Genet 2008, 82:834–848.
ing of a critical exon of human survival motor neuron
is regulated by a unique silencer element located in the 64. Lim SR, Hertel KJ. Modulation of survival motor
last intron. Mol Cell Biol 2006, 26:1333–1346. neuron pre-mRNA splicing by inhibition of alter-
native 3 splice site pairing. J Biol Chem 2001,
50. Singh NN, Androphy EJ, Singh RN. In vivo selection
276:45476–45483.
reveals combinatorial controls that define a critical
exon in the spinal muscular atrophy genes. RNA 2004, 65. Madocsai C, Lim SR, Geib T, Lam BJ, Hertel KJ. Cor-
10:1291–1305. rection of SMN2 pre-mRNA splicing by antisense U7
small nuclear RNAs. Mol Ther 2005, 12:1013–1022.
51. Singh NN, Androphy EJ, Singh RN. An extended
inhibitory context causes skipping of exon 7 of SMN2 66. Marquis J, Meyer K, Angehrn L, Kampfer SS, Rothen-
in spinal muscular atrophy. Biochem Biophys Res Rutishauser B, Schumperli D. Spinal muscular atro-
Commun 2004, 315:381–388. phy: SMN2 pre-mRNA splicing corrected by a
U7 snRNA derivative carrying a splicing enhancer
52. Kashima T, Manley JL. A negative element in SMN2 sequence. Mol Ther 2007, 15:1479–1486 (Epub ahead
exon 7 inhibits splicing in spinal muscular atrophy. of print; June 5, 2007).
Nat Genet 2003, 34:460–463.
67. Meyer K, Marquis J, Trub J, Nlend Nlend R, Verp S,
53. Khoo B, Krainer AR. Splicing therapeutics in SMN2 Ruepp MD, Imboden H, Barde I, Trono D, Schumperli
and APOB. Curr Opin Mol Ther 2009, 11:108–115. D. Rescue of a severe mouse model for spinal muscular
54. Wirth B, Brichta L, Hahnen E. Spinal muscular atro- atrophy by U7 snRNA-mediated splicing modulation.
phy and therapeutic prospects. Prog Mol Subcell Biol Hum Mol Genet 2009, 18:546–555.
2006, 44:109–132. 68. Singh NN, Shishimorova M, Cao LC, Gangwani L,
55. Baughan T, Shababi M, Coady TH, Dickson AM, Singh RN. A short antisense oligonucleotide mask-
Tullis GE, Lorson CL. Stimulating full-length SMN2 ing a unique intronic motif prevents skipping of a

558  2011 Jo h n Wiley & So n s, L td. Vo lu me 2, Ju ly/Au gu s t 2011


WIREs RNA SMN in spinal muscular atrophy and snRNP biogenesis

critical exon in spinal muscular atrophy. RNA Biol 82. Charroux B, Pellizzoni L, Perkinson RA, Yong J,
2009, 6:341–350. Shevchenko A, Mann M, Dreyfuss G. Gemin4. A novel
69. Skordis LA, Dunckley MG, Yue B, Eperon IC, Mun- component of the SMN complex that is found in both
toni F. Bifunctional antisense oligonucleotides provide gems and nucleoli. J Cell Biol 2000, 148:1177–1186.
a trans-acting splicing enhancer that stimulates SMN2 83. Gubitz AK, Mourelatos Z, Abel L, Rappsilber J, Mann
gene expression in patient fibroblasts. Proc Natl Acad M, Dreyfuss G. Gemin5, a novel WD repeat protein
Sci U S A 2003, 100:4114–4119. component of the SMN complex that binds Sm pro-
70. Williams JH, Schray RC, Patterson CA, Ayitey SO, teins. J Biol Chem 2002, 277:5631–5636.
Tallent MK, Lutz GJ. Oligonucleotide-mediated sur- 84. Pellizzoni L, Baccon J, Rappsilber J, Mann M, Drey-
vival of motor neuron protein expression in CNS fuss G. Purification of native survival of motor neurons
improves phenotype in a mouse model of spinal mus- complexes and identification of Gemin6 as a novel
cular atrophy. J Neurosci 2009, 29:7633–7638. component. J Biol Chem 2002, 277:7540–7545.
71. Geib T, Hertel KJ. Restoration of full-length SMN 85. Carissimi C, Baccon J, Straccia M, Chiarella P, Maio-
promoted by adenoviral vectors expressing RNA anti- lica A, Sawyer A, Rappsilber J, Pellizzoni L. Unrip
sense oligonucleotides embedded in U7 snRNAs. PLoS is a component of SMN complexes active in snRNP
One 2009, 4:e8204. assembly. FEBS Lett 2005, 579:2348–2354.
72. Lefebvre S, Burlet P, Liu Q, Bertrandy S, Clermont O, 86. Otter S, Grimmler M, Neuenkirchen N, Chari A, Sick-
Munnich A, Dreyfuss G, Melki J. Correlation between mann A, Fischer U. A comprehensive interaction map
severity and SMN protein level in spinal muscular
of the human survival of motor neuron (SMN) com-
atrophy. Nat Genet 1997, 16:265–269.
plex. J Biol Chem 2007, 282:5825–5833.
73. Fischer U, Liu Q, Dreyfuss G. The SMN-SIP1 complex
87. Lorson CL, Strasswimmer J, Yao JM, Baleja JD, Hah-
has an essential role in spliceosomal snRNP biogenesis.
nen E, Wirth B, Le T, Burghes AH, Androphy EJ. SMN
Cell 1997, 90:1023–1029.
oligomerization defect correlates with spinal muscular
74. Liu Q, Fischer U, Wang F, Dreyfuss G. The spinal atrophy severity. Nat Genet 1998, 19:63–66.
muscular atrophy disease gene product, SMN, and its
associated protein SIP1 are in a complex with spliceo- 88. Rossoll W, Kroning AK, Ohndorf UM, Steegborn C,
somal snRNP proteins. Cell 1997, 90:1013–1021. Jablonka S, Sendtner M. Specific interaction of SMN,
the spinal muscular atrophy determining gene prod-
75. Meister G, Buhler D, Pillai R, Lottspeich F, Fischer U. uct, with hnRNP-R and gry-rbp/hnRNP-Q: a role for
A multiprotein complex mediates the ATP-dependent SMN in RNA processing in motor axons? Hum Mol
assembly of spliceosomal U snRNPs. Nat Cell Biol Genet 2002, 11:93–105.
2001, 3:945–949.
89. Todd AG, Morse R, Shaw DJ, McGinley S, Stebbings
76. Meister G, Eggert C, Fischer U. SMN-mediated assem-
H, Young PJ. SMN, Gemin2 and Gemin3 associate
bly of RNPs: a complex story. Trends Cell Biol 2002,
with β-actin mRNA in the cytoplasm of neuronal cells
12:472–478.
in vitro. J Mol Biol 2010, 401:681–689.
77. Ogawa C, Usui K, Ito F, Itoh M, Hayashizaki Y,
90. Todd AG, Morse R, Shaw DJ, Stebbings H, Young PJ.
Suzuki H. Role of survival motor neuron complex
Analysis of SMN-neurite granules: core Cajal body
components in small nuclear ribonucleoprotein assem-
components are absent from SMN-cytoplasmic com-
bly. J Biol Chem 2009, 284:14609–14617.
plexes. Biochem Biophys Res Commun 2010,
78. Pellizzoni L, Charroux B, Dreyfuss G. SMN mutants 397:479–485.
of spinal muscular atrophy patients are defective in
binding to snRNP proteins. Proc Natl Acad Sci U S A 91. Todd AG, Shaw DJ, Morse R, Stebbings H, Young PJ.
1999, 96:11167–11172. SMN and the Gemin proteins form sub-complexes
that localise to both stationary and dynamic neu-
79. Baccon J, Pellizzoni L, Rappsilber J, Mann M, Drey-
rite granules. Biochem Biophys Res Commun 2010,
fuss G. Identification and characterization of Gemin7,
394:211–216.
a novel component of the SMN complex. J Biol Chem
2002, 13:13. 92. Zhang H, Xing L, Rossoll W, Wichterle H, Singer RH,
Bassell GJ. Multiprotein complexes of the survival of
80. Carissimi C, Saieva L, Baccon J, Chiarella P, Maiolica
motor neuron protein SMN with Gemins traffic to neu-
A, Sawyer A, Rappsilber J, Pellizzoni L. Gemin8 is a
ronal processes and growth cones of motor neurons.
novel component of the survival motor neuron com-
J Neurosci 2006, 26:8622–8632.
plex and functions in small nuclear ribonucleoprotein
assembly. J Biol Chem 2006, 281:8126–8134. 93. Zhang HL, Pan F, Hong D, Shenoy SM, Singer RH,
81. Charroux B, Pellizzoni L, Perkinson RA, Shevchenko Bassell GJ. Active transport of the survival motor neu-
A, Mann M, Dreyfuss G. Gemin3: a novel DEAD box ron protein and the role of exon-7 in cytoplasmic
protein that interacts with SMN, the spinal muscular localization. J Neurosci 2003, 23:6627–6637.
atrophy gene product, and is a component of gems. 94. Friesen WJ, Dreyfuss G. Specific sequences of the Sm
J Cell Biol 1999, 147:1181–1194. and Sm-like (Lsm) proteins mediate their interaction

Vo lu me 2, Ju ly/Au gu s t 2011  2011 Jo h n Wiley & So n s, L td. 559


Advanced Review wires.wiley.com/rna

with the spinal muscular atrophy disease gene product protein with two major non-ribosomal nucleolar pro-
(SMN). J Biol Chem 2000, 275:26370–26375. teins and its implication in spinal muscular atrophy.
95. Brahms H, Meheus L, de Brabandere V, Fischer U, Hum Mol Genet 2002, 11:1017–1027.
Luhrmann R. Symmetrical dimethylation of arginine 107. Espert L, Eldin P, Gongora C, Bayard B, Harper F,
residues in spliceosomal Sm protein B/B’ and the Sm- Chelbi-Alix MK, Bertrand E, Degols G, Mechti N.
like protein LSm4, and their interaction with the SMN The exonuclease ISG20 mainly localizes in the nucleo-
protein. RNA 2001, 7:1531–1542. lus and the Cajal (coiled) bodies and is associated with
96. Narayanan U, Ospina JK, Frey MR, Hebert MD, Mat- nuclear SMN protein-containing complexes. J Cell
era AG. SMN, the spinal muscular atrophy protein, Biochem 2006, 98:1320–1333.
forms a pre-import snRNP complex with snur- 108. Hua Y, Zhou J. Survival motor neuron protein facil-
portin1 and importin β. Hum Mol Genet 2002, itates assembly of stress granules. FEBS Lett 2004,
11:1785–1795. 572:69–74.
97. Mouaikel J, Narayanan U, Verheggen C, Matera AG, 109. Wang IF, Reddy NM, Shen CK. Higher order arrange-
Bertrand E, Tazi J, Bordonne R. Interaction between ment of the eukaryotic nuclear bodies. Proc Natl Acad
the small-nuclear-RNA cap hypermethylase and the Sci U S A 2002, 99:13583–13588.
spinal muscular atrophy protein, survival of motor 110. Saunders LR, Perkins DJ, Balachandran S, Michaels
neuron. EMBO Rep 2003, 4:616–622. R, Ford R, Mayeda A, Barber GN. Characteriza-
98. Giesemann T, Rathke-Hartlieb S, Rothkegel M, Bar- tion of two evolutionarily conserved, alternatively
tsch JW, Buchmeier S, Jockusch BM, Jockusch H. spliced nuclear phosphoproteins, NFAR-1 and -2, that
A role for polyproline motifs in the spinal muscular function in mRNA processing and interact with the
atrophy protein SMN. Profilins bind to and colocal- double-stranded RNA-dependent protein kinase PKR.
ize with SMN in nuclear gems. J Biol Chem 1999, J Biol Chem 2001, 276:32300–32312.
274:37908–37914. 111. Hebert MD, Szymczyk PW, Shpargel KB, Matera AG.
99. Oprea GE, Krober S, McWhorter ML, Rossoll W, Coilin forms the bridge between Cajal bodies and
Muller S, Krawczak M, Bassell GJ, Beattie CE, SMN, the spinal muscular atrophy protein. Genes
Wirth B. Plastin 3 is a protective modifier of autoso- Dev 2001, 15:2720–2729.
mal recessive spinal muscular atrophy. Science 2008, 112. Zou J, Barahmand-pour F, Blackburn ML, Matsui Y,
320:524–527. Chansky HA, Yang L. Survival motor neuron
100. Mourelatos Z, Abel L, Yong J, Kataoka N, Dreyfuss (SMN) protein interacts with transcription corepressor
G. SMN interacts with a novel family of hnRNP and mSin3A. J Biol Chem 2004, 279:14922–14928.
spliceosomal proteins. EMBO J 2001, 20:5443–5452. 113. Young PJ, Francis JW, Lince D, Coon K, Androphy
101. Piazzon N, Rage F, Schlotter F, Moine H, Branlant C, EJ, Lorson CL. The Ewing’s sarcoma protein interacts
Massenet S. In vitro and in cellulo evidences for asso- with the Tudor domain of the survival motor neuron
ciation of the survival of motor neuron complex with protein. Brain Res Mol Brain Res 2003, 119:37–49.
the fragile X mental retardation protein. J Biol Chem 114. Strasswimmer J, Lorson CL, Breiding DE, Chen JJ,
2008, 283:5598–5610. Le T, Burghes AH, Androphy EJ. Identification of
102. Tadesse H, Deschenes-Furry J, Boisvenue S, Cote J. survival motor neuron as a transcriptional activator-
KH-type splicing regulatory protein interacts with binding protein. Hum Mol Genet 1999, 8:1219–1226.
survival motor neuron protein and is misregulated 115. Barth S, Liss M, Voss MD, Dobner T, Fischer U,
in spinal muscular atrophy. Hum Mol Genet 2008, Meister G, Grasser FA. Epstein-Barr virus nuclear
17:506–524. antigen 2 binds via its methylated arginine-glycine
103. Park JW, Voss PG, Grabski S, Wang JL, Patterson RJ. repeat to the survival motor neuron protein. J Virol
Association of galectin-1 and galectin-3 with Gemin4 2003, 77:5008–5013.
in complexes containing the SMN protein. Nucleic 116. Young PJ, Jensen KT, Burger LR, Pintel DJ, Lorson
Acids Res 2001, 29:3595–3602. CL. Minute virus of mice small nonstructural protein
104. Pellizzoni L, Charroux B, Rappsilber J, Mann M, Dre- NS2 interacts and colocalizes with the SMN protein.
yfuss G. A functional interaction between the survival J Virol 2002, 76:6364–6369.
motor neuron complex and RNA polymerase II. J Cell 117. Young PJ, Jensen KT, Burger LR, Pintel DJ, Lor-
Biol 2001, 152:75–85. son CL. Minute virus of mice NS1 interacts with the
105. Hua Y, Zhou J. Rpp20 interacts with SMN and is re- SMN protein, and they colocalize in novel nuclear
distributed into SMN granules in response to stress. bodies induced by parvovirus infection. J Virol 2002,
Biochem Biophys Res Commun 2004, 314:268–276. 76: 3892–3904.
106. Lefebvre S, Burlet P, Viollet L, Bertrandy S, Huber C, 118. Young PJ, Day PM, Zhou J, Androphy EJ, Morris GE,
Belser C, Munnich A. A novel association of the SMN Lorson, CL. A direct interaction between the survival

560  2011 Jo h n Wiley & So n s, L td. Vo lu me 2, Ju ly/Au gu s t 2011


WIREs RNA SMN in spinal muscular atrophy and snRNP biogenesis

motor neuron protein and p53 and its relation- U snRNP assembly: implications for spinal muscular
ship to spinal muscular atrophy. J Biol Chem 2002, atrophy. Hum Mol Genet 1999, 8:2351–2357.
277:2852–2859. 131. Mohaghegh P, Rodrigues NR, Owen N, Ponting CP,
119. Kurihara N, Menaa C, Maeda H, Haile DJ, Reddy SV. Le TT, Burghes AH, Davies KE. Analysis of mutations
Osteoclast-stimulating factor interacts with the spinal in the tudor domain of the survival motor neuron
muscular atrophy gene product to stimulate osteoclast protein SMN. Eur J Hum Genet 1999, 7:519–525.
formation. J Biol Chem 2001, 276:41035–41039.
132. Selenko P, Sprangers R, Stier G, Buhler D, Fischer U,
120. Trinkle-Mulcahy L, Boulon S, Lam YW, Urcia R, Sattler M. SMN tudor domain structure and its inter-
Boisvert FM, Vandermoere F, Morrice NA, Swift S, action with the Sm proteins. Nat Struct Biol 2001,
Rothbauer U, Leonhardt H, et al. Identifying specific 8:27–31.
protein interaction partners using quantitative mass
spectrometry and bead proteomes. J Cell Biol 2008, 133. Sprangers R, Groves MR, Sinning I, Sattler M. High-
183:223–239. resolution X-ray and NMR structures of the SMN
Tudor domain: conformational variation in the bind-
121. Carnegie GK, Sleeman JE, Morrice N, Hastie CJ, Peg-
ing site for symmetrically dimethylated arginine
gie MW, Philp A, Lamond AI, Cohen PT. Protein
residues. J Mol Biol 2003, 327:507–520.
phosphatase 4 interacts with the survival of motor neu-
rons complex and enhances the temporal localisation 134. Sprangers R, Selenko P, Sattler M, Sinning I, Gro-
of snRNPs. J Cell Sci 2003, 116:1905–1913. ves MR. Definition of domain boundaries and crystal-
122. Claus P, Doring F, Gringel S, Muller-Ostermeyer F, lization of the SMN Tudor domain. Acta Crystallogr
Fuhlrott J, Kraft T, Grothe C. Differential intranuclear D Biol Crystallogr 2003, 59:366–368.
localization of fibroblast growth factor-2 isoforms and 135. Bowerman M, Shafey D, Kothary R. SMN depletion
specific interaction with the survival of motoneuron alters profilin II expression and leads to upregulation
protein. J Biol Chem 2003, 278:479–485. of the RhoA/ROCK pathway and defects in neuronal
123. Gangwani L, Mikrut M, Theroux S, Sharma M, Davis integrity. J Mol Neurosci 2007, 32:120–131.
RJ. Spinal muscular atrophy disrupts the interaction 136. Sharma A, Lambrechts A, Hao le T, Le TT, Sewry CA,
of ZPR1 with the SMN protein. Nat Cell Biol 2001, Ampe C, Burghes AH, Morris GE. A role for com-
3:376–383. plexes of survival of motor neurons (SMN) protein
124. Stelzl U, Worm U, Lalowski M, Haenig C, Brembeck with gemins and profilin in neurite-like cytoplasmic
FH, Goehler H, Stroedicke M, Zenkner M, Schoenherr extensions of cultured nerve cells. Exp Cell Res 2005,
A, Koeppen S, et al. A human protein-protein interac- 309:185–197.
tion network: a resource for annotating the proteome. 137. Talbot K, Ponting CP, Theodosiou AM, Rodrigues
Cell 2005, 122:957–968.
NR, Surtees R, Mountford R, Davies KE. Missense
125. Sun Y, Grimmler M, Schwarzer V, Schoenen F, Fis- mutation clustering in the survival motor neuron gene:
cher U, Wirth B. Molecular and functional analysis a role for a conserved tyrosine and glycine rich region
of intragenic SMN1 mutations in patients with spinal of the protein in RNA metabolism? Hum Mol Genet
muscular atrophy. Hum Mutat 2005, 25:64–71. 1997, 6:497–500.
126. Wirth B. An update of the mutation spectrum of the 138. Morse R, Shaw DJ, Todd AG, Young PJ. Targeting of
survival motor neuron gene (SMN1) in autosomal SMN to Cajal bodies is mediated by self-association.
recessive spinal muscular atrophy (SMA). Hum Mutat Hum Mol Genet 2007, 16:2349–2358.
2000, 15:228–237.
139. Hua Y, Zhou J. Modulation of SMN nuclear foci and
127. Bertrandy S, Burlet P, Clermont O, Huber C, Fon-
cytoplasmic localization by its C-terminus. Cell Mol
drat C, Thierry-Mieg D, Munnich A, Lefebvre S. The
Life Sci 2004, 61:2658–2663.
RNA-binding properties of SMN: deletion analysis of
the zebrafish orthologue defines domains conserved in 140. Mattis VB, Bowerman M, Kothary R, Lorson CL. A
evolution. Hum Mol Genet 1999, 8:775–782. SMN7 read-through product confers functionality to
128. Lorson CL, Androphy EJ. The domain encoded by the SMN7 protein. Neurosci Lett 2008, 442:54–58.
exon 2 of the survival motor neuron protein medi- 141. Vitte J, Fassier C, Tiziano FD, Dalard C, Soave S,
ates nucleic acid binding. Hum Mol Genet 1998, Roblot N, Brahe C, Saugier-Veber P, Bonnefont JP,
7:1269–1275. Melki J, et al. Refined characterization of the expres-
129. Young PJ, Man NT, Lorson CL, Le TT, Androphy EJ, sion and stability of the SMN gene products. Am
Burghes AH, Morris GE. The exon 2b region of the J Pathol 2007, 171:1269–1280.
spinal muscular atrophy protein, SMN, is involved in 142. Wolstencroft EC, Mattis V, Bajer AA, Young PJ, Lor-
self-association and SIP1 binding. Hum Mol Genet son CL. A non-sequence-specific requirement for SMN
2000, 9:2869–2877. protein activity: the role of aminoglycosides in induc-
130. Buhler D, Raker V, Luhrmann R, Fischer U. Essential ing elevated SMN protein levels. Hum Mol Genet
role for the tudor domain of SMN in spliceosomal 2005, 14:1199–1210.

Vo lu me 2, Ju ly/Au gu s t 2011  2011 Jo h n Wiley & So n s, L td. 561


Advanced Review wires.wiley.com/rna

143. Zhang H, Xing L, Singer RH, Bassell GJ. QNQKE heteromeric complexes and an snRNP subcore particle
targeting motif for the SMN-Gemin multiprotein com- in vitro. EMBO J 1996, 15:2256–2269.
plex in neurons. J Neurosci Res 2007, 85:2657–2667. 159. van Dam A, Winkel I, Zijlstra-Baalbergen J, Smeenk
144. Heier CR, Gogliotti RG, DiDonato CJ. SMN tran- R, Cuypers HT. Cloned human snRNP proteins B and
script stability: could modulation of messenger RNA B differ only in their carboxy-terminal part. EMBO J
degradation provide a novel therapy for spinal muscu- 1989, 8:3853–3860.
lar atrophy? J Child Neurol 2007, 22:1013–1018.
160. Friesen WJ, Massenet S, Paushkin S, Wyce A, Drey-
145. Pellizzoni L, Kataoka N, Charroux B, Dreyfuss G. A fuss G. SMN, the product of the spinal muscular
novel function for SMN, the spinal muscular atro- atrophy gene, binds preferentially to dimethylarginine-
phy disease gene product, in pre-mRNA splicing. Cell containing protein targets. Mol Cell 2001, 7:
1998, 95:615–624. 1111–1117.
146. Will CL, Luhrmann R. Spliceosomal U snRNP biogen-
161. Meister G, Eggert C, Buhler D, Brahms H, Kambach
esis, structure and function. Curr Opin Cell Biol 2001,
C, Fischer U. Methylation of Sm proteins by a complex
13:290–301.
containing PRMT5 and the putative U snRNP assem-
147. Hamm J, Darzynkiewicz E, Tahara SM, Mattaj IW. bly factor pICln. Curr Biol 2001, 11:1990–1994.
The trimethylguanosine cap structure of U1 snRNA
is a component of a bipartite nuclear targeting signal. 162. Meister G, Fischer U. Assisted RNP assembly: SMN
Cell 1990, 62:569–577. and PRMT5 complexes cooperate in the forma-
tion of spliceosomal U snRNPs. EMBO J 2002,
148. Hamm J, Mattaj IW. Monomethylated cap structures
21:5853–5863.
facilitate RNA export from the nucleus. Cell 1990,
63:109–118. 163. Chari A, Golas MM, Klingenhager M, Neuenkirchen
N, Sander B, Englbrecht C, Sickmann A, Stark H, Fis-
149. Fischer U, Huber J, Boelens WC, Mattaj IW, Luhr-
mann R. The HIV-1 Rev activation domain is a nuclear cher U. An assembly chaperone collaborates with the
export signal that accesses an export pathway used by SMN complex to generate spliceosomal snRNPs. Cell
specific cellular RNAs. Cell 1995, 82:475–483. 2008, 135:497–509.
150. Fornerod M, Ohno M, Yoshida M, Mattaj IW. CRM1 164. Neuenkirchen N, Chari A, Fischer U. Deciphering the
is an export receptor for leucine-rich nuclear export assembly pathway of Sm-class U snRNPs. FEBS Lett
signals. Cell 1997, 90:1051–1060. 2008, 582:1997–2003.
151. Izaurralde E, Lewis J, Gamberi C, Jarmolowski A, 165. Gubitz AK, Feng W, Dreyfuss G. The SMN complex.
McGuigan C, Mattaj IW. A cap-binding protein com- Exp Cell Res 2004, 296:51–56.
plex mediating U snRNA export. Nature 1995, 166. Pellizzoni L, Yong J, Dreyfuss G. Essential role for the
376:709–712. SMN complex in the specificity of snRNP assembly.
152. Izaurralde E, Lewis J, McGuigan C, Jankowska M, Science 2002, 298:1775–1779.
Darzynkiewicz E, Mattaj IW. A nuclear cap binding 167. Kambach C, Walke S, Nagai K. Structure and assem-
protein complex involved in pre-mRNA splicing. Cell
bly of the spliceosomal small nuclear ribonucleopro-
1994, 78:657–668.
tein particles. Curr Opin Struct Biol 1999, 9:222–230.
153. Izaurralde E, McGuigan C, Mattaj IW. Nuclear local-
168. Kambach C, Walke S, Young R, Avis JM, de la
ization of a cap-binding protein complex. Cold Spring
Fortelle E, Raker VA, Luhrmann R, Li J, Nagai K.
Harb Symp Quant Biol 1995, 60:669–675.
Crystal structures of two Sm protein complexes and
154. Nakielny S, Dreyfuss G. Transport of proteins and their implications for the assembly of the spliceosomal
RNAs in and out of the nucleus. Cell 1999, snRNPs. Cell 1999, 96:375–387.
99:677–690.
169. Ma Y, Dostie J, Dreyfuss G, Van Duyne GD. The
155. Ohno M, Segref A, Bachi A, Wilm M, Mattaj IW.
Gemin6-Gemin7 heterodimer from the survival of
PHAX, a mediator of U snRNA nuclear export whose
motor neurons complex has an Sm protein-like struc-
activity is regulated by phosphorylation. Cell 2000,
ture. Structure (Camb) 2005, 13:883–892.
101:187–198.
156. Shaw DJ, Eggleton P, Young PJ. Joining the dots: 170. Newman AJ, Nagai K. Structural studies of the
production, processing and targeting of U snRNP spliceosome: blind men and an elephant. Curr Opin
to nuclear bodies. Biochim Biophys Acta 2008, Struct Biol 2010, 20:82–89.
1783:2137–2144. 171. Lau CK, Bachorik JL, Dreyfuss G. Gemin5-snRNA
157. Kataoka N, Ohno M, Kangawa K, Tokoro Y, Shimura interaction reveals an RNA binding function for
Y. Cloning of a complementary DNA encoding an 80 WD repeat domains. Nat Struct Mol Biol 2009,
kilodalton nuclear cap binding protein. Nucleic Acids 16:486–491.
Res 1994, 22:3861–3865. 172. Yong J, Kasim M, Bachorik JL, Wan L, Dreyfuss G.
158. Raker VA, Plessel G, Luhrmann R. The snRNP core Gemin5 delivers snRNA precursors to the SMN com-
assembly pathway: identification of stable core protein plex for snRNP biogenesis. Mol Cell 38:551–562.

562  2011 Jo h n Wiley & So n s, L td. Vo lu me 2, Ju ly/Au gu s t 2011


WIREs RNA SMN in spinal muscular atrophy and snRNP biogenesis

173. Mouaikel J, Verheggen C, Bertrand E, Tazi J, Bordon- 188. Sharp PA, Burge CB. Classification of introns: U2-type
ne R. Hypermethylation of the cap structure of both or U12-type. Cell 1997, 91:875–879.
yeast snRNAs and snoRNAs requires a conserved 189. Wu Q, Krainer AR. Splicing of a divergent subclass
methyltransferase that is localized to the nucleolus. of AT-AC introns requires the major spliceosomal
Mol Cell 2002, 9:891–901. snRNAs. RNA 1997, 3:586–601.
174. Palacios I, Hetzer M, Adam SA, Mattaj IW. Nuclear 190. Gabanella F, Butchbach ME, Saieva L, Carissimi C,
import of U snRNPs requires importin β. EMBO J Burghes AH, Pellizzoni L. Ribonucleoprotein assembly
1997, 16:6783–6792. defects correlate with spinal muscular atrophy sever-
175. Rexach M, Blobel G. Protein import into nuclei: asso- ity and preferentially affect a subset of spliceosomal
ciation and dissociation reactions involving transport snRNPs. PLoS One 2007, 2:e921.
substrate, transport factors, and nucleoporins. Cell 191. Zhang Z, Lotti F, Dittmar K, Younis I, Wan L, Kasim
1995, 83:683–692. M, Dreyfuss G. SMN deficiency causes tissue-
176. Matera AG, Shpargel KB. Pumping RNA: nuclear specific perturbations in the repertoire of snR-
bodybuilding along the RNP pipeline. Curr Opin Cell NAs and widespread defects in splicing. Cell 2008,
Biol 2006, 18:317–324. 133:585–600.
177. Sleeman JE, Lamond AI. Nuclear organization of pre- 192. Baumer D, Lee S, Nicholson G, Davies JL, Parkin-
mRNA splicing factors. Curr Opin Cell Biol 1999, son NJ, Murray LM, Gillingwater TH, Ansorge O,
11:372–377. Davies KE, Talbot K, et al. Alternative splicing
events are a late feature of pathology in a mouse
178. Sleeman JE, Lamond AI. Newly assembled snRNPs
model of spinal muscular atrophy. PLoS Genet 2009,
associate with coiled bodies before speckles, suggest-
5:e1000773.
ing a nuclear snRNP maturation pathway. Curr Biol
1999, 9:1065–1074. 193. Bevan AK, Hutchinson KR, Foust KD, Braun L,
McGovern VL, Schmelzer L, Ward JG, Petruska JC,
179. Nesic D, Tanackovic G, Kramer A. A role for Cajal
Lucchesi PA, Burghes AH. Early heart failure in the
bodies in the final steps of U2 snRNP biogenesis.
SMN7 model of spinal muscular atrophy and correc-
J Cell Sci 2004, 117:4423–4433.
tion by postnatal scAAV9-SMN delivery. Hum Mol
180. Schaffert N, Hossbach M, Heintzmann R, Achsel T, Genet 2010, 19:3895–3905 (Epub ahead of print;
Luhrmann R. RNAi knockdown of hPrp31 leads to July 16, 2010).
an accumulation of U4/U6 di-snRNPs in Cajal bodies.
194. Heier CR, Hampton TG, Wang D, Didonato CJ.
EMBO J 2004, 23:3000–3009.
Development of electrocardiogram intervals during
181. Stanek D, Neugebauer KM. Detection of snRNP growth of FVB/N neonate mice. BMC Physiol 2010,
assembly intermediates in Cajal bodies by fluores- 10:16.
cence resonance energy transfer. J Cell Biol 2004,
195. Shababi M, Habibi J, Yang HT, Vale SM, Sewell WA,
166:1015–1025.
Lorson CL. Cardiac defects contribute to the pathol-
182. Stanek D, Rader SD, Klingauf M, Neugebauer KM. ogy of spinal muscular atrophy models. Hum Mol
Targeting of U4/U6 small nuclear RNP assembly fac- Genet 2010, 19:4059–4071 (Epub ahead of print;
tor SART3/p110 to Cajal bodies. J Cell Biol 2003, August 9, 2010).
160:505–516.
196. Wishart TM, Huang JP, Murray LM, Lamont DJ,
183. Patel AA, Steitz JA. Splicing double: insights from the Mutsaers CA, Ross J, Geldsetzer P, Ansorge O, Talbot
second spliceosome. Nat Rev Mol Cell Biol 2003, K, Parson SH, et al. SMN deficiency disrupts brain
4:960–970. development in a mouse model of severe spinal mus-
184. Will CL, Luhrmann R. Splicing of a rare class of cular atrophy. Hum Mol Genet 2010.
introns by the U12-dependent spliceosome. Biol Chem 197. Wu Q, Krainer AR. AT-AC pre-mRNA splicing mech-
2005, 386:713–724. anisms and conservation of minor introns in voltage-
185. Will CL, Schneider C, MacMillan AM, Katopodis NF, gated ion channel genes. Mol Cell Biol 1999,
Neubauer G, Wilm M, Luhrmann R, Query CC. A 19:3225–3236.
novel U2 and U11/U12 snRNP protein that asso- 198. Rossoll W, Bassell GJ. Spinal muscular atrophy and a
ciates with the pre-mRNA branch site. EMBO J 2001, model for survival of motor neuron protein function
20:4536–4546. in axonal ribonucleoprotein complexes. Results Probl
186. Hall SL, Padgett RA. Conserved sequences in a class Cell Differ 2009, 48:289–326.
of rare eukaryotic nuclear introns with non-consensus 199. Fan L, Simard LR. Survival motor neuron (SMN) pro-
splice sites. J Mol Biol 1994, 239:357–365. tein: role in neurite outgrowth and neuromuscular
187. Dietrich RC, Incorvaia R, Padgett RA. Terminal maturation during neuronal differentiation and devel-
intron dinucleotide sequences do not distinguish opment. Hum Mol Genet 2002, 11:1605–1614.
between U2- and U12-dependent introns. Mol Cell 200. Rossoll W, Jablonka S, Andreassi C, Kroning AK,
1997, 1:151–160. Karle K, Monani UR, Sendtner M. SMN, the spinal

Vo lu me 2, Ju ly/Au gu s t 2011  2011 Jo h n Wiley & So n s, L td. 563


Advanced Review wires.wiley.com/rna

muscular atrophy-determining gene product, modu- 209. McWhorter ML, Monani UR, Burghes AH, Beattie
lates axon growth and localization of β-actin mRNA CE. Knockdown of the survival motor neuron
in growth cones of motoneurons. J Cell Biol 2003, (SMN) protein in zebrafish causes defects in motor
163:801–812. axon outgrowth and pathfinding. J Cell Biol 2003,
201. Williams BY, Hamilton SL, Sarkar HK. The survival 162:919–931.
motor neuron protein interacts with the transactivator 210. McGovern VL, Gavrilina TO, Beattie CE, Burghes
FUSE binding protein from human fetal brain. FEBS AH. Embryonic motor axon development in the severe
Lett 2000, 470:207–210. SMA mouse. Hum Mol Genet 2008, 17:2900–2909.
202. Eom T, Antar LN, Singer RH, Bassell GJ. Localiza- 211. Murray LM, Lee S, Baumer D, Parson SH, Talbot K,
tion of a β-actin messenger ribonucleoprotein complex et al. Pre-symptomatic development of lower motor
with zipcode-binding protein modulates the density of neuron connectivity in a mouse model of severe
dendritic filopodia and filopodial synapses. J Neurosci spinal muscular atrophy. Hum Mol Genet 2010,
2003, 23:10433–10444. 19:420–433.
203. Jablonka S, Wiese S, Sendtner M. Axonal defects in 212. Winkler C, Eggert C, Gradl D, Meister G, Giegerich
mouse models of motoneuron disease. J Neurobiol M, Wedlich D, Laggerbauer B, Fischer U. Reduced U
2004, 58:272–286. snRNP assembly causes motor axon degeneration in
204. Pan F, Huttelmaier S, Singer RH, Gu W. ZBP2 facil- an animal model for spinal muscular atrophy. Genes
itates binding of ZBP1 to β-actin mRNA during Dev 2005, 19:2320–2330.
transcription. Mol Cell Biol 2007, 27:8340–8351.
213. Bretscher A. Fimbrin is a cytoskeletal protein that
205. Tiruchinapalli DM, Oleynikov Y, Kelic S, Shenoy SM, crosslinks F-actin in vitro. Proc Natl Acad Sci U S A
Hartley A, Stanton PK, Singer RH, Bassell GJ. 1981, 78:6849–6853.
Activity-dependent trafficking and dynamic localiza-
214. Glenney JR Jr, Kaulfus P, Matsudaira P, Weber K.
tion of zipcode binding protein 1 and β-actin mRNA
F-actin binding and bundling properties of fimbrin,
in dendrites and spines of hippocampal neurons.
a major cytoskeletal protein of microvillus core fila-
J Neurosci 2003, 23:3251–3261.
ments. J Biol Chem 1981, 256:9283–9288.
206. Zhang HL, Eom T, Oleynikov Y, Shenoy SM, Liebelt
DA, Dictenberg JB, Singer RH, Bassell GJ. 215. Bowerman M, Anderson CL, Beauvais A, Boyl PP,
Neurotrophin-induced transport of a β-actin mRNP Witke W, Kothary R. SMN, profilin IIa and plastin
complex increases β-actin levels and stimulates growth 3: a link between the deregulation of actin dynam-
cone motility. Neuron 2001, 31:261–275. ics and SMA pathogenesis. Mol Cell Neurosci 2009,
42:66–74.
207. Glinka M, Herrmann T, Funk N, Havlicek S, Rossoll
W, Winkler C, Sendtner M. The heterogeneous 216. Bowerman M, Beauvais A, Anderson CL, Kothary R.
nuclear ribonucleoprotein-R is necessary for axonal Rho-kinase inactivation prolongs survival of an inter-
β-actin mRNA translocation in spinal motor neurons. mediate SMA mouse model. Hum Mol Genet 2010,
Hum Mol Genet 2010, 19:1951–1966. 19:1468–1478.
208. Boon KL, Xiao S, McWhorter ML, Donn T, Wolf- 217. Workman E, Saieva L, Carrel TL, Crawford TO, Liu
Saxon E, Bohnsack MT, Moens CB, Beattie CE. D, Lutz C, Beattie CE, Pellizzoni L, Burghes AH. A
Zebrafish survival motor neuron mutants exhibit SMN missense mutation complements SMN2 restor-
presynaptic neuromuscular junction defects. Hum Mol ing snRNPs and rescuing SMA mice. Hum Mol Genet
Genet 2009, 18:3615–3625. 2009, 18:2215–2229.

564  2011 Jo h n Wiley & So n s, L td. Vo lu me 2, Ju ly/Au gu s t 2011

You might also like