ArticlePDF Available

Minor spliceosome and disease

Authors:
  • All India Institute of Medical Sciences New Delhi India

Abstract and Figures

The U12-type dependent (minor) spliceosome excises a rare group of introns that are characterized by a highly conserved 5' splice site and branch point sequence. Several new congenital or somatic diseases have recently been associated with mutations in components of the minor spliceosome. A common theme in these diseases is the detection of elevated levels of transcripts containing U12-type introns, of which a subset is associated with other splicing defects. Here we review the present understanding of the 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 diseases.
Content may be subject to copyright.
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
ARTICLE IN PRESS
G Model
YSCDB-2404;
No.
of
Pages
10
Seminars
in
Cell
&
Developmental
Biology
xxx
(2017)
xxx–xxx
Contents
lists
available
at
ScienceDirect
Seminars
in
Cell
&
Developmental
Biology
j
ourna
l
h
o
me
page:
www.elsevier.com/locate/semcdb
Review
Minor
spliceosome
and
disease
Bhupendra
Verma1,
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
Article
history:
Received
4
July
2017
Received
in
revised
form
21
September
2017
Accepted
27
September
2017
Available
online
xxx
Keywords:
U12-type
introns
Minor
spliceosome
Human
diseases
Pre-mRNA
splicing
Cryptic
splice
sites
Exon
skipping
Intron
retention
a
b
s
t
r
a
c
t
The
U12-dependent
(minor)
spliceosome
excises
a
rare
group
of
introns
that
are
characterized
by
a
highly
conserved
5splice
site
and
branch
point
sequence.
Several
new
congenital
or
somatic
diseases
have
recently
been
associated
with
mutations
in
components
of
the
minor
spliceosome.
A
common
theme
in
these
diseases
is
the
detection
of
elevated
levels
of
transcripts
containing
U12-type
introns,
of
which
a
subset
is
associated
with
other
splicing
defects.
Here
we
review
the
present
understanding
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
diseases.
©
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/).
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:
Mikko.Frilander@Helsinki.Fi
(M.J.
Frilander).
1Present
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
ARTICLE IN PRESS
G Model
YSCDB-2404;
No.
of
Pages
10
2
B.
Verma
et
al.
/
Seminars
in
Cell
&
Developmental
Biology
xxx
(2017)
xxx–xxx
1.
Introduction
Pre-mRNA
splicing
is
an
essential
step
in
the
gene
expression
pathway
of
all
eukaryotes.
During
splicing,
the
non-coding
intron
sequences
are
recognized
and
removed
from
precursor
mRNA
(pre-
mRNA)
by
the
spliceosome,
a
large
ribonucleoprotein
complex.
Defects
in
splicing
of
one
or
more
mRNA
species
are
a
major
cause
of
human
diseases
and
present
estimates
suggest
that
up
to
60%,
are
associated
with
pre-mRNA
splicing
[1,2].
In
addition
to
mono-
genic
disorders,
genetic
variants
altering
splicing
are
also
thought
to
be
an
important
contributor
to
complex
diseases
and
cancer
[3,4].
The
majority
of
disease-associated
splicing
defects
are
cis-
acting
mutations
within
a
single
gene
that
disrupt
either
the
splice
site
consensus
sequences
or
splicing
regulatory
elements
located
in
introns
or
exons
[5].
Notably,
exonic
mutations
interpreted
as
mis-
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
of
the
affected
individual
mRNAs.
In
contrast,
mutations
in
splicing
factors,
spliceosome
components
or
spliceosome
assembly
factors
often
lead
to
widespread
defects
in
the
processing
of
large
numbers
of
pre-mRNAs
[8–10].
Most
metazoan
species
contain
two
distinct
pre-mRNA
splicing
machineries
known
as
the
major
(U2-dependent)
and
minor
(U12-
dependent)
spliceosomes,
which
recognize
and
excise
either
the
major
(U2-type)
or
minor
(U12-type)
class
of
introns,
respectively.
In
contrast
to
the
major
introns,
U12-type
introns
are
character-
ized
by
divergent
and
highly
conserved
5splice
site
(5ss)
and
branch
point
sequences
(BPS)
(Fig.
1A;
[11]).
These
introns
also
lack
the
characteristic
polypyrimidine
tract
(PPT)
that
is
present
in
U2-type
introns
immediately
upstream
of
the
3splice
site
(3ss).
Minor
introns
constitute
only
0.35%
of
all
human
introns
and
have
been
reported
to
be
present
in
700–800
genes,
each
of
which
typ-
ically
carry
only
a
single
U12-type
intron
and
multiple
U2-type
introns.
U12-type
introns
are
enriched
in
genes
that
represent
a
rather
restricted
set
of
functional
classes
and
pathways.
Particu-
larly
they
are
present
in
genes
related
to
‘information
processing
functions’,
such
as
DNA
replication
and
repair,
transcription,
RNA
processing,
and
translation,
but
can
also
be
found
in
genes
related
to
cytoskeletal
organization,
vesicular
transport,
and
voltage-gated
ion
channel
activity,
as
suggested
originally
by
Burge
et
al.,
[12]
and
verified
later
[13,14].
Both
the
identities
of
genes
carrying
U12-
type
introns
and
their
positions
within
the
genes
are
evolutionarily
conserved
[15].
The
overall
organisational
and
functional
features
of
both
spliceosomes
are
highly
similar.
Both
are
composed
of
five
small
nuclear
RNA
(snRNA)
molecules
that
associate
with
protein
factors
to
give
rise
to
small
nuclear
ribonucleoproteins
(snRNPs).
Within
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
is
shared
between
the
two
spliceosomes.
Of
the
200–300
pro-
teins
associated
with
spliceosomes,
most
are
thought
to
be
shared
between
the
two
systems
and
only
7
proteins,
associated
with
U11
and
U12
snRNPs,
are
unique
to
the
minor
spliceosome
[16,17].
The
highly
similar
spliceosome
composition
is
reflected
in
the
conserved
assembly
pathway
and
catalytic
mechanism.
Both
spliceosomes
are
assembled
sequentially
starting
from
intron
recognition,
followed
by
formation
of
a
catalytically
active
spliceo-
some
and
joining
of
the
exons
flanking
the
excised
intron.
With
minor
introns,
the
5ss
and
BPS
are
co-operatively
recognized
by
a
pre-formed
U11/U12
di-snRNP
[18],
contrary
to
the
sequential
recognition
of
these
sequences
by
individual
U1
and
U2
snRNPs
in
major
introns.
Since
the
PPT
is
lacking
in
minor
introns,
the
U2AF1/2
heterodimer
that
recognizes
the
PPT
and
3ss
of
major
introns
does
not
associate
with
minor
introns.
Instead,
an
integral
U11/U12
di-snRNP
protein
component,
Urp/ZRSR2
takes
up
the
role
of
3ss
recognition
with
minor
introns
[19].
Following
this
initial
recognition,
both
splicing
pathways
proceed
with
association
of
a
tri-snRNP,
either
U4atac/U6atac.U5
or
U4/U6.U5
(Fig.
1B;
[20,21]).
Further
rearrangements
in
RNA–RNA
and
RNA–protein
interac-
tions
lead
to
the
formation
of
a
catalytically
active
spliceosome
and
catalytic
excision
of
the
intron
[20,22,23].
Here,
we
review
the
role
of
the
minor
spliceosome
in
human
diseases,
with
a
specific
focus
on
diseases
caused
by
mutations
in
the
integral
components
of
the
minor
spliceosome.
Given
that
most
protein
components
and
U5
snRNA
are
shared
with
the
major
spliceosome,
there
are
also
several
diseases
where
mutations
dis-
rupting
shared
components
can
potentially
affect
the
functions
of
both
spliceosomes.
Those
have
been
discussed
in
detail
elsewhere
[24].
2.
Minor
spliceosome
in
human
disease
The
direct
targets
for
human
diseases
specific
for
the
minor
spliceosome
are
the
unique
snRNA
and
protein
components.
This
includes
several
components
of
the
U11/U12
intron
recognition
complex
and
the
U4atac
and
U6atac
snRNAs
in
the
minor
tri-snRNP.
The
U11/U12
di-snRNP
contains,
in
addition
to
the
U11
and
U12
snRNAs,
seven
integral
proteins
(65K,
48K,
59K,
35K,
31K,
25K
and
20K)
that
are
unique
to
the
minor
spliceosome
[25,26].
Addition-
ally,
the
Urp/ZRSR2
protein
associated
with
the
U11/U12
di-snRNP
has
been
reported
to
function
in
both
minor
and
major
spliceo-
somes,
with
an
essential
role
for
U12-type
intron
3ss
recognition
[19].
To
date
five
human
diseases
with
mutations
in
the
spe-
cific
components
of
the
minor
spliceosome
have
been
described
(Table
1).
Three
of
them
affect
the
components
of
the
U11/U12
di-
snRNP,
namely
the
U11/U12-65K
protein
(RNPC3;
[27]),
U12
snRNA
(RNU12;
[28])
and
Urp
protein
(ZRSR2;
[29]);
while
two
diseases
are
attributed
to
mutations
in
the
U4atac
snRNA
(RNU4ATAC;
[30–32]).
Each
of
these
diseases
is
hypomorphic,
leading
only
to
a
partial
loss
of
minor
spliceosome
function
because
correctly
spliced
mRNAs
can
be
detected
in
the
patient
cells.
We
briefly
introduce
each
disease
and
later
discuss
the
impact
of
disease
mutations
on
the
assembly
of
the
minor
spliceosome
and
the
fate
of
affected
mRNAs.
Apart
from
mutations
in
the
core
minor
spliceosome
compo-
nents,
we
also
describe
the
few
reported
human
diseases
caused
by
mutations
at
the
splice
sites
of
U12-type
introns,
though
we
note
that
this
appears
to
be
underexplored
territory
given
the
small
number
of
cases
reported
so
far.
Finally,
we
mention
the
emerging
role
for
the
minor
spliceosome
in
cancer
and
autoimmune
disorders
as
well
as
neurodegenerative
diseases.
2.1.
Diseases
affecting
the
intron
recognition
step
2.1.1.
Isolated
growth
hormone
deficiency
Recessive
mutations
in
the
RNPC3
gene,
encoding
U11/U12-
65K,
one
of
the
seven
minor
spliceosome-specific
proteins,
have
been
associated
with
isolated
growth
hormone
deficiency
(IGHD)
and
associated
pituitary
hypoplasia.
The
65K
protein
is
part
of
a
molecular
bridge
that
connects
the
U11
and
U12
snRNPs
into
a
di-
snRNP
(Fig
1B;
[33]).
Initially,
RNPC3
mutations
were
detected
in
a
single
family
only
[27],
but
additional
cases
with
overlapping
muta-
tions
and
similar
phenotypes
have
been
described
subsequently
[34].
IGHD
is
a
condition
characterized
by
a
shortage
or
absence
of
growth
hormone,
with
the
absence
of
associated
pituitary
hor-
mone
deficiencies.
Genetically,
IGHD
is
a
diverse
disease
and
can
result
from
either
recessive
or
dominant
mutations
in
various
genes
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
ARTICLE IN PRESS
G Model
YSCDB-2404;
No.
of
Pages
10
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
compound
heterozygous
with
a
missense
P474T
mutation
com-
bined
with
either
R502X
or
R205X
nonsense
mutation
[27,34].
At
the
protein
level
the
P474T
and
R502X
mutations
map
to
the
C-
terminal
RNA
recognition
motif
(RRM)
of
the
protein,
while
the
R205X
maps
to
the
proline-rich
region
between
the
two
RRMs
(Fig.
2A).
Although
the
tissue
affected
in
IGHD,
the
pituitary
gland,
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
ARTICLE IN PRESS
G Model
YSCDB-2404;
No.
of
Pages
10
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
minor
snRNPs
RNPC3
Isolated
Growth
Hormone
Deficiency
IGHD
[27]
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
5ss 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-
tent
with
the
function
of
the
65K
protein
in
the
intron
recognition
complex,
several
types
of
aberrant
splicing
events
associated
with
a
subset
of
U12-type
introns
were
detected,
including
increased
retention
of
U12-type
introns,
activation
of
nearby
cryptic
U2-
type
splice
sites
and
exon
skipping
events.
Biochemical
analysis
of
patient
cells
revealed
a
reduction
in
levels
of
the
65K
protein,
as
well
as
reduced
stability
of
the
U11/U12
di-snRNP
complex.
This
result
is
consistent
with
the
function
of
the
65K
protein
as
a
component
of
themolecular
bridge
between
U11
and
U12
snRNPs
through
interactions
with
the
U11-59K
protein
and
U12
snRNA
[33].
The
65K-U12
snRNA
interaction
is
mediated
by
the
C-terminal
RRM
domain
and,
as
predicted,
the
IGHD
mutations
were
shown
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
ARTICLE IN PRESS
G Model
YSCDB-2404;
No.
of
Pages
10
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
is
emerging
from
these
studies
is
that
the
IGHD
mutations
lead
to
impaired
interaction
between
the
65K
protein
and
U12
snRNA,
which
in
turn
causes
destabilization
or
reduced
formation
of
the
U11/U12
di-snRNP.
Additionally,
it
is
likely
that
both
RNPC3
non-
sense
mutations
described
for
IGHD
lead
to
allele-specific
decay
of
the
65K
mRNA
via
nonsense
mediated
decay
(NMD)
pathway,
as
shown
recently
for
the
R502X
allele
[36].
This
is
expected
to
signif-
icantly
reduce
the
levels
of
65K
proteins
encoded
by
the
nonsense
alleles.
However,
we
note
that
additional
mechanisms
may
be
at
play,
as
it
is
not
currently
known
whether
the
65K
protein
is
purely
a
structural
component
of
the
U11/U12
di-snRNP,
or
whether
it
has
additional
functions
in
the
splicing
process.
A
zebrafish
study
indi-
cated
that
the
65K
protein
may
have
other
functions
later
in
the
spliceosome
assembly
[37].
Consistently,
recent
work
reported
an
interaction
between
65K
and
the
3terminal
stem-loop
of
U6atac
snRNA
(Fig.
2C;
[38]),
which
is
similarly
impaired
by
the
IGHD
mutations
[36].
2.1.2.
Cerebellar
ataxia
A
mutation
in
the
noncoding
U12
snRNA
gene
(RNU12)
has
recently
been
associated
with
an
early-onset
cerebellar
ataxia
(EOCA)
[28].
In
general,
ataxias
are
a
diverse
group
of
disorders
characterized
by
defects
in
muscle
coordination
which,
in
the
case
of
cerebellar
ataxias,
results
from
abnormal
development
and/or
degeneration
of
the
cerebellum.
The
patients
in
this
single
study
are
homozygous
for
a
point
mutation
(84C
>
T;
see
Fig.
2B)
with
symptoms
of
hypotonia
at
infancy,
delayed
motor
development,
abnormal
gait,
speech
and
learning
difficulties
[28].
RNAseq
and
RT-PCR
analyses
of
patient
mononuclear
cells
showed
increased
U12-type
intron
retention
and
upregulation
of
the
U12
snRNA.
The
84C
>
T
mutation
is
located
at
the
base
of
U12
snRNA
stem-
loop
III,
and
is
predicted
to
weaken
the
stem-closing
G-C
base-pair
by
converting
it
to
a
weaker
G-U
base-pair
(Fig.
2B).
Mechanistic
understanding
of
how
this
mutation
leads
to
a
defect
in
minor
spliceosome
function
is
currently
lacking.
The
mutation
does
not
directly
overlap
with
any
known
functional
elements,
such
as
sequences
involved
in
known
RNA–RNA
or
RNA–protein
interac-
tions;
however,
the
apical
part
of
this
long
stem-loop
acts
as
the
binding
site
for
the
65K
protein
(Fig
2B;
[33])
and
the
assembly
site
for
the
Sm
protein
ring,
an
integral
part
of
all
snRNPs,
is
located
only
two
nucleotides
away.
Furthermore,
as
the
U12
snRNA
not
only
recognizes
the
branch
point
sequence
during
intron
recogni-
tion,
but
is
also
part
of
the
catalytic
core
of
the
minor
spliceosome,
several
additional
mechanistic
scenarios
are
conceivable.
Further
functional
studies
are
needed
to
uncover
the
mechanistic
basis
of
this
disease.
2.1.3.
Myelodysplastic
syndrome
Myelodysplastic
syndrome
(MDS;
myelodysplasia)
is
a
term
for
a
group
of
disorders
characterized
by
inefficient
hematopoiesis,
with
a
risk
of
progression
to
acute
myeloid
leukemia
(AML).
In
recent
years,
several
whole-exome
sequencing
studies
have
revealed
frequent
somatic
mutations
in
genes
encoding
splicing
factors,
including
U2AF1,
ZRSR2,
SRSF2
and
SF3B1
in
patients
with
MDS
reviewed
by
[9,39].
Although
patients
with
a
combination
of
different
splicing
factor
mutations
have
been
reported,
typically
only
one
splicing
factor
gene
in
each
patient
has
been
mutated.
Fur-
thermore,
the
splicing
factor
mutations
frequently
co-occur
with
mutations
in
genes
encoding
epigenetic
factors,
cell
signaling
reg-
ulators
and
transcriptional
regulators
[39,40].
Here,
we
focus
on
the
Urp/ZRSR2
protein
involved
in
U12-type
3ss
recognition.
The
ZRSR2
protein
shares
both
primary
structure
and
functional
features
with
the
splicing
factor
U2AF1,
that
facilitates
3ss
recogni-
tion
with
the
major
introns.
Both
contain
a
central
U2AF-homology
domain
(UHM)
flanked
on
each
side
by
a
CCCH-type
zinc
finger,
and
an
RS
domain
near
the
C-terminus
(Fig.
2D).
ZRSR2
is
an
integral
component
of
the
U11/U12
di-snRNP
and
functions
in
the
recogni-
tion
of
the
U12-type
3splice
site,
but
it
has
also
been
reported
to
function
during
the
second
catalytic
step
of
U2-type
intron
splicing
[19,25].
MDS-associated
mutations
in
ZRSR2
include
a
wide
spec-
trum
of
nonsense,
missense,
frameshift
and
splice
site
mutations
which
are
uniformly
scattered
across
the
gene
(Fig.
2D).
This
distri-
bution
is
in
striking
contrast
to
MDS
cases
with
mutations
in
U2AF1,
SRSF2
and
SF3B1
genes
that
show
a
narrower
mutation
spectrum,
with
particular
preference
for
missense
mutations
that
are
located
mostly
within
a
small
number
of
hotspot
positions.
Of
the
numerous
somatic
mutations
reported
in
the
ZRSR2
(Fig.
2D),
only
few
have
been
studied
in
detail
[29].
In
the
Madan
et
al.
[29]
RNAseq
study,
bone
marrow
samples
from
eight
ZRSR2-
mutant
MDS
patients,
each
carrying
a
different
mutation,
were
compared
to
both
ZRSR2
wild-type
MDS
patients
and
healthy
indi-
viduals.
The
analysis
revealed
a
global
increase
in
U12-type
intron
retention,
as
well
as
activation
of
cryptic
U2-type
splice
sites
only
in
MDS
patients
carrying
the
mutated
ZRSR2
allele.
Importantly,
the
effect
of
ZRSR2
mutations
and
knockdown
of
ZRSR2
on
U2-type
intron
splicing
were
comparatively
small.
The
affected
U2-type
introns
were
mostly
in
genes
containing
U12-type
introns.
Similar
conclusions
have
also
been
reported
in
plants
[41].
These
findings
are
in
stark
contrast
to
the
earlier
biochemical
experiments
which
suggested
that
an
essential
role
for
ZRSR2
was
in
splicing
of
both
U2-
and
U12-type
introns
[19].
2.2.
Diseases
affecting
the
formation
of
catalytically
active
spliceosome
2.2.1.
MOPD1/TALS
Recessive
mutations
in
the
U4atac
snRNA
gene
(RNU4atac)
associated
with
MOPD1/TALS,
a
rare
autosomal
recessive
disease,
represent
the
first
human
disease
with
mutations
in
a
specific
component
of
the
minor
spliceosome
[31,32,42].
U4atac
snRNA
is
an
essential
component
of
the
U4atac/U6atac.U5
tri-snRNP
(minor
tri-snRNP),
which
joins
the
nascent
spliceosome
after
the
intron
boundaries
have
been
recognized
by
the
U11/U12
di-snRNP
(Fig.
1B).
During
the
assembly
of
the
minor
tri-snRNP,
U4atac
forms
an
extensively
base-paired
duplex
with
the
U6atac
snRNA
(Figs.
1B
and
2C).
This
U4atac/U6atac
di-snRNA
complex
forms
a
platform
for
the
binding
of
tri-snRNP-specific
proteins
that
are
needed
for
the
association
of
the
U5
snRNP
to
the
complex.
MOPD1/TALS
in
its
severe
form
is
characterized
by
intrauterine
and
postnatal
growth
retardation,
developmental
defects
in
sev-
eral
organs,
including
microcephaly,
and
death
typically
within
3
years
after
birth.
However,
milder
forms
of
the
disease
have
been
reported
displaying
more
subtle
growth
retardation,
less
severe
developmental
defects
and/or
survival
to
at
least
to
12
years
of
age
or
even
adulthood
[43,44].
Presently,
individual
single-nucleotide
point
mutations
at
ten
different
positions
within
the
U4atac
snRNA
have
been
associated
with
MOPD1/TALS
(Fig.
2C);
in
addition,
one
patient
with
a
duplication
of
U4atac
nucleotides
16–100
has
been
reported
[44].
All
patients
are
either
homozygous
or
compound
heterozygous
for
the
disease
mutations.
In
contrast,
heterozy-
gous
parents
carrying
only
a
single
disease-causing
mutation
are
phenotypically
normal,
suggesting
that
a
loss
of
a
single
allele
is
well-tolerated
at
the
cellular
level.
The
majority
of
disease-causing
mutations,
particularly
those
leading
to
severe
forms
of
the
disease,
are
found
in
the
U4atac
5stemloop,
with
only
few
pathogenic
mutations
located
else-
where
in
the
U4atac
snRNA
(Fig.
2C).
Earlier
studies
from
the
major
spliceosome
have
indicated
that
this
region
is
recognized
by
a
group
of
proteins
(Fig.
2C),
particularly
the
15.5K
(Snu13)
and
PRPF31
pro-
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
ARTICLE IN PRESS
G Model
YSCDB-2404;
No.
of
Pages
10
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
region
reduce
the
15.5K
binding,
which
in
turn
decreases
the
cellu-
lar
levels
of
U4atac/U6atac.U5
tri-snRNP
[46].
Importantly,
there
is
a
correlation
between
disease
severity
and
the
affinity
of
the
15.5K
protein.
Mutations
on
the
tip
of
the
5stemloop
that
reduce
most
the
15.5K
binding,
also
lead
to
severe
forms
of
the
disease.
In
con-
trast,
the
more
distal
mutations
(such
as
U4atac
positions
30,
53
and
55–see
Fig.
2C)
have
little
effect
on
the
binding
of
the
15.5K
pro-
tein
and
are
consequently
associated
with
milder
symptoms,
such
as
longer
life
expectancy
[43,46].
Additionally,
at
least
one
of
the
mutations
(124G
>
A)
reduces
the
U4atac
snRNA
expression
levels
[46].
No
transcriptome
studies
have
yet
been
conducted
for
MOPD1/TALS,
but
qPCR
analyses
have
shown
increased
U12-type
intron
retention
in
patient
cells
that
can
be
rescued
by
expression
of
wild
type
U4atac
snRNA
[31,32].
Significantly,
an
analysis
with
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
iPS
cells
carrying
the
mutation
show
much
milder
splicing
defects
[31,32,46].
2.2.2.
Roifman
syndrome
In
addition
to
MOPD1/TALS,
mutations
in
the
RNU4atac
locus
have
been
associated
with
a
phenotypically
different
Roifman
syndrome
(RFMN)
[30].
Patients
suffering
from
RFMN
are
char-
acterized
by
poor
pre-
and
post-natal
growth,
distinct
facial
dystrophies,
cognitive
delay
and
immunological
defects
which,
with
the
exception
of
growth
defects,
are
distinct
to
Roifman
patients
[30].
Interestingly,
all
RFMN
patients
are
compound
het-
erozygotes,
with
one
mutation
shared
with
MOPD1/TALS
patients
with
severe
forms
of
the
diseases
(Fig.
2C).
The
other
mutation
is
predominantly
located
among
the
highly
conserved
positions
of
the
RNU4atac
stem
II,
except
for
a
single
case
with
a
mutation
within
the
Sm
site.
No
functional
studies
have
been
performed
with
the
RFMN
mutations,
but
it
is
safe
to
assume
that
the
mutations
shared
with
the
MOPD1/TALS
lead
to
a
similar
defect
in
minor
tri-snRNP
assem-
bly
[46].
In
contrast,
the
stem
II
mutations
presumably
lead
to
a
milder
functional
defect
in
minor
splicing,
given
the
overall
less
severe
disease
phenotype.
While
the
molecular
defects
resulting
from
the
stem
II
mutations
remain
to
be
determined,
the
investiga-
tion
from
the
major
spliceosome
assembly
tentatively
suggest
that
the
association
of
the
PRPF4/PRPF3/PPIH
complex
(Fig.
2C)
with
the
stem
II
may
be
compromised
[30,47,48].
In
addition,
RNAseq
analyses
of
the
patient
cells
revealed
a
widespread
retention
of
U12-type
introns.
Significantly,
neither
increased
retention
of
U2-type
introns,
nor
other
types
of
alter-
native
splicing
changes
were
detected
in
the
patient
cells
[30],
suggesting
a
very
specific
splicing
defect
affecting
U12-type
introns
only.
2.3.
Diseases
with
mutations
in
the
U12-type
splice
sites
To
our
knowledge
there
are
only
two
reported
cases
in
lit-
erature
that
specifically
attribute
U12-type
splice
site
mutations
with
human
disease.
These
include
the
autosomal
dominant
dis-
order
Peutz-Jeghers
syndrome
(PJS),
characterized
by
the
presence
of
multiple
intestinal
polyps
and
a
high
risk
for
cancer
[49],
and
Spondyloepiphyseal
dysplasia
tarda
(SEDT),
an
X-linked
recessive
disorder
[50].
Both
are
caused
by
mutations
in
a
U12-type
5ss
that
lead
to
incorrect
splicing
of
the
respective
mRNAs.
Additionally,
the
Motor
endplate
disease
in
mouse
reports
a
mutation
of
a
U2-type
5ss
in
a
downstream
intron,
leading
to
skipping
of
the
upstream
U12-type
intron
[51].
These
three
cases
do
not
only
demonstrate
that
a
single
point
mutation
in
the
highly
conserved
U12-type
5ss
sequence
can
efficiently
disable
the
U12-type
intron
recognition,
but
also
that
splicing
of
a
U12-type
intron
is
linked
to
other
splicing
events
in
the
same
transcript
and
that
in
some
cases
the
outcome
may
be
difficult
to
predict.
PJS,
in
which
a
mutation
at
the
+1
position
of
an
AT–AC-type
U12-type
intron
is
changed
to
G,
is
a
particu-
larly
illuminating
case
[49].
At
the
sequence
level,
it
appears
to
be
a
simple
U12-type
5ss
subtype
change,
but
the
experimentally
determined
outcome
is
in
fact
complex,
showing
activation
of
sev-
eral
non-canonical
3splice
sites
that
illuminate
the
flexibility
of
the
U12-dependent
spliceosome
in
3ss
recognition.
Even
though
the
outcome
in
each
case
is
the
same,
i.e.
introduction
of
a
pre-
mature
STOP-codon
and
mRNA
decay
via
the
NMD
pathway,
this
particular
mutation
demonstrates
that
simple
point
mutations
can
lead
to
unexpected
consequences.
2.4.
Other
spliceosome
mutations
affecting
minor
spliceosome
activity
In
addition
to
diseases
caused
by
mutations
in
the
specific
components
of
the
minor
spliceosome,
there
are
several
addi-
tional
human
diseases
caused
by
congenital
or
somatic
mutations
in
either
the
shared
components
of
the
two
spliceosomes
or
the
assembly
factors
necessary
for
both
major
and
minor
snRNPs.
Examples
of
the
former
include
Retinitis
pigmentosa
(RP)
mutations
in
genes
encoding
the
tri-snRNP
specific
proteins
PRPF3,
PRPF4,
PRPF6,
PRPF8,
PRPF31
and
BRR2
[Reviewed
in
8],
and
MDS
muta-
tions
in
PRPF8
[52],
in
SF3b1,
a
component
of
both
U2
snRNP
and
U11/U12
di-snRNP
[26],
and
in
SRSF2,
a
splicing
activator
poten-
tially
affecting
both
spliceosomes
[53].
Spinal
muscular
atrophy
(SMA)
represents
the
latter
group
and
is
caused
by
mutations
in
the
SMN1
gene,
coding
for
an
essential
factor
for
the
assembly
of
all
Sm-class
snRNPs,
including
components
of
both
major
and
minor
spliceosomes
[54].
With
RP,
the
shared
protein
composition
between
the
minor
and
major
tri-snRNPs
[17]
and
functional
studies
[55,56]
predict
that
the
mutations
should
have
an
equal
effect
on
both
the
major
and
minor
tri-snRNPs.
With
SMA,
however,
several
investigations
using
different
model
systems
have
reported
a
preferential
reduc-
tion
in
the
cellular
levels
of
minor
snRNPs
[10,57–61].
In
a
subset
of
the
studies
this
has
been
reported
to
lead
to
an
increased
retention
U12-type
introns,
suggesting
that
defects
in
the
splicing
of
minor
introns
may
contribute
to
the
SMA
symptoms.
In
a
Drosophila
model
of
SMA
the
effects
on
neuronal
cells
were
attributed
to
a
single
U12-type
intron
containing
gene,
Stasimon,
encoding
a
transmem-
brane
protein
required
for
axonal
growth
and
regulation
of
synaptic
transmission
of
motor
neurons
[60].
However,
some
aspects
of
this
study
have
been
challenged
subsequently
[62].
Finally,
two
independent
studies
have
linked
minor
spliceo-
some
to
amyotrophic
lateral
sclerosis
(ALS).
Ishihara
et
al.
[63]
reported
that
ALS-associated
depletion
of
TDP-43
protein
leads
to
a
co-depletion
of
U12
snRNA
both
in
cultured
cells
and
in
patient
samples,
presumably
due
to
defects
in
snRNP
assembly.
In
another
study
[64],
ALS-associated
mutations
in
the
Fused
in
sarcoma
(FUS)
protein
lead
to
reduced
nuclear
import
of
FUS
and
its
accumulation
in
the
cytoplasm.
FUS
is
a
multifunctional
RNA-binding
protein
that
interacts
with
several
splicing
factors,
including
U11
snRNP
and
is
needed
for
the
splicing
of
at
least
a
subset
of
minor
introns
[64].
Sig-
nificantly,
the
cytoplasmic
accumulation
of
a
particular
FUS
mutant
leads
to
a
concomitant
mislocalization
of
U11
and
U12
snRNPs
in
the
cytoplasm,
which
in
turn
leads
to
defects
in
the
splicing
of
U12-type
introns.
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
ARTICLE IN PRESS
G Model
YSCDB-2404;
No.
of
Pages
10
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
In
addition
to
being
a
target
for
disease-causing
mutations,
sev-
eral
components
of
the
minor
spliceosome
have
been
reported
as
cancer
biomarkers.
A
recent
study
by
Xu
et
al.
[65]
using
serum
from
patients
with
scleroderma
and
coincident
cancer
reported
autoantibodies
targeted
to
components
of
the
U11/U12
di-snRNP,
particularly
the
U11/U12-65K
protein,
but
also
the
25K,
35K
and
59K
proteins.
Of
these,
the
U11/U12-65K
autoantibodies
appear
to
show
a
reliable
association
with
an
increased
risk
of
cancer
occurring
within
2
years
of
the
onset
of
scleroderma
[66].
Similar
associations
have
been
described
for
U11
snRNA
in
familial
prostate
cancers
[67]
and
U11-59K
protein
in
Acute
Myeloid
Leukemia
[68].
However,
the
role
of
minor
spliceosome-specific
autoantibodies
in
the
pathogenesis
of
scleroderma
and/or
coincident
cancer,
or
the
expression
level
changes
of
other
minor
spliceosome
components
in
other
diseases
is
presently
unclear.
4.
Consequences
of
minor
spliceosome-specific
mutations
Presently,
mutations
in
four
specific
components
of
the
minor
spliceosome
have
been
associated
with
human
diseases
showing
very
different
disease
phenotypes.
RT-PCR
and
RNAseq
analyses
have
demonstrated
that
each
of
the
five
human
diseases
show
the
expected
molecular
level
phenotype,
i.e.
the
increased
levels
of
unspliced
U12-type
introns
in
the
patient
cells.
With
a
subset
of
diseases,
additional
splicing
defects,
particularly
activation
of
cryp-
tic
U2-type
splice
sites
and
exon
skipping
events
have
also
been
described.
Presently,
there
has
not
been
clear
consensus
on
how
to
interpret
the
molecular
consequences
of
minor
spliceosome
muta-
tions.
Here,
we
propose
a
simple
classification
of
disease-causing
mutations
according
to
their
impact
on
the
recognition
of
the
U12-
type
introns.
This
provides
a
simple
framework
for
predicting
the
molecular
consequences
of
the
mutations,
but
may
also
help
to
interpret
the
subsequent
disease
phenotypes.
4.1.
The
fate
of
the
mRNA:
intron
retention
vs
cryptic
splicing
According
to
this
classification
the
mutations
associated
with
both
the
MOPD1/TALS
and
RFMN
affect
the
formation
of
the
U4atac/U6atac.U5
tri-SNRP
(see
2.2.1
and
2.2.2),
but
not
the
initial
intron
recognition
step.
Therefore,
in
these
diseases
the
spliceo-
some
assembly
is
arrested
at
the
pre-spliceosome
stage
where
the
U11/U12
di-snRNP
is
bound
to
the
intron
but
the
subsequent
assembly
steps
are
inhibited.
Consequently,
the
bound
U11/U12
di-snRNP
can
be
expected
to
maintain
exon-definition
interactions
with
the
surrounding
(U2-type)
introns
via
the
RS
domain
contain-
ing
U11-35K
[69]
and
possibly
Urp/ZRSR2
proteins.
Furthermore,
the
U11/U12
di-snRNP
bound
to
the
partially
spliced
mRNA
can
trap
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
U12-type
introns
in
IGHD
and
MDS
(and
possibly
EOCA)
leads
to
a
different
outcome.
Due
to
the
loss
of
U11/U12
binding,
transcripts
containing
unspliced
U12-type
introns
are
free
to
be
exported
to
the
cytoplasm.
Concomitantly,
the
loss
of
local
exon
definition
interac-
tions
may
lead
to
increased
activation
of
cryptic
splice
sites
or
exon
skipping
events
near
the
U12-type
introns
[73].
The
outcome
can
be
either
truncated
or
altered
protein,
or
mRNA
decay
via
the
NMD
quality
control
pathway
(Fig.
3).
Is
there
evidence
supporting
this
model?
RNAseq
analyses
from
both
IGHD
and
MDS
patient
cells
provide
extensive
evidence
of
acti-
vation
of
cryptic
U2-type
splice
sites
and
increased
levels
of
exon
skipping
near
U12-type
introns
in
addition
to
the
expected
reten-
tion
of
U12-type
introns
[27,29].
In
contrast,
besides
the
increased
intron
retention
of
U12-type
introns
seen
with
both
MOPD1/TALS
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
ARTICLE IN PRESS
G Model
YSCDB-2404;
No.
of
Pages
10
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
detailed
RNAseq
analysis
of
RFMN
patient
cells.
Together,
present
evidence
supports
the
proposal
that
the
consequences
of
the
minor
spliceosome
mutations
are
linked
to
the
type
of
assembly
defect
introduced.
However,
with
both
MOPD1/TALS
and
RFMN
it
is
pos-
sible
that
some
arrested
transcripts
trapped
in
the
nucleus
may
either
lose
the
bound
U11/U12
di-snRNP
or
undergo
the
splicing
process
post-transcriptionally
(see
Fig.
3).
4.2.
Fate
of
the
mRNA
and
disease
severity
It
is
somewhat
puzzling
to
note
that
the
severity
of
congen-
ital
minor
spliceosome
diseases
and
the
level
of
mRNA
splicing
defect
follow
a
counterintuitive
association
as
described
above.
The
two
diseases
showing
only
relatively
mild
intron
retention
defects
(MOPD1/TALS
and
RFMN)
show
pleiotropic,
and
often
severe,
defects
in
many
tissues
[30–32].
In
contrast,
IGHD
and
EOCA
show
large
splicing
defects
that
were
detected
presumably
ubiq-
uitously
also
from
cells/tissues
not
associated
with
the
disease
[27,28].
Even
though
the
present
small
dataset
does
not
allow
gen-
eralization,
it
is
tempting
to
speculate
that
the
difference
may
be
linked
to
the
fate
of
the
unspliced
mRNAs.
Specifically,
it
is
possible
that
the
unspliced
U12-type
introns
or
transcripts
con-
taining
cryptic
splicing
events
are
better
tolerated
when
exported
to
cytoplasm
where
they
can
be
targeted
by
the
cytoplasmic
qual-
ity
control
mechanisms.
Transcripts
trapped
in
the
nucleus
may,
in
contrast,
accumulate
in
some
tissues
or
lead
to
other
detrimen-
tal
downstream
effects.
This
hypothesis
receives
tentative
support
from
a
zebrafish
study
where
a
specific
65K
mutation
arrests
the
spliceosome
assembly
at
a
late
stage
and
presumably
traps
such
mRNAs
in
the
nucleus.
Unlike
the
more
restricted
phenotype
seen
with
IGHD
65K
mutations,
the
zebrafish
phenotype
is
severe,
with
developmental
defects
in
multiple
organs
[37].
4.3.
Defects
with
individual
genes
vs
a
global
splicing
defect
One
of
the
outstanding
questions
is
whether
the
pathologi-
cal
consequences
of
the
minor
spliceosome
diseases
are
caused
by
a
global
defect
in
the
splicing
of
U12-type
introns
or
whether
the
various
effects
seen
in
different
tissues
are
caused
by
mis-
splicing
of
a
small
subset
of
genes
containing
U12-type
introns.
Gene
expression
analyses
have
suggested
various
candidate
genes
responsible
for
the
observed
disease
phenotypes.
These
must,
how-
ever,
be
interpreted
with
caution
since
in
all
cases
except
MDS,
the
gene
expression
analyses
have
been
done
using
cells
other
than
the
actual
affected
tissue.
Furthermore,
it
is
not
yet
possible
to
conclude
whether
a
par-
ticular
tissue
or
cell
type
would
be
more
susceptible
to
defects
in
the
U12-dependent
spliceosome.
Of
the
present
congenital
minor
spliceosome
diseases,
the
only
common
target
appears
to
be
the
nervous
system.
Specifically,
microcephaly
has
been
reported
for
three
of
the
diseases
(severe
for
MOPD1/TALS,
mild
for
RFMN
and
IGHD),
which
with
MOPD1/TALS
is
associated
with
struc-
tural
abnormalities
of
brain.
Additionally,
RFMN
and
EOCA
patients
display
cognitive
delay
and
cerebellar
ataxia,
respectively.
The
observed
tissue-specific
effects
may
be
linked
to
the
associations
of
the
minor
spliceosome
activity
with
cell
proliferation
[37,74]
or
cel-
lular
response
to
stress
[75].
Consistently,
recent
work
has
reported
that
components
of
the
U11/U12
di-snRNP,
including
the
65K,
48K
and
Urp/ZRSR2
proteins
are
downregulated
during
neuron
terminal
differentiation
[70].
5.
Future
perspective
The
recent
discoveries
of
human
diseases
affecting
the
minor
spliceosome
have
significantly
advanced
our
understanding
of
the
significance
and
function
of
the
U12-dependent
spliceosome.
How-
ever,
many
aspects
of
the
disease
process,
including
the
basis
of
the
tissue
specificity
and
the
detailed
mechanism
of
the
disease
process
are
yet
to
be
determined
for
most
diseases,
requiring
animal
models
of
each
disease.
These
will
also
help
to
address
the
more
fundamen-
tal
question
of
the
existence
of
two
separate
spliceosomes.
Acknowledgements
This
work
was
supported
by
the
Academy
of
Finland
(Grant
140087
to
MJF
and
Grant
278798
to
BV)
and
Sigrid
Jusélius
Foun-
dation
(MJF).
AJN
was
supported
by
the
Integrative
Life
Science
doctoral
program
at
the
University
of
Helsinki.
References
[1]
N.
Lopez-Bigas,
B.
Audit,
C.
Ouzounis,
G.
Parra,
R.
Guigo,
Are
splicing
mutations
the
most
frequent
cause
of
hereditary
disease?
FEBS
Lett.
579
(9)
(2005)
1900–1903.
[2]
K.H.
Lim,
L.
Ferraris,
M.E.
Filloux,
B.J.
Raphael,
W.G.
Fairbrother,
Using
positional
distribution
to
identify
splicing
elements
and
predict
pre-mRNA
processing
defects
in
human
genes,
Proc.
Natl.
Acad.
Sci.
U.
S.
A.
108
(27)
(2011)
11093–11098.
[3]
Y.I.
Li,
B.
van
de
Geijn,
A.
Raj,
D.A.
Knowles,
A.A.
Petti,
D.
Golan,
Y.
Gilad,
J.K.
Pritchard,
RNA
splicing
is
a
primary
link
between
genetic
variation
and
disease,
Science
352
(6285)
(2016)
600–604.
[4]
F.
Supek,
B.
Mi˜
nana,
J.
Valcárcel,
T.
Gabaldón,
B.
Lehner,
Synonymous
mutations
frequently
act
as
driver
mutations
in
human
cancers,
Cell
156
(6)
(2014)
1324–1335.
[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.
[6]
H.Y.
Xiong,
B.
Alipanahi,
L.J.
Lee,
H.
Bretschneider,
D.
Merico,
R.K.C.
Yuen,
Y.
Hua,
S.
Gueroussov,
H.S.
Najafabadi,
T.R.
Hughes,
Q.
Morris,
Y.
Barash,
A.R.
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
347
(6218)
(2015).
[7]
R.
Soemedi,
K.J.
Cygan,
C.L.
Rhine,
J.
Wang,
C.
Bulacan,
J.
Yang,
P.
Bayrak-Toydemir,
J.
McDonald,
W.G.
Fairbrother,
Pathogenic
variants
that
alter
protein
code
often
disrupt
splicing,
Nat.
Genet.
49
(6)
(2017)
848–855.
[8] ˇ
S.
R˚
uˇ
ziˇ
cková,
D.
Stanˇ
ek,
Mutations
in
spliceosomal
proteins
and
retina
degeneration,
RNA
Biol.
14
(5)
(2017)
544–552.
[9]
D.
Inoue,
R.K.
Bradley,
O.
Abdel-Wahab,
Spliceosomal
gene
mutations
in
myelodysplasia:
molecular
links
to
clonal
abnormalities
of
hematopoiesis,
Genes
Dev.
30
(9)
(2016)
989–1001.
[10]
Z.
Zhang,
F.
Lotti,
K.
Dittmar,
I.
Younis,
L.
Wan,
M.
Kasim,
G.
Dreyfuss,
SMN
deficiency
causes
tissue-specific
perturbations
in
the
repertoire
of
snRNAs
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
by
the
minor
spliceosome,
Wiley
Interdiscip.
Rev.
RNA
4
(2013)
61–76.
[12]
C.B.
Burge,
R.A.
Padgett,
P.A.
Sharp,
Evolutionary
fates
and
origins
of
U12-type
introns,
Mol.
Cell
2
(1998)
773–785.
[13]
Q.
Wu,
A.R.
Krainer,
AT-AC
pre-mRNA
splicing
mechanism
and
conservation
of
minor
introns
in
voltage-gated
ion
channel
genes,
Mol.
Cell
Biol.
19
(1999)
3225–3236.
[14]
G.W.
Yeo,
E.L.
Van
Nostrand,
T.Y.
Liang,
Discovery
and
analysis
of
evolutionarily
conserved
intronic
splicing
regulatory
elements,
PLoS
Genet.
3
(5)
(2007)
e85.
[15]
M.K.
Basu,
W.
Makalowski,
I.B.
Rogozin,
E.V.
Koonin,
U12
intron
positions
are
more
strongly
conserved
between
animals
and
plants
than
U2
intron
positions,
Biol.
Direct.
3
(2008)
19.
[16]
C.L.
Will,
R.
Lührmann,
Splicing
of
a
rare
class
of
introns
by
the
U12-dependent
spliceosome,
Biol.
Chem.
386
(8)
(2005)
713–724.
[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
compositions,
Mol.
Cell
Biol.
22
(10)
(2002)
3219–3229.
[18]
M.J.
Frilander,
J.A.
Steitz,
Initial
recognition
of
U12-dependent
introns
requires
both
U11/5’
splice-site
and
U12/branchpoint
interactions,
Genes
Dev.
13
(1999)
851–863.
[19]
H.
Shen,
X.
Zheng,
S.
Luecke,
M.R.
Green,
The
U2AF35-related
protein
Urp
contacts
the
3’
splice
site
to
promote
U12-type
intron
splicing
and
the
second
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
for
splicing
rare
AT-AC
introns,
Science
273
(1996)
1824–1832.
[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
catalytic
core
formation
in
the
U12-dependent
spliceosome,
Mol.
Cell
7
(1)
(2001)
217–226.
[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.
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
ARTICLE IN PRESS
G Model
YSCDB-2404;
No.
of
Pages
10
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)
(2016)
19–32.
[25]
C.L.
Will,
C.
Schneider,
M.
Hossbach,
H.
Urlaub,
R.
Rauhut,
S.
Elbashir,
T.
Tuschl,
R.
Lührmann,
The
human
18S
U11/U12
snRNP
contains
a
set
of
novel
proteins
not
found
in
the
U2-dependent
spliceosome,
RNA
10
(6)
(2004)
929–941.
[26]
C.L.
Will,
C.
Schneider,
R.
Reed,
R.
Lührmann,
Identification
of
both
shared
and
distinct
proteins
in
the
major
and
minor
spliceosomes,
Science
284
(1999)
2003–2005.
[27]
J.
Argente,
R.
Flores,
A.
Gutiérrez-Arumí,
B.
Verma,
G.A.
Martos-Moreno,
I.
Cuscó,
A.
Oghabian,
J.A.
Chowen,
M.J.
Frilander,
L.A.
Pérez-Jurado,
Defective
minor
spliceosome
mRNA
processing
results
in
isolated
familial
growth
hormone
deficiency,
EMBO
Mol.
Med.
6
(2014)
299–306.
[28]
M.F.
Elsaid,
N.
Chalhoub,
T.
Ben-Omran,
P.
Kumar,
H.
Kamel,
K.
Ibrahim,
Y.
Mohamoud,
E.
Al-Dous,
I.
Al-Azwani,
J.A.
Malek,
K.
Suhre,
M.E.
Ross,
A.A.
Aleem,
Mutation
in
noncoding
RNA
RNU12
causes
early
onset
cerebellar
ataxia,
Ann.
Neurol.
81
(1)
(2017)
68–78.
[29]
V.
Madan,
D.
Kanojia,
J.
Li,
R.
Okamoto,
A.
Sato-Otsubo,
A.
Kohlmann,
M.
Sanada,
V.
Grossmann,
J.
Sundaresan,
Y.
Shiraishi,
S.
Miyano,
F.
Thol,
A.
Ganser,
H.
Yang,
T.
Haferlach,
S.
Ogawa,
H.P.
Koeffler,
Aberrant
splicing
of
U12-type
introns
is
the
hallmark
of
ZRSR2
mutant
myelodysplastic
syndrome,
Nat.
Commun.
6
(2015)
6042.
[30]
D.
Merico,
M.
Roifman,
U.
Braunschweig,
R.K.C.
Yuen,
R.
Alexandrova,
A.
Bates,
B.
Reid,
T.
Nalpathamkalam,
Z.
Wang,
B.
Thiruvahindrapuram,
P.
Gray,
A.
Kakakios,
J.
Peake,
S.
Hogarth,
D.
Manson,
R.
Buncic,
S.L.
Pereira,
J.-A.
Herbrick,
B.J.
Blencowe,
C.M.
Roifman,
S.W.
Scherer,
Compound
heterozygous
mutations
in
the
noncoding
RNU4ATAC
cause
Roifman
Syndrome
by
disrupting
minor
intron
splicing,
Nat.
Commun.
6
(2015)
8718.
[31]
H.
He,
S.
Liyanarachchi,
K.
Akagi,
R.
Nagy,
J.
Li,
R.C.
Dietrich,
W.
Li,
N.
Sebastian,
B.
Wen,
B.
Xin,
J.
Singh,
P.
Yan,
H.
Alder,
E.
Haan,
D.
Wieczorek,
B.
Albrecht,
E.
Puffenberger,
H.
Wang,
J.A.
Westman,
R.A.
Padgett,
D.E.
Symer,
A.
de
la
Chapelle,
Mutations
in
U4atac
snRNA,
a
component
of
the
minor
spliceosome,
in
the
developmental
disorder
MOPD
I,
Science
332
(6026)
(2011)
238–240.
[32]
P.
Edery,
C.
Marcaillou,
M.
Sahbatou,
A.
Labalme,
J.
Chastang,
R.
Touraine,
E.
Tubacher,
F.
Senni,
M.B.
Bober,
S.
Nampoothiri,
P.-S.
Jouk,
E.
Steichen,
S.
Berland,
A.
Toutain,
C.A.
Wise,
D.
Sanlaville,
F.
Rousseau,
F.
Clerget-Darpoux,
A.-L.
Leutenegger,
Association
of
TALS
developmental
disorder
with
defect
in
minor
splicing
component
U4atac
snRNA,
Science
332
(6026)
(2011)
240–243.
[33]
H.
Benecke,
R.
Lührmann,
C.L.
Will,
The
U11/U12
snRNP
65K
protein
acts
as
a
molecular
bridge,
binding
the
U12
snRNA
and
U11-59K
protein,
EMBO
J.
24
(17)
(2005)
3057–3069.
[34]
M.
Guceva,
V.
Polenakovicb,
Y.
Tasica,
J.
LeBoucc,
J.
Klammtd,
A.
Pfaeffled,
Severe
isolated
growth
hormone
deficiency
and
myopathy
in
two
brothers
with
RNPC3
mutation,
Horm.
Res.
Paediatr.
84
(Suppl.
1)
(2015)
447.
[35]
K.S.
Alatzoglou,
M.T.
Dattani,
Genetic
causes
and
treatment
of
isolated
growth
hormone
deficiency—an
update,
Nat.
Rev.
Endocrinol.
6
(10)
(2010)
562–576.
[36]
A.J.
Norppa,
T.
Kauppala,
H.A.
Heikkinen,
B.
Verma,
H.
Iwai,
M.J.
Frilander,
Mutations
in
the
U11/U12-65
K
protein
associated
with
isolated
growth
hormone
deficiency
lead
to
structural
destabilization
and
impaired
binding
of
U12
snRNA,
RNA,
in
press.
[37]
S.
Markmiller,
N.
Cloonan,
R.M.
Lardelli,
K.
Doggett,
M.-C.
Keightley,
Y.
Boglev,
A.J.
Trotter,
A.Y.
Ng,
S.J.
Wilkins,
H.
Verkade,
E.A.
Ober,
H.A.
Field,
S.M.
Grimmond,
G.J.
Lieschke,
D.Y.R.
Stainier,
J.K.
Heath,
Minor
class
splicing
shapes
the
zebrafish
transcriptome
during
development,
Proc.
Natl.
Acad.
Sci.
U.
S.
A.
111
(2014)
3062–3067.
[38]
J.
Singh,
K.
Sikand,
H.
Conrad,
C.L.
Will,
A.A.
Komar,
G.C.
Shukla,
U6atac
snRNA
stem-loop
interacts
with
U12
p65
RNA
binding
protein
and
is
functionally
interchangeable
with
the
U12
apical
stem-loop
III,
Sci.
Rep.
6
(2016)
31393.
[39]
B.
Saez,
M.J.
Walter,
T.A.
Graubert,
Splicing
factor
gene
mutations
in
hematologic
malignancies,
Blood
129
(10)
(2017)
1260–1269.
[40]
S.A.
Mian,
A.E.
Smith,
A.G.
Kulasekararaj,
A.
Kizilors,
A.M.
Mohamedali,
N.C.
Lea,
K.
Mitsopoulos,
K.
Ford,
E.
Nasser,
T.
Seidl,
G.J.
Mufti,
Spliceosome
mutations
exhibit
specific
associations
with
epigenetic
modifiers
and
proto-oncogenes
mutated
in
myelodysplastic
syndrome,
Haematologica
98
(7)
(2013)
1058–1066.
[41]
C.M.
Gault,
F.
Martin,
W.
Mei,
F.
Bai,
J.B.
Black,
W.B.
Barbazuk,
A.M.
Settles,
Aberrant
splicing
in
maize
rough
endosperm3
reveals
a
conserved
role
for
U12
splicing
in
eukaryotic
multicellular
development,
Proc.
Natl.
Acad.
Sci.
U.
S.
A.
114
(11)
(2017)
E2195–E2204.
[42]
H.K.J.
Pessa,
M.J.
Frilander,
Minor
splicing,
disrupted,
Science
332
(6026)
(2011)
184–185.
[43]
G.M.H.
Abdel-Salam,
M.S.
Abdel-Hamid,
M.
Issa,
A.
Magdy,
A.
El-Kotoury,
K.
Amr,
Expanding
the
phenotypic
and
mutational
spectrum
in
microcephalic
osteodysplastic
primordial
dwarfism
type
I,
Am.
J.
Med.
Genet.
158A
(6)
(2012)
1455–1461.
[44]
A.B.
Krøigård,
A.P.
Jackson,
L.S.
Bicknell,
E.
Baple,
K.
Brusgaard,
L.K.
Hansen,
L.B.
Ousager,
Two
novel
mutations
in
RNU4ATAC
in
two
siblings
with
an
atypical
mild
phenotype
of
microcephalic
osteodysplastic
primordial
dwarfism
type
1,
Clin.
Dysmorphol.
25
(2)
(2016)
68–72.
[45]
O.V.
Makarova,
E.M.
Makarov,
S.
Liu,
H.-P.
Vornlocher,
R.
Lührmann,
Protein
61K,
encoded
by
a
gene
(PRPF31)
linked
to
autosomal
dominant
retinitis
pigmentosa,
is
required
for
U4/U6·U5
tri-snRNP
formation
and
pre-mRNA
splicing,
EMBO
J.
21
(5)
(2002)
1148–1157.
[46]
F.
Jafarifar,
R.C.
Dietrich,
J.M.
Hiznay,
R.A.
Padgett,
Biochemical
defects
in
minor
spliceosome
function
in
the
developmental
disorder
MOPD
I,
RNA
20
(7)
(2014)
1078–1089.
[47]
K.
Nottrott,
P.
Hartmuth,
H.
Fabrizio,
I.
Urlaub,
R.
Vidovic,
R.
Ficner,
Functional
interaction
of
a
novel
15.5kD
[U4/U6
U5]
tri-snRNP
protein
with
the
5’
stem-loop
of
U4
snRNA,
EMBO
J.
18
(21)
(1999)
6119–6133.
[48]
J.W.
Hardin,
C.
Warnasooriya,
Y.
Kondo,
K.
Nagai,
D.
Rueda,
Assembly
and
dynamics
of
the
U4/U6
di-snRNP
by
single-molecule
FRET,
Nucleic
Acids
Res.
43
(22)
(2015)
10963–10974.
[49]
M.L.
Hastings,
N.
Resta,
D.
Traum,
A.
Stella,
G.
Guanti,
A.R.
Krainer,
An
LKB1
AT-AC
intron
mutation
causes
Peutz-Jeghers
syndrome
via
splicing
at
noncanonical
cryptic
splice
sites,
Nat.
Struct.
Mol.
Biol.
12
(1)
(2005)
54–59.
[50]
M.A.
Shaw,
N.
Brunetti-Pierri,
L.
Kádasi,
V.
Kovácová,
L.
Van
Maldergem,
D.
De
Brasi,
M.
Salerno,
J.
Gécz,
Identification
of
three
novel
SEDL
mutations,
including
mutation
in
the
rare,
non-canonical
splice
site
of
exon
4,
Clin.
Genet.
64
(3)
(2003)
235–242.
[51]
D.C.
Kohrman,
J.B.
Harris,
M.H.
Meisler,
Mutation
detection
in
the
med
and
medJ
alleles
of
the
sodium
channel
scn8a:
unusual
splicing
due
to
a
minor
class
AT-AC
intron,
J.
Biol.
Chem.
271
(29)
(1996)
17576–17581.
[52]
A.
Kurtovic-Kozaric,
B.
Przychodzen,
J.
Singh,
M.M.
Konarska,
M.J.
Clemente,
Z.K.
Otrock,
M.
Nakashima,
E.D.
Hsi,
K.
Yoshida,
Y.
Shiraishi,
K.
Chiba,
H.
Tanaka,
S.
Miyano,
S.
Ogawa,
J.
Boultwood,
H.
Makishima,
J.P.
Maciejewski,
R.A.
Padgett,
PRPF8
defects
cause
missplicing
in
myeloid
malignancies,
Leukemia
29
(1)
(2015)
126–136.
[53]
M.L.
Hastings,
A.R.
Krainer,
Functions
of
SR
proteins
in
the
U12-dependent
AT-AC
pre-mRNA
splicing
pathway,
RNA
7
(3)
(2001)
471–482.
[54]
D.K.
Li,
S.
Tisdale,
F.
Lotti,
L.
Pellizzoni,
SMN
control
of
RNP
assembly:
from
post-transcriptional
gene
regulation
to
motor
neuron
disease,
Semin.
Cell
Dev.
Biol.
32
(2014)
22–29.
[55]
H.R.
Luo,
G.A.
Moreau,
N.
Levin,
M.J.
Moore,
The
human
Prp8
protein
is
a
component
of
both
U2-
and
U12-dependent
spliceosomes,
RNA
5
(1999)
893–908.
[56]
M.-C.
Keightley,
M.O.
Crowhurst,
J.E.
Layton,
T.
Beilharz,
S.
Markmiller,
S.
Varma,
B.M.
Hogan,
T.A.
de
Jong-Curtain,
J.K.
Heath,
G.J.
Lieschke,
In
vivo
mutation
of
pre-mRNA
processing
factor
8
(Prpf8)
affects
transcript
splicing,
cell
survival
and
myeloid
differentiation,
FEBS
Lett.
587
(14)
(2013)
2150–2157.
[57]
M.
Jangi,
C.
Fleet,
P.
Cullen,
S.V.
Gupta,
S.
Mekhoubad,
E.
Chiao,
N.
Allaire,
C.F.
Bennett,
F.
Rigo,
A.R.
Krainer,
J.A.
Hurt,
J.P.
Carulli,
J.F.
Staropoli,
SMN
deficiency
in
severe
models
of
spinal
muscular
atrophy
causes
widespread
intron
retention
and
DNA
damage,
Proc.
Natl.
Acad.
Sci.
U.
S.
A.
114
(12)
(2017)
E2347–E2356.
[58]
F.
Gabanella,
M.E.R.
Butchbach,
L.
Saieva,
C.
Carissimi,
A.H.M.
Burghes,
L.
Pellizzoni,
Ribonucleoprotein
assembly
defects
correlate
with
spinal
muscular
atrophy
severity
and
preferentially
affect
a
subset
of
spliceosomal
snRNPs,
PLoS
One
2
(9)
(2007)
e921.
[59]
N.
Boulisfane,
M.
Choleza,
F.
Rage,
H.
Neel,
J.
Soret,
R.
Bordonné,
Impaired
minor
tri-snRNP
assembly
generates
differential
splicing
defects
of
U12-type
introns
in
lymphoblasts
derived
from
a
type
I
SMA
patient,
Hum.
Mol.
Genet.
20
(4)
(2011)
641–648.
[60]
F.
Lotti,
Wendy
L.
Imlach,
L.
Saieva,
Erin
S.
Beck,
Le
T.
Hao,
Darrick
K.
Li,
W.
Jiao,
George
Z.
Mentis,
Christine
E.
Beattie,
Brian
D.
McCabe,
L.
Pellizzoni,
An
SMN-dependent
U12
splicing
event
essential
for
motor
circuit
function,
Cell
151
(2)
(2012)
440–454.
[61]
T.K.
Doktor,
Y.
Hua,
H.S.
Andersen,
S.
Brøner,
Y.H.
Liu,
A.
Wieckowska,
M.
Dembic,
G.H.
Bruun,
A.R.
Krainer,
B.S.
Andresen,
RNA-sequencing
of
a
mouse-model
of
spinal
muscular
atrophy
reveals
tissue-wide
changes
in
splicing
of
U12-dependent
introns,
Nucleic
Acids
Res.
45
(2016)
395–416.
[62]
E.L.
Garcia,
Z.
Lu,
M.P.
Meers,
K.
Praveen,
A.G.
Matera,
Developmental
arrest
of
Drosophila
survival
motor
neuron
(Smn)
mutants
accounts
for
differences
in
expression
of
minor
intron-containing
genes,
RNA
19
(11)
(2013)
1510–1516.
[63]
T.
Ishihara,
Y.
Ariizumi,
A.
Shiga,
T.
Kato,
C.-F.
Tan,
T.
Sato,
Y.
Miki,
M.
Yokoo,
T.
Fujino,
A.
Koyama,
A.
Yokoseki,
M.
Nishizawa,
A.
Kakita,
H.
Takahashi,
O.
Onodera,
Decreased
number
of
Gemini
of
coiled
bodies
and
U12
snRNA
level
in
amyotrophic
lateral
sclerosis,
Hum.
Mol.
Genet.
22
(20)
(2013)
4136–4147.
[64]
S.
Reber,
J.
Stettler,
G.
Filosa,
M.
Colombo,
D.
Jutzi,
S.C.
Lenzken,
C.
Schweingruber,
R.
Bruggmann,
A.
Bachi,
S.M.
Barabino,
O.
Mühlemann,
M.D.
Ruepp,
Minor
intron
splicing
is
regulated
by
FUS
and
affected
by
ALS-associated
FUS
mutants,
EMBO
J.
35
(14)
(2016)
1504–1521.
[65]
G.J.
Xu,
A.A.
Shah,
M.Z.
Li,
Q.
Xu,
A.
Rosen,
L.
Casciola-Rosen,
S.J.
Elledge,
Systematic
autoantigen
analysis
identifies
a
distinct
subtype
of
scleroderma
with
coincident
cancer,
Proc.
Natl.
Acad.
Sci.
U.
S.
A.
113
(47)
(2016)
E7526–E7534.
[66]
G.
Shah,
A.
Xu,
L.K.
Rosen,
F.M.
Hummers,
S.J.
Wigley,
L.
Elledge,
Anti–RNPC-3
antibodies
as
a
marker
of
cancer-associated
scleroderma,
Arthritis
Rheumatol.
69
(6)
(2017)
1306–1312.
[67]
D.
Fischer,
T.
Wahlfors,
H.
Mattila,
H.
Oja,
T.L.J.
Tammela,
J.
Schleutker,
MiRNA
profiles
in
lymphoblastoid
cell
lines
of
finnish
prostate
cancer
families,
PLoS
One
10
(5)
(2015)
e0127427.
[68]
Y.
Tian,
Z.
Huang,
Z.
Wang,
C.
Yin,
L.
Zhou,
L.
Zhang,
K.
Huang,
H.
Zhou,
X.
Jiang,
J.
Li,
L.
Liao,
M.
Yang,
F.
Meng,
Identification
of
novel
molecular
markers
for
prognosis
estimation
of
acute
myeloid
leukemia:
over-expression
of
PDCD7,
FIS1
and
ang2
may
indicate
poor
prognosis
in
pretreatment
patients
with
acute
myeloid
leukemia,
PLoS
One
9
(1)
(2014)
e84150.
[69]
E.H.
Niemelä,
J.
Verbeeren,
P.
Singha,
V.
Nurmi,
M.J.
Frilander,
Evolutionarily
conserved
exon
definition
interactions
with
U11
snRNP
mediate
alternative
splicing
regulation
on
U11-48K
and
U11/U12-65K
genes,
RNA
Biol.
12
(2015)
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
ARTICLE IN PRESS
G Model
YSCDB-2404;
No.
of
Pages
10
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,
Alternative
exon
definition
events
control
the
choice
between
nuclear
retention
and
cytoplasmic
export
of
U11/U12-65K
mRNA,
PLoS
Genet.
13
(5)
(2017)
e1006824.
[71]
M.K.
Doma,
R.
Parker,
RNA
quality
control
in
eukaryotes,
Cell
131
(4)
(2007)
660–668.
[72]
E.H.
Niemelä,
A.
Oghabian,
R.H.J.
Staals,
G.J.M.
Pruijn,
M.J.
Frilander,
Global
analysis
of
the
nuclear
processing
of
unspliced
U12-type
introns
by
the
exosome,
Nucleic
Acids
Res.
42
(2014)
7358–7369.
[73]
J.J.
Turunen,
C.L.
Will,
M.
Grote,
R.
Lührmann,
M.J.
Frilander,
The
U11-48K
protein
contacts
the
5’
splice
site
of
U12-type
introns
and
the
U11-59K
protein,
Mol.
Cell
Biol.
28
(10)
(2008)
3548–3560.
[74]
H.
König,
N.
Matter,
R.
Bader,
W.
Thiele,
F.
Müller,
Splicing
segregation:
the
minor
spliceosome
acts
outside
the
nucleus
and
controls
cell
proliferation,
Cell
131
(4)
(2007)
718–729.
[75]
I.
Younis,
K.
Dittmar,
W.
Wang,
S.W.
Foley,
M.G.
Berg,
K.Y.
Hu,
Z.
Wei,
L.
Wan,
G.
Dreyfuss,
Minor
introns
are
embedded
molecular
switches
regulated
by
highly
unstable
U6atac
snRNA,
eLife
2
(2013)
e00780.
[76]
S.A.
Forbes,
D.
Beare,
H.
Boutselakis,
S.
Bamford,
N.
Bindal,
J.
Tate,
C.G.
Cole,
S.
Ward,
E.
Dawson,
L.
Ponting,
R.
Stefancsik,
B.
Harsha,
C.Y.
Kok,
M.
Jia,
H.
Jubb,
Z.
Sondka,
S.
Thompson,
T.
De,
P.J.
Campbell,
COSMIC:
somatic
cancer
genetics
at
high-resolution,
Nucleic
Acids
Res.
45
(D1)
(2017)
D777–D783.
... The minor U12-type spliceosome excises introns that are characterized by a highly conserved 5′ splice site and branch point sequence. Mutations in components of the minor U12-type spliceosome have been reported in several congenital or somatic diseases including PJS [10][11][12]. ...
Preprint
Full-text available
Peutz–Jeghers syndrome is an autosomal dominant disease characterized by intestinal polyposis, mucocutaneous pigmentation, and an increased risk of various types of cancer. Germline mutations in STK11 ( LKB1 ), which encodes serine/threonine kinase 11, have been identified as the major cause of Peutz–Jeghers syndrome. Here, we detected a rare variant of undetermined significance in intron 2 of STK11 using multi-gene panel analysis in a girl with clinically suspected Peutz–Jeghers syndrome based on family history and characteristic mucocutaneous pigmentation. We confirmed this variant caused abnormal splicing in exons 2 and 3 using reverse transcription-PCR and Sanger sequencing. To validate the predicted impact of this variant on splicing, we performed functional analysis using a minigene assay. The functional analysis experiments demonstrated that this variant suppressed normal splicing, and the clinical significance of the STK11 variant, which was initially thought to be a variant of “Uncertain Significance,” was determined to be “Likely Pathogenic.” Functional analysis and confirming the genetic diagnosis of cases with actionable hereditary diseases would be helpful for surveillance decisions and family diagnosis.
... RBPs are finely modulated at the expression level, localization and activity in a cell and tissue-specific manner (Baralle and Giudice, 2017;Paronetto et al., 2016). For these reasons, defects in the spliceosome machinery or in auxiliary RBPs are key factors driving disease susceptibility and severity (Verma et al., 2018). ...
... U12type introns are especially present in genes related to DNA replication and repair, transcription, RNA processing, and translation. However, they can also work on genes related to cytoskeletal organization, vesicular transport, and voltage-gated ion channel activity [6]. The U12-type spliceosome comprises several small nuclear RNAs (snRNA) and associated proteins [7]. ...
Article
Full-text available
Introduction: Pathogenic biallelic RNPC3 variants cause congenital hypopituitarism (CH) with congenital cataracts, neuropathy, developmental delay/intellectual disability, primary ovarian insufficiency, and pituitary hypoplasia. Here, we aimed to evaluate the clinical and molecular characteristics of two patients with CH and neuropathy. Material and methods: Proband was evaluated by clinical, laboratory, and radiological exams followed by exome sequencing (ES). Clinical investigation of an affected sibling and variant segregation in the family was performed by Sanger sequencing. A three-dimensional protein model study was conducted to predict the effect of the variant on the function of the RNPC3 peptide. Results: Proband was a 16-month-old girl who was referred for the evaluation of failure to thrive. Her height, weight, and head circumference were 55.8 cm (-7.6 SDS), 6.5kg (-3.6 SDS), and 41.8 cm (-3.82), respectively. She had a developmental delay and intellectual disability. Central hypothyroidism, growth hormone, and prolactin deficiencies were identified, and MRI revealed pituitary hypoplasia. Electroneuromyography performed for the gait abnormality revealed peripheral neuropathy. A homozygous novel variant c.484C>T/p.(Pro162Ser) in the RNPC3 was detected in the ES. Her brother had the same genotype, and he similarly had pituitary hormone deficiencies with polyneuropathy. Conclusion: Expanding our knowledge of the spectrum of RNPC3 variants, and apprehending clinical and molecular data of additional cases, is decisive for accurate diagnosis and genetic counseling.
Preprint
Full-text available
Around 60% of individuals with neurodevelopmental disorders (NDD) remain undiagnosed after comprehensive genetic testing, primarily of protein-coding genes1. Increasingly, large genome-sequenced cohorts are improving our ability to discover new diagnoses in the non-coding genome. Here, we identify the non-coding RNA RNU4-2 as a novel syndromic NDD gene. RNU4-2 encodes the U4 small nuclear RNA (snRNA), which is a critical component of the U4/U6.U5 tri-snRNP complex of the major spliceosome2. We identify an 18 bp region of RNU4-2 mapping to two structural elements in the U4/U6 snRNA duplex (the T-loop and Stem III) that is severely depleted of variation in the general population, but in which we identify heterozygous variants in 119 individuals with NDD. The vast majority of individuals (77.3%) have the same highly recurrent single base-pair insertion (n.64_65insT). We estimate that variants in this region explain 0.41% of individuals with NDD. We demonstrate that RNU4-2 is highly expressed in the developing human brain, in contrast to its contiguous counterpart RNU4-1 and other U4 homologs, supporting RNU4-2s role as the primary U4 transcript in the brain. Overall, this work underscores the importance of non-coding genes in rare disorders. It will provide a diagnosis to thousands of individuals with NDD worldwide and pave the way for the development of effective treatments for these individuals.
Article
Full-text available
Minor intron-containing genes (MIGs) account for less than 2% of all human protein coding genes and are uniquely dependent on the minor spliceosome for proper excision. Despite their low numbers, we surprisingly found significant enrichment of MIG-encoded proteins (MIG-Ps) in protein-protein interactomes and host factors of positive sense RNA viruses including, SARS-CoV-1, SARS-CoV-2, MERS-CoV, and Zika virus. Similarly, we observed significant enrichment of MIG-Ps in the interactomes and sets of host factors of negative sense RNA viruses such as Ebola virus, influenza A virus, and the retrovirus HIV-1. We also found enrichment of MIG-Ps in double stranded DNA viruses such as Epstein-Barr virus, human papillomavirus and herpes simplex viruses. In general, MIG-Ps were highly connected and placed in central positions in a network of human host protein interactions. Moreover, MIG-Ps that interact with viral proteins were enriched with essential genes. We also provide evidence that viral proteins interact with ancestral MIGs, that date back to unicellular organisms and are mainly involved in basic cellular functions, such as cell cycle, cell division, and signal transduction. Our results suggest that MIG-Ps form a stable, evolutionarily conserved backbone that viruses putatively tap to invade and propagate in human host cells.
Preprint
Full-text available
The minor spliceosome catalyses the excision of U12-dependent introns from pre-mRNAs. These introns are rare, but their removal is critical for cell viability. We obtained a cryo-EM reconstruction of the 13-subunit U11 snRNP complex, revealing structures of U11 snRNA and five minor spliceosome-specific factors. U11 snRNP appears strikingly different from the equivalent major spliceosome U1 snRNP. SNRNP25 and SNRNP35 form a dimer, which specifically recognises U11 snRNA. PDCD7 forms extended helices, which bridge SNRNP25 and SNRNP48, located at the distal ends of the particle. SNRNP48 forms multiple interfaces with U11 snRNP and, together with ZMAT5, are positioned near the 5′-end of the U11 snRNA and likely stabilise the binding of the incoming 5′-SS. Our structure provides mechanistic insights into U12-dependent intron recognition and the evolution of the splicing machinery.
Article
Full-text available
Small nuclear RNAs (snRNAs) are structural and functional cores of the spliceosome. In metazoan genomes, each snRNA has multiple copies/variants, up to hundreds in mammals. However, the expressions and functions of each copy/variant in one organism have not been systematically studied. Focus on U1 snRNA genes, we investigated all five copies in Drosophila melanogaster using two series of constructed strains. Analyses of transgenic flies that each have a U1 promoter-driven gfp revealed that U1 : 21D is the major and ubiquitously expressed copy, and the other four copies have specificities in developmental stages and tissues. Mutant strains that each have a precisely deleted copy of U1-gene exhibited various extents of defects in fly morphology or mobility, especially deletion of U1 : 82Eb . Interestingly, splicing was changed at limited levels in the deletion strains, while large amounts of differentially-expressed genes and alternative polyadenylation events were identified, showing preferences in the down-regulation of genes with 1–2 introns and selection of proximal sites for 3’-end polyadenylation. In vitro assays suggested that Drosophila U1 variants pulled down fewer SmD2 proteins compared to the canonical U1. This study demonstrates that all five U1-genes in Drosophila have physiological functions in development and play regulatory roles in transcription and 3’-end formation.
Article
The minor spliceosome regulates the removal of a conserved subset of introns present in genes with regulatory functions. In this issue of Molecular Cell, Augspach et al.1 report that elevated levels of U6atac snRNA, a key minor spliceosome component, contribute to prostate cancer cell growth and can be a novel therapeutic target.
Article
Full-text available
Mutations in the components of the minor spliceosome underlie several human diseases. A subset of patients with isolated growth hormone deficiency (IGHD) harbor mutations in the RNPC3 gene, which encodes the minor spliceosome-specific U11/U12-65K protein. Although a previous study showed that IGHD patient cells have defects in U12-type intron recognition, the biochemical effects of these mutations on the 65K protein have not been characterized. Here, we show that a proline-to-threonine missense mutation (P474T) and a nonsense mutation (R502X) in the C-terminal RNA recognition motif (C-RRM) of the 65K protein impair the binding of 65K to U12 and U6atac snRNAs. We further show that the nonsense allele is targeted to the nonsense-mediated decay (NMD) pathway, but in an isoform-specific manner, with the nuclear-retained 65K long-3'UTR isoform escaping NMD pathway. In contrast, the missense P474T mutation leads, in addition to the RNA binding defect, to a partial defect in the folding of the C-RRM and reduced stability of the full-length protein, thus reducing the formation of U11/U12 di-snRNP complexes. We propose that both the C-RRM folding defect and NMD-mediated decrease in the levels of the U11/U12-65K protein lead to reduced formation of the U12-type intron recognition complex and missplicing of a subset of minor introns, as reported by Argente et al. (2014), leading to pituitary hypoplasia and a subsequent defect in growth hormone secretion.
Article
Full-text available
Cellular homeostasis of the minor spliceosome is regulated by a negative feed-back loop that targets U11-48K and U11/U12-65K mRNAs encoding essential components of the U12-type intron-specific U11/U12 di-snRNP. This involves interaction of the U11 snRNP with an evolutionarily conserved splicing enhancer giving rise to unproductive mRNA isoforms. In the case of U11/U12-65K, this mechanism controls the length of the 3' untranslated region (3'UTR). We show that this process is dynamically regulated in developing neurons and some other cell types, and involves a binary switch between translation-competent mRNAs with a short 3'UTR to non-productive isoforms with a long 3'UTR that are retained in the nucleus or/and spliced to the downstream amylase locus. Importantly, the choice between these alternatives is determined by alternative terminal exon definition events regulated by conserved U12- and U2-type 5' splice sites as well as sequence signals used for pre-mRNA cleavage and polyadenylation. We additionally show that U11 snRNP binding to the U11/U12-65K mRNA species with a long 3'UTR is required for their nuclear retention. Together, our studies uncover an intricate molecular circuitry regulating the abundance of a key spliceosomal protein and shed new light on the mechanisms limiting the export of non-productively spliced mRNAs from the nucleus to the cytoplasm.
Article
Full-text available
Significance The last eukaryotic common ancestor had two spliceosomes. The major spliceosome acts on nearly all introns, whereas the minor spliceosome removes rare, U12-type introns. Based on in vitro RNA-splicing assays, the RGH3/ZRSR2 RNA-splicing factor has functions in both spliceosomes. Here, we show that the maize rgh3 mutant allele primarily disrupts U12 splicing, similar to human ZRSR2 mutants, indicating a conserved in vivo function in the minor spliceosome. These mutant alleles block cell differentiation leading to overaccumulation of stem cells in endosperm and blood, respectively. We found extensive conservation between maize and human U12-type intron-containing genes, demonstrating that a common genetic architecture controls at least a subset of cell differentiation pathways in both plants and animals.
Article
Full-text available
COSMIC, the Catalogue of Somatic Mutations in Cancer (http://cancer.sanger.ac.uk) is a high-resolution resource for exploring targets and trends in the genetics of human cancer. Currently the broadest database of mutations in cancer, the information in COSMIC is curated by expert scientists, primarily by scrutinizing large numbers of scientific publications. Over 4 million coding mutations are described in v78 (September 2016), combining genome-wide sequencing results from 28 366 tumours with complete manual curation of 23 489 individual publications focused on 186 key genes and 286 key fusion pairs across all cancers. Molecular profiling of large tumour numbers has also allowed the annotation of more than 13 million non-coding mutations, 18 029 gene fusions, 187 429 genome rearrangements, 1 271 436 abnormal copy number segments, 9 175 462 abnormal expression variants and 7 879 142 differentially methylated CpG dinucleotides. COSMIC now details the genetics of drug resistance, novel somatic gene mutations which allow a tumour to evade therapeutic cancer drugs. Focusing initially on highly characterized drugs and genes, COSMIC v78 contains wide resistance mutation profiles across 20 drugs, detailing the recurrence of 301 unique resistance alleles across 1934 drug-resistant tumours. All information from the COSMIC database is available freely on the COSMIC website.
Article
The lack of tools to identify causative variants from sequencing data greatly limits the promise of precision medicine. Previous studies suggest that one-third of disease-associated alleles alter splicing. We discovered that the alleles causing splicing defects cluster in disease-associated genes (for example, haploinsufficient genes). We analyzed 4,964 published disease-causing exonic mutations using a massively parallel splicing assay (MaPSy), which showed an 81% concordance rate with splicing in patient tissue. Approximately 10% of exonic mutations altered splicing, mostly by disrupting multiple stages of spliceosome assembly. We present a large-scale characterization of exonic splicing mutations using a new technology that facilitates variant classification and keeps pace with variant discovery.
Article
Significance Spinal muscular atrophy is the leading monogenic cause of infant mortality and is caused by homozygous loss of the survival of motor neuron 1 ( SMN1 ) gene. We investigated global transcriptome changes in the spinal cord of inducible SMA mice. SMN depletion caused widespread retention of introns with weak splice sites or belonging to the minor (U12) class. In addition, DNA double strand breaks accumulated in the spinal cord of SMA mice and in human SMA cell culture models. DNA damage was partially rescued by suppressing the formation of R-loops, which accumulated over retained introns. We propose that instead of single gene effects, pervasive splicing defects caused by severe SMN deficiency trigger a global DNA damage and stress response, thus compromising motor neuron survival.
Article
Introduction: Prior studies have demonstrated an increased risk of cancer-associated scleroderma in patients with RNA polymerase III (POL) autoantibodies and in patients negative for anti-centromere (CENP), anti-topoisomerase-1 (TOPO), and anti-POL antibodies (referred to as CENP/TOPO/POL (CTP)-Negative). In a recent study of 16 CTP-negative scleroderma patients with coincident cancer, we found that 25% had autoantibodies to RNPC3, a member of the minor spliceosome complex. In this investigation, we validated the relationship between anti-RNPC3 antibodies and cancer and examined the associated clinical phenotype in a large sample of scleroderma patients. Methods: Scleroderma patients with cancer were assayed for CENP, TOPO, POL and RNPC3 autoantibodies. Disease characteristics and the cancer-scleroderma interval were compared across autoantibody groups. The relationship between autoantibody status and cancer-associated scleroderma was assessed by logistic regression. Results: Of 318 patients with scleroderma and cancer, 70 (22.0%) were positive for anti-POL, 54 (17.0%) for anti-TOPO, and 96 (30.2%) for anti-CENP. Twelve patients (3.8% of overall group or 12.2% of CTP-negatives) were positive for anti-RNPC3. Patients with anti-RNPC3 had a short cancer-scleroderma interval (median 0.9 years). Relative to patients with anti-CENP, patients with anti-RNPC3 (OR 4.3; 95%CI 1.10-16.9; p=0.037) and anti-POL (OR 4.49; 95%CI 1.98-10.2; p<0.001) had a >4-fold increased risk of cancer within 2 years of scleroderma onset. Patients with anti-RNPC3 had severe restrictive lung and gastrointestinal disease, Raynaud's, and myopathy. Conclusion: Anti-RNPC3 autoantibodies associate with an increased risk of cancer at scleroderma onset, similar to POL autoantibodies. These data suggest the possibility of cancer-induced autoimmunity in this scleroderma subset. This article is protected by copyright. All rights reserved.
Article
Alternative splicing generates a diversity of mRNA transcripts from a single mRNA precursor and contributes to the complexity of our proteome. Splicing is perturbed by a variety of mechanisms in cancer. Recurrent mutations in splicing factors have emerged as a hallmark of several hematologic malignancies. Splicing factor mutations tend to occur in the founding clone of myeloid cancers and these mutations have recently been identified in blood cells from normal healthy elderly individuals with clonal hematopoiesis who are at increased risk of subsequently developing a hematopoietic malignancy, suggesting these mutations contribute to disease initiation. Splicing factor mutations change the pattern of splicing in primary patient and mouse hematopoietic cells and alter hematopoietic differentiation and maturation in animal models. Recent developments in this field are reviewed here, with an emphasis on the clinical consequences of splicing factor mutations, mechanistic insights from animal models, and implications for development of novel therapies targeting the pre-mRNA splicing pathway.
Article
Objective: Exome sequences account for only 2% of the genome and may overlook mutations causing disease. To obtain a more complete view, whole genome sequencing (WGS) was analyzed in a large consanguineous family in which members displayed autosomal recessively inherited cerebellar ataxia manifesting before two years of age. Methods: WGS from blood derived gDNA was used for homozygosity mapping and a rare variant search. RNA from isolated blood leukocytes was used for quantitative PCR, RNA sequencing and comparison of the transcriptomes of affected and unaffected family members. Results: WGS revealed a point mutation in non-coding RNA, RNU12, that was associated with early-onset cerebellar ataxia. The U12-dependent minor spliceosome edits 879 known transcripts. RT-PCR demonstrated minor intron retention in all of nine randomly selected RNAs from this group, and RNAseq showed splicing disruption specific to all U12-type introns detected in blood monocytes from affected individuals. Moreover, 144 minor intron containing RNAs were differentially expressed, including transcripts for three genes previously associated with cerebellar neurodegeneration. Interpretation: Interference with particular spliceosome components, including snRNAs, cause reproducible uniquely distributed phenotypic and transcript specific effects, making this an important category of disease associated mutation. Our approach to differential expression analysis of minor intron-containing genes is applicable to other diseases involving altered transcriptome processing. This article is protected by copyright. All rights reserved.
Article
Significance In this study, we created a barcoded whole-genome ORF mRNA display library and combined it with phage-immunoprecipitation sequencing to look for autoantibodies in sera from patients with scleroderma who also had coincident cancer without a known autoantibody biomarker. Using these two technologies, we found that 25% of these patients had autoantibodies to RNA Binding Region Containing 3 (RNPC3) and multiple other components of the minor spliceosome. There was evidence of intra- and intermolecular epitope spreading within RNPC3 and the complex. These combined technologies are highly effective for rapidly discovering autoantibodies in patient subgroups, which will be useful tools for patient stratification and discovery of pathogenic pathways.