Recognition of the Pyrimidine-Tract of the pre-mRNA PUF60

advertisement
Recognition of the Pyrimidine-Tract of the pre-mRNA
by U2AF and a Novel Splicing Factor PUF60
by
Patrick Schonleber McCaw
B.A. in Biology
Haverford College
Submitted to the Department of Biology
in partial fulfillment of the requirements for the degree of
Doctor of Philosophy in Biology
at
The Massachusetts Institute of Technology
June 1998
© 1998 Massachusetts Institute of Technology.
All rights reserved.
Signature of Author
Department of Biology
May 29, 1998
Certified by
Dr. Phillip A. Sharp
Professor of Biology
Thesis Supervisor
Accepted by
Dr. Frank Solomon
Chair, Biology Graduate Committee
Recognition of the Pyrimidine-Tract of the Pre-mRNA
by U2AF and a Novel Splicing Factor PUF60
by
Patrick Schonleber McCaw
Submitted to the Department of Biology on May 29, 1998
in partial fulfillment of the requirements for the degree of
Doctor of Philosophy in Biology
ABSTRACT
The genetic information required for encoding functional protein molecules is interrupted in
eukaryotes by non-coding sequences. These non-coding sequences, called introns, must be
removed from the transcribed RNA molecule by the spliceosome before the genetic information,
encoded in exons, can be used by the cell. The process of removing introns and joining exons is
called splicing. The splice site sequences are recognized multiple times during splicing of the
intron, presumably as a mechanism to ensure high fidelity splicing. Recognition of the intron by
the spliceosome requires the interaction of many protein and snRNP components. The first intron
recognition events are the recognition of the 5' splice site sequence by U 1 snRNP, recognition of
the branch sequence by SF1/BBP, and recognition of the pyrimidine-tract sequence by U2AF.
The 5' splice site and the branch sequence are subsequently recognized by U5 and U6 snRNPs and
by U2 snRNP, respectively. The PUF splicing activity was identified as a second pyrimidinetract binding factor that is required for efficient splicing in vitro. The PUF activity is required for
efficient formation of the stable, ATP-independent U2 snRNP:pre-mRNA complex, An . U2AF
is not required for splicing under certain conditions. However, the PUF activity is required for
splicing even in the absence of U2AF.
The PUF activity is composed of the previously described splicing factor p54 and the novel protein
PUF60. PUF60 binds specifically to pyrimidine-tract RNA, is conserved between vertebrates and
the invertebrate Drosophila,and forms SDS-resistant dimers. The SDS-resistant dimerization of
PUF60 is mediated by the C-terminal domain, the PUMP domain. Several other proteins
containing PUMP-domain homologies have been identified. Among these PUMP-domain
homologies is the domain of the small subunit of U2AF which is required for the stable proteinprotein interaction between the small subunit of U2AF and the large subunit of U2AF. The PUMP
domain may generally mediate protein-protein interactions.
Thesis Supervisor: Phillip A. Sharp
Title: Salvador E. Luria Professor of Biology and Head, Department of Biology
ACKNOWLEDGMENTS
There are many people who have made this thesis possible. I would like to start by
thanking the teachers. First and foremost I would like to thank Mr. Werner Feig who taught
critical thinking by way of American History at Scarsdale High School; among many great
teachers, I have had he remains the best. Dom Castillo taught chemistry in seventh and eighth
grade and it is to him that I owe my interest in biochemistry. He was the first to teach
thermodynamics and AG, and I thank him for having the courage to introduce the idea of Gibb's
free energy to a pre-adolescent. Dom was followed by Mr. Moffett and Professor John Chesick in
teaching me the mysteries of thermodynamics. Professor Chesick warned us often, "word to the
wise, cudgel to the obtuse: study, practice, learn." An arrogant young biologist wannabe, I
thought the gas laws a bit arcane, and did not take the wise word quite seriously enough. The
cudgel did not hit until David Baltimore made a passing remark that thermodynamics was the most
important course for a biologist. Now, years later, I have no doubt he was right, and I hope that
some of the work presented here reflects an attempt to understand thermodynamics in the context
of splicing.
I have been generously invited to work in many labs and I owe many people thanks for
teaching me the doing of science. I would like to thank Robert Wooley at the Albert Einstein
College of Medicine for taking me in as a high school student and teaching me how to isolate nuclei
from tumor samples, so that he could determine the ploidy of the tumor cells with a wonderful
Rube Goldberg flow cytometer. It was from Karl Pfenninger and Marie-France Mayli6-Pfenninger
that I first really learned experimental biology. Karl drove in to work on the Bronx River Parkway
at heart-stopping speeds, filling my sleep-addled brain (I had not yet discovered coffee--thanks
Charlotte) with the power of good experimentation and the fun of doing good science. I only
wish I had been more awake. Marie-France taught me perspective and embryology. Edie Abreu
showed me how to use a pipetman and much more.
David Baltimore and Kees Murre taught me how to take an idea and turn it into good
science, a lesson I am still trying to learn. The Baltimore lab, and the third floor of the Whitehead
Institute, was a fantastic place to learn science and there are too many people to thank for my time
there. I would especially like to thank the technicians with whom I worked: Carolyn Gorka, Anne
Gifford, and Mike Paskind in the Baltimore lab; Annie Smith and Mitch Walkowicz in the Korman
lab; Melissa Woodrow and Geoff Parsons in the Mulligan lab; and Lorene Lanier, Rebecca Riehl
and Sallie Smith of the Weinberg lab.
I would like to thank my great friends and classmates Jonathan Loeb, Julie Segre, and
Brenda Schulman; also, Chris Stipp and Mary Herndon; Neil Silverman, Greg Marcus, Rachel
Kindt, Jen Mach, Annie Williamson, and Leisa Johnson. My good friends from my days in the
fly labs: Charlotte Wang, Paul Kauffman and Kathy Collins, Mandy Hannaford, Lulu Fresco,
Tau-Mu Yi (honorary member), Chris Seibel and Sima Misra, Fay Shemanski, Jan Carminati, and
Dan Moore. With co-workers and friends like these its a wonder we ever left the lab and what a
place it was to work and learn: from meiosis to nuclear overhauser effect, rotisserie baseball to
feminism, and genetics to gel filtration, all before lunch.
I would especially like to thank Phillip Sharp and Margarita Siafaca for all their support and
patience. I would like to thank the many people in the Sharp lab who have made my time here an
enjoyable one and have made this work possible in so many ways: Maggie Beddall, Ben
Blencowe, Chris Burge, Helen Cargill, Karyn Cepek, Dan Chasman, John Crispino, Gene Hunh,
Robbyn Issner, Jae-Sang Kim, Lee Lim, Andrew MacMillan, Joel Pomerantz, Barbara Panning,
Rock Pulak, Yubin Qiu, Charles Query, Erica Reifenberg, Tom Tuschl and my baymates, Ben
Shykind, Grace Jones and Akira Mitsui. Kevin Amonlirdviman worked with me in the summer of
1997 and without him the PUF60 binding studies would not have been done and the summer
would not have been nearly as much fun.
I would like to thank my family, my parents Jack and Maura McCaw and John Schonleber
for their support and understanding. Thanks also to my brothers and sisters: Chris, Michael, Kath,
and Anna; and to my cousin and her husband Maura McMillin and Terefe Kerse.
Finally, and most importantly, I would like to thank Andrea Page for making my life happy
and complete.
TABLE OF CONTENTS
CHAPTER 1: RECOGNITION OF THE SPLICE SITE SEQUENCES IN pre-mRNA
SPLICING AND THEIR ROLE IN MEDIATING SPLICEOSOME ASSEMBLY...... 10
INTRODUCTION.............
.
...................
..........
THE SPLICING REACTION .........................................
.................... 11
....................................
12
INFORMATION AND THE SPLICE SITE DETERMINATION PROBLEM ....................................... 12
RECOGNITION OF THE 5' SPLICE SITE ..........................................................
.................... 15
U snRNP................................... .................... .................................................... 15
RECOGNITION OF THE 3' SPLICE SITE ....................
......................................... 18
Three distinct recognitionelements at the 3' end of the intron.................................
.... 18
U 2AF ..................................... .................
......
20
SF /B BP ..........................................................
ASSEMBLY OF THE SPLICEOSOME .........
...
.......... .... ........
............................................
23
....................................... 2 4
E comp lex ...............................................................
A complex..................................................
26
The spliceosomalcomplexes B and C.....................................
MY CONTRIBUTIONS TO THIS PROJECT ............................
REFERENCES..........................................
FIGURES ...........
23
..........................................
..................
27
.........................
28
................................
30
......
..........
..............
44
Figure1. The splicing reaction is a two step transesterificationreaction........................ 44
Figure2. Splice site sequences do not alone determine splice sites.................................
46
Figure 3. Cartoon of the spliceosome assembly pathway .................
48
....................
CHAPTER 2: IDENTIFICATION, PURIFICATION AND CHARACTERIZATION OF
A NEW PYRIMIDINE-TRACT BINDING SPLICING ACTIVITY......................... 50
ABSTRACT.....................................
INTRODUCTION ..............
RESULTS
.........
........
...........................................
51
.........................................................................
........................................................................................
55
Depletion ofNE for pyrimidine tractsplicing .........................
Role for U2AF35..............................
52
........
55
.........................
55
Purificationof the PUFactivity ............................................................
................ 56
IdentificationofPUF60 andp54 as the predominantproteins in the activefraction.......
The PUFactivity purifies as 400 kD complex ......................................
. 56
58
PUFis requiredfor efficient A3' complex assembly................................... 58
RNA crosslinking of the PUFfraction...................................................................... 58
PUFdoes not detectably interactwith the branchsequence or AG dinucleotide....................
59
PUFand U2AF do not bind to the same pyrimidine-tractRNA ........................................ 59
Interactionwith the branch sequence bindingprotein SF1/BBP.....................
.. 60
Subcellular localizationofPUF60....................................................
DISCUSSION .........................................................................................................
60
...... 62
Splicing in vitro requires two pyrimidine-tractRNA binding activities......................... 62
The PUFproteins: p54 and PUF60.......
........
..........
.............. 63
Sub-cellularlocalization ofPUF60....................................................
64
M ETHODS AND M ATERIALS............................................................................... ............ 65
Preparationofpoly[U] depleted nuclearextract ..........
.........
...............
65
Purificationof the PUFactivity................................................................................ 66
RNAs used in this study.............................................................................................
66
Sp licing in vitro ..................................................................................................... 67
Complex assembly assays.............................................................................
Gel shift...........................................................
Crosslinking ........................................................
A ntibodies .........................................
.........
...........
........... 67
............................. 67
..............................
....................
68
....................................................... 68
Immunoblotting........................................
................
...................... 68
Immunoprecipitationsand Gstpull-downs.................................................................. 68
Im munofluorescence ..............................................................................................
68
ACKNOW LEDGMENTS...................................................................................................
69
R EFERENCES...............................................................................................................
70
FIGURE LEGENDS .......................................................................................................
75
Figure1. Poly[U]-depletednuclearextract requiresboth U2AF andPUF......................... 75
Figure2. Purificationof the PUFactivity...
...............................
78
Figure3. PUF60 is ubiquitously expressed in humans as a 2.0 kb mRNA ................... 83
Figure4. NEAU is depleted of both PUF60and p54 ....................................................
85
Figure5. The PUFfactorp54 can form complexes with PUF60.................................. 88
Figure6. PUFactivity is requiredfor efficient A3 'spliceosome assembly. ...................... 90
Figure7. The PUFfraction has three species that crosslink to pre-mRNA................... 92
Figure8. PUFdoes not bind the AG dinucleotide or the branch sequence ......................
94
Figure9. PUFpyrimidine-tractbindingin the presence of either U2AF or U2AF65............. 96
Figure10. PUF60interacts with the branchsequence bindingprotein SF1/BBP........
. 98
Figure 11. PUF60 localizes to a non-speckle domain of the nucleus...............................100
6
CHAPTER 3: PUF60 A NOVEL PYRIMIDINE TRACT BINDING FACTOR WITH
HOMOLOGY TO THE SPLICING FACTORS U2AF65 AND MUD2P DIMERIZES VIA
ITS C-TERMINAL RRM-LIKE DOMAIN, THE PUMP DOMAIN ....................... 102
ABSTRACT ...........................................................................................
INTRODUCTION .....................................................................................
R ESULTS......
.................................
103
..................... 104
....................................................................... 106
PUF60,conserved in evolution, is related to the yeast splicingfactorMud2p...........
106
The PUMP domain is a distinctsubfamily of the large RRM domain family .....................
107
The PUMPdomain is aprotein-proteininteractiondomain........................................... 108
The PUMP domain does not contribute to RNA binding..............................................109
DISCUSSIoN.........................................................................................................
.. 111
RNA binding activity of PUF60............................................................................. 111
PUF60 is a U2AF65 homologue.................................
.........................................
112
The PUMP domain is a subset of the RRM domainfamily............................................ 112
The PUMP domain is a protein-proteininteractiondomain.........................................114
PUMP domain interactions: otherproteins................................................................ 114
M ETHODS AND M ATERIALS.......................................................................
................. 116
Identification of ESTs, sequencing,and alignments..................................................... 116
Expression andpurificationof His6PUF60and His6PUF60AC........................116
Translationin vitro................................................................................................117
Dim erization assay ...............................................................................................
117
RNA binding assay.................................................................................................117
REFERENCES..........
..............................
.................................................................. 119
FIGURE LEGENDS...........................................
........................................................ 122
Figure1. Sequence ofPUF60and DPUF68; comparisonofPUF60, U2AF65 and DPUF68.122
Figure2. The PUMP domain is a distinct subset of the RRM domainfamily ............... 125
Figure3. HPUF60forms SDS-resistantdimers...........................................................128
Figure 4. RNA binding activity of His6PUF60........................................
...... 133
CHAPTER 4: SC35 MEDIATED RECONSTITUTION OF SPLICING IN U2AFDEPLETED NUCLEAR EXTRACT .........................................
ABSTRACT.................................................
..................
................................. 138
INTRODUCTION .........................................................................................
M ATERIALS AND M ETHODS..................................................................
RNA Transcription................................................
137
...............
............... 139
...................... 141
..................... 141
NuclearExtracts.....................................................................................................141
U2AF and SC35 Preparation.................................................................................... 141
Splicing Assays............................................................................................142
R ESU LTS................................................................................. ................................ 142
U2AF6 5 and SC35 Mediated Reconstitution of Splicing in U2AF-DepletedExtracts...........142
FactorDependent Splicing in U1-Blocked Extracts......................................................143
DISCUSSION...................................................
........................................
......... 143
ACKNOWLEDGMENTS ..................................................................................................
146
REFERENCES .............................................................................................
147
FIGURE LEGENDS ..........................................................................................
Figure 1. SC35functionally substitutesfor U2AF6 5 ......................
........... 149
.......................... .
149
Figure2. SC35 reconstitution ofsplicing in U2AF-depletedreactions is substratespecific..151
Figure3. SC35 reconstitutespre-mRNA splicing dependent on the presence of U1 snRNP. 153
Figure4. Three distinctpathways resulting in spliceosome assembly. ............................. 155
CHAPTER 5: A MINIMAL SPLICEOSOMAL A COMPLEX RECOGNIZES BRANCH
SITE AND POLYPYRIMIDINE TRACTS................................................
157
A BSTRACT ..........................................
................................................................... 158
INTRODUCTION ........................................................................................................
MATERIALS AND METHODS ......................................
.
159
................................ 160
RNA transcriptionand synthesis ofsubstrates.............................................................160
Formationand native gel analysis of splicing complexes. .......................................
Nuclearextracts andpurificationofsplicingfactors...........................
161
....................161
Photo-cross-linkingassays...................................................................................
162
RE SULTS ..........................................
....................................................................... 162
A short oligonucleotide canform complexes with U2 snRNP.....................................162
Both branchsequence andpolypyrimidine tract are required............................................ 164
Similarities of A,,m to complex A containing U2 snRNP ........................................... 165
Amin complexformation is ATP independentand undergoes an ATP-dependent dissociation.167
Effects of 2'-Hsubstitutions...................................................................................168
D ISCUSSION ............................................................................................................... 169
A more sensitive system - 2'-OHand adenine interactions..........................................
169
Amin complex forms independently of Ul snRNP and ATP.......................................170
An active mechanism of U2 snRNP removal............................................................... 171
ACKNOWLEDGMENTS..........
..........................
...........
......................... 172
REFERENCES.............................................................................................................
172
LEGENDS TO FIGURES...............................................................................................180
FIGURE 1. BS-PPT RNA forms an A-like complex with U2 snRNP .......................... 180
FIGURE2. Both branch sequence andpolypyrimidinetractare requiredin cis................... 182
FIGURE 3. CharacteristicsofAmin complex. .........................................
...... 184
FIGURE 4. Amin complex formation in the presence ofATP...................................186
FIGURE 5. Time course of complex assembly ................
.................
..
189
FIGURE 6. Summary offormation of complexes A (left) and Amin (right)....................... 191
TABLES ....................................
.......................
193
TABLE 1. Relative yields for Amin complex formation of modified substrates.................. 193
SPECULATIVE APPENDIX: A PROPOSED INTERACTION BETWEEN THE
BRANCH ADENOSINE AND THE U5 LOOP NUCLEOTIDE URIDINE 4, A
MECHANISM TO JUXTAPOSE THE SUBSTRATES OF FIRST STEP OF pre-mRNA
SPLIC IN G ........................................................................................
195
A NEW REPRESENTATION OF THE SPLICEOSOMAL SECONDARY STRUCTURE ............................ 196
DESCRIPTION OF THE MODEL...................................
.............
......................
197
STRUCTURAL CONSIDERATIONS................................................................
198
GENETIC EXPERIMENTS THAT ARE CONSISTENT WITH THE MODEL ........................................ 199
BIOCHEMICAL EVIDENCE ...................
.
.......
.
........................
201
202
EXPERIMENTS THAT TEST THE MODEL ................................................
ACKNOWLEDGMENTS..........................
REFERENCES..............................
FIGURE LEGENDS .............................
.............................
...............
................... 202
.............
...............
203
.................................
............
........ 206
Figure1. Known base-pairinginteractionsof U1, U2, U5, and U6 ............................... 206
Figure2. The rearrangedbase-pairinginteractions ....................................
208
Figure3. The proposedbase-pairinginteractionbetween the branch adenosineand Uridine4..210
Figure4. Photographsof the model of the proposedstructure....................................... 212
Figure 5. Proposedbase-pairinginteractions ............................................................ 214
AFTERW ORD ....................................................................................
217
PUF60:a splicingfactor? ............................................................. 217
Experiments that addressfunctional issues...............................
................. 218
PUF60 immunolocalization
is the native protein detectable?.................. ............................................
Is PUF60a dimer in the native state? ........................................
...... 219
...................
219
CHAPTER 1:
RECOGNITION OF THE SPLICE SITE
SEQUENCES IN pre-mRNA SPLICING AND THEIR ROLE IN
MEDIATING
SPLICEOSOME ASSEMBLY
INTRODUCTION
Without the ability to accurately and reproducibly access and manipulate information the
cell would be unable to function. Information in a biological context exists in many forms, and
biological information can be carried by many kinds of biomolecules. Most famously,
biological information is carried by DNA in the form of genes, but information can also be
carried by proteins. For example, G proteins carry information about the state of membrane
trafficking by carrying either GTP or GDP in their guanine nucleotide binding pocket (Boguski
and McCormick, 1993). Other proteins carry information in their phosphorylation states:
phosphorylation of the retinoblastoma protein is a marker of cell cycle state (Sherr, 1996). The
pre-mRNA molecule carries information beyond the genetic information required for protein
synthesis. This pre-mRNA specific information is required for appropriate processing of the
pre-mRNA such as splicing and polyadenylation.
Splicing removes introns from the pre-mRNA, a key step in the formation of the mRNA
molecule. The unique gene structure of eukaryotes, first described over 20 years ago (Berget et
al., 1977; Chow et al., 1977), in which the protein coding information is interrupted by noncoding sequence, requires that the pre-mRNA be accurately processed to remove each of the
non-coding (intronic) sequences, leaving only the coding (exonic) sequences required for
translation or other mRNA function. In the nucleus pre-mRNA is processed to remove introns,
yielding mRNA which is exported from the nucleus and is competent for translation by the
ribosome. Recognition of an intronic sequence element by a newly recognized splicing factor is
the focus of this thesis. A description of the sequence elements required for intron/exon
recognition and the trans-acting factors that recognize these elements will be the subject of this
introductory chapter. Formation and constitution of the catalytically competent splicing enzyme,
the spliceosome, will be more briefly described. This thesis concludes with a speculative
appendix in which I discuss one aspect of the structure of the catalytic spliceosome. I shall
make a distinction between the catalytically active splicesome, the catalytic spliceosome, and the
early spliceosomal complexes that appear to play a role in splice site sequence recognition and
not in catalysis. These problems, the splice site definition problem and the catalytic problem,
are distinct intellectually; and I would argue that they are distinct, though related, problems
biochemically.
THE SPLICING REACTION
The splicing reaction is indicated diagrammatically in Figure 1. Exons are indicated by
boxes and the intron by a line. Removal of the intron is a two-step transesterification reaction,
catalyzed by a large multi-subunit ribonucleoprotein particle, the spliceosome (Brody and
Abelson, 1985; Frendeway and Keller, 1985; Grabowski et al., 1984; Grabowski et al., 1985;
Padgett et al., 1984; Ruskin et al., 1984). In the first step, a 2' hydroxyl group of an intronic
adenosine nucleotide, referred to as the branch adenosine, attacks the phosphodiester bond
separating the 5' exon from the intron, referred to as the 5' splice site. This leads to the release
of the 5' exon and the formation of a branched 2'-5' adenosine within the intron-3' exon
fragment (Padgett et al., 1984; Ruskin et al., 1984; Wallace and Edmonds, 1983). This
branched intron-3' exon first step product is often referred to as the lariat intron-3' exon because
of its circular structure. In the second transesterification reaction the 3' hydroxyl of the 5' exon
attacks the phosphodiester bond at the boundary between the intron and 3' exon, referred to as
the 3' splice site, forming ligated exon product and free lariat intron product. How this
chemical reaction is accomplished, both in terms of what are the catalytically important groups
and in how those catalytically important groups come to be arrayed about the splice sites remain
important and unanswered questions.
INFORMATION AND THE SPLICE SITE DETERMINATION PROBLEM
An important question arises from consideration of the information necessary to
accomplish accurate, high-fidelity removal of the intronic sequence. Inaccurate splicing would
be disastrous for the cell as most genes (with the exception of S. cerevisiaegenes) have multiple
introns and even a relatively accurate splicing reaction, 90%, would lead to very few, 35%,
accurately spliced mRNA molecules in a pre-mRNA with ten introns. Most of these
inaccurately spliced introns may be functionally unimportant as they are subject to non-sense
mediated decay, but some would have severe functional consequences (Hodgkin et al., 1989;
Pulak and Anderson, 1993).
A typical mammalian pre-mRNA will have thousands of nucleotides, thousands of
phosphodiester bonds and 2' hydroxyls. How does the spliceosome know which ones to use?
Sequences at the intron-exon boundary and the sequence near the branch adenosine, the branch
sequence, are partially conserved. Are these sequences sufficient to uniquely define the splice
sites? For the yeast Saccharomyces cerevisiae,the conserved sequences at the 5' splice site, the
3' splice site and the branch sequence are probably sufficient to define each intron uniquely, but
for mammalian cells it is clear that there is insufficient information within these sequences to
uniquely define intron position (Stephens and Schneider, 1992). In yeast the splice site
sequences are precisely defined and little variation is found or tolerated in these sequences
(Rymond and Rosbash, 1992). In vertebrates, as for the metazoans generally, the sequences
are less well defined and variation of the sequence is better tolerated (see for example; Reed and
Maniatis, 1985; Senapathy et al., 1990). The splice site consensus sequences have recently
been refined by C. Burge and P. A. Sharp (to be published in Burge and Sharp, 1998; also see
Mount, 1982) from a database of over 1600 mammalian introns. For these introns, the 5' splice
site sequence consensus has been determined to be aG/GURrG, the branch sequence consensus
is YTrAy and the pyrimidine tract-3' splice site sequence consensus is yyyyyyyynCAG/r
(where / marks the splice site phosphodiester bonds, A is the position of the branch adenosine,
R is a conserved purine, Y is a conserved pyrimidine and lower case letters are less conserved).
The insufficiency of splice site sequences is illustrated in figure 2 which shows the first 7210
nucleotides of the Human CREB-RP transcript (chosen at random, Min et al., 1995). The 5'
and 3' terminal nucleotides of the introns are indicated in bold. Note that the sequence of the
fourth 5' splice site (underlined) is also found in the fifth intron (also underlined); however in
this position this sequence is not used as a 5' splice site. How is the identical sequence used as
a 5' splice site in one position but not at another?
More information must be available to the spliceosome than is found in the splice site
and branch sequences. This missing information is often referred to as the "context" of the
splice site. Experimentally, the importance of context is demonstrated by the observation that
mutation of a 5' or 3' splice site sequence activates cryptic splice sites. Activation of cryptic
splice sites is seen in vivo (Treisman et al., 1983) and in vitro (Krainer et al., 1984). While
cryptic splice site sequences fit the splice site sequence consensus, they are generally poorer
matches to the consensus than the endogenous splice site sequence. Cryptic splice sites are not
known to be used unless the endogenous splice site sequence is inactivated by mutation
(Treisman et al., 1983). Many naturally occurring mutations of the globin loci have been
identified, and mutations of splice sites cause both skipping and cryptic splice site activation
(see for example; Treisman et al., 1983; Faustino, 1998). This result argues that information
defining a splice site is not constrained to splice site sequences themselves, but can be encoded
elsewhere and hence, context is important.
The presence of information outside of the splice site sequence, the existence of splice
site context, raises the question of whether the splice site sequence is not only not sufficient but
also perhaps not necessary for splice site determination. In one sense, the splice site sequence
is clearly necessary for splice site determination as all splice sites identified bear some
resemblance to the splice site sequence consensus' and so the splice site sequence must
determine which phosphodiester bond is targeted by the spliceosome. In a broader sense it is
not as clear that the splice site sequence is necessary for determining that a splice site exists in a
given region, as the frequency with which these cryptic splice sites are predicted to occur is
quite high. Put another way, the splice site sequence is necessary to define the precise
phosphodiester bond used as the splice site, but the splice site sequence is not sufficient to
identify a region of the pre-mRNA as having a splice site (Green, 1986; Maniatis and Reed,
1987). Also the splice site sequence is not often necessary to identify a region as having a
splice site, as inactivation of the wild-type splice site by mutation of the splice site sequence
leads to activation of other splice sites that would not otherwise be used (Treisman et al., 1983;
Wieringa et al., 1983). The source of the information that is necessary for determining a region
as having an exon-intron border, the context, is not known. For pre-mRNAs that have
regulated introns or exons this information may be contained in enhancer elements that can be
found in either the intron or the exon (Robberson et al., 1990; Talerico and Berget, 1994).
For constitutive exons and introns, the situation is less clear and has not been as well
studied, although there is recent work on trans-acting factors that may recognize context (Sun et
al., 1993; Tacke et al., 1997; Tacke and Manley, 1995; Wang and Manley, 1997). However,
there appears to be broad similarity between the constitutive and regulated splicing systems. It
is not clear whether the information required for regulated splicing and context information is
encoded primarily in the exonic or in the intronic sequence. Rather it appears that location of the
information varies between species: invertebrates may use information within introns (Talerico
and Berget, 1994), while vertebrates use information primarily within exons (Robberson et al.,
1990). Localization of the information may also vary from intron to intron as small mammalian
exons have sequence elements in adjacent introns which regulate their recognition (see for
example, Chan and Black, 1995), while others clearly have information within the exon (see for
example, Lavigueur et al., 1993).
In the following pages I will discuss recognition of the splice site sequences focusing on
the recognition of the 5' splice site sequence by the Ul small nuclear ribonucleoprotein particle
(U1 snRNP) and recognition of the pyrimidine tract by U2 snRNP auxiliary factor (U2AF). I
will also discuss, more briefly, the increasing understanding we have of how the non-splice site
sequence information is recognized and the protein factors that are implicated in this process.
Assembly of the catalytic spliceosome will then be briefly discussed. The mammalian splicing
' Many of the exceptions to this statement are now known to be splice sites that are spliced by a new
class of spliceosome, the U12 spliceosome (Hall and Padgett, 1996; Tarn et al., 1995).
system is the model for these discussions, but it is not possible to discuss splicing without
reference to the yeast Saccharomycescerevisiae. Whenever possible mammalian nomenclature
will be used in preference to yeast nomenclature.
RECOGNITION OF THE 5' SPLICE SITE
The 5' splice site sequence consists of the consensus sequence aG/GURrG and this
sequence is complementary to the 5' end of U1 snRNA (Lerner et al., 1980; Rogers and Wall,
1980). The sequence is not well conserved in vertebrates and variations are readily used in vivo
(see for example Treisman et al., 1983). In the yeast, S. cerevisiae,the 5' splice site consensus
sequence is more strictly conserved, and mutations are more detrimental to splicing (Rymond
and Rosbash, 1992).
U1 snRNP
Ul snRNP is the most abundant U snRNP in the cell with approximately 106 copies per
cell. Ul snRNP is a 17S protein-RNA complex (Reddy and Busch, 1988) consisting of Ul
snRNA, a 164 nucleotide RNA transcribed by RNA polymerase II, and multiple proteins that
are both specific to Ul snRNP and specific to the snRNPs generally (Reddy and Busch, 1988).
U1 snRNP specific protein factors include U1A, UlB, UlC, and Ul 70k. The function of
some of these factors is known and will be discussed below. The principle function of Ul
snRNP is binding to 5' splice site sequences. This binding event likely identifies these
sequences as potential targets of spliceosome complex formation.
Interaction of Ul snRNP with the 5' splice site occurs via a base-pairing interaction
between the 5' end of Ul snRNA and the 5' splice site. This interaction was first proposed
based on the complementarity of the 5' splice site sequence and the 5' end of Ul snRNA
(Lerner et al., 1980; Rogers and Wall, 1980) and binding was demonstrated fifteen years ago
(Mount et al., 1983). While base-pairing has been demonstrated between the 5' splice site and
the 5' end of U1 (the 5' splice site sequence complementarity region, Zhuang and Weiner,
1986), interaction of Ul snRNP with a 5' splice site RNA does not require the 5' splice site
complementarity region (Rossi et al., 1996). This result suggests that protein components of
U1 snRNP play a role in mediating pre-mRNA association, but that specificity and stability may
require the Ul snRNA 5' end. In support of this hypothesis, it has been found that the Ul
snRNP-specific protein Ul C is required for formation of the Ul snRNP-pre-mRNA complex,
known as E, in Ul snRNP reconstitution experiments and is sufficient for mediating this
interaction in the absence of the 5' splice site complementarity region (Heinrichs et al., 1990;
Jamison et al., 1995; Will et al., 1996). U1C has additionally been shown to crosslink to 5'
splice site sequence containing RNA oligonucleotides indicating a physical interaction between
the pre-mRNA 5' splice site and U 1C (Rossi et al., 1996). In yeast, the role U 1C, YU 1C,
plays in mediating pre-mRNA binding has been investigated genetically (Tang et al., 1997).
YU1C, is required for viability and in the absence of YU1C formation of the Ul snRNPcontaining complexes CC1 and CC2 is inhibited. Further, the 5' end of U snRNA is
hypersensitive to RNase digestion in YU 1C depleted strains (Tang et al., 1997).
Other U 1 snRNP-specific proteins have been tested for their role in mediating Ul
snRNP association with the pre-mRNA. Both the U1A and Ul 70k proteins are not required
for binding to the 5' splice site sequence (Jamison et al., 1995; Will et al., 1996) and a
functional role for U1A remains elusive. UlA is highly homologous to the U2 snRNP specific
protein U2B" and both have two RNA recognition motif (RRM) domains. U1A binds stem
loop II of U1 snRNA via its N-terminal RRM domain (Lutz-Freyermuth et al., 1990; Query et
al., 1989b). The interaction between this RNA and RRM domain are among the best studied
RNA-protein interactions (Nagai, 1996). The biochemical function of the Ul A C-terminal
RRM domain is not known, but this domain is not thought to bind RNA (Lu and Hall, 1995).
Yeast U A was identified in a screen for enhancers of a Ul snRNA temperature sensitive
mutation and is called MUD1 (Liao et al., 1992). MUDI is not an essential gene, but mudlA is
synthetic lethal with the U1 snRNA mutant. Surprisingly, the C-terminal RRM domain of
Mudlp is more conserved than the N-terminal domain suggesting that it is functionally
important (Tang and Rosbash, 1996). The decreased conservation of the N-terminal RRM
compared to the C-terminal RRM may be due to the divergence of the U snRNA binding
element rather than a difference in function. Mutations in MUDI have only mild effects in vitro.
Decreased splicing efficiency is observed only for pre-mRNA substrates that already splice
inefficiently in vitro (Liao et al., 1992; Tang and Rosbash, 1996).
The Ul 70k protein binds to stem-loop I of Ul snRNA via its single RRM domain
(Query et al., 1989a; Query et al., 1989b; Surowy et al., 1989). Ul 70k has a C-terminal
degenerate RS domain. RS domains are regions rich in the dipeptide arginine-serine or serinearginine and are found on many splicing factors including both subunits of U2AF, p54 and Ul
70k as well as the SR proteins. The RS domain of U1 70k, sometimes referred to as the RD/E
domain, is unusual in that many of the serines are replaced by aspartate or glutamate in the
dipeptide repeats. In this respect, the RS domain of Ul 70k is similar to the RS domains of
both U2AF and p54. Although Ul 70k does not play an essential role in mediating Ul snRNP
binding to the pre-mRNA, it may play a role in stabilizing that interaction. Several studies have
shown, that the RS-domains of ASF/SF2, SC35 and other SR proteins bind the RS domain of
U1 70k (Jamison et al., 1995; Kohtz et al., 1994; Wu and Maniatis, 1993). Subsequently, it
has been shown that phosphorylation of ASF/SF2 is important in mediating the ASF/SF2-U 1
70k association. ASF/SF2 binding to Ul 70k does not disrupt Ul 70k-U 1 snRNA binding
(Xiao and Manley, 1997). It has been shown that SR proteins stabilize the interaction of Ul
snRNP with the pre-mRNA (Kohtz et al., 1994). This interaction is specific for the SR protein
and pre-mRNA and is presumably due to the interaction between Ul 70k and an SR protein
bound to the pre-mRNA (Zahler and Roth, 1995).
U1 70 k is well conserved in evolution; the RRM domain and RD/E domain are
conserved to the invertebrate Drosophila(Mancebo et al., 1990). However, the yeast Ul 70k
homologue, SNP 1, is not as well conserved. SNP 1 has been found to be non-essential in
yeast; however, snpl cells are slow growing and exhibit a severe temperature sensitive
phenotype (Hilleren et al., 1995). Remarkably, only the N-terminal domain, and not the RRM
domain, is required to rescue these phenotypes (Hilleren et al., 1995). A partial deletion of the
SNP 1 open reading frame is lethal in at least one strain; this mutation can be rescued with a
yeast-human chimeric protein suggesting that the functional interactions of U1 70 k/SNP 1 are
conserved between yeast and humans (Smith and Barrell, 1991).
Yeast has two additional Ul snRNP specific genes, PRP39 (Lockhart and Rymond,
1994) and PRP40 (Kao and Siliciano, 1996). PRP39 is an essential gene, prp39 mutants show
defects in splicing both in vivo and in vitro. Prp39p has no obvious structural similarities to
other proteins. Absence of Prp39p from extracts prevents formation of spliceosomal
complexes. Prp39p is associated with U1 snRNP and associates with spliceosomes as
demonstrated by immunoprecipitation (Lockhart and Rymond, 1994). PRP40 was identified as
a U 1 snRNP 5' splice site sequence mutation suppressor and is an essential gene. PRP40 is
stoichiometrically associated with U1 snRNP and is required for the first catalytic step of
splicing (Kao and Siliciano, 1996). PRP40 is also known to interact with the branch sequence
binding protein SF 1/BBP and with the MUD2, the probable U2AF homologue in yeast
(Abovich and Rosbash, 1997).
Despite the clear role U 1 snRNP plays in binding the 5' splice site and the requirement
in yeast for several Ul snRNP specific factors for viability (UI snRNA, YU1C, PRP39, and
PRP40), the importance of Ul snRNP has been called into question by three observations.
First, U snRNP appears to disassemble, or be destabilized, from the spliceosome before
catalysis occurs and so is not present at the in the catalytic spliceosome (Konarska and Sharp,
1986; Michaud and Reed, 1993). Second, in yeast some mutations in the 5' splice site sequence
lead to the activation of aberrant 5' splice site selection events, but compensatory mutagenesis of
Ul snRNA does not correct this defect (Fouser and Friesen, 1986; Jacquier et al., 1985; Parker
and Guthrie, 1985). This suggests that the 5' splice site sequence is recognized by two
independent factors, Ul snRNP and a second factor that determines the position of the 5' splice
site phophodiester bond (Rymond and Rosbash, 1992). Third, splicing in vitro can occur in the
absence of U1 snRNP. This has been demonstrated both by affinity depletion of Ul snRNP
from HeLa nuclear extract (Crispino et al., 1994; Crispino et al., 1996; Crispino and Sharp,
1995) and by specific nuclease digestion of the 5' end of Ul snRNP (Tam and Steitz, 1994).
Most tested pre-mRNAs can only splice in U -depleted or Ul-knockout extracts in the presence
of exogenously added SR proteins at high concentrations (Crispino et al., 1996). Cryptic 5'
splice site sequences are activated in U 1 snRNP depleted extract suggesting that 5' splice site
sequence determination in the absence of Ul snRNP is less specific (Crispino et al., 1996).
These results clearly demonstrate that Ul snRNP is not required for formation of the catalytic
spliceosome. Some molecule other than Ul snRNP must be responsible for recognition of the
5' splice site at the catalytic steps. The second recognition 5' splice site recognition event is
believed to be mediated by U6 snRNP and by U5 snRNP (Kandels-Lewis and Seraphin, 1993;
Lesser and Guthrie, 1993; Sawa and Abelson, 1992; Sawa and Shimura, 1992; Sontheimer and
Steitz, 1993; Wyatt et al., 1992). These results suggest that Ul snRNP determines which 5'
splice site sequence is chosen for spliceosome formation. Presumably, in the absence ofU 1
snRNP, 5' splice site sequences are chosen based on the specificity of the catalytic spliceosome
and not on the splice site determination functions of the early spliceosomal complexes.
RECOGNITION OF THE 3' SPLICE SITE
Three distinct recognition elements at the 3' end of the intron
The 3' splice site consists of three RNA elements, the branch sequence, the pyrimidine
tract and the AG dinucleotide. The vertebrate 3' splice site sequence elements are similar to
those used in the invertebrate Drosophila(Mount et al., 1992). The branch sequence,
canonically YTrAY, contains the branch adenosine (underlined) and the 2' hydroxyl that is the
nucleophile for the first catalytic step. In vertebrates, sequence requirements for recognition of
the branch sequence are not as stringent as the requirements for recognition of the 5' splice site
and the pyrimidine tract. In vertebrates, mutation of the branch sequence has little effect on
splicing efficiency (Wieringa et al., 1983); however, splicing in vitro of these pre-mRNAs is
less efficient than in vivo (Padgett et al., 1985). Mapping of the branch adenosine used in these
mutant introns showed that the branch sequence used did not match the consensus, suggesting
that the branch sequence was not an essential recognition element (Padgett et al., 1985). These
data suggest that branch sequence recognition is not an essential feature of intron recognition in
vertebrates. There is, however, a clear constraint on the branch nucleotide in that the branch
nucleotide must be an adenosine for both steps of splicing to be completed (Query et al., 1994).
In the yeast S. cerevisiae, in contrast, recognition of the branch sequence is probably the
critical recognition event in mediating spliceosome assembly, and the branch sequence is both
highly conserved and very sensitive to mutation (Parker et al., 1987). There is probably
sufficient information in the yeast branch sequence to uniquely identify all but the longest yeast
introns (Rymond and Rosbash, 1992), strongly implicating the branch sequence in being the
critical mediator of intron recognition in yeast. Mutation of the branch sequence in S. cerevisiae
leads either to a block in splicing or to inefficient splicing (Cellini et al., 1986; Parker et al.,
1987) and can also lead to the accumulation of first step products (Fouser and Friesen, 1986).
The pyrimidine tract in vertebrates generally lies between the branch sequence and the
AG dinucleotide. Recognition of a pyrimidine tract is important for splicing in vertebrates as
mutations of the pyrimidine tract decrease or block splicing (Reed and Maniatis, 1985; Roscigno
et al., 1993; Ruskin and Green, 1985a; Ruskin and Green, 1985b; Smith et al., 1989). In
vertebrates, recognition of the pyrimidine tract appears to be the primary recognition event at the
3' end of the intron as the branch sequence is so poorly conserved and the AG dinucleotide, 3'
splice site, appears to be defined in relation to the pyrimidine tract (Smith et al., 1989).
The pyrimidine tract and AG dinucleotide consensus sequence of both C. elegans
(Zhang and Blumenthal, 1996) and the yeast S. cerevisiae (Parker et al., 1987) is quite
different from the vertebrate consensus. The C. elegans 3' splice site consensus is UUUCAG/R
with no discernible branch sequence and no pyrimidine tract (Zhang and Blumenthal, 1996).
The yeast 3' splice site consensus is quite different in that most of the sequence conservation is
found at the branch sequence with no apparent pyrimidine tract. Mutations or deletions in the
region between the branch sequence and the AG dinucleotide generally have little effect on
splicing in yeast (Fouser and Friesen, 1987). The important sequence recognition elements of
the 3' splice site region appear to have switched between yeast and vertebrate introns. For yeast
the presence of a pyrimidine tract is not important, but the branch sequence is essential, while
for vertebrates the pyrimidine tract is important while the branch sequence is not.
The AG dinucleotide is not an essential recognition element in vertebrates as deletion of
the AG dinucleotide does not effect intron recognition. Both spliceosome assembly and first
step catalysis can occur on pre-mRNAs that lack the AG dinucleotide (Anderson and Moore,
1997; Frendeway and Keller, 1985; Reed and Maniatis, 1985). The AG dinucleotide is,
however, required for the second step, but it remains unclear whether the AG dinucleotide is
selected by a scanning mechanism or by some other mechanism (Anderson and Moore, 1997)
and whether this recognition event generally occurs before or after the first step.
The following sections will discuss the pyrimidine tract recognition factor U2AF and the
branch sequence binding protein SF 1/BBP.
U2AF
U2AF (U2 snRNP auxiliary factor) was first identified in an experiment that
demonstrated that a protein factor was essential for the stable ATP-dependent association of U2
snRNP with the branch sequence (Ruskin et al., 1988). U2AF is a heterodimeric protein
splicing factor consisting of a small subunit, U2AF35, and a large subunit, U2AF65 (Zamore
and Green, 1989). Both subunits of U2AF have been shown to be highly conserved through
evolution and both subunits are found in the fission yeast S. pombe (Potashkin et al., 1993;
Wentz and Potashkin, 1996), in the plant A. thaliana (Accession number: AC002332) and in
the invertebrates C. elegans (a small subunit orthologue has not been identified in C. elegans;
Zorio et aL., 1997) and D. melanogaster(Kanaar et al., 1993). U2AF was purified using a U2
snRNP complex assembly assay. This assay demonstrated the critical importance of the
pyrimidine tract in mediating U2 snRNP association with the pre-mRNA (Zamore and Green,
1989). The 5' splice site is not required for the formation of this complex; however, Ul
snRNP is required (Barabino et al., 1990).
U2AF65
U2AF was found to bind poly[U] RNA at high salt concentrations (Zamore and Green,
1989) and this allowed U2AF activity to be efficiently and specifically depleted from nuclear
extract (Zamore and Green, 1991). U2AF65 was found to be necessary and sufficient to
reconstitute in vitro splicing activity to these depleted extracts (Zamore and Green, 1991).
The Drosophilalarge subunit of U2AF has been shown to be essential for viability and
temperature sensitive mutations of the S. pombe homologue of the large subunit of U2AF are
inviable (Kanaar et al., 1993; Potashkin et al., 1993). A gene with limited sequence similarity
to U2AF65 is found in the yeast S. cerevisiae,Mud2p; despite the limited sequence similarity
this gene is likely to be the S. cerevisiae orthologue of U2AF65 due to the striking functional
similarity of Mud2p to the U2AF65. Mud2p will be discussed separately below. The domain
structure of the large subunit of U2AF65 is conserved throughout these proteins. The Nterminal region of the protein consists of three parts, a short N-terminal peptide not conserved
between invertebrates and vertebrates (Zorio et al., 1997), an RS-dipeptide rich region of
variable length reminiscent of the RS domain of Ul 70k in its high RE and RD content, and a
"hinge" region which has been shown to interact with the small subunit of U2AF in both S.
pombe and H. sapiens. The C-terminal two thirds of U2AF65 is the RNA binding region of the
protein and consists of two RRM domains and a third C-terminal RRM-like domain. The
RRM-like domain is the most conserved portion of these proteins. This domain will be
described in more detail in chapter 3.
The C-terminal RRM-like domain of U2AF65 has additionally been shown to interact
with the branch-sequence binding protein SF1/BBP. The RRM-like domain is required for the
association of U2AF with SF1/BBP. Constructs missing 15 amino acids of the N-terminus of
this domain cannot associate with SF1/BBP (Berglund et al., 1997). The RS domain of
U2AF65 interacts weakly with SR proteins (Wu and Maniatis, 1993) and the RS domain may
contact the branch sequence (Valcarcel et al., 1996). The interaction of the human U2AF
subunits is stable to extremely high salt concentrations (at least 2M KCl; Zamore and Green,
1989). U2AF65 also contacts the DEAD-box protein UAP56 and recruits it to the assembling
spliceosome (Fleckner et al., 1997). The N-terminal region of U2AF65, including the RS
domain, the hinge domain, and a portion of the first RRM, can confer U2AF65 activity on a
heterologous RNA binding protein, Sex lethal, in a splicing reaction with the U2AF depleted
extract NEAU2AF (Valcarcel et al., 1993).
The S. cerevisiae splicing factor Mud2p
MUD2 is a clear functional homologue to the U2AF large subunit family. MUD2 has,
however, diverged in sequence, and the amino acid sequence similarity between U2AF65 and
Mud2p is difficult to detect (Abovich et al., 1994). Nonetheless, Mud2p has the same domain
structure and organization as U2AF65 and is the most similar molecule to the U2AF65 in the S.
cerevisiaegenome (Abovich et al., 1994). Like U2AF65, Mud2p interacts with the branch
sequence binding protein SF1/BBP (Berglund et al., 1998). Mud2p plays a role in the early
steps of spliceosome assembly, as does U2AF65 (Abovich et al., 1994).
MUD2 has been shown to interact genetically, but not biochemically, with the yeast
homologue of the U2 snRNP protein U2B ". YU2B " is not essential and mutants do not show
a growth defect, however the U2 snRNP-containing spliceosome complex is compromised in
yu2b" mutant extracts (Tang et al., 1996). Mud2p is present in the Ul snRNP complex CC2,
but is not present in CC1. Mud2p does not interact with Ul snRNP in the absence of premRNA (Abovich et al., 1994). Interaction of Mud2p with pre-mRNA is dependent on the
presence of a consensus branch sequence, but only moderately effected by mutation of the 5'
splice site. Mud2p has been shown to interact with PRP 11 both by synthetic lethality and yeast
two-hybrid analysis. PRP11 is the SAP62 component of the U2 snRNP component SF3a and
associates with the pre-mRNA only in the presence of U1 and ATP (Abovich et al., 1994).
Similarly, it has been shown that in the vertebrate equivalent of the yeast CC2 complex, the E
complex, U2AF interacts with SAP62 and U2 snRNP (Hong et al., 1997).
U2AF35
The small subunit of U2AF is also conserved between the fission yeast S. pombe
(Wentz and Potashkin, 1996) and Drosophila(Rudner et al., 1996) and consists of a conserved
N-terminal domain and less well conserved C-terminal RS domain. The N-terminal region of
the protein consists of a conserved PUMP domain (chapter 3) flanked by two regions of near
identity of about 60 and 20 amino acids each, between S. pombe, D. melanogaster,and humans
(Wentz and Potashkin, 1996). Interaction with the large subunit of U2AF has been shown to
be through this central domain. The U2AF small subunit has been shown to be required for
viability in Drosophila(Rudner et al., 1996)
Extracts depleted of U2AF, NEAU2AF, require the addition of U2AF65, but not
U2AF35, for reconstitution of splicing activity (Zamore and Green, 1991). A role for U2AF35
in splicing in vitro was found by using extracts that had been co-depleted of U2AF65 and
U2AF35 using polyclonal U2AF35 serum (Zuo and Maniatis, 1996). The remaining U2AF in
the extract is approximately stoichiometric for both subunits. In contrast, there is approximately
the same amount of U2AF35 remaining in NEAU2AF as in the antibody depleted extract, but
substantially less U2AF65 (Zuo and Maniatis, 1996). These antibody depleted extracts show
partial restoration of in vitro splicing activity with addition of either U2AF65 or U2AF35 and
substantially more splicing activity when both proteins are present. The function of U2AF35 in
these extracts is suggested by experiments in which the association of U2AF65 with the
pyrimidine tract of a pre-mRNA that required an enhancer sequence for in vitro splicing was
monitored. U2AF65 was shown to interact with this pyrimidine tract only in the presence of
SR proteins, which bound the enhancer, and U2AF35. This suggests that U2AF35 acts to
bridge U2AF65 and SR proteins bound to the pre-mRNA (Zuo and Maniatis, 1996). This
interpretation is supported by the observation that SR proteins interact preferentially with
U2AF35, and not U2AF65, in a two-hybrid assay (Wu and Maniatis, 1993). The interactions
between the SR proteins, bound to the pre-mRNA, and U2AF65, mediated by U2AF35, is
paralleled by the interaction between SR proteins, bound to the pre-mRNA, and Ul snRNP,
mediated by Ul 70k. This interaction may be functionally significant in formation of the E
complex and in splice site determination (Staknis and Reed, 1994b).
Paralogues2 of U2AF
The previous section has described the probable orthologues of U2AF65 and U2AF35;
both of these molecules also have paralogues. U2AF65 has two paralogues; the first is HCC,
which was identified as a human autoimmune antigen in hepatocarcinoma patients (Imai et al.,
1993); the second, PUF60 is the subject of chapters 2 and 3. U2AF35 also has a paralogue,
Urp or U2AFrsl (Tronchere et al., 1997). Urp is required for in vitro splicing and the Urp
deficit can not be complemented with U2AF35. Urp binds U2AF65 in the same region that
U2AF35 does. The region of Urp that interacts with U2AF65 contains an RRM-like domain
(discussed in chapter 3; Tronchere et al., 1997).
SF1/BBP
SF1 was first identified as a biochemical fraction that was required for spliceosomal
complex assembly together with other protein splicing factors, including U2AF and the U2
snRNP associated factors SF3, and purified snRNP particles (Brosi et al., 1993; Kramer and
Utans, 1991; Utans and Kramer, 1990). Subsequently it has been shown that SF1 has a branch
sequence binding activity and is the orthologue of the yeast protein BBP (Abovich and
Rosbash, 1997; Berglund et al., 1997). SF1/BBP interacts with Mud2p and the Ul snRNP
protein PRP40 in the CC2 complex (Abovich and Rosbash, 1997).
ASSEMBLY OF THE SPLICEOSOME
Spliceosome assembly is a multi-step process in which five snRNPs assemble on the
pre-mRNA in an ordered fashion and numerous proteins are found in the spliceosomal
complex, some are specific for particular complexes while others are found in more than one.
This discussion will focus on the assembly of the earliest complex and on the formation of
transitional complexes to the catalytic spliceosome. More complete descriptions of spliceosome
2
Paralogues are genes that show similarity in sequence, occur within the same genome and derive from
a common ancestral gene (this definition is from Kendrew, 1994). The prototypical example of a paralogue is
alpha and beta globin which are paralogues arising from the duplication of an ancestral globin gene. Paralogues
are contrasted to orthologues; while paralogues are observed within a single genome, within a single species,
orthologues are observed between species. Orthologous genes are related by similarity between species and are
related by descent to an ancestral molecule with no duplication events. Thus, the alpha globin genes of horse and
human are orthologues of one another, but the alpha globin gene and the beta globin gene are not orthologues
but paralogues.
assembly, the protein factors found in spliceosomal complexes and the RNA secondary
structures found in the spliceosomal complexes can be found elsewhere (Madhani and Guthrie,
1994; Moore et al., 1993; Reed, 1996). Spliceosome assembly is shown diagrammatically in
figure 3.
E complex
The first stable splicing complex that can be identified is the E, or early, complex (Kohtz
et al., 1994; Michaud and Reed, 1993) which is analogous to the commitment complexes, CC1
and CC2, seen in S. cerevisiae (Abovich and Rosbash, 1997; S6raphin and Rosbash, 1989;
S6raphin and Rosbash, 1991). E complex forms in the absence of ATP. E complexes can form
on both 5' and 3' half substrates; while not requiring an intact pre-mRNA, E does require the
presence of splice site sequences. Surprisingly, exonic sequences are also important in E5' and
E3' complex formation (Michaud and Reed, 1993). The yeast CC1 complex requires the 5'
splice site, but does not require the branch sequence for formation (S6raphin and Rosbash,
1989; S6raphin and Rosbash, 1991). It is likely that Ul snRNP binds to the pre-mRNA at
many 5' splice site sequence consensus sites that may not correspond to the 5' splice site and
that only those Ul snRNP complexes that bind near to SR proteins bound to enhancer
sequences are stabilized and can initiate formation of the ATP-dependent spliceosomal
complexes. Consistent with this there appears to be a higher stoichiometry of Ul snRNP in E
complex than there is for the other snRNAs in later complexes (Michaud and Reed, 1991;
Michaud and Reed, 1993). In one case it has been shown that Ul snRNP binds to a nonfunctional 5' splice site sequence (Siebel et al., 1992). However, in this case binding of U1
snRNP to this sequence is important in regulating this intron. It is also likely that U2AF binds
to pyrimidine tracts some of which may not be 3' splice site sequences. So although Ul
snRNP and U2AF have clear roles in splice site sequence recognition, they are probably not
sufficient, in themselves, to determine the splice site. This is consistent with the previous
discussion arguing that the splice site sequences themselves are insufficient to determine splice
sites. Transition from the E complex to the A complex, which contains a stably bound U2
snRNP, requires ATP (Konarska and Sharp, 1986); ATP hydrolysis may, then, be required for
the splice site determination step.
There are two candidate classes of ATPases that may be responsible for the ATPdependent E to A complex transition. The first class of ATPases is the DEAD-box family of
proteins which are believed to be ATP-dependent helicases (Will and Luhrmann, 1997).
Although there are currently no helicases known to function at this step in spliceosome
assembly, U2AF65 is known to bind to the UAP56 DEAD box protein and this protein may
play a role in this transition (Fleckner et al., 1997). The Prp5p DEAD-box protein may also be
required for this step in yeast (Wiest et aL, 1996). The second class of ATPases that may play a
role in this process are the RS-domain kinases. Two RS-domain kinases have been purified
based on substrate specificity (Colwill et al., 1996; Gui et al., 1994) and another RS domain
kinase activity associates with Ul snRNP (Tazi et al., 1993). DNA topoisomerase I is also a
candidate RS kinase (Rossi et al., 1996). These kinases have not been demonstrated to have an
essential role in splicing, but it is known that dephosphorylation is essential for splicing
(Mermoud et al., 1994), suggesting that a phosphorylation-dephosphorylation cycle is
important in splicing (Cao et al., 1997). In these experiments phosphatase inhibitors blocked
splicing, however spliceosomal complexes formed (Mermoud et al., 1994).
Thiophosphorylation, known to inhibit phosphatases, of the Ul 70k protein also leads to
accumulation of spliceosomal complexes and to a block to splicing (Tazi et al., 1993).
The SR proteins have been shown to be present in the E complex and to play a role in
intron recognition (Staknis and Reed, 1994b). It is interesting in this respect to recall that
exonic sequences are important for the formation of the E complex (Michaud and Reed, 1993).
SR proteins generally have two N-terminal RNA recognition motif (RRM) domains that are
important for both sequence specific binding (Manley and Tacke, 1996) and intron specific
splicing (Chandler et al., 1997; Tacke and Manley, 1995). SR proteins have also been shown
to bind splicing regulatory elements (Lynch and Maniatis, 1996) and are likely to bind enhancer
elements more generally. SR proteins would, therefore, be well suited to act as mediators in
recognition of the context information that is required in addition to the splice site sequence
information.
These results suggest a model in which Ul snRNP and U2AF may be stabilized by
interaction with SR proteins bound to nearby enhancer sequences. This interaction is likely to
require phosphorylation of the SR domains of one or both components of the interaction (Xiao
and Manley, 1997). The biochemical interaction between Ul snRNP or U2AF65 and the SR
proteins would link splice site sequence information, in the form of the enhancer sequences, to
context information, and also link ATP hydrolysis (phosphorylation of SR domains) to the
transition from E to A complex. The complexes observed in the presence of phosphatase
inhibitors and thiophosphorylated Ul 70k may represent complexes that are blocked in the
transition from the U1 snRNP containing complexes to the later spliceosomal complexes. A
model in which factors that associate with the splice site sequences, such as Ul 70k and U2AF,
also interact with factors, such as SR proteins, associated with exonic or intronic sequence
elements may allow for greater flexibility and fidelity than a model in which the factors bind to
only one or two sequences. Herschlag argues that multiple, weak interactions can be more
specific than a single, strong interaction (Herschlag, 1991).
This hypothesis also predicts that phosphatases and kinases may play a role in regulating
splicing. Interestingly, recent work suggests that a phosphatase interacts with PSF, a
pyrimidine tract-associated splicing factor (Hirano et al., 1996; Patton et al., 1993). The PSFassociated protein PTB has been shown to play a role in inhibiting exon inclusion (Ashiya and
Grabowski, 1997; Gooding et al., 1998).
It is interesting to contrast the splice site determination-system in vertebrates to that
found in yeast where there are no clear orthologues to the SR proteins. SR proteins may not be
required in yeast as there is much more information present in the yeast splice site sequences
than in vertebrate sequences, perhaps enough to uniquely define each intron (C. Burge,
personal communication). In the case of yeast splice site determination, additional enhancer
information, or context, may not be required and all the splice site determination information
may be found within the splice site sequences themselves. Notably, there are few long introns
in yeast and for these introns an RNA duplex formed between intronic sequences, near the
intron ends, is thought to assist splicing efficiency (Charpentier and Rosbash, 1996). These
duplex sequences may be the functional equivalent to splicing enhancer sequences found in the
more complicated vertebrate splicing systems.
A complex
U2 snRNP has recently been shown to associate with E complex; the SF3a component
of 17S U2 snRNP is likely to be important for this interaction (Hong et al., 1997). The S.
cerevisiaeequivalent of SF3a is also required for the binding of U2 snRNP to the CC complex
(Ruby et al., 1993). SF3a is composed of three proteins, SAP61, SAP62 and SAP114, and
these proteins are the orthologues of PRP9, PRP11 and PRP21 of S. cerevisiae (Behrens et al.,
1993; Bennett and Reed, 1993; Brosi et al., 1993). The yeast gene PRP5 is a DEAD-box
protein and interacts with PRP9, 11 and 21 and so may be responsible for mediating the ATPdependent association of U2 snRNP with the branch sequence in yeast (Dalbadie-McFarland
and Abelson, 1990; Ruby et al., 1993). The A. complex may also represent an early stage in
the association of U2 snRNP with the pre-mRNA (Query et al., 1997). Assembly of this
complex requires the activity of the PUF fraction that is the subject of chapters 2 and 3 (Query et
al., 1997).
U2 snRNP has been shown to base-pair with the branch sequence (Parker et al., 1987;
Wu and Manley, 1989; Zhuang and Weiner, 1989), leaving the branch adenosine unpaired and
bulged in A complex (see for example; Query et al., 1994). The SF3a components have been
shown to contact the pre-mRNA both 5' and 3' to the branch sequence in this complex. This
interaction does not occur prior to the ATP-dependent stable association of U2 snRNP to the
branch sequence (Gozani et al., 1996).
The spliceosomal complexes B and C
Stable binding of U2 snRNP to the pre-mRNA is rapidly followed by the association of
the tri-snRNP complex U4/5/6 snRNP (Konarska and Sharp, 1986). Factors required for the
association of the tri-snRNP with the pre-mRNA and the structure of this complex have been
described elsewhere (Moore et al., 1993). The catalytically active spliceosome, C, is thought to
result from a conformational change in the RNA and protein components present in the B
complex (Moore et al., 1993). Some second-step specific splicing factors may associate at this
time as well (Umen and Guthrie, 1995). The B and C complexes and protein factors and genes
responsible for their assembly and function have been reviewed elsewhere (Moore et al., 1993).
Few experiments address the question of how the transition from the A to B/C complexes is
accomplished. I will describe an unusual complex identified by Ast and colleagues that may
represent an intermediate transitional complex between the A and the B/C complexes (Ast and
Weiner, 1997a; Ast and Weiner, 1997b; Ast and Weiner, 1996).
Ast and co-workers have shown that a U1/4/5 snRNP complex is detectable under some
conditions (Ast and Weiner, 1996). This was unexpected as both Ul and U4 are destabilized
from the spliceosome before splicing catalysis (Blencowe et al., 1989; Cheng and Abelson,
1987; Lamond et al., 1988; Michaud and Reed, 1993; Pikielny et al., 1986; Yean and Lin,
1991), but U5 is associated with the spliceosome at the time of catalysis (Sontheimer and Steitz,
1993). A function for this complex seemed, at first, obscure. In more detail, this work shows
that addition of an oligo, BUSAe, complementary to the 3' end of the U5 stem induces a
conformational change in U5 snRNP (Ast and Weiner, 1997a). The conformational change
leads to the formation of a U5 complex with Ul and U4 snRNPs and this U1/4/5 complex has
5' splice site binding properties. Addition of a pyrimidine-tract 3' splice site oligo to the 5'
splice site binding reaction increases association of the 5' splice site oligo by 8-fold.
Association of the pyrimidine tract occurs in a complex containing U2 and U6 snRNPs to the
U1/4/5 complex by protein factors. The interaction of U2 and U6 snRNPs with the U1/4/5
complex enhanced by the presence of pre-mRNA suggests that a factor mediates a bridging
interaction (Ast and Weiner, 1996). Such a bridging interaction could be mediated by
interaction with U2 via the pyrimidine tract and with U1 via Ul 70 kD. The formation of a
U 1/4/5 complex and its possible association with a U2/6 complex is suggestive of a transitional
complex that may form between the commitment complex and the catalytic spliceosome. It is
tempting to speculate that a U 1/4-containing complex disassembles from the pre-mRNA in a
concerted fashion as the U2/5/6 spliceosomal components bind to the pre-mRNA. The U 1/4/5
complex may then represent the final complex formed before U5 binds the exonic region of the
5' splice site and Ul and U4 dissociate from the pre-mRNA.
The presence of the BU5Ae oligo is required for the formation of these complexes.
While the addition of this oligo does not inhibit splicing and can associate with U5 snRNP in
the spliceosome through both catalytic steps, it is not clear whether the observed changes in U5
snRNP caused by BU5Ae faithfully model the changes that occur normally during spliceosome
complex assembly (Ast and Weiner, 1996). The nature of these changes, if shown to occur in
the spliceosome, will be of great interest. It will also be of interest to determine whether the
complexes observed in the presence of the BU5Ae oligo can be observed in its absence.
MY CONTRIBUTIONS TO THIS PROJECT:
Chapter two presents the identification, purification, and characterization of a new
pyrimidine tract binding factor, poly[U] factor (PUF), that is required for efficient splicing in
vitro. Chapter three describes a novel pyrimidine tract binding protein present in the PUF factor
that associates with p54, an SR-protein splicing factor, and identifies a domain found of that
protein as a protein-protein interaction domain.
Chapter four describes experiments by Andrew MacMillan, John Crispino and myself
(MacMillan et al., 1997) that demonstrate that U2AF is not required for splicing in vitro. John
had previously demonstrated that U 1 snRNP is not required for splicing in vitro and Andrew
determined using the poly[U]-depleted splicing extract, described in chapter 2 (NEAU), that
U2AF was not required for splicing in vitro in the presence of high concentrations of added
SC35, an SR protein. As described above, neither Ul snRNP nor U2AF are thought to be
present in the catalytic spliceosome, these experiments and John Crispino's work (Crispino et
al., 1994; Crispino and Sharp, 1995) suggest that the spliceosomes formed in the absence of
U1 snRNP or U2AF have bypassed the splice site determination step. It is interesting to note
that PUF is required in the U2AF-depleted SC35 reconstitution experiments; this suggests that
PUF acts at a post-U2AF dependent step in spliceosome assembly. Alternatively, under these
conditions, PUF may substitute for U2AF function.
Chapter 5 describes the identification of a U2 snRNP complex that forms on a minimal
RNA ligand (Query et al., 1997). Charles Query demonstrated that this complex uncovers
sequence binding specificities of U2 snRNP that are masked in the U2 snRNP complexes that
form on larger RNAs. These specificities are presumably masked when U2 snRNP binds to
longer RNAs due to the RNA binding activities of U2 snRNP associated proteins (Staknis and
Reed, 1994a). Together, Charles and I, showed that this U2 snRNP complex requires the
PUF and U2AF activity to form in NEAU. This demonstrates that PUF activity is required
prior to, or coincident with, U2 snRNP complex formation at the branch sequence.
The thesis includes a speculative appendix that describes a model for the interaction of
the branch adenosine with the conserved U5 snRNA loop. This base-pairing interaction
juxtaposes the 5' splice site phosphodiester bond with the 2' hydroxyl nucleophile of the branch
adenosine and so may be an interaction that aligns these substrates for the first chemical step.
The model is discussed in the context of the genetic and biochemical evidence describing the
interaction between the U5 snRNA loop and the 5' splice site sequence (Newman, 1997;
O'Keefe et al., 1996; Sontheimer and Steitz, 1993; Wyatt et al., 1992) and found to be
consistent with this interaction. Experiments that test the model are suggested.
In an afterword, experiments are discussed that will help to clarify the many outstanding
issues regarding the PUF factor and PUF60. Particular emphasis will be placed on determining
if PUF60 is required for splicing in vitro.
REFERENCES
Abovich, N., Liao, X. C., and Rosbash, M. (1994). The yeast MUD2 protein: an interaction
with PRP 11 defines a bridge between commitment complexes and U2 snRNP addition. Genes
Dev 8, 843-54.
Abovich, N., and Rosbash, M. (1997). Cross-intron bridging interactions in the yeast
commitment complex are conserved in mammals. Cell 89, 403-12.
Anderson, K., and Moore, M. J. (1997). Bimolecular exon ligation by the human spliceosome.
Science 276, 1712-6.
Ashiya, M., and Grabowski, P. J. (1997). A neuron-specific splicing switch mediated by an
array of pre-mRNA repressor sites: evidence of a regulatory role for the polypyrimidine tract
binding protein and a brain-specific PTB counterpart. RNA 3, 996-1015.
Ast, G., and Weiner, A. M. (1997a). Antisense oligonucleotide binding to U5 snRNP induces
a conformational change that exposes the conserved loop of U5 snRNA. Nucleic Acids Res 25,
3508-13.
Ast, G., and Weiner, A. M. (1997b). A novel U1/U5 interaction indicates proximity between
U1 and U5 snRNAs during an early step of mRNA splicing. RNA 3, 371-81.
Ast, G., and Weiner, A. M. (1996). A U1/U4/U5 snRNP complex induced by a 2'-O-methyloligoribonucleotide complementary to U5 snRNA. Science 272, 881-4.
Barabino, S. M. L., Blencowe, B. J., Ryder, U., Sproat, B. S., and Lamond, A. I. (1990).
Targeted snRNP depletion reveals an additional role for mammalian Ul snRNP in spliceosome
assembly. Cell 63, 293-302.
Behrens, S. E., Galisson, F., Legrain, P., and Luhrmann, R. (1993). Evidence that the 60-kDa
protein of 17S U2 small nuclear ribonucleoprotein is immunologically and functionally related
to the yeast PRP9 splicing factor and is required for the efficient formation of prespliceosomes.
Proc Natl Acad Sci U S A 90, 8229-33.
Bennett, M., and Reed, R. (1993). Correspondence between a mammalian spliceosome
component and an essential yeast splicing factor. Science 262, 105-8.
Berget, S. M., Moore, C., and Sharp, P. A. (1977). Spliced segments at the 5' terminus of
adenovirus 2 late mRNA. Proc. Natl. Acad. Sci. USA 74, 3171-3175.
Berglund, J. A., Abovich, N., and Rosbash, M. (1998). A cooperative interaction between
U2AF65 and mBBP/SF1 facilitates branchpoint region recognition. Genes Dev 12, 858-67.
Berglund, J. A., Chua, K., Abovich, N., Reed, R., and Rosbash, M. (1997). The splicing
factor BBP interacts specifically with the pre-mRNA branchpoint sequence UACUAAC. Cell
89, 781-7.
Birney, E., Kumar, S., and Krainer, A. R. (1993). Analysis of the RNA-recognition motif and
RS and RGG domains: conservation in metazoan pre-mRNA splicing factors. Nucleic Acids
Res 21, 5803-16.
Blencowe, B. J., Sproat, B. S., Ryder, U., Barabino, S., and Lamond, A. I. (1989).
Antisense probing of the human U4/U6 snRNP with biotinylated 2'-OMe RNA
oligonucleotides. Cell 59, 531-539.
Boguski, M. S., and McCormick, F. (1993). Proteins regulating Ras and its relatives. Nature
366, 643-54.
Brody, E., and Abelson, J. (1985). The 'spliceosome': Yeast premessenger RNA associates
with a 40 S complex in a splicing-dependent reaction. Science 228, 963-967.
Brosi, R., Groning, K., Behrens, S. E., Luhrmann, R., and Krimer, A. (1993). Interaction of
mammalian splicing factor SF3a with U2 snRNP and relation of its 60-kD subunit to yeast
PRP9. Science 262, 102-5.
Burge, C., and Sharp, P. A. (1998). Manuscript in preparation. In The RNA World II.
Cao, W.,Jamison, S. F., and Garcia-Blanco, M. A. (1997). Both phosphorylation and
dephosphorylation of ASF/SF2 are required for pre-mRNA splicing in vitro. RNA 3, 14561467.
Cellini, A., Parker, R., McMahon, J., Guthrie, C., and Rossi, J. (1986). Activation of a
cryptic TACTAAC box in the Saccharomyces cerevisiae actin intron. Mol Cell Biol 6, 1571-8.
Chan, R. C., and Black, D. L. (1995). Conserved intron elements repress splicing of a neuronspecific c-src exon in vitro. Mol Cell Biol 15, 6377-85.
Chandler, S. D., Mayeda, A., Yeakley, J. M., Krainer, A. R., and Fu, X. D. (1997). RNA
splicing specificity determined by the coordinated action of RNA recognition motifs in SR
proteins. Proc Natl Acad Sci U S A 94, 3596-601.
Charpentier, B., and Rosbash, M. (1996). Intramolecular structure in yeast introns aids the
early steps of in vitro spliceosome assembly. RNA 2, 509-22.
Cheng, S. C., and Abelson, J. (1987). Spliceosome assembly in yeast. Genes Dev 1, 10141027.
Chow, L. T., Gelinas, R. E., Broker, T. R., and Roberts, R. J. (1977). An amazing sequence
arrangement at the 5' ends of adenovirus 2 messenger RNA. Cell 12, 1-8.
Colwill, K., Pawson, T., Andrews, B., Prasad, J., Manley, J. L., Bell, J. C., and Duncan, P.
I. (1996). The Clk/Sty protein kinase phosphorylates SR splicing factors and regulates their
intranuclear distribution. EMBO J 15, 265-75.
Crispino, J. D., Blencowe, B. J., and Sharp, P. A. (1994). Complementation by SR proteins
of pre-mRNA splicing reactions depleted of U1 snRNP. Science 265, 1866-9.
Crispino, J. D., Mermoud, J. E., Lamond, A. I., and Sharp, P. A. (1996). Cis-acting elements
distinct from the 5' splice site promote U -independent pre-mRNA splicing. RNA 2, 664-73.
Crispino, J. D., and Sharp, P. A. (1995). A U6 snRNA:pre-mRNA interaction can be ratelimiting for Ul-independent splicing. Genes & Dev. 9, 2314-2323.
Dalbadie-McFarland, G., and Abelson, J. (1990). PRP5: A helicase-like protein required for
mRNA splicing in yeast. Proc. Natl. Acad. Sci. USA 87, 4236-4240.
Faustino, P., Osorio-Almeida, L., Romao, L., Barbot, J., Fernandes, B., Justica, B., and
Lavinha, J. (1998). Dominantly transmitted beta-thalassemia arising from the production of
several aberrant mRNA species and one abnormal peptide. Blood 91, 685-90.
Fleckner, J., Zhang, M., Valcarcel, J., and Green, M. R. (1997). U2AF65 recruits a novel
human DEAD box protein required for the U2 snRNP-branchpoint interaction. Genes Dev 11,
1864-72.
Fouser, L. A., and Friesen, J. D. (1987). Effects on mRNA splicing of mutations in the 3'
region of the Saccharomyces cerevisiae actin intron. Mol Cell Biol 7, 225-30.
Fouser, L. A., and Friesen, J. D. (1986). Mutations in a yeast intron demonstrate the
importance of specific conserved nucleotides for the two stages of nuclear mRNA splicing. Cell
45, 81-93.
Frendeway, D., and Keller, W. (1985). Stepwise assembly of a pre-mRNA splicing complex
requires U-snRNAs and specific intron sequences. Cell 42, 355-367.
Gooding, C., Roberts, G. C., and Smith, C. W. (1998). Role of an inhibitory pyrimidine
element and polypyrimidine tract binding protein in repression of a regulated alpha-tropomyosin
exon. RNA 4, 85-100.
Gozani, O., Feld, R., and Reed, R. (1996). Evidence that sequence-independent binding of
highly conserved U2 snRNP proteins upstream of the branch site is required for assembly of
spliceosomal complex A. Genes Dev 10, 233-43.
Grabowski, P. J., Padgett, R. A., and Sharp, P. A. (1984). Messenger RNA splicing in vitro:
An excised intervening sequence and a potential intermediate. Cell 37, 415-427.
Grabowski, P. J., Seiler, S. R., and Sharp, P. A. (1985). A multicomponent complex is
involved in the splicing of messenger RNA precursors. Cell 42, 345-353.
Green, M. R. (1986). Pre-mRNA splicing. Annu. Rev. Genet. 20, 671-708.
Gui, J. F., Tronchere, H., Chandler, S. D., and Fu, X. D. (1994). Purification and
characterization of a kinase specific for the serine- and arginine-rich pre-mRNA splicing factors.
Proc Natl Acad Sci U S A 91, 10824-8.
Hall, S. L., and Padgett, R. A. (1996). Requirement of U12 snRNA for in vivo splicing of a
minor class of eukaryotic nuclear pre-mRNA introns. Science 271, 1716-8.
Heinrichs, V., Bach, M., Winkelmann, G., and Luhrmann, R. (1990). Ul-specific protein C
needed for efficient complex formation of U1 snRNP with a 5' splice site. Science 247, 69-72.
Herschlag, D. (1991). Implications of ribozyme kinetics for targeting the cleavage of specific
RNA molecules in vivo: more isn't always better. Proc Natl Acad Sci U S A 88, 6921-6925.
Hilleren, P. J., Kao, H. Y., and Siliciano, P. G. (1995). The amino-terminal domain of yeast
U 1-70K is necessary and sufficient for function. Mol Cell Biol 15, 6341-50.
Hirano, K., Erdodi, F., Patton, J. G., and Hartshorne, D. J. (1996). Interaction of protein
phosphatase type 1 with a splicing factor. FEBS Lett 389, 191-4.
Hodgkin, J., Papp, A., Pulak, R., Ambros, V., and Anderson, P. (1989). A new kind of
informational suppression in the nematode Caenorhabditis elegans. Genetics 123, 301-13.
Hong, W., Bennett, M., Xiao, Y., Feld Kramer, R., Wang, C., and Reed, R. (1997).
Association of U2 snRNP with the spliceosomal complex E. Nucleic Acids Res 25, 354-61.
Imai, H., Chan, E. K., Kiyosawa, K., Fu, X. D., and Tan, E. M. (1993). Novel nuclear
autoantigen with splicing factor motifs identified with antibody from hepatocellular carcinoma. J
Clin Invest 92, 2419-26.
Jacquier, A., Rodriguez, J. R., and Rosbash, M. (1985). A quantitative analysis of the effects
of 5' junction and TACTAAC box mutants and mutant combinations on yeast mRNA splicing.
Cell 43, 423-30.
Jamison, S. F., Pasman, Z., Wang, J., Will, C., Luhrmann, R., Manley, J. L., and GarciaBlanco, M. A. (1995). U snRNP-ASF/SF2 interaction and 5' splice site recognition:
characterization of required elements. Nucleic Acids Res 23, 3260-7.
Kanaar, R., Roche, S. E., Beall, E. L., Green, M. R., and Rio, D. C. (1993). The conserved
pre-mRNA splicing factor U2AF from Drosophila: requirement for viability. Science 262, 56973.
Kandels-Lewis, S., and Seraphin, B. (1993). Involvement of U6 snRNA in 5' splice site
selection. Science 262, 2035-9.
Kao, H. Y., and Siliciano, P. G. (1996). Identification of Prp40, a novel essential yeast
splicing factor associated with the Ul small nuclear ribonucleoprotein particle. Mol Cell Biol
16, 960-7.
Kendrew, J. (1994). The Encyclopedia of Molecular Biology (Oxford: Blackwell Science Ltd.).
Kohtz, J. D., Jamison, S. F., Will, C. L., Zuo, P., Luhrmann, R., Garcia-Blanco, M. A., and
Manley, J. L. (1994). Protein-protein interactions and 5'-splice-site recognition in mammalian
mRNA precursors. Nature 368, 119-24.
Konarska, M. M., and Sharp, P. A. (1986). Electrophoretic separation of complexes involved
in the splicing of precursors to mRNAs. Cell 46, 845-855.
Krainer, A. R., Maniatis, T., Ruskin, B., and Green, M. R. (1984). Normal and mutant
human beta-globin pre-mRNAs are faithfully and efficiently spliced in vitro. Cell 36, 993-1005.
Kramer, A., and Utans, U. (1991). Three protein factors (SF1, SF3 and U2AF) function in
pre-splicing complex formation in addition to snRNPs. EMBO J 10, 1503-9.
Lamond, A. I., Konarska, M. M., Grabowski, P. J., and Sharp, P. A. (1988). Spliceosome
assembly involves binding and release of U4 small nuclear ribonucleoprotein. Proc. Natl. Acad.
Sci. USA 85, 411-415.
Lavigueur, A., LaBranche, H., Komblihtt, A. R., and Chabot, B. (1993). A splicing enhancer
in the human fibronectin alternate ED 1 exon interacts with SR proteins and stimulates U2
snRNP binding. Genes & Dev. 7, 2405-2417.
Lerner, M. R., Boyle, J. A., Mount, S. M., Wolin, S. L., and Steitz, J. A. (1980). Are
snRNPs involved in splicing? Nature 283, 220-224.
Lesser, C. F., and Guthrie, C. (1993). Mutations in U6 snRNA that alter splice site specificity:
implications for the active site. Science 262, 1982-8.
Liao, X. C., Colot, H. V., Wang, Y., and Rosbash, M. (1992). Requirements for U2 snRNP
addition to yeast pre-mRNA. Nucleic Acids Res. 20, 4237-4245.
Lockhart, S. R., and Rymond, B. C. (1994). Commitment of yeast pre-mRNA to the splicing
pathway requires the novel Ul snRNP polypeptide, Prp39p. Mol. Cell. Biol., in press.
Lu, J., and Hall, K. B. (1995). An RBD that does not bind RNA: NMR secondary structure
determination and biochemical properties of the C-terminal RNA binding domain from the
human U1A protein. J Mol Biol 247, 739-52.
Lutz-Freyermuth, C., Query, C. C., and Keene, J. D. (1990). Quantitative determination that
one of two potential RNA-binding domains of the A protein component of the Ul small nuclear
ribonucleoprotein complex binds with high affinity to stem-loop II of U1 RNA. Proc. Natl.
Acad. Sci. USA 87, 6393-6397.
Lynch, K. W., and Maniatis, T. (1996). Assembly of specific SR protein complexes on distinct
regulatory elements of the Drosophila doublesex splicing enhancer. Genes & Dev. 10, 20892101.
MacMillan, A. M., McCaw, P. S., Crispino, J. D., and Sharp, P. A. (1997). SC35-mediated
reconstitution of splicing in U2AF-depleted nuclear extract. Proc Natl Acad Sci U S A 94, 1336.
Madhani, H. D., and Guthrie, C. (1994). Dynamic RNA-RNA interactions in the spliceosome.
Annu Rev Genet 28, 1-26.
Mancebo, R., Lo, P. C. H., and Mount, S. M. (1990). Structure and expression of the
Drosophilamelanogastergene for the Ul small nuclear ribonucleoprotein particle 70K protein.
Mol. Cell. Biol. 10, 2492-2502.
Maniatis, T., and Reed, R. (1987). The role of small nuclear ribonucleoprotein particles in premRNA splicing. Nature 325, 673-8.
Manley, J. L., and Tacke, R. (1996). SR proteins and splicing control. Genes Dev 10, 156979.
Mermoud, J. E., Cohen, P. T., and Lamond, A. I. (1994). Regulation of mammalian
spliceosome assembly by a protein phosphorylation mechanism. EMBO J 13, 5679-88.
Michaud, S., and Reed, R. (1991). An ATP-independent complex commits pre-mRNA to the
mammalian spliceosome assembly pathway. Genes & Dev. 5, 2534-2546.
Michaud, S., and Reed, R. (1993). A functional association between the 5' and 3' splice site is
established in the earliest prespliceosome complex (E) in mammals. Genes Dev 7, 1008-20.
Min, J., Shukla, H., Kozono, H., Bronson, S. K., Weissman, S. M., and Chaplin, D. D.
(1995). A novel Creb family gene telomeric of HLA-DRA in the HLA complex. Genomics 30,
149-56.
Moore, M. J., Query, C. C., and Sharp, P. A. (1993). Splicing of precursors to mRNA by the
spliceosome. In The RNA World, R. Gesteland and J. Atkins, eds. (New York: Cold Spring
Harbor Laboratory Press), pp. 303-357.
Mount, S. M. (1982). A catalogue of splice junction sequences. Nucleic Acids Res 10, 459-72.
Mount, S. M., Burks, C., Hertz, G., Stormo, G. D., White, 0., and Fields, C. (1992).
Splicing signals in Drosophila:intron size, information content, and consensus sequences.
Nucleic Acids Res. 20, 4255-4262.
Mount, S. M., Pettersson, I., Hinterberger, M., Karmas, A., and Steitz, J. A. (1983). The Ul
small nuclear RNA-protein complex selectively binds a 5' splice site in vitro. Cell 33, 509-18.
Nagai, K. (1996). RNA-protein complexes. Curr. Opin. Struct. Biol. 6, 53-61.
Newman, A. J. (1997). The role of U5 snRNP in pre-mRNA splicing. EMBO J 16, 5797-800.
O'Keefe, R. T., Norman, C., and Newman, A. J. (1996). The invariant U5 snRNA loop 1
sequence is dispensable for the first catalytic step of pre-mRNA splicing in yeast. Cell 86, 67989.
Padgett, R. A., Konarska, M. M., Aebi, M., Hornig, H., Weissmann, C., and Sharp, P. A.
(1985). Nonconsensus branch-site sequences in the in vitro splicing of transcripts of mutant
rabbit 3-globin genes. Proc. Natl. Acad. Sci. 82, 8349-8353.
Padgett, R. A., Konarska, M. M., Grabowski, P. J., Hardy, S. F., and Sharp, P. A. (1984).
Lariat RNA's as intermediates and products in the splicing of messenger RNA precursors.
Science 225, 898-903.
Parker, R., and Guthrie, C. (1985). A point mutation in the conserved hexanucleotide at a yeast
5' splice junction uncouples recognition, cleavage, and ligation. Cell 41, 107-18.
Parker, R., Siliciano, P. G., and Guthrie, C. (1987). Recognition of the TACTAAC box
during mRNA splicing in yeast involves base pairing to the U2-like snRNA. Cell 49, 229-39.
Patton, J. G., Porro, E. B., Galceran, J., Tempst, P., and Nadal-Ginard, B. (1993). Cloning
and characterization of PSF, a novel pre-mRNA splicing factor. Genes & Dev. 7, 393-406.
Pikielny, C. W., Rymond, B. C., and Rosbash, M. (1986). Electrophoresis of
ribonucleoproteins reveals an ordered assembly pathway of yeast splicing complexes. Nature
324, 341-345.
Potashkin, J., Naik, K., and Wentz, H. K. (1993). U2AF homolog required for splicing in
vivo. Science 262, 573-5.
Pulak, R., and Anderson, P. (1993). mRNA surveillance by the Caenorhabditis elegans smg
genes. Genes Dev 7, 1885-97.
Query, C. C., Bentley, R. C., and Keene, J. D. (1989a). A common RNA recognition motif
identified within a defined Ul RNA binding domain of the 70K Ul snRNP protein. Cell 57,
89-101.
Query, C. C., Bentley, R. C., and Keene, J. D. (1989b). A specific 31 nucleotide domain of
Ul RNA directly interacts with the 70K Ul snRNP protein. Mol. Cell. Biol. 9, 4872-4881.
Query, C. C., McCaw, P. S., and Sharp, P. A. (1997). A minimal spliceosomal complex A
recognizes the branch site and polypyrimidine tract. Mol Cell Biol 17, 2944-53.
Query, C. C., Moore, M. J., and Sharp, P. A. (1994). Branch nucleophile selection in premRNA splicing: evidence for the bulged duplex model. Genes & Dev. 8, 587-597.
Reddy, R., and Busch, H. (1988). Small Nuclear RNAs: RNA Sequences, Structure and
Modifications. In Small Ribonucleoprotein Particles, M. L. Birnsteil, ed. (Berlin: SpringerVerlag), pp. 1-37.
Reed, R. (1996). Initial splice-site recognition and pairing during pre-mRNA splicing. Curr
Opin Genet Dev 6, 215-20.
Reed, R., and Maniatis, T. (1985). Intron sequences involved in lariat formation during premRNA splicing. Cell 41, 95-105.
Robberson, B. L., Cote, G. J., and Berget, S. M. (1990). Exon definition may facilitate splice
site selection in RNAs with multiple exons. Mol. Cell. Biol. 10, 84-94.
Rogers, J., and Wall, R. (1980). A mechanism for RNA splicing. Proc. Natl. Acad. Sci. USA
77, 1877-1879.
Roscigno, R. F., Weiner, M., and Garcia-Blanco, M. A. (1993). A mutational analysis of the
polypyrimidine tract of introns. Effects of sequence differences in pyrimidine tracts on splicing.
J Biol Chem 268, 11222-9.
Rossi, F., Forne, T., Antoine, E., Tazi, J., Brunel, C., and Cathala, G. (1996). Involvement
of Ul small nuclear ribonucleoproteins (snRNP) in 5' splice site-U1 snRNP interaction. J Biol
Chem 271, 23985-91.
Rossi, F., Labourier, E., Forne, T., Divita, G., Derancourt, J., Riou, J. F., Antoine, E.,
Cathala, G., Brunel, C., and Tazi, J. (1996). Specific phosphorylation of SR proteins by
mammalian DNA topoisomerase I. Nature 381, 80-2.
Ruby, S. W., Chang, T. H., and Abelson, J. (1993). Four yeast spliceosomal proteins (PRP5,
PRP9, PRP11, and PRP21) interact to promote U2 snRNP binding to pre-mRNA. Genes Dev
7, 1909-25.
Rudner, D. Z., Kanaar, R., Breger, K. S., and Rio, D. C. (1996). Mutations in the small
subunit of the Drosophila U2AF splicing factor cause lethality and developmental defects. Proc
Natl Acad Sci U S A 93, 10333-7.
Ruskin, B., and Green, M. R. (1985a). Role of the 3' splice site consensus sequence in
mammalian pre-mRNA splicing. Nature 317, 732-4.
Ruskin, B., and Green, M. R. (1985b). Specific and stable intron-factor interactions are
established early during in vitro pre-mRNA splicing. Cell 43, 131-42.
Ruskin, B., Krainer, A. R., Maniatis, T., and Green, M. R. (1984). Excision of an intact
intron as a novel lariat structure during pre- mRNA splicing in vitro. Cell 38, 317-31.
Ruskin, B., Zamore, P. D., and Green, M. R. (1988). A factor, U2AF, is required for U2
snRNP binding and splicing complex assembly. Cell 52, 207-19.
Rymond, B. C., and Rosbash, M. (1992). Yeast pre-mRNA splicing. In The Molecular and
Cellular Biology of the Yeast Saccharomyces: Gene Expression, E. W. Jones, J. R. Pringle and
J. R. Broach, eds. (Cold Spring Harbor, New York: Cold Spring Harbor Laboratory Press),
pp. 143-192.
Sawa, H., and Abelson, J. (1992). Evidence for a base-pairing interaction between U6 snRNA
and the 5' splice site during the splicing reaction in yeast. Proc. Natl. Acad. Sci. USA 89,
11269-11273.
Sawa, H., and Shimura, Y. (1992). Association of U6 snRNA with the 5'-splice site region of
pre-mRNA in the spliceosome. Genes & Dev. 6, 244-254.
Senapathy, P., Shapiro, M. B., and Harris, N. L. (1990). Splice junctions, branch point sites,
and exons: sequence statistics, identification, and applications to genome project. Methods
Enzymol. 183, 252-278.
S6raphin, B., and Rosbash, M. (1989). Identification of functional Ul snRNP-pre-mRNA
complexes committed to spliceosome assembly and splicing. Cell 59, 349-358.
S6raphin, B., and Rosbash, M. (1991). The yeast branchpoint sequence is not required for the
formation of a stable Ul snRNP pre-mRNA complex and is recognized in the absence of U2
snRNA. EMBO J. 10, 1209-1216.
Sherr, C. J. (1996). Cancer cell cycles. Science 274, 1672-7.
Siebel, C. W., Fresco, L. D., and Rio, D. C. (1992). The mechanism of somatic inhibition of
DrosophilaP-element pre-mRNA splicing: multiprotein complexes at an exon pseudo-5' splice
site control Ul snRNP binding. Genes & Dev. 6, 1386-1401.
Smith, C. W. J., Porro, E. B., Patton, J. G., and Nadal-Ginard, B. (1989). Scanning from an
independently specified branch point defines the 3' splice site of mammalian introns. Nature
342, 243-247.
Smith, V., and Barrell, B. G. (1991). Cloning of a yeast U1 snRNP 70K protein homologue:
functional conservation of an RNA-binding domain between humans and yeast. EMBO J 10,
2627-34.
Sontheimer, E. J., and Steitz, J. A. (1993). The U5 and U6 small nuclear RNAs as active site
components of the spliceosome. Science 262, 1989-96.
Staknis, D., and Reed, R. (1994a). Direct interactions between pre-mRNA and six U2 small
nuclear ribonucleoproteins during spliceosome assembly. Mol Cell Biol 14, 2994-3005.
Staknis, D., and Reed, R. (1994b). SR proteins promote the first specific recognition of PremRNA and are present together with the U1 small nuclear ribonucleoprotein particle in a general
splicing enhancer complex. Mol Cell Biol 14, 7670-82.
Stephens, R. M., and Schneider, T. D. (1992). Features of spliceosome evolution and function
inferred from an analysis of the information at human splice sites. J Mol Biol 228, 1124-36.
Sun, Q., Hampson, R. K., Krainer, A. T., and Rottman, F. M. (1993). General splicing factor
SF2/ASF promotes alternative splicing by binding to an exonic splicing enhancer. Genes &
Dev. 7, 2598-2608.
Surowy, C. S., van Santen, V. L., Scheib-Wixted, S. M., and Spritz, R. A. (1989). Direct,
sequence-specific binding of the human U1-70K ribonucleoprotein antigen protein to loop I of
U1 small nuclear RNA. Mol Cell Biol 9,4179-86.
Tacke, R., Chen, Y., and Manley, J. L. (1997). Sequence-specific RNA binding by an SR
protein requires RS domain phosphorylation: creation of an SRp40-specific splicing enhancer.
Proc Natl Acad Sci U S A 94, 1148-53.
Tacke, R., and Manley, J. L. (1995). The human splicing factors ASF/SF2 and SC35 possess
distinct, functionally significant RNA binding specificities. EMBO J 14, 3540-51.
Talerico, M., and Berget, S. M. (1994). Intron definition in splicing of small Drosophila
introns. Mol Cell Biol 14, 3434-45.
Tang, J., Abovich, N., Fleming, M. L., Seraphin, B., and Rosbash, M. (1997). Identification
and characterization of a yeast homolog ofU snRNP-specific protein C. EMBO J 16,408291.
Tang, J., Abovich, N., and Rosbash, M. (1996). Identification and characterization of a yeast
gene encoding the U2 small nuclear ribonucleoprotein particle B" protein. Mol Cell Biol 16,
2787-95.
Tang, J., and Rosbash, M. (1996). Characterization of yeast U1 snRNP A protein:
identification of the N- terminal RNA binding domain (RBD) binding site and evidence that the
C- terminal RBD functions in splicing. RNA 2, 1058-70.
Tarn, W. Y., and Steitz, J. A. (1994). SR proteins can compensate for the loss of Ul snRNP
functions in vitro. Genes Dev 8, 2704-17.
Tam, W. Y., Yario, T. A., and Steitz, J. A. (1995). U12 snRNA in vertebrates: evolutionary
conservation of 5' sequences implicated in splicing of pre-mRNAs containing a minor class of
introns. RNA 1, 644-56.
Tazi, J., Kornstadt, U., Rossi, F., Jeanteur, P., Cathala, G., Brunel, C., and Luhrmann, R.
(1993). Thiophosphorylation of Ul-70K protein inhibits pre-mRNA splicing. Nature 363, 2836.
Treisman, R., Orkin, S. H., and Maniatis, T. (1983). Specific transcription and RNA splicing
defects in five cloned beta- thalassaemia genes. Nature 302, 591-6.
Tronchere, H., Wang, J., and Fu, X. D. (1997). A protein related to splicing factor U2AF35
that interacts with U2AF65 and SR proteins in splicing of pre-mRNA. Nature 388, 397-400.
Umen, J. G., and Guthrie, C. (1995). The second catalytic step of pre-mRNA splicing. RNA
1, 869-885.
Utans, U., and Kramer, A. (1990). Splicing factor SF4 is dispensable for the assembly of a
functional splicing complex and participates in the subsequent steps of the splicing reaction.
EMBO J 9, 4119-26.
Valcarcel, J., Gaur, R. K., Singh, R., and Green, M. R. (1996). Interaction of U2AF 6 5 RS
region with pre-mRNA of branch point and promotion base pairing with U2 snRNA. Science
273, 1706-9.
Valcarcel, J., Singh, R., Zamore, P. D., and Green, M. R. (1993). The protein Sex-lethal
antagonizes the splicing factor U2AF to regulate alternative splicing of transformer pre-mRNA.
Nature 362, 171-5.
Wallace, J. C., and Edmonds, M. (1983). Polyadenylated nuclear RNA contains branches.
Proc. Natl. Acad. Sci. USA 80, 950-954.
Wang, J., and Manley, J. L. (1997). Regulation of pre-mRNA splicing in metazoa. Curr Opin
Genet Dev 7, 205-11.
Wentz, H. K., and Potashkin, J. (1996). The small subunit of the splicing factor U2AF is
conserved in fission yeast. Nucleic Acids Res 24, 1849-54.
Wieringa, B., Meyer, F., Reiser, J., and Weissmann, C. (1983). Unusual splice sites revealed
by mutagenic inactivation of an authentic splice site of the rabbit beta-globin gene. Nature 301,
38-43.
Wiest, D. K., O'Day, C. L., and Abelson, J. (1996). In vitro studies of the Prp9.Prp 11.Prp21
complex indicate a pathway for U2 small nuclear ribonucleoprotein activation. J Biol Chem
271, 33268-76.
Will, C. L., and Luhrmann, R. (1997). Protein functions in pre-mRNA splicing. Curr Opin
Cell Biol 9, 320-8.
Will, C. L., Rumpler, S., Klein, G. J., van, V. W., and Luhrmann, R. (1996). In vitro
reconstitution of mammalian Ul snRNPs active in splicing: the U1-C protein enhances the
formation of early (E) spliceosomal complexes. Nucleic Acids Res 24, 4614-23.
Wu, J., and Manley, J. (1989). Mammalian pre-mRNA branch site selection by U2 snRNP
involves base pairing. Genes & Dev. 3, 1553-1561.
Wu, J. Y., and Maniatis, T. (1993). Specific interactions between proteins implicated in splice
site selection and regulated alternative splicing. Cell 75, 1061-70.
Wyatt, J. R., Sontheimer, E. J., and Steitz, J. A. (1992). Site-specific cross-linking of
mammalian U5 snRNP to the 5' splice site before the first step of pre-mRNA splicing. Genes
Dev 6, 2542-53.
Xiao, S. H., and Manley, J. L. (1997). Phosphorylation of the ASF/SF2 RS domain affects
both protein-protein and protein-RNA interactions and is necessary for splicing. Genes Dev 11,
334-44.
Yean, S.-L., and Lin, R.-J. (1991). U4 small nuclear RNA dissociates from a yeast
spliceosome and does not participate in the subsequent splicing reaction. Mol. Cell. Biol. 11,
5571-5577.
Zahler, A. M., and Roth, M. B. (1995). Distinct functions of SR proteins in recruitment of U1
small nuclear ribonucleoprotein to alternative 5' splice sites. Proc Natl Acad Sci U S A 92,
2642-6.
Zamore, P. D., and Green, M. R. (1991). Biochemical characterization of U2 snRNP auxiliary
factor: an essential pre-mRNA splicing factor with a novel intranuclear distribution. EMBO J
10, 207-14.
Zamore, P. D., and Green, M. R. (1989). Identification, purification, and biochemical
characterization of U2 small nuclear ribonucleoprotein auxiliary factor. Proc Natl Acad Sci U S
A 86, 9243-7.
Zhang, H., and Blumenthal, T. (1996). Functional analysis of an intron 3' splice site in
Caenorhabditis elegans. RNA 2, 380-8.
Zhang, M., Zamore, P. D., Carmo, F. M., Lamond, A. I., and Green, M. R. (1992). Cloning
and intracellular localization of the U2 small nuclear ribonucleoprotein auxiliary factor small
subunit. Proc Natl Acad Sci U S A 89, 8769-73.
Zhuang, Y., and Weiner, A. M. (1989). A compensatory base change in human U2 snRNA can
suppress a branch site mutation. Genes Dev. 3, 1545-1552.
Zhuang, Y., and Weiner, A. M. (1986). A compensatory base change in Ul snRNA
suppresses a 5' splice site mutation. Cell 46, 827-35.
Zorio, D. A., Lea, K., and Blumenthal, T. (1997). Cloning of Caenorhabditis U2AF65: an
alternatively spliced RNA containing a novel exon. Mol Cell Biol 17, 946-53.
Zuo, P., and Maniatis, T. (1996). The splicing factor U2AF35 mediates critical protein-protein
interactions in constitutive and enhancer-dependent splicing. Genes Dev 10, 1356-68.
FIGURES
Figure 1.
The splicing reaction is a two step transesterification reaction.
The exons are indicated by boxes and the introns by lines. The 2' hydroxyl that is the first step
substrate is indicated by the :OH. The phosphodiester bonds that are the splice site substrates of
the first and second steps are at the junction between exon and intron. The first and second
steps are indicated.
3'
51
:OH
I
ntron
EXON I
IZ: +
2
I
LI
EXON 2
1
I
\\4 ,
I +
Figure 2.
Splice site sequences do not alone determine splice sites.
The first 7210 nucleotides of the CREB-RS gene pre-mRNA are shown. The sequences of the
terminal dinucleotides of each intron are indicated in bold. The 5' splice site sequence of the
fourth intron is underlined. The identical sequence is found in the middle of the fourth intron;
however, this sequence is not known to be used as a 5' splice site.
oobbbpnonppnbnooBbnpbbnoonoooobnoebnbnc)no.6,eobpbnbb-eobeoo.6,ebebbobbbbobvppvbobbpnopobppbbbpp
bnbnonnbLveo-eb-eni2-ebn5nenonn-enoobb-eono5noooonnopoobnc)ooooo-eb-ebbnobn-e-eBnbneoobbbnnn-elebbieni2b-ee
obpoonpupbbnbbppnpbbb-eobbbnn-eobn-eb-eb.bn-eobeonbnnbbobbnnoeorobnebpbbBbbeoobeoD-V-5 =DVobnbbno
nleiebeonbvebbnnob-eobbleob-eooabbbpeb.bn-e-e.6-ebnleobble.B".Bno-eBnbbbnnnebL;nnf3,eooBbn-ennn-eonoobe-ebbeon
,eeobbbboebnbnbnnoepeiebbebbbnoo5n,6bnnnbnobnnnobbebno-ebnbb-eB-eonobbebf)npoobbebbnn-ennnieb-eo.bblep
-enronb-e-eo-e-eon-ebnb-e.bbn-eonnnonnnn-eoB-enoobbbbn-eo-eboo-eonnniennnnbbb-e-eb-eonooobbnon-e-ebnobbbnbnooe
bnnbnoebbnbbnb-enbbebpoopBbnenbbo-e-ebnb-e-e-eooienb-epnn-e-en-enec)n-ebnB'ennonLooobeeie-e-ennnnble-ebnn-e-e-ene
npep-eeonn-enon-enn-enbno-ebnieonenbnnn-ebbnnnbonnbnnnoonnnn-e-enno-e.bnnnnnnnnnnpbeeooboooobnBoopoob
vbnbobbpopnnpbbbnobnbepeooonoobbonoobnoopoonvbnbonoopbnoononebononBbne5broobvnnbnbooponnnB
bbboieb-eb-enb-ennnnnnn-enonnnnnL*-eno.bbnoobo-eoovooboonbobb-eo-eno-e.5b.bnobnnb-eeooononB-eonooonc)ononne
oobneonnbbbnoooooboonoBLuobno-eonof)bonon-e-eobobbnBpobnbebbnobbLooo'enonobononbeb-eo-eb-ebnnnnnnn
nnnnnnnnnnnnnb-epbbbleebbboo-ebb-ebbnoiebnb-ebnon-e-ebbb-enbnbnononiennbbnb-en-ebnonnf5-en-e-e-ennbbn-eoob-eo
oobnvoopoobpbnbobbvopnnpbbbnobnbpppooonoobbonbnbbbnnbobnoovoonpvobpponnobbnoonorrvonbnbBno
bb-eonnbnnbneoobnnnnbbb-eoeb*eb-enbbnnnnn-enbnnnnnnn-enobboon5n-eoo-eoobnbo-eo.Bb-eo-eno-ebbienob-enlepnc)o
oooobeonoobnoonbnnienob-elennnbbbnoonnnboonoo-e-eobno-eono-eoononlebnbobbnnieobn.6-ebpno.6bvoooeonbnoo
obnnnnbebieoeblebnnnnnnnnnnnnnnnnnnnnnnnnnooonnb-en-e-elennbbn-eon-eo-en-e-en-ennbbb-ebnbnnnnb-eb-pbbnnnn
obnnonnb-en-eoeb-ebn-eo-ebbn-eeonooo-ennno-enbnooibbb-ebnnnnnn-enb.6-enno-en-eoonoononn-eb-eonb-eb-enn-ennbnbp
npobnnpobnnnobbnbbnnbnnpbbppnBnnpbbvpppobvobpnnpnobnorooobonpbpbooBpbnb-eobnnob-ebbobbpbbboo
opvbnbbBbnvvbpbbpobbpbnobbvbbbonovnobrononb-enbnoo-eobb-eobbnbbn-eobb-eoobenn-ee-e-e-e-eo-en-ev-e-e-eno-en
ononvoobobpvbnbropovvnobbbonnvoo-ebe-eon-eb-ebb-eonbbeboeoneB-eobbbobbe-eoobbpbbbnnno-eob-eooon-eenB
noovovonnbbnbvorobb'eoo.bb-eobenn-eoo-en-e-e-ef)nn-e.B.B-een-e-e'e-eoLDopnLob-e-epeL-en-ennbn-eo'en-e-ennpbLDobienonienp
nnLobLDoeeeopBbb-ebbnnnLobbn.Bevebbn*ebbb-ebbb-eob-eo-eob-ebbb-eonon-e.bbnooononnebeonn-eoonb-eobn-ef)nobooo
bpbbbpooppoonovvbnobbnoponononvoorobbnoononiebbnonnoo'enb-e-e-eonnnLbnnbnnLf)nnf)n-eo-ebnnb-eooobn-eo
...squ TSETIII
Pnoopobpvnnpnnnpnpppbppppppnpnnppnppnpnbbvbnon-en-eon-ebnbn-ebnn'ennnbn'e2Lnb-ennn-enbnbnobpnbonop
nnnpnnnbeleooonnbnbeeLDnnnnnenonnf)nnn-ennieoie'enn-e-eo'enonneb-eno'e'e'eno'enpnpooLoepf)ienobb-eeebeo-eneoe
vvnnnnnpopppnpooobpooo5obonvoobubnbobbpounn-eb-ebnobnbe-eeooonooboonoopnoobonbpbnponoovbnoono
eebnnonbbnobbL:)oobbnnf)n-eooeonnnbbbbn-e.B-eb-enb-ennnnn-enbnnnnnn-elenobvoooboeoc),eoo-eooo-eobbeo-ennebb
bnobgnBvpooonoobvononbnoononnebnb-e-eonn.Bb-eooonooLoonoo'eleobnoeo'nob*eononbbnbn.bbnb-eobnbepbnobb
bonobonbnovopononBvbpopbvbnnbnnbnnbnnnnnpovvvnvnnpnnnnpoppooopoonoooonnpbnjbnbnon-en5n-enpobb
bennnbieob"ebnbnno'e'eepennnnno-nno-eLonn-eoobnLonbnonn-enb-enoonnbnnonnnnnonononnnnn-enn-en-ebbn-ebpb
b-ebnobnb-eobbbe-eob-eoeoo'eebbb-eonnn-eo-e-e-ebb-eb-ebb-e-ebbvobb-eebbbiebb-eooLD-eLDLDbLobbn-e-e-eibbBb-eb.bbbnbb-ee
PepbvbbbpopbbvobvobBeobnbeebbbb-ebbn-ebvnbnbnbnvnbnbnbnBnjBnbnennbnbneebebbbo-eno-enonobbnnoonp
Pooooobnoobrovppenpenbbponoonn-ebn7ebnonooeoooonennbn-eeonebeoonboor-e-ebnnneonoon-eo-e-eooo-ebn-ebe
bBbnoononbnbnopooooopob5nnoonbpBeopbvvbnbnpoonoBnbbpbbbBpnbbbbnnonobbvooopononoppoovopoooo
Bnooooo5nonnnonnooobnnnnnnbnnooononpo-e-eonbno-ennonobononnbnnnnobp-e-enonnoee-enooob-e-enbnnobbne
bnnppnvpponbnppbpoopbponnpoopnbponbbnpbpppbpnnbbvopvbnppbpppbpnbpppnpppbvooononnsponvonnop
bebneonbeeBnnnenvoobbiebnonoeoonbnnf)-e-eooobnnonnbnoie-ebbnonn-e-ebebnobnoLbnn-ennf:)-eb-ebpiebppbBbnc)b
bbbnbnpnnbLobpe-en-enLLonnnnnonbbbnnonebnooboL*oo-eoob-eien-enbbpo-enn-ebbnnob",eepooon-eobbnnoo-eooono
onebnbe-eonobbbnoonoeepnnoubbnb-ebeooobnnbn'enoonnonbbbbnL2-e-e'e-enbnnnbnniennnnnnnneienof)bnonbneoo
Poobnpopopbpopoovbbbnnbenbebnoonoob-eonoobnoonoone.6nbbLonob-ebnoonoo-ebnnoobpnbno-eonobeo-eonbb
beoebnbenf)nbeo'eno-eveoooeonbnonooonoo.6bbvnebvbnnnnnnnnobnnnnnnbnnnnnnbnnnbnnnbnnnnnnnEbLovibn
Brobpnonpnnbbnnnoooonropoonnnnponnonpneennoonnnboo5nnnoopnopoononnoobbbbbbnbnb.bebnbnbnbbieb
nponb-ebn-eno.bnbnbnnniebn-elepnnn-ebn-e-elef)-e-e-eo-ebTibbbnbooo-eonnooonneonnobnonoebbbbo-ebnj5nonono-ennon
ooon-en-eoob-eb-eDVDLj E)DVbobvoon-eoob-eb-eo-eoonononboboneonbeboonob"eonooonoonoonnonobnooooonnon"eo
obpbnonbppbnbBponnonebvoonoonnnooonoernonnnpenvbnoonnobnonobppnnonnonovbbnnnnnopnbbbnnoonb
npopboopoooonppbbvoLnbbobnobnnooononnonnnbnnnnnenoooon-e-eonnoonononoonpnoobnb-e-envo-enonnobo-e
onoonpnpnoonnobpnbopnopppbpooonnnbnopooonnonvonooovvvbvoonnnpbvonooonnbonbnnnonbpnbbvpnbbp
ooonnonpboobnoonc),epBbbnpooooob-ebnonoooob-ec)nbnpbbn-ebbb.bnbo-ebbnooonobeobb:)PbnnnbLnnnpnnooono
onpnbnnonooobnoonoropononnnoonvononnnognooooponbebbnonronovenvoobbroponnpooovbbnooevbpooob
vbnbppvbbnbbbnbbnnooopvnononnnoobvoooonbnbnnertBbooonbnpbbuobpbbooobnnboonnonobrobopbpobpbb
Pboobbnbppbnpbpnoobbnonnpnbnnoorobroebrobnnonnn-eob-eonobo-eboooo;oorbopuboopoobbbbnobbvnbnonp
Bobnbnbnnobbbbbobbbbnpbbbpbnppoobpbbobpboobbnonbbbnbbobbbobbbnbvponnnnoponpnbn"bnooobobpp
bnobbbbpbbpvbbbbonbbbboonnbnnbpbonbovvbbnobnooonobnonopebvvbb5ponbpeppopnpbbbbbppnnpobnbnn
bonobbbbbpobpbbobvbLobbppbnon-eooboon-e-ebbo'en-e-e-eobf)bnonbeLbb-ebppL*noobbbbbobpebpvpBbbo5opobbpb
nooonbnopprobbnbnobbbpbpbbbbpBbbboo-eobb-ebnb-eB-eobnon6biBiBno-ebb-ebbooob-ennobnoo-eeo-ebooeonnonnn
Figure 3.
Cartoon of the spliceosome assembly pathway.
Assembly of the spliceosome on the pre-mRNA is shown. The exons are shown as boxes, the
intron as a line. The 5' GU dinucleotide; the 3' AG dinucleotide; the branch adenosine, A; and
the pyrimidine tract, PYR, are indicated on the intron. E complex contains Ul snRNP, U2AF
and the SR proteins and is the first spliceosomal complex to form on the pre-mRNA. Ul
snRNP is shown base-paired to the 5' splice site sequence. The SR proteins are shown
interacting via RS domains interactions with U1 snRNP and U2AF. The SR proteins are not
shown bound to the pre-mRNA as binding can occur to either the intron or exon sequences.
Following E complex is A complex, the first ATP-dependent complex that forms on the premRNA. U1 snRNP and U2AF are believed to be dissociated or destabilized from the premRNA at this step and are shown in gray. The tri-snRNP is bound to the assembling
spliceosome and then base-pairing with the 5' splice site sequence in complexes B and C.
A
PYR
f
E
+
U-
ul
U2 AF
A
+
i
U1
ri
1
*s~
i
U2 AF
U2
~~~~u
5
B,C
CHAPTER 2:
IDENTIFICATION, PURIFICATION AND
CHARACTERIZATION OF A NEW PYRIMIDINE-TRACT
BINDING SPLICING ACTIVITY.
Patrick Schonleber McCaw, Andrew MacMillan, Charles Query, Barbara Panning,
and Phillip A. Sharp
ABSTRACT
We have identified a new pyrimidine-tract binding splicing factor (PUF) that is required,
together with U2AF, for the efficient reconstitution of in vitro splicing activity to a poly[U]
depleted nuclear extract. The activity has been purified to near homogeneity and found to consist
of at least two proteins; PUF60 and the previously described splicing factor p54 (Zhang and
Wu, 1996). p54 and PUF60 form a complex in vitro and PUF 60 interacts weakly, but
detectably, with the branch sequence binding protein SF1/BBP. While PUF is required for the
formation of the U2 snRNP-branch sequence complex, A. (Query et al., 1997), it is not
absolutely required for the formation of the U2 snRNP complex formed on a 3' half substrate,
A3'. This A3' complex forms with diminished efficiency in the absence of the PUF activity.
PUF binds to pyrimidine-tract RNAs, but does not appreciably interact with either the branch
sequence or the AG dinucleotide of the 3' splice site. PUF60, unlike p54, appears to localize
to speckle-like structures in the nucleus that are distinct from the nuclear speckle domain to
which many splicing factors localize.
INTRODUCTION
Four sequence elements found in the mammalian intron are known to be essential for
recognition of the intron. These are the 5' splice site sequence, the branch sequence, the
pyrimidine tract and the 3' splice site AG dinucleotide. The 5' splice site sequence is bound by
Ul snRNP early in spliceosome assembly and ATP is not required for this interaction (Mount et
al., 1983; Siliciano and Guthrie, 1988; Zhuang and Weiner, 1986). The branch sequence is
recognized at least twice during spliceosome assembly, first by the branch sequence binding
protein SF1/BBP and, subsequently, by U2 snRNP (Berglund et al., 1997). The stable
binding of U2 snRNP to the branch sequence requires recognition of the pyrimidine tract by the
splicing factor U2 snRNP Auxiliary Factor, U2AF, and ATP (Roscigno et al., 1993; Ruskin et
al., 1988; Zamore and Green, 1989). Ul snRNP and U2AF appear to be the primary
determinants of splice site sequence binding early in spliceosome assembly (Bennett et al.,
1992; Reed, 1996). The factors required for recognition of the 3' splice site AG dinucleotide
are not known; however, spliceosome assembly and the first step of splicing can occur in the
absence of an AG dinucleotide (Anderson and Moore, 1997; Frendeway and Keller, 1985;
Ruskin and Green, 1985) indicating that recognition of the AG dinucleotide is not required for
spliceosome assembly.
The pyrimidine-tract binding splicing factor U2AF was identified as an activity required
for the association of U2 snRNP with the pre-mRNA. U2AF was purified and found to consist
of two proteins of 65 and 35 kD (Zamore and Green, 1989). U2AF can be depleted from
extracts by taking advantage of the very stable interaction of U2AF has with poly[U] RNA.
U2AF binds poly[U] RNA in the presence of 1 M KCl and requires either 2 M Guanidine HCl
(Zamore and Green, 1989) or 3 M KC1 (this work) to be dissociated from a poly[U] Sepharose
column. The large subunit of U2AF, U2AF65, has been shown to be essential for the splicing
of pre-mRNA in nuclear extract depleted by this method (Zamore and Green, 1991), while the
small subunit appears to act as an enhancer of this activity (Zuo and Maniatis, 1996). U2AF65
is the pyrimidine-tract binding component of U2AF. U2AF is highly conserved in evolution:
the invertebrates D. melanogaster(Kanaar et al., 1993) and C. elegans (Zorio et al., 1997) and
the fission yeast S. pombe (Potashkin et al., 1993) have clear orthologs of the large subunit of
U2AF, while the yeast S. cerevisiae gene MUD2 is a probable orthologue of U2AF65. The
sequence, but not the function, of MUD2 has diverged sufficiently that this can not be
determined with certainty (Abovich et al., 1994) and the plant A. thalianahas a similar protein
identified in the sequence project (Rounsley et al., 1997). While the small subunit of U2AF
may not be conserved in C. elegans, orthologs of this protein can be found in D. melanogaster
(Rudner et al., 1996) and S. pombe (Wentz and Potashkin, 1996) as well as vertebrates.
U2AF, bound to the pyrimidine tract, and Ul snRNP, bound to the 5' splice site,
together with several other proteins, including SR proteins bound to exon sequences, form the
E complex prior to the ATP-dependent spliceosome assembly step (Michaud and Reed, 1991;
Staknis and Reed, 1994). Following the formation of E complex, U2 snRNP assembles on the
branch sequence in an ATP-dependent step, the resulting complex is known as spliceosomal
complex A. An analogue of A complex can form on 3' half substrate pre-mRNA consisting of
the branch sequence, pyrimidine tract, 3' splice site and 3' exon, known as A3' (Barabino et
al., 1990; Zamore and Green, 1991). U1 snRNP and U2AF are dissociated or destabilized
from the pre-mRNA upon formation of A complex. The pyrimidine tract may remain associated
with other pyrimidine-tract splicing factors such as PTB and PSF. PSF is known to be required
for the second chemical step of splicing (Patton et al., 1993). Whether the pyrimidine tract is
recognized between these two steps is not known. Following U2 snRNP association with the
pre-mRNA, the tri-snRNP, U4/5/6 snRNP, binds the pre-mRNA and the complete spliceosome
is formed and the chemical steps of splicing take place.
In order to better understand the substrate binding requirements for U2 snRNPs binding
to the pre-mRNA, a complex on a minimal U2 snRNP binding RNA has been described (Query
et al., 1997). This U2 snRNP complex, An n, shows additional sequence specificity
requirements and is, therefore, an important tool in understanding what the requirements are for
stable U2 snRNP binding. A~ n forms many of the same protein-RNA contacts that complex A
forms as judged by photochemical crosslinking.
Splicing factors have been shown to localize to discrete structures of the nucleus known
as nuclear speckles. Many splicing factors are known to localize to these domains including the
SR protein p54 (Chaudhary et al., 1991). p54 is conserved between vertebrates and the
invertebrates D. melanogaster(Kennedy and Berget, 1998) and C. elegans (McCombie et al.,
1993). The function of p54 is not known, although like the general class of SR proteins it
activates the cytoplasmic extract S 100 for in vitro splicing (Zhang and Wu, 1996). Unlike the
general class of SR proteins, p54 has an SR domain that has sequence characteristics that are
reminiscent ofUl1 70 kD and U2AF.
We have identified a new pyrimidine-tract binding splicing factor (PUF) that is required,
together with U2AF, for the efficient reconstitution of in vitro splicing to a poly[U] depleted
nuclear extract. The activity has been purified to near homogeneity and found to consist of at
least two proteins; PUF60 and the previously described splicing factor p54. This chapter
compiles the current state of our understanding of the PUF splicing activity and the PUF60
protein. Several lines of evidence suggest that the PUF activity is required after the U2AFdependent step in spliceosome assembly. However, we have been unable to conclusively
demonstrate that p54, PUF60 or both proteins are responsible for the PUF splicing activity.
RESULTS
Depletion of NE for pyrimidine tract splicing
Splicing extracts can be depleted of the pyrimidine-tract binding factor, U2 snRNP
Auxiliary Factor (U2AF) by passing the extract over a poly[U] Sepharose or oligo dT column at
high salt concentrations. Extracts depleted in this way (NEAU2AF) are unable to catalyze the
splicing reaction and are blocked at an early step in spliceosome assembly. Spliceosome
assembly and in vitro splicing activity can be restored to these extracts through the addition of
purified U2AF or recombinant U2AF65 (Zamore and Green, 1991). Small modifications of
this depletion protocol (discussed in the Methods section) lead to the co-depletion of a second
pyrimidine-tract associated splicing activity. Nuclear extract depleted using this method
(NEAU) is inactive for splicing in vitro (figure lA, lane 2). Addition of either recombinant
U2AF65 or the 2.0 M KCl eluate of the poly[U] column leads to a partial restoration of splicing
activity (figure 1A, lanes 3 and 4). Efficient restoration of in vitro splicing activity requires the
addition of both fractions (figure 1A, lane 5). The activity present in the 2.0 M KCl eluate is
referred to as PUF (Poly[U] Eactor).
To determine if the PUF activity was required for splicing of multiple introns or was
specific to the PIP85a intron, two other pre-mRNAs were tested for their requirements for the
PUF activity. We tested the in vitro splicing activity in the reconstituted system of the AD 10
pre-mRNA and a PIPI3G chimeric intron. Both introns required both PUF activity and U2AF65
for efficient reconstitution of in vitro splicing activity (figure IB compare lanes 4 with 5, and 18
with 19).
Role for U2AF35
Recombinant U2AF65 is sufficient to reconstitute splicing activity to extracts depleted of
both U2AF65 and U2AF35 (Zamore and Green, 1991). To determine if the PUF activity
substituted for U2AF35 in NEAU reconstituted splicing, U2AF purified from nuclear extract,
containing both U2AF35 and U2AF65, was compared to recombinant U2AF65 in the
reconstituted splicing reaction. For each pre-mRNA tested, PUF activity stimulated splicing
when compared to purified U2AF or recombinant U2AF65. In the case of PIP85a, purified
U2AF had additional stimulatory activity when compared to recombinant U2AF65, presumably
due to the presence of U2AF35 in this fraction. For the PIPBG and the AD10 pre-mRNAs
purified U2AF and recombinant U2AF65 had comparable activities either in the presence of the
PUF activity (figure IB compare lane 5 to 7 and 19 to 21) or in its absence (compare lane 4 with
6 and 18 to 20). In contrast to the results obtained with PIPBG and AD10, purified U2AF had
more activity than recombinant U2AF65 on the PIP85a substrate in both the presence and the
absence of the PUF activity (compare lanes 12 to 14 and 11 to 13). This result argues that PUF
does not functionally substitute for U2AF35 in these reactions.
Purification of the PUF activity
The PUF activity was purified to near homogeneity using the reconstituted in vitro
splicing reaction as an assay. Purification of the activity was by the scheme shown in figure
2A. The final purification step on S Sepharose is shown in figure 2B. The top panel shows the
silver-stained gel of eluted fractions, while the bottom panel shows the activity of each eluted
fraction. Quantitation of the PUF activity recovered during fractionation has been difficult, and
we believe that this was due to three factors. First, the in vitro reconstituted splicing reaction
had a limited linear range. Second, the presence of multiple poly[U] binding activities that copurified with the activity may have interfered with the assay by competing for pyrimidine tract
binding. Finally, the presence of contaminating poly[U] RNA from the poly[U] Sepharose
affinity chromatography may have competed for PUF and U2AF activity.
A Coomassie stained gel showing fraction 8 of this purification is shown in figure 2C.
Bands present in this fraction are of apparent molecular weight 130 kD, 80 kD, 60 kD and 48
kD. All four bands were excised from the gel, digested with trypsin and peptides were
sequenced.
Identification of PUF60 and p54 as the predominant proteins in the active
fraction
The four proteins identified in figure 2C were digested with trypsin and the resulting
peptides were sequenced. Three peptides were sequenced from the 60 kD band and were
identified as the previously described splicing factor p54 (Chaudhary et al., 1991; Zhang and
Wu, 1996). Fourteen peptides were sequenced from the 130 kD band and found to be encoded
by a previously undescribed gene. No tryptic peptides were identified from the 80 kD or 48 kD
bands. cDNAs that encoded all fourteen peptides found in the 130 kD protein were identified in
the EST database, and the cloning and characterization of this factor (PUF60) and its domain
structure will be the subject of the next chapter. PUF60 forms SDS-resistant dimers with an
apparent molecular weight of 130 kD explaining the aberrant mobility of this protein on SDSpolyacrylamide gels (discussed in chapter 3).
PUF60 is a ubiquitously expressed and abundant mRNA of 2.0 kb (figure 3) as would
be expected of a splicing factor. The differences in expression between tissues seen on this blot
mirror those seen for the nuclear-matrix associated splicing factor SRml60 (BlC8, (Blencowe
et al., 1995).
Rabbit polyclonal antibodies raised to PUF60, demonstrate that NEAU is depleted of
PUF60 as shown in figure 4A. Equal volumes of different batches of NEAU (lanes 2, 4, and
5) and control extract, NE (lanes 3 and 7) were compared by immunoblotting. NEAU was
estimated to be depleted at least 90% of PUF60 by this method. Antibodies raised against the
splicing factor p54 demonstrate that p54 is also depleted from NEAU to a similar extent (figure
4B, lanes 1 and 2). It should be noted that p54 has an aberrantly slow mobility in SDS PAGE
gels (figure 4B compare lane 2 with lane 4). When depleted extracts boiled in SDS were added
to the purified or partially purified PUF activity (also boiled in SDS sample buffer), p54 comigrated with p54 found endogenously in the extract (figure 4B, lanes 2, 3 and 4). As both the
extract and the p54 containing sample had been boiled in SDS prior to mixing we do not believe
that the change in mobility represents a covalent modification of p54, but rather represents a gel
artifact. (The band marked x is a cross-reacting band that is not detected with other p54
antisera.) p54 translated in vitro, in the presence of the proteins found reticulocyte lysate, runs
as a discrete band of 65 kD (figure 5, lane 1). However, when immunoprecipitated from the
translation reaction p54 migrates aberrantly as a diffuse band of approximately 65 kD, as well as
bands at the interface between the stacking and resolving gel and at the origin (figure 5, lane 5).
Presumably, the high protein concentration found in nuclear extract and in reticulocyte lysate
acts as a carrier, allowing resolution of the protein in SDS-polyacrylamide gels. Similar
aberrantly slow migration of proteins is observed for the highly charged protein PACT and for
the SR proteins. PACT is known to behave aberrantly on SDS-polyacrylamide gels without
prior covalent modification of the lysine residues of this protein (Simons et al., 1997). The
arginine-rich SR proteins also migrate aberrantly slowly on SDS-polyacrylamide gels (Zahler et
al., 1993), suggesting that aberrantly slow migration in SDS-polyacrylamide gels may be a
common property of proteins with arginine and lysine-rich domains.
To determine if PUF60 and p54 can form a complex in vitro, co-translation of PUF60
and p54 was performed in vitro. Antibody to p54 (Chaudhary et al., 1991) immunoprecipitates
p54 (figure 5, lane 6). Immunoprecipitation of the co-translation reaction of p54 and PUF60
led to the immunoprecipitation of PUF60, whereas control immunoprecipitations with no p54
present does not lead to the precipitation of PUF60. The co-immunoprecipitation of PUF60
with p54 strongly suggests that p54 and PUF60 form a complex in vitro.
The PUF activity purifies as 400 kD complex
Application of the purified PUF fraction to a gel filtration column allowed further
purification of the splicing activity and resolution of the size of the active PUF complex. The
peak of PUF activity eluted from the column with an estimated size of 400 kD (figure 2D,
bottom panel lanes 5 and 6). The proteins present in the adjacent fractions included the 130 kD
band and the 60 kD band (figure 2D, top panel lanes 6, 7 and 8). Presumably, these represent
PUF60 monomer and dimer and p54. Notably, the 48 kD protein does not co-purify with the
activity; the 48 kD protein was found in fractions 20-22 and presumably is a distinct protein
complex from the PUF activity (top panel, lanes 5 and 6). Other proteins of about 125 kD and
48 kD eluted earlier and later than the PUF activity.
PUF is required for efficient A3' complex assembly
U2AF depleted extract, NEAU2AF, is blocked prior to the first step in splicing and does
not form spliceosomal complexes (Zamore and Green, 1991). NEAU was blocked prior to the
first step of splicing as is NEAU reconstituted with U2AF alone (figure lA and lB) suggesting
that the PUF activity, like U2AF, acts early in spliceosome complex assembly. To determine if
this was the case, spliceosomal complex formation on 3' half pre-mRNAs consisting of a
branch sequence, pyrimidine tract, 3' splice site and 3' exon was tested. A time course of A3'
complex assembly is shown in figure 6. While PUF activity is not absolutely required for
formation of A3' complex (figure 6 lanes 5 and 6 have about 30% of the A3' found in lanes 11
and 12), it is required for efficient formation of this complex. In the experiments shown in
figure 6, the formation of A3' was particularly robust in the absence of PUF. More generally,
in the absence of PUF the A3' complex formed at about 10% the levels seen in the presence of
both factors, rather than the 30% shown here (not shown). In contrast to these results,
formation of the ATP-independent U2 snRNP complex, An, is more dependent on the
presence of PUF activity (Query et al., 1997).
RNA crosslinking of the PUF fraction
To determine which proteins in the active fraction contact RNA a crosslinking assay was
performed. Purified PUF fraction was incubated with full length PIP85a pre-mRNA under
splicing conditions and subsequently irradiated with ultraviolet light to crosslink the proteins to
the RNA. Crosslinked bands are apparent at 130 kD, 60 kD and 48 kD (figure 7, lane 1),
indicating that PUF60 and the 48 kD band contact RNA. It is not clear from this experiment
whether p54 also contacts the RNA as p54 runs as a diffuse band of approximately 60 kD at the
protein concentrations used here and so may not be resolvable under these conditions. The
PUF60 protein should be monomeric at the concentrations used in this experiment and so
should migrate primarily as a 60 kD band; the 60 kD crosslinked product is presumed to be
PUF60. To determine the specificity of PUF protein binding, the binding reaction was carried
out in the presence of RNA homo- and heteropolymers. As expected, poly[U] RNA efficiently
competed for the RNA binding activity of the PUF fraction, more surprisingly, poly[C] did not
compete, but poly[G] did compete at high concentrations. Because p48 does not co-purify with
the splicing activity, but does crosslink to RNA we suggest that p48 fortuitously co-purified
with the PUF activity through its ability to bind poly[U] RNA.
PUF does not detectably interact with the branch sequence or AG dinucleotide
Both the UV crosslinking assay and PUF's affinity for poly[U] Sepharose strongly
suggested that PUF activity bound to the pyrimidine tract. To determine whether the
predominant RNA binding activity in the purified extract interacted with other 3' splice site
sequences, mobility shift assays were performed. Purified PUF fractions form a discrete
complex on pyrimidine tract containing RNAs (figure 8). To determine if the PUF activity
interacted with the AG dinucleotide found at the 3' splice site, the affinity of PUF for a
pyrimidine-tract RNA or a pyrimidine-tract RNA-AG dinucleotide RNA was compared (figure
8, compare lanes 9 to 14 with 15 to 20). No difference in affinity was detected. Similarly, to
determine if the presence of a branch sequence 5' to the pyrimidine tract increases RNA-binding
affinity of the purified PUF activity, an RNA with the branch sequence 5' to the pyrimidine tract
was compared to a mutant RNA in which the branch sequence was placed 3' to the pyrimidine
tract. No difference in RNA binding affinity was observed between these two RNAs (figure 8,
compare lanes 1 to 4 with lanes 5 to 8). These results suggest that PUF interacts solely with
the pyrimidine tract and not with other 3' splice site sequences.
PUF and U2AF do not bind to the same pyrimidine-tract RNA
To determine if the pyrimidine tract binding factors U2AF and PUF cooperate in binding
the pyrimidine tract, binding reactions were performed in the presence of PUF and either
U2AF65 or U2AF. Comparison was made between three identical binding titrations of PUF
(from 2.15 nM to 1 tM, figure 9, lanes 12-20) in the presence U2AF65 (figure 9, lanes 2-10,
U2AF65 alone is in lane 1) or U2AF (figure 9, lanes 22-30, U2AF alone in lane 21, RNA alone
is shown in lanes 11 and 31). Both the change in apparent binding affinity and the appearance
of super-shifted bands was assessed. Neither a super-shifted band nor a change in apparent
PUF affinity for the pyrimidine-tract RNA was observed, strongly suggesting that PUF and
U2AF do not interact on pyrimidine-tract RNA incubated under splicing conditions. It was also
noted that PUF bound to the RNA cooperatively, with a Hill coefficient of close to 2 (for a more
complete description of the RNA binding activity of PUF60 see the next chapter) and that this
RNA-binding activity was not effected by the presence of either U2AF65 or U2AF.
Interaction with the branch sequence binding protein SF1/BBP
To determine if PUF60 interacts with the branch sequence binding protein SF1/BBP, a
GstSF l/BBP fusion protein was bound to glutathione-agarose beads and then incubated with
nuclear extract. The associated proteins were detected by immunoblotting. A distinct, but
weak, PUF60 band was observed in the GstSFl/BBP pull down from the nuclear extract
(figure 10, top panel, lane 2). Although the signal observed with this pull-down was very faint
when compared with the signal observed with a U2AF65 antibody on a duplicate immunoblot
(figure 10, bottom panel, lane 2), we believe that the inefficient binding observed represents
specific binding, as the result has been reproduced multiple times and as no PUF60 was
observed in binding reactions with Gst (lane 8), GstPTB (lane 4) or GstU2AF65 (lane 6).
Further, as a control for antibody specificity each Gst fusion protein was incubated with NEAU
(odd numbered lanes); no PUF60 binding was observed in these lanes. The Gst fusion proteins
(marked with arrowheads), present in vast excess in each of these binding reactions, crossreacts
to the PUF60 primary antibody. This weak, but specific, interaction between GstSF 1/BBP and
PUF60 is also observed with in vitro translated PUF60 (data not shown).
Subcellular localization of PUF60
To investigate the cellular localization of the PUF factors immunoflourescence was
performed both on a spontaneously transformed mouse cell line and the human cell line 293
using PUF60 affinity-purified antiserum. p54 is known to co-localize with the splicing factors
in nuclear speckle bodies (Chaudhary et al., 1991). Because p54 and PUF60 associate in vitro
and co-purify, we expected that PUF60 would localize with p54 to nuclear speckles. To
determine if PUF60 co-localized with these bodies we co-stained with the nuclear-speckle
marker antibody BlC8 (Blencowe et al., 1994; Wan et al., 1994). Typical results from such an
experiment are shown in figure 11. Surprisingly, PUF60 does not co-localize with B 1C8 in
this assay (compare panels B and C, merged in panel E). Instead, PUF60 localizes to a small
number of discrete structures, only some of which overlap with or are adjacent to the speckle
bodies. These PUF 60 staining structures are substantially less abundant and of lower intensity
than the nuclear speckles and their identity is currently unknown. The antibodies used in these
experiments are not known to recognize PUF60 in the native state (as determined by
immunoprecipitation experiments, data not shown); it is possible then, that the PUF60 staining
structures represent denatured PUF60 and are not indicative of the subcellular localization of
native PUF60 protein.
DISCUsSION
We have identified a new pyrimidine-tract binding splicing factor (PUF) that is required,
together with U2AF, for the efficient reconstitution of in vitro splicing of multiple pre-mRNAs
to a poly[U] depleted nuclear extract. The activity has been purified to near homogeneity and is
found to consist of at least two proteins PUF60, and the previously described splicing factor
p54 (Zhang and Wu, 1996).
Splicing in vitro requires two pyrimidine-tract RNA binding activities
PUF was purified to near homogeneity and found to consist of at least two proteins, the
previously described splicing factor p54 (Chaudhary et al., 1991; Zhang and Wu, 1996) and a
novel protein with striking similarity to U2AF65, PUF60 (see next chapter for a description of
this protein). The PUF activity eluted from a gel filtration column with an apparent size
equivalent to a 400 kD protein complex. The size of this complex suggests that the PUF60/p54
complex is a higher order oligomer in the purified fraction. It is also possible that this
unexpectedly large complex is the result of oligomerization of the PUF activity through proteinpoly[U] RNA interactions. RNA from the poly[U] Sepharose column co-elutes with the PUF
activity. The gel filtration experiment suggests that PUF activity does not depend on the
presence of the 48 kD protein found in the S Sepharose purified fraction. However as the 80
kD protein, also found in the S Sepharose purified fraction, was not detected in any of the gel
filtration fractions its importance to the activity could not be evaluated, but it is unlikely to be an
important component of the activity.
The PUF activity together with U2AF is required for the efficient splicing of several
introns. For the PIPBG and AD10 pre-mRNAs the presence of U2AF35 was not required for
maximal splicing activity. For the PIP85a substrate, maximal activity was obtained only in the
presence of U2AF35. This is reminiscent of the U2AF35 splicing-enhancer activity detected in
U2AF35 depleted nuclear extracts and is presumably due to interactions between U2AF35 and
SR proteins bound to an enhancer element found in the pre-mRNA (Zuo and Maniatis, 1996).
PUF activity was required for the efficient assembly of A3' complex, the U2AF and ATPdependent U2 snRNP-containing complex that forms on branch-sequence, pyrimidine-tract, 3'
exon RNAs. In contrast, PUF activity was more stringently required for the formation of the
An n complex (Query et al., 1997). This complex, a model U2 snRNP-branch sequence
complex, is more sensitive to the sequence and functional groups of the branch sequence than A
or A3', revealing specificities to the interaction of U2 snRNPs with the branch sequence that are
masked when U2 snRNP binds to longer RNAs (Query et al., 1997). Factor requirements for
U2 snRNP's interaction with the branch sequence are also revealed in this complex that are not
as apparent for the A or A3' complexes: the PUF activity is more stringently required for A,,
than for A3' or A complex formation. This may be due to the absence of binding sites for the
SF3a factors on the pre-mRNA 5' to the branch sequence (Chiara et al., 1994; Gozani et al.,
1996).
What role PUF plays in mediating spliceosome complex assembly is not known, but it
is interesting to speculate that PUF may be required after the U2AF-dependent step. Three
arguments support this view. First, it is observed that U2AF or U2AF65 and PUF can not bind
to the same RNA molecules and so it seems unlikely that they act at the same point in
spliceosome assembly. Second, in the absence of PUF activity, A3' spliceosomal complexes
can form, but they do not form in the absence of U2AF, suggesting that PUF may play a role in
the transition from A or A3' to later spliceosomal complexes. Third, U2AF is not absolutely
required for the splicing reaction, but PUF activity is required under these conditions
(MacMillan et al., 1997). Furthermore, it is known that U2AF becomes destabilized from the
pre-mRNA, possibly with Ul snRNP, at the transition between the commitment complex and
formation of the spliceosome (Michaud and Reed, 1993). PUF could replace U2AF at the
pyrimidine tract during this transition and might act as a U2AF-like factor for assembly of the
tri-snRNP complex or other splicing factors on the pre-mRNA. It also remains a possibility that
PUF acts as an intron-specific splicing factor. At least for the limited number of pre-mRNAs
tested this does not appear to be the case.
The PUF proteins: p54 and PUF60
The purified PUF activity consists of at least four polypeptides, two of which, PUF60
and p54 have been identified in this work. We do not believe that the other two polypeptides,
the 48 kD and 80 kD bands, are likely to be important for the PUF activity as they did not copurify with PUF activity upon gel filtration chromatography. p54 has been described
previously as a protein that co-localizes with splicing factors, has an SR domain that is similar
in sequence to the Ul 70k SR domain, and reconstitutes splicing activity to an S100 extract
(Chaudhary et al., 1991; Zhang and Wu, 1996).
PUF60 interacts weakly with the splicing factor SFl/BBP, the branch sequence binding
protein. This interaction is substantially weaker than the interaction between U2AF65 and
SF /BBP. This may be indicative of functional differences between PUF60 and U2AF65,
either in regulating splice site use or in mediating spliceosome complex assembly.
PUF60 has been identified in unpublished work in two other experiments. The first
experiment to identify PUF60 was a yeast two-hybrid experiment; PUF60 was identified as a
protein that interacts with the mouse homologue of the Drosophilaprotein seven-in-absentia (D.
Bowtell, personal communication, PUF60 appears in the Genbank database as SIAH-BP1
based on this work). Seven-in-absentia, sina,is known to target the product of the tramtrack
(ttk) gene for degradation. Ttk is a transcription factor that prevents cell-fate determination and
its degradation is essential for cell fate determination of the R7 cell (Dickson, 1998; Li et al.,
1997). Strikingly, ttk protein exists in two alternatively spliced forms, at least one of which has
been determined to be degraded through the sina pathway (ttk88B; Li et al., 1997). It is
interesting to speculate that the D. melanogaster homologue of PUF60, DPUF68 (chapter 3),
may also interact with sina protein and so may regulate the ttk alternative splicing pathway.
The second experiment to identify PUF60 was an immunoprecipitation experiment; PUF60 was
identified as a protein that co-immunoprecipitated with the human auto-immune antigen Ro
(Pascal Bouffard, personal communication). The function of the Ro auto-antigen is not known,
but Ro is known to localize to both nuclear and cytoplasmic compartments (Peek et al., 1993).
Nuclear Ro may be associated with the nuclear speckle domain (Wahren et al., 1996). The
functional significance of the association between Ro and PUF60 remains to be determined.
Sub-cellular localization of PUF60
Antibodies raised against PUF60 have proven unable to recognize the protein in native
form in immunoprecipitation experiments (data not shown). Although this has greatly hindered
the biochemical characterization of this protein, the antibodies do recognize faint speckle-like
structures in the nucleus. These structures are reminiscent of the PML-staining bodies found in
the nucleus (Lin et al., 1998; Sternsdorf et al., 1997). PML, a protein commonly found to be a
fusion protein with the retinoic acid receptor in promyelocytic leukemia has been shown to be
covalently modified by ubiquitin-like molecules (Kamitani et al., 1998; Muller et al., 1998). It
will be of great interest to determine if the PUF60-staining structures are related to the PML
structures. It is intriguing that two avenues of study, the localization to PML-like bodies and
the association with the degradation-targeting protein sina, suggest that PUF60 may be targeted
for degradation or modification by ubiquitin or ubiquitin-like molecules. As PUF60 antibodies
do not recognize the native protein in immunoprecipitation assays, it is possible that the majority
of the PUF60 immunostaining observed represents denatured protein or peptide fragments of
PUF60 and that the bulk of the PUF60 remains undetected by the methods used. We are
currently pursuing methods to detect the localization of the native protein.
METHODS AND MATERIALS
Preparation of poly[U] depleted nuclear extract:
HeLa cell nuclear extract was prepared using standard protocols (Dignam et al., 1983).
Briefly, 15-30 liters HeLa cells grown in suspension culture were harvested by centrifugation,
washed in ice cold PBS and resuspended in five packed cell volumes of ice cold Buffer A (10
mM Hepes [from 1.0 M stock pH 7.9, with KOH at room temperature], 1.5 mM MgCl 2, 10
mM KC1, 5 mM dithiothreitol), proteinase inhibitors were included in all buffers and included
50 ptg/ml of PMSF and Leupeptin, Pepstatin A and Aprotinin (Boehringer Mannheim, used at
the concentration recommended by the manufacturer), and incubated for 10 minutes on ice. All
subsequent steps were carried out on ice or at 40 C. Swollen cells were centrifuged at 1200g,
resuspended to two swollen cell pellet volumes of Buffer A and dounced 10 strokes with a
Hamilton B dounce to lyse the cells. Lysed cells were centrifuged for 10 minutes at 1200g and
the supernatant was decanted off the loosely packed nuclei and discarded. Nuclei were packed
at 25,000 g for 20 minutes and the supernatant was discarded. Nuclei were resuspended in
1.25 volumes Buffer C (20 mM Hepes pH 7.9, 0.42 M NaCl, 1.5 mM MgCl 2, 0.2 mM EDTA,
5 mM dithiothreitol, 0.5 mM PMSF) per volume of packed cells with a Hamilton A dounce and
incubated tumbling at 40 C for 45 minutes. Extracted nuclei were pelleted at 20,000 g for 10
minutes and the pellet was discarded. Nuclear extract supernatant was twice dialyzed against
1.0 M KCl HENG10 (20 mM Hepes pH 7.9, 0.2 mM EDTA, 0.05% Np40, 10% glycerol) for
1 hour each on ice, until the conductivity of the extract matched that of the dialysis buffer.
Control nuclear extract (NE) dialyzed to 1.0 M KCl was removed and set aside at 40 C during the
depletion procedure.
The high salt nuclear extract was depleted by application of the extract to a poly[U]
Sepharose column resuspended in H20 on ice, and washed several times in batch in H20
followed by equilibration in 1.0 M KC1 HENG10. A volume of poly[U] Sepharose
approximately equal to that of the high salt extract was used for the depletion. Extract was
applied to the column at approximately one column volume per hour. The flow through of the
column (NEAU) was detected by Bradford, pooled and dialyzed immediately against three
changes of 0.1 M KC1 HENG20 (HENG10 but with 20% glycerol) on ice for a total of three
hours. Extract was centrifuged at 25,000g for 15 minutes to pellet insoluble material. The
protein concentration of NEAU consistently was two thirds to one half the protein concentration
of the control extract (NE) due to loss during depletion and dialysis. Extract was frozen in small
aliquots in liquid nitrogen and stored at -80 0 C. The entire procedure was routinely performed in
a single, long day for best results.
The critical difference between this protocol, which uncovers the PUF requirement, and
the U2AF depletion protocol (Zamore and Green, 1991) lies in the dialysis of the nuclear extract
directly into high salt buffer. If the extract is dialyzed into low salt buffer (0.1 M KC1) prior to
dialysis into high salt, the extract is not depleted of PUF activity, nor is it depleted of the
PUF60 and p54 proteins as determined by immunoblotting analysis (data not shown). The
reason for this difference is not known, but it is interesting to speculate that the prolonged highsalt dialysis used in making NEAU may disrupt an interaction with a protein or RNA that
associates with the PUF activity under low salt conditions and prevents its interaction with
poly[U] RNA. This interaction might not be disrupted if the extract were dialyzed first into low
salt buffer.
Purification of the PUF activity
PUF activity eluted from the poly[U] Sepharose column with 2.0 M KCl HENG10.
U2AF was eluted with 3.0 M KC1. Both factors eluted in a broad peak of 1.5 to 2 column
volumes and were dialyzed against 0.1 M KC1 HENG20. The 2.0 M KCl eluate was applied to
a phosphocellulose (Whatmann P 11) column according to manufacturers instructions and
washed at 0.1 M and 0.3 M and eluted in batch at 0.6 and 1.0 M KC1. The 0.6 and 1.0 M KCl
eluates were pooled and dialyzed to 0.9 M KCl with 1.0 M HENG20 and applied to a 5 ml
poly[U] Sepharose (Pharmacia) column at 0.25 ml/min and eluted with 1.0 M KC1. The
poly[U] Sepharose eluate (360 gtg) was dialyzed against 0.1 M KC1 HENG20 and applied to a
5 ml S Sepharose column (Pharmacia). The S Sepharose column was step eluted at 0.1 M KC1,
0.55 M KCl and 1.0 M KC1. Activity eluted at 0.55 M KC1.
Further purification of the activity was performed by gel filtration chromatography on
Superose 6 (Pharmacia) by directly applying the S Sepharose eluate in 0.55 M KCl to the
Superose column equilibrated at 0.55 M KCl at 0.25 ml/min KC1 HENG20. Elution of the PUF
activity was compared to that of markers (Boehringer Mannheim Biochemicals, HPLC markers,
#1213 776).
RNAs used in this study
The PIP85a, PIPBG and AD10 RNA substrates were transcribed from plasmids
pPIP85a (Moore and Sharp, 1992), pPIPBG (Crispino et al., 1996), and pAD 10 (Konarska and
Sharp, 1986) using T7 RNA polymerase (United States Biochemicals)and c32P UTP (New
England Nuclear) under standard conditions (Query et al., 1996). The 3' half substrate
PIP85aARX was transcribed from pPIP85aARX which is a deletion of PIP85a. pPIP85aARX
was constructed by digesting pPIP85a with Eco RI and Xho I, the 5' overhang ends of the
DNA were filled-in with Klenow and the DNA was ligated. Pyrimidine-tract RNA, bs-ppt and
ppt-bs RNAs and ppt-AG were previously described RNA oligomers (Query et al., 1997) and
were labeled at their 5' ends.
Splicing in vitro
In vitro splicing reactions were performed under standard conditions (Grabowski et al.,
1984) and were 24% NE or NEAU supplemented with recombinant U2AF65 or the 3 M KCI
eluate of the poly[U] Sepharose column and the PUF fraction in 60 to 100 mM KC1, 20 mM
Hepes pH 7.9, 5 mM MgCl 2, 2 mM ATP and 5 mM creatine phosphate for 1.5 hours at 300.
The reaction was stopped with the addition of 250 pl of 2 x Proteinase K buffer (10 mM Tris
pH7.8, 10 mM MgCl 2, 0.5% SDS), 2jig of glycogen (Boehringer Mannheim Biochemicals)
and 0.5 mg/ml Proteinase K (Boehringer Mannheim Biochemicals). Reactions were digested
for 30 minutes at 650 C and precipitated with the addition of 4 volumes of 95% ethanol, 1.5 M
ammonium acetate, 5 mM MgC12 . 70% ethanol washed pellets were resuspended in 5 tl 8 M
Urea, 1 X TBE, heated to 1000 for three minutes and resolved on 20% Acrylamide (19:1)/50%
Urea (Natural Diagnostics) gels in lx TBE.
Complex assembly assays
A3' complex assembly was performed under standard splicing conditions (above) using
an RNA transcribed from PIP85aARX (an Eco RI to Xho I deletion of PIP85a) and resolved on
4% Acrylamide (60:1) Tris-glycine gels (Konarska and Sharp, 1986). Binding reactions were
stopped with the addition of heparin to 5 mg/ml.
Gel shift
Binding reactions were carried out under splicing conditions at 80 mM KCl to CQ58-19
(Query et al., 1997), labeled at the 5' end, for fifteen minutes at 30 'C, heparin was added to
0.7 mg/ml and reactions were incubated on ice for the remainder of the time course. Binding
reactions were resolved on 8% acrylamide (60:1) Tris-glycine gels. 10 V/cm 2.5 h at room
temperature.
Crosslinking
Binding reactions were carried out under splicing conditions for 10 minutes at 300 C and
crosslinking was carried out on ice for 15 minutes as previously described. The reaction was
digested for lh at 370 with 10 ltg of RNaseA (Calbiochem), trichloroacetic acid precipitated in
the presence of deoxycholate and resolved on a 7.5% polyacrylamide gel.
Antibodies
PUF60 antibodies were generated in two rabbits by inoculation of bacterially expressed
PUF60(P) (Covance). PUF60(P) is the Pst I internal fragment of PUF60 inserted into the Pst I
site of pQE31 (Qiagen) and expressed and purified in 6M Urea according to the manufacturers
instructions. Protein was renatured on the Ni-NTA resin (Qiagen) according to manufacturers
instructions. PUF60(P) protein was further purified on mono S Sepharose. For
immunofluorescence experiments, PUF60 antibodies were affinity purified against PUF60(P)
coupled to CNBr Sepharose (Pharmacia) and eluted sequentially with 100 mM glycine pH 2.0
and 100 mM Triethylamine pH 11.5 and neutralized with Tris to pH 7.
Immunoblotting
The gel that was blotted for figure 4B was run in the presence of 2M Urea to yield a
sharper p54 band. All other gels were standard SDS 4-20% polyacrylamide gels (BioRad).
p54 antibody (anti pep C) was the generous gift of Nilabh Chaudhary (Chaudhary et al., 1991).
p54 immunoblotting was detected using 125 -protein A using standard techniques (Harlow and
Lane). PUF60 immunoblot was detected using the ECL reagent (Amersham).
Immunoprecipitations and Gst pull-downs
GstSF1 and GstU2AF was prepared as previously described (Berglund et al., 1997).
GstU2AF was prepared using standard techniques except that the protein was ammonium
sulfate precipitated and then purified on S Sepharose (Pharmacia). GstPTB and Gst were the
kind gifts of Anna Gil.
Immunofluorescence
Spontaneously transformed mouse fibroblasts (B. Panning, unpublished observations)
were fixed as described by (Lawrence et al., 1989), and immunofluorescence was carried out as
described by (Leonhardt et al., 1992).
ACKNOWLEDGMENTS:
I would like to thank Nilabh Chaudhary for generously contributing p54 antibodies and
his substantial enthusiasm, Nadja Abovich for GstSF1 and SF1/BBP antisera, Helen Cargill,
Margaret Beddall, and Yubin Qiu for expert technical assistance and Robbyn Issner for so much
including growing the cells used in the purification of PUF and especially for general lab sanity
maintenance. GstU2AF was the kind gift of Anna Gil, Phil Zamore and Michael Green. The
protein sequencing of p54 and PUF60 was expertly performed by MIT Biopolymers.
REFERENCES
Abovich, N., Liao, X. C., and Rosbash, M. (1994). The yeast MUD2 protein: an interaction
with PRP 11 defines a bridge between commitment complexes and U2 snRNP addition. Genes
Dev 8, 843-54.
Anderson, K., and Moore, M. J. (1997). Bimolecular exon ligation by the human spliceosome.
Science 276, 1712-6.
Barabino, S. M. L., Blencowe, B. J., Ryder, U., Sproat, B. S., and Lamond, A. I. (1990).
Targeted snRNP depletion reveals an additional role for mammalian Ul snRNP in spliceosome
assembly. Cell 63, 293-302.
Bennett, M., Michaud, S., Kingston, J., and Reed, R. (1992). Protein components specifically
associated with prespliceosome and spliceosome complexes. Genes Dev. 6, 1986-2000.
Berglund, J. A., Chua, K., Abovich, N., Reed, R., and Rosbash, M. (1997). The splicing
factor BBP interacts specifically with the pre-mRNA branchpoint sequence UACUAAC. Cell
89, 781-7.
Blencowe, B. B., Nickerson, J. A., Issner, R., Penman, S., and Sharp, P. A. (1994).
Association of nuclear matrix antigens with exon-containing splicing complexes. J. Cell Biol.
127, 593-607.
Blencowe, B. J., Issner, R., Kim, J., McCaw, P., and Sharp, P. A. (1995). New proteins
related to the Ser-Arg family of splicing factors. RNA 1, 852-65.
Chaudhary, N., McMahon, C., and Blobel, G. (1991). Primary structure of a human argininerich nuclear protein that colocalizes with spliceosome components. Proc. Natl. Acad. Sci. USA
88, 8189-8193.
Chiara, M. D., Champion-Arnaud, P., Buvoli, M., Nadal-Ginard, B., and Reed, R. (1994).
Specific protein-protein interactions between the essential mammalian spliceosome-associated
proteins SAP 61 and SAP 114. Proc Natl Acad Sci U S A 91, 6403-7.
Crispino, J. D., Mermoud, J. E., Lamond, A. I., and Sharp, P. A. (1996). Cis-acting elements
distinct from the 5' splice site promote Ul-independent pre-mRNA splicing. RNA 2, 664-73.
Dickson, B. J. (1998). Photoreceptor development: breaking down the barriers. Curr Biol 8,
R90-2.
Dignam, J. D., Lebovitz, R. M., and Roeder, R. D. (1983). Accurate transcription initiation by
RNA polymerase II in a soluble extract from isolated mammalian nuclei. Nucleic Acids Res. 11,
1475-1489.
Frendeway, D., and Keller, W. (1985). Stepwise assembly of a pre-mRNA splicing complex
requires U-snRNAs and specific intron sequences. Cell 42, 355-367.
Gozani, O., Feld, R., and Reed, R. (1996). Evidence that sequence-independent binding of
highly conserved U2 snRNP proteins upstream of the branch site is required for assembly of
spliceosomal complex A. Genes Dev 10, 233-43.
Grabowski, P. J., Padgett, R. A., and Sharp, P. A. (1984). Messenger RNA splicing in vitro:
An excised intervening sequence and a potential intermediate. Cell 37, 415-427.
Kamitani, T., Nguyen, H. P., Kito, K., Fukuda-Kamitani, T., and Yeh, E. T. (1998).
Covalent modification of PML by the sentrin family of ubiquitin-like proteins. J Biol Chem
273, 3117-20.
Kanaar, R., Roche, S. E., Beall, E. L., Green, M. R., and Rio, D. C. (1993). The conserved
pre-mRNA splicing factor U2AF from Drosophila:requirement for viability. Science 262, 56973.
Kennedy, C. F., and Berget, S. M. (1998). Direct Submission to GENBANK: accession
AF055719. Unpublished.
Konarska, M. M., and Sharp, P. A. (1986). Electrophoretic separation of complexes involved
in the splicing of precursors to mRNAs. Cell 46, 845-855.
Lawrence, J. B., Singer, R. H., and Marselle, L. M. (1989). Highly localized tracks of specific
transcripts within interphase nuclei visualized by in situ hybridization. Cell 57, 493-502.
Leonhardt, H., Page, A. W., Weier, H. U., and Bestor, T. H. (1992). A targeting sequence
directs DNA methyltransferase to sites of DNA replication in mammalian nuclei. Cell 71, 86573.
Li, S., Li, Y., Carthew, R. W., and Lai, Z. C. (1997). Photoreceptor cell differentiation
requires regulated proteolysis of the transcriptional repressor Tramtrack. Cell 90, 469-78.
Lin, R. J., Nagy, L., Inoue, S., Shao, W., Miller, W. H., Jr., and Evans, R. M. (1998). Role
of the histone deacetylase complex in acute promyelocytic leukaemia. Nature 391, 811-4.
MacMillan, A. M., McCaw, P. S., Crispino, J. D., and Sharp, P. A. (1997). SC35-mediated
reconstitution of splicing in U2AF-depleted nuclear extract. Proc Natl Acad Sci U S A 94, 1336.
McCombie, W. R., Kelley, J. M., Aubin, L., Goscoechea, M., FitzGerald, M. G., Wu, A.,
Adams, M. D., Dubnick, M., Kerlavage, A. R., Venter, J. C., and Fields, C. A. (1993).
Caenorhabditiselegans cDNAs. Genbank: TO 1856.
Michaud, S., and Reed, R. (1991). An ATP-independent complex commits pre-mRNA to the
mammalian spliceosome assembly pathway. Genes & Dev. 5, 2534-2546.
Michaud, S., and Reed, R. (1993). A functional association between the 5' and 3' splice site is
established in the earliest prespliceosome complex (E) in mammals. Genes Dev 7, 1008-20.
Moore, M. J., and Sharp, P. A. (1992). Site-specific modification ofpre-mRNA: the 2'
hydroxyl groups at the splice sites. Science 256, 992-997.
Muller, S., Matunis, M. J., and Dejean, A. (1998). Conjugation with the ubiquitin-related
modifier SUMO-1 regulates the partitioning of PML within the nucleus. EMBO J 17, 61-70.
Patton, J. G., Porro, E. B., Galceran, J., Tempst, P., and Nadal-Ginard, B. (1993). Cloning
and characterization of PSF, a novel pre-mRNA splicing factor. Genes & Dev. 7, 393-406.
Peek, R., Pruijn, G. J., van der Kemp, A. J., and van Venrooij, W. J. (1993). Subcellular
distribution of Ro ribonucleoprotein complexes and their constituents. J Cell Sci 106, 929-35.
Potashkin, J., Naik, K., and Wentz, H. K. (1993). U2AF homolog required for splicing in
vivo. Science 262, 573-5.
Query, C. C., McCaw, P. S., and Sharp, P. A. (1997). A minimal spliceosomal complex A
recognizes the branch site and polypyrimidine tract. Mol Cell Biol 17, 2944-53.
Query, C. C., Strobel, S. A., and Sharp, P. A. (1996). Three recognition events at the branchsite adenine. EMBO J 15, 1392-402.
Reed, R. (1996). Initial splice-site recognition and pairing during pre-mRNA splicing. Curr.
Opin. Genet. Dev. 6, 215-220.
Roscigno, R. F., Weiner, M., and Garcia-Blanco, M. A. (1993). A mutational analysis of the
polypyrimidine tract of introns. Effects of sequence differences in pyrimidine tracts on splicing.
J Biol Chem 268, 11222-9.
Rounsley, S. D., Lin, X., Ketchum, K. A., Crosby, M. L., Brandon, R. C., Sykes, S. M.,
Mason, T. M., Kerlavage, A. R., Adams, M. D., Somerville, C. R., and Venter, J. C. (1997).
Arabidopsis thalianachromosome II BAC F4P9 genomic sequence. Unpublished AC002332.
Rudner, D. Z., Kanaar, R., Breger, K. S., and Rio, D. C. (1996). Mutations in the small
subunit of the DrosophilaU2AF splicing factor cause lethality and developmental defects. Proc
Natl Acad Sci U S A 93, 10333-7.
Ruskin, B., and Green, M. R. (1985). Role of the 3' splice site consensus sequence in
mammalian pre-mRNA splicing. Nature 317, 732-4.
Ruskin, B., Zamore, P. D., and Green, M. R. (1988). A factor, U2AF, is required for U2
snRNP binding and splicing complex assembly. Cell 52, 207-19.
Simons, A., Melamed-Bessudo, C., Wolkowicz, R., Sperling, J., Sperling, R., Eisenbach,
L., and Rotter, V. (1997). PACT: cloning and characterization of a cellular p53 binding protein
that interacts with Rb. Oncogene 14, 145-55.
Staknis, D., and Reed, R. (1994). SR proteins promote the first specific recognition of PremRNA and are present together with the U1 small nuclear ribonucleoprotein particle in a general
splicing enhancer complex. Mol Cell Biol 14, 7670-82.
Sternsdorf, T., Grotzinger, T., Jensen, K., and Will, H. (1997). Nuclear dots: actors on many
stages. Immunobiology 198, 307-31.
Wahren, M., Mellqvist, E., Vene, S., Ringertz, N. R., and Pettersson, I. (1996). Nuclear
colocalization of the Ro 60 kDa autoantigen and a subset of U snRNP domains. Eur J Cell Biol
70, 189-97.
Wan, K., Nickerson, J. A., Krockmalnic, G., and Penman, S. (1994). The B1C8 protein is in
the dense assemblies of the nuclear matrix and relocates to the spindle and pericentriolar
filaments at mitosis. Proc. Natl. Acad. Sci. USA 91, 594-598.
Wentz, H. K., and Potashkin, J. (1996). The small subunit of the splicing factor U2AF is
conserved in fission yeast. Nucleic Acids Res 24, 1849-54.
Zahler, A. M., Neugebauer, K. M., Stolk, J. A., and Roth, M. B. (1993). Human SR proteins
and isolation of a cDNA encoding SRp75. Mol Cell Biol 13, 4023-8.
Zamore, P. D., and Green, M. R. (1991). Biochemical characterization of U2 snRNP auxiliary
factor: an essential pre-mRNA splicing factor with a novel intranuclear distribution. EMBO J
10, 207-14.
Zamore, P. D., and Green, M. R. (1989). Identification, purification, and biochemical
characterization of U2 small nuclear ribonucleoprotein auxiliary factor. Proc Natl Acad Sci U S
A 86, 9243-7.
Zhang, W. J., and Wu, J. Y. (1996). Functional properties of p54, a novel SR protein active in
constitutive and alternative splicing. Mol Cell Biol 16, 5400-8.
Zorio, D. A., Lea, K., and Blumenthal, T. (1997). Cloning of Caenorhabditis U2AF65: an
alternatively spliced RNA containing a novel exon. Mol Cell Biol 17, 946-53.
Zuo, P., and Maniatis, T. (1996). The splicing factor U2AF35 mediates critical protein-protein
interactions in constitutive and enhancer-dependent splicing. Genes Dev 10, 1356-68.
FIGURE LEGENDS
Figure 1.
Poly[U]-depleted nuclear extract requires both U2AF and PUF.
A. in vitro splicing reaction on the PIP85a pre-mRNA. Nuclear extract, high salt treated but
not depleted (NE), is compared to nuclear extract that has been depleted at high salt by passage
over a poly[U] Sepharose column (NEAU, compare lanes 1 and 2). Addition of either the 2.0
M KCl eluate fraction of the poly U depletion column (the PUF containing fraction) or
recombinant U2AF65 to the depleted extract does not efficiently restore splicing activity to the
depleted extract (lanes 3 and 4). Addition of both recombinant U2AF65 and PUF restores
splicing activity to the extract. The positions of the pre-mRNA, mRNA and lariat product and
intermediate are indicated schematically to the right of the figure.
B. PUF activity is required for efficient splicing of at least three pre-mRNAs. U2AF purified
from nuclear extract (the 3 M KC1 eluate fraction of the poly[U] depletion column) was
compared to U2AF65 in the reconstituted splicing system. Splicing in vitro of PIPBG (lanes 17), PIP85a (lanes 8 to 14) and AD10 (lanes 15 to 21) with or without added purified PUF
activity were compared in the presence of U2AF (lanes 6 and 7, 13 and 14, and 20 and 21) or
recombinant U2AF65 (lanes 4 and 5, 11 and 12, and 18 and 19). The activity supplied by the
PUF fraction does not substitute for U2AF35. If PUF activity substituted for U2AF35, then
PUF60 and U2AF35 containing fractions should have equivalent activities, while this may be
true for PIP85a (compare lanes 12 and 13) it is not true for PIPBG or for AD10 (compare lane 5
with 6 and lane 19 with 20).
A
NE NEAU
+
His 6 U2AF65
PUF
+
+
+
O-r
0-
rn-E
EL
12345
B
PIPBG
PIP85a
AD10O
NE NEAU
NE NEAU
NE NEAU
+
+
+
PUF
U2AF65
+
+
++
++
+
U2AF
+
+
+
+
+
+
+
+
+
+
I
EID15 16 17 18 19 20 21
I IE I
1
2
3
4
5
6
7
8
9
10 11 12 13 14
I
Figure 2.
Purification of the PUF activity.
A. Schematic diagram of the purification procedure, described in the Methods section.
B. Chromatography of the PUF activity on S Sepharose. Top panel, silver stained protein gel
of the fractionation. Bottom panel, reconstitution of in vitro splicing activity to NEAU
supplemented with recombinant U2AF65, PIP85a substrate. Load on the column (LD) and
flowthrough (FT) are indicated as are fraction numbers.
C. Coomassie stained gel of the highly purified PUF activity fraction (fraction 8, in figure 2B,
lane 2) and Perfect Protein markers (Novagen, lanes 1 and 3).
D. Purification of the S Sepharose purified material on Superose 6 gel filtration
chromatography. Top panel, silver stained gel of the even numbered column fractions, peak
fraction of the of the gel filtration standards (BioRad; 415 kD, fraction 23; 150 kD, fraction 27;
50 kD, fraction 33) is indicated above the gel. Bottom panel, reconstitution of in vitro splicing
assay of odd numbered fractions.
A
NE
I
poly U Sepharose
I
2
2.0
1.0
3.0 M KCI
(NEAU)
Phosphocellulose
I
1 I
0.6
FT (0.1 M) 0.3
1.0 M KCI
poly U
2.0 M KCI
1.0
S Sepharose
1.0 M
0.55
(PUF)
FT (0.1 M)
FT (0.9 M)
B
1.0 M
mM 500 mM
100 -m
fraction:
3 i 6 7 8 9 171819 20
LD FT 1 2.....
ii:i-:i::
!
~ ::':':::''i"
ii
U
-
:
5?
*
LD
1
FT
.0)
C,
0
-
75 kD
2
........
' ::::
...
....
: i::::: I :: s : : .:.:.::..::
50
7::::::::::':::':
':
:-:-:::i--:-::::i:--::::i:i::-S~
::::::
3
7
kD
i
8
...........
.
.j
150 kD
. ::'.
i~
::: : I?[:
IX[!!!i[!:
ii?i ii[[
!!!!::i
-
35 kD
-
25 kD
1.0 M
500 mM
100 mM
fraction:
-
:-'!'''''''';::::::::':: iiiiili'iiiiiiiiii
iiii~
i - 100 kD
9
17 18
oC1E
o)
0
O
0
0O
.L
u
d o it
N
0
IC,
0
CV)
0
IC
CN
0
cm
150 kD
1
1
50 kD
415415 I
16 18 20 22 24 26 28 30 32 34
II
LD
2
3
4
5
6
7 8
9
10 11 12 13 14
LL.
IL.
o
12345678910
........
Figure 3.
PUF60 is ubiquitously expressed in humans as a 2.0 kb mRNA.
A northern blot of human tissues (tissues indicated, Clontech) was probed with an antisense
PUF60 RNA probe. A ubiquitously expressed transcript of 2.0 kb is observed and shows
similar expression levels in tissue as SRm160/B 1C8 on the same blot (Blencowe et al., 1995).
I
PLACENTA
LUNG
LIVER
MUSCLE
KIDNEY
PANCREAS
") :'iFii':i:ii:c;i
i
BRAIN
HEART
Figure 4.
NEAU is depleted of both PUF60 and p54.
A. PUF60 immunoblotting analysis of three different NEAU (lanes 2, 4 and 5) and the
corresponding NE for two of those extracts (lanes 3 and 7). A cytoplasmic extract, S 100, is
also shown (lane 1). The 55 kD band is a non-specific band that appears in immunoblots
developed with pre-immune serum.
B. Immunoblotting analysis of NE (lane 2) and NEAU (lane 1) detected with an antibody
directed against a p54 peptide (Chaudhary et al., 1991). The p54 in the purified PUF fraction is
of slower mobility than the p54 found in nuclear extract (compare lane 4 with lane 2). This
slower mobility of purified p54 is a gel artifact as mixing SDS-treated and boiled NEAU to
SDS-treated and boiled PUF fraction followed by immediate loading on the gel, led to the
comigration of the purified activity with the endogenous activity (compare lane 3 with lanes 2
and 4). x marks a cross-reacting species.
4
-le,
44*
IL
V0
co
LL
0
C
D
"0
o.4
PUF
NEApU + PUF
NE
jNEApU
Figure 5.
The PUF factor p54 can form complexes with PUF60.
Translation in vitro of p54, PUF60, and both p54 and PUF60 (lanes 1-3 respectively) were
immunoprecipitated with p54 antibody and the pellets were resolved by SDS PAGE (lanes 5-7).
Immunoprecipitation of unprogrammed lysate (UN) is shown in lane 4. Control
immunoprecipitation with no antibody is shown in lane 8.
TOTAL
IP
no
anti-p54
p54
p54 PUF PUF
UN
p54 p54
p54 PUF PUF PUF
well (/)
00a,
co,
PUF60 dimer
ci
0)
03)
o
0i
p54
I-
PUF60
1
2
3
4
5
6
7
8
Figure 6.
PUF activity is required for efficient A3' spliceosome assembly.
A time course (0 minutes, 10 minutes and 30 minutes) of complex assembly in NEAU alone
(lanes 1-3), in the presence of PUF activity (lanes 4-6), in the presence of recombinant U2AF65
(lanes 7-9), or in the presence of both activities (lanes 10-12). The complexes formed in NE
(lanes 13-15) are shown for comparison.
NE
NEAU
His 6U2AF65
PUF
time
+ + +
+ + +
+++
+++
0 10 30 0 10 30 0 10 300 10 30 0 10 30
A3'
H
1 2 3 4 5 6 7 8
9 10 11 12 13 14 15
Figure 7. The PUF fraction has three species that crosslink to pre-mRNA.
PIP85a RNA was incubated with purified PUF fraction under splicing conditions except that
ATP and creatine phosphate were omitted. The reaction was exposed to ultraviolet light,
digested with RNaseA and cross-linked proteins were resolved by SDS-PAGE (lane 1).
Specificity of this interaction was tested by including 0.5 and 5 gg of homo- or heteropolymeric
RNAs in the binding reaction. poly[G] RNA competed poorly for PUF binding (lanes 2 and
3), poly[I] and poly[C] did not compete at either concentration tested (lanes 4 and 5, 8 and 9),
poly[U] competed at both concentrations tested (lanes 6 and 7) and poly [ACU] competed only
at the higher concentration tested (10 and 11).
00.5 5 0.5 5 0.5 5 05 5 0.5 5
12
3 4
5
6
7
8
9
pg competitor
10 11
Figure 8.
PUF does not bind the AG dinucleotide or the branch sequence.
An electrophoretic mobility shift analysis of a binding titration of PUF on pyrimidine-tract
RNAs with or without an AG dinucleotide is shown (lanes 9-14 and lanes 15 to 20). A similar
titration is shown with a pyrimidine-tract RNA containing either a 3' branch sequence or a 5'
branch sequence (lanes 1 to 4 and lanes 5 to 8). No difference in PUF60 binding was observed
for any of these RNAs.
ppt-bs
bs-ppt
ppt AG
ppt
PUF60
,, . . : ::
...., .
......
:,"::':
,........,:.-..:.,:
!~:::.,:..:::•.
i~i:
•i! :,.:,,q
i~ ii• i!.............................
ii~i:"
BOUNDiiii
BOUND
BOUND
FREE
FREE
1234
5678
9 1011121314
15 16 17 18 19 20
Figure 9. The binding of PUF to pyrimidine-tract RNA is unaffected by the
presence of either U2AF or U2AF65.
Serial dilutions of PUF (lanes 2 to 10, 12 to 20, and 22 to 30) were added to constant amounts
of buffer alone (lanes 12 to 20), U2AF (lanes 2 to 11), or U2AF65 (lanes 22 to 31) and then
mixed with pyrimidine-tract RNA probe. U2AF in the absence of PUF is shown in lane 11 and
U2AF65 in the absence of PUF is shown in lane 31. Pyrimidine-tract RNA probe is shown in
lanes 1 and 21.
U2AF65
U2AF
PUF
U2AF I
U2AF65
PUF60
PUF60
U2AF65
FREE
FREE
1 2 3 4 5
6
7
8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31
Figure 10.
SF1/BBP.
PUF60 interacts with the branch sequence binding protein
GstSF1/BBP fusion protein (lanes 1 and 2) was immobilized on glutathione agarose and
incubated with NE (even numbered lanes) or NEAU (odd numbered lanes) and associated
proteins were detected by immunoblotting using antiserum to PUF60 (top panel) or to U2AF65
(bottom panel). As controls for specificity the pyrimidine-tract binding proteins GstPTB (lanes
3 and 4, Anna Gil and P. A. S., unpublished) and GstU2AF65 (lanes 5 and 6) and the Gst
protein alone (lanes 7 and 8) were used. Arrow indicates the PUF60 signal Lanes marked TTL
are the signal obtained from 20% of input NE.
4%
1I.t
0
(P
E
u
AU NE AU NE
E
Cm
u"
AU NE AU NE TTL
oaPUF60
TTL
1
2
3
4
i~oiiIU2AF65
5
6
7
8
TTL
Figure 11. PUF60 localizes to a non-speckle domain of the nucleus.
A. DAPI stained nucleus of a spontaneously transformed mouse fibroblast cell
B. The same nucleus stained with the B1C8 antibody which recognizes SRml60, a nuclear
speckle component (Blencowe et al., 1994).
C. Staining with anti-PUF60 antibody, the PUF60 signal localizes to a discrete nuclear body.
All of the available PUF60 antibodies are not observed to recognize native protein and the
PUF60 staining structures seen here may reflect only the non-native PUF60 protein present in
the cell.
D. Merge of the DAPI, B C8 and PUF60 signals and E. merge of the B1C8 and PUF60 signal
showing that some PUF60 staining bodies co-localize with B 1C8-stained nuclear speckles.
100
CHAPTER 3: PUF60 A NOVEL PYRIMIDINE TRACT
BINDING FACTOR WITH HOMOLOGY TO THE SPLICING
FACTORS U2AF65 AND MUD2P DIMERIZES VIA ITS CTERMINAL RRM-LIKE I)OMAIN, THE PUMP DOMAIN
Patrick Schonleber McCaw, Kevin Amonlirdviman, and
Phillip A. Sharp
Center for Cancer Research
Massachusetts Institute of Technology
102
ABSTRACT
We have identified a new member of the U2AF65 family of splicing factors, PUF60.
PUF60 was identified in highly purified fractions that, together with U2AF, restored splicing
activity to poly[U] depleted nuclear extracts (see chapter 2). Homologues of PUF60 are found
in vertebrates (human and mouse) and in the invertebrate Drosophila,but not in C. elegans.
The most similar protein in the budding yeast, S. cerevisiae, is Mud2p, which is the probable
orthologue of U2AF65 based on functional and structural similarities. PUF60 has the unusual
property of forming an SDS-resistant dimer. This dimerization is mediated by a C-terminal
RRM-like domain, the PUMP domain. The SDS-resistant dissociation constant of dimerization
by the PUMP domain is approximately 1 tM. The PUMP domain homology is found in
several other proteins, some of which have been shown to mediate protein-protein interactions.
103
INTRODUCTION
Binding of many proteins to RNA occurs via the RNA recognition motif (RRM)
domain. The RRM domain has been found in many splicing and polyadenylation factors,
hnRNP proteins, and other proteins and is considered a hallmark of one class of RNA-binding
proteins (Kenan et al., 1991; Nagai et al., 1995). While many RRM domains have been found
to interact with RNA, others have no known RNA ligand. For instance the U A protein has
two RRM domains: the first RRM mediates interaction with the Ul stem loop, while the second
RRM domain has no known RNA binding activity. Despite the fact that this RRM domain has
no known RNA ligand, it is the most conserved domain of the protein and has been shown to
be functionally important in the yeast S. cerevisiae(Tang and Rosbash, 1996). It has been
known for some time that some RRM domains can mediate protein-protein interactions. The
best known example of this is the interaction of the first RRM of U2B" with U2A' (Scherly et
al., 1990).
The RRM domain is a compact domain of 80 or more amino acids that is characterized
by two conserved motifs: the N-terminal RNP2 or hexamer motif and the central RNP 1 or
octamer motif (Kenan et al., 1991). These motifs are characterized by the presence of
conserved aromatic residues, but the RRM domain family is extremely diverse in sequence.
Aside from the RNP2 and RNP 1 motifs, the RRM domains has few conserved positions,
although there are conserved hydrophobic residues that constitute the hydrophobic core of the
domain (Nagai et al., 1995). The structure of the U1A RRM1 domain has been solved both
bound to the Ul snRNA stem-loop (Oubridge et al., 1994) and unbound (Avis et al., 1996;
Hoffman et al., 1991; Nagai et al., 1995). The RRM domain consists of four stranded
antiparallel B sheet backed by two a helices in a P a P P a P structure (Hoffman et al., 1991;
Nagai et al., 1990). The tertiary structure of the RRM domain explains the high degree of
conservation of the aromatic residues found in the RNP2 and RNP I motifs. These residues are
found on the surface of the protein in the B1 and B3 strands that compose the RNP motifs. The
conserved aromatic residues of U1A are known to make base-stacking interactions with the
stem loop I RNA to which U1A binds. Many other conserved positions are found to constitute
the hydrophobic core (Hoffman et al., 1991; Nagai et al., 1990; Oubridge et al., 1994).
The pre-mRNA is recognized by the spliceosome through recognition of the 5' splice
site, the branch sequence and the pyrimidine tract. Recognition of the 5' splice site is
accomplished by means of base pairing to U1 snRNA, while recognition of the pyrimidine tract
is accomplished by the binding of the pyrimidine-tract binding splicing factor U2AF (U2
104
snRNP Auxiliary Factor). U2AF is a heterodimer of 65 and 35 kD proteins and is highly
conserved in all splicing organisms. U2AF65 is required for the stable, ATP-dependent
association of U2 snRNP with the branch sequence. However, neither Ul snRNP nor U2AF
is required for splicing in vitro. Splicing in Ul snRNP and U2AF depleted extracts is
dependent on the addition of large quantities of SR proteins and is also dependent on the
presence of the PUF pyrimidine-tract binding factor (MacMillan et al., 1997).
The PUF pyrimidine-tract binding factor has been purified and consists of the splicing
factor p54 and at least one other protein, poly[U] binding factor 60 kD (PUF60; chapter 2).
PUF60 is a member of the U2AF/Mud2 family of pyrimidine tract binding proteins. In this
chapter we show that PUF60 is conserved from insects to man. The Drosophilahomologue of
PUF60, DPUF68, has been cloned and sequenced. Like U2AF65 and Mud2p, PUF60 has a
central domain containing two RRM domains and a C-terminal domain consisting of a
degenerate RRM domain. We have found that this C-terminal domain of PUF60 mediates an
unusually stable homodimerization reaction and does not contribute to the RNA-binding affinity
of PUF60. This domain is also shown to be a distinct subfamily of the RRM domain homology
group. Because of the unusual properties of this domain and its status as a distinct subfamily of
the RRM domain homology, we have termed this domain the PUMP (PUF60, U2AF65 and
Mud2p Protein-Protein interaction) domain.
105
RESULTS
PUF60, conserved in evolution, is related to the yeast splicing factor Mud2p
We have purified a complex of proteins from HeLa cell nuclear extracts that bind at high
salt concentrations to poly[U] Sepharose and promote the efficiency of RNA splicing in vitro
(chapter 2). A 130 kD protein was selected for tryptic digestion and peptide sequencing.
Thirteen peptides were sequenced yielding 169 amino acids of sequence. A cDNA in the human
EST database was identified which encoded all thirteen sequenced peptides in a 559 amino acid
open reading frame. This cDNA encoded a predicted protein of 59.9 kD with overall similarity
to the pyrimidine-tract binding protein U2AF65, the human auto-immune antigen HCC (Imai et
al., 1993) and the S. cerevisiaesplicing factor Mud2p. The protein encoded by this cDNA is
referred to as PUF60 (poly[_] binding Factor-60 kD). A search of the EST database identified
numerous mouse cDNA sequences allowing reconstruction of the murine ORF from the
sequences available in the database. Only 12 amino acids were found to differ between the
human and murine PUF60 homologues. These differences consisted of an insertion of five
alanines in the N-terminal poly-alanine repeat, and the remaining seven differences were
conservative substitutions (data not shown). A PUF60 homologue was identified in the
DrosophilaEST database and has been sequenced. The DrosophilacDNA was sequenced and
is 45.3% similar to PUF60 over its entire length (figure lA). The Drosophilahomologue
(DPUF68) contains an ORF of 570 amino acids, encoding a predicted protein of 68 kD.
Comparison of the hypothetical translation products of the human, mouse, and
Drosophilahomologues identified four domains of this protein (figure 1). The N-terminal
domain is not well conserved between PUF60 and DPUF68, and DPUF68 contains four RS or
SR dipeptides which are not present in the mammalian proteins. The conserved regions begin
with a segment of approximately 40 amino acids immediately upstream of the first RNA
recognition motif (RRM) domain. The central domain consists of two RRM domains with good
matches to the RNP 1 and RNP2 consensus motifs; these domains are similar to the RRM
domains of U2AF65 and Mud2p. The C-terminal domain is preceded by a variable domain
that, in Drosophila,contains a polyalanine motif similar to the polyalanine motif found near the
N-terminus of the mammalian PUF60 homologue. The C-terminal domain is similar to the
RRM domain, however it is unusual in that it is characterized by a poor match to the N-terminal
RNP2 consensus motif, but does have a good match to the central RNP I1consensus motif.
This C-terminal domain is the most conserved part of the protein when compared among the
106
PUF60 homologues U2AF65, Mud2p and HCC. Because this domain has both sequence
features and biochemical activities (discussed below) that differ from the general class of RRM
domains it is referred to as the PUMP (PUF60, U2AF65, Mud2p protein-protein interaction)
domain.
The PUMP domain is a distinct subfamily of the large RRM domain family
To determine if the unusual sequence characteristics of the PUMP domain of PUF60
and its homologues U2AF65 and Mud2p are found in other proteins we performed a database
search by building a motif block of the PUMP domains of these proteins using the
BLOCKMAKER algorithm (Henikoff et al., 1995). The block was then used to search the
non-redundant Genbank database for potential homologues using the MAST program (Bailey
and Gribskov, 1998; Bailey and Gribskov, 1997). Several PUMP domain homologues were
identified that had not previously been identified as RRM domains (for example, U2AF35),
while others had previously been identified as potential RRM domains (for example, kis/PCIP2). Alignment of the potential PUMP domain homologues (using ClustalW; Thompson et
al., 1994) with all of the RRM family of domains found in the PFAM database (Sonnhammer et
al., 1998) clearly demonstrated that the PUMP domain is a distinct subset of the RRM domain
family (figure 2). Notably, alignment of the PUMP domain with the RRM domain family
showed that the conserved hydrophobic residues of the RRM domain were conserved in the
PUMP domain as well. However, the RNP2 motif was absent and may be replaced instead by
a cluster of hydrophobic amino acids. In contrast to the PUMP RNP2 motif, an RNP1 motif
was readily identifiable in the PUMP homology, but the RNP 1 motif showed remarkable
differences from that of the RRM domain family consensus. In the PUMP domains the
conserved basic residue ofRNP1 was replaced with a hydrophobic residue and the first
aromatic residue was replaced with lysine, asparagine or arginine. C-terminal to the RNP 1
motif a conserved hydrophobic residue in the RRM family is found to be an aromatic residue,
while C-terminal to the suspected position of the RNP2 motif there is often a clustering of acidic
residues in the PUMP family that is not present in the RRM family. Together these features
form a signature for the PUMP domain.
Proteins identified as containing PUMP homologies by this method include the
cytoplasmic kinase, P-CIP2, a protein previously identified as having homology with the Cterminus of U2AF65, U2AF35 and its homologue Urp, and Tat-SF 1, a protein implicated in the
transcriptional elongation activity of the HIV protein Tat. Several other examples were
identified that are implicated in splicing including the S. cerevisiae factor Cus2 (Wells et al.,
1996) and the S. pombe U2AF interacting protein Uap2 (McKinney et al., 1997). Domains
107
that are likely to be PUMP homologues based on this criteria include the splicing factor Prp24,
and the yeast genes Not4, Nrdl and Ylfl which are involved in RNA metabolism.
The PUMP domain is a protein-protein interaction domain
Because PUF60 is encoded by an open reading frame of 559 amino acids, it is expected
to have a mass of 59.9 kD and is expected to migrate on an SDS-polyacrlyamide gel at 60 kD.
Instead, PUF60 was identified as a 130 kD product (chapter 2, figure 2C). The paradox of
how a 130 kD protein purified from nuclear extract could be encoded by an open reading frame
of 559 residues was resolved when PUF60 was produced in vitro by a translation reaction. The
predominant translation product migrated as a 60 kD polypeptide, but a small amount migrated
as a 130 kD polypeptide. This suggested that the 60 kD primary translation product could form
SDS-resistant dimers with an apparent mobility of a 130 kD polypeptide. To test whether the
130 kD form is a dimer of PUF60, full-length PUF60 and N-terminally truncated PUF60
(deleted from 1 to 76, PUF60AN) were co-translated (figure 3A). Translation in vitro of the
full length PUF60 produced a predominant 60 kD product and a small amount of a 130 kD
product. Translation in vitro of PUF60AN produced a predominant band of approximately 55
kD and a small amount of a 120 kD species. Co-translation of both the full length and
PUF60AN produced the expected 60 kD and 55 kD bands as well as bands of 130, 125, and
120 kD. These latter bands were present in approximately the ratio of 1:2:1, consistent with the
hypothesis that the 130 kD and 125 kD species were homodimers of the input forms of PUF60
while the 125 kD species was a heterodimer of these two forms. Other experiments indicate that
dimerization of PUF60 was stable in high concentrations of the reducing reagents dithiothreitol
(100 mM) or 2-mercaptoethanol (280 mM) and was stable to the addition of 4-vinylpyridine, a
thiol-alkylating reagent, when added in excess after reduction with 2-mercaptoethanol,
indicating that dimerization was not dependent on disulfide bond formation (data not shown).
Consistent with the hypothesis that PUF60 forms SDS-resistant dimers, we observe in
immunoblot experiments that the PUF60 protein in nuclear extract occurs in both monomer (60
kD) and dimer (130 kD) form (chapter 2, figure 3A).
Bacterially expressed PUF60 also formed SDS-resistant dimers (figure 3B). Further,
this dimerization was found to be concentration dependent; high concentrations of monomers
formed dimers more efficiently than low concentrations. When the concentration of PUF60
protein, which was synthesized in bacteria as a His6 fusion protein (His 6PUF60), was increased
by two-fold increments from 70 nM to 2.3 gM the fraction of protein migrating as a dimer
dramatically increased. It was estimated that an approximately equal fraction of PUF60 was
108
monomer and dimer at between 0.75 to 1.5 gM suggesting a dissociation constant for the SDSresistant form of about 1 jtM.
The PUMP domain at the carboxyl terminus of PUF60 was necessary for formation of
the dimer. PUF60AC protein, which lacks this domain (amino acids 516 to 559), does not
form dimers even at high concentrations of protein. This C-terminal truncation deletes the last
44 amino acids containing the RNPI motif of the PUMP domain. The PUF60AC version of the
protein was produced as a His 6 fusion protein in parallel with His 6PUF60. Low and high
concentrations of these proteins were incubated and then analyzed by SDS-polyacrylamide gel
electrophoresis (SDS-PAGE)(Figure 6). His6PUF60 formed SDS-resistant dimers while
His 6PUF60AC did not. It should be noted that there are no cysteines in the deleted region of the
protein which could stabilize dimerization by disulfide bond formation.
Formation of SDS-resistant dimers is a property of the PUMP domain alone. Different
portions of PUF60 were expressed in translation reactions in vitro and in bacterial expression
systems. These proteins were individually tested for formation of SDS-resistant dimerization at
high concentrations (figure 3D). Subregions of PUF60 that included the PUMP domain formed
dimers, while subregions that did not include the PUMP domain did not form dimers. A Gst
fusion protein with the C-terminal 94 amino acids of PUF60, encoding the entire PUMP
domain, formed SDS-resistant dimers at approximately the same efficiency and concentration as
the full length protein.
The PUMP domain does not contribute to RNA binding
To test if the PUF60 PUMP domain contributed significantly to RNA-binding activity,
the affinities of bacterially expressed PUF60 and PUF60AC for a pyrimidine-tract RNA were
determined. A series of different length pyrimidine-tract RNAs were synthesized using
sequences from the PIP85a substrate, and RNA binding was evaluated using an electrophoretic
mobility shift assay. Pyrimidine-tract RNAs of 14 (pyrl4) and 23 (pyr23) nucleotides were
bound to approximately the same extent by PUF60. In contrast, RNAs of 11 (pyrl 1) and 7
(pyr7) nucleotides were not bound by PUF60. This suggested that optimal binding required
sequences longer than 11 nucleotides. The specificity of PUF60 binding to pyr23 was
examined by competition with RNA homo- and heteropolymers. Pyr23 binding was competed
only with RNA polymers containing uridine, i.e., with poly(U), poly(C,U), and poly (G,U).
This binding could not be competed with homopolymers of poly[C], poly[G], poly[A] or
poly[I].
109
When the binding activity of His 6PUF60 was compared to that of His 6PUF60AC for the
substrate pyr23, the two proteins had indistinguishable affinities (Kd obs = 138 nM and 122 nM
respectively). The Hill coefficients, which indicate degree of cooperativity during binding,
were evaluated (Creighton, 1984). The coefficients were found to be 2.8 and 1.8, respectively.
This difference in Hill coefficients suggests that the PUMP domain of PUF60 mediates a
cooperative pyrimidine tract binding; however, as these proteins are not monodisperse (data not
shown) the Hill coefficients are difficult to interpret. Under these same conditions a His 6 fusion
of U2AF65 had a Kd obs of 300 nM and a Hill coefficient of 1.1. Zamore et al. (1992) report
values of GstU2AF65 binding ranging from 10 nM to 2 tM for wild-type pyrimidine tracts.
These latter affinity measurements were made with RNAs that differ in two respects from
pyr23. First, they contain a branch sequence and 3' splice site, and second, the pyrimidine
tracts tend to be shorter than those of either pyr23 or pyrl4. These measurements were made
with the GstU2AF65 fusion protein rather than the His, fusions used here; glutathione-Stransferase dimerization may also have affected affinity measurements.
110
DIscusSION
We have identified a new member of the U2AF65 family of pyrimidine-tract binding
factors, PUF60. PUF60 was identified in highly purified fractions that, together with U2AF,
restored splicing activity to poly[U] depleted nuclear extracts. Orthologues of the PUF60 are
found in vertebrates (human and mouse) and in the invertebrate Drosophila. The closest
homologue in the budding yeast, S. cerevisiae, is Mud2p, which is structurally similar and
approximately equally distant in sequence similarity to both PUF60 and U2AF65 (data not
shown). PUF60 has the unusual property of forming SDS-resistant dimers. This dimerization
is mediated by a C-terminal RRM-like domain, the PUMP domain. The dissociation constant
for formation of the SDS-resistant dimers was approximately 1 ptM. The PUMP domain
homology is found in several other proteins, some of which have been shown to mediate
protein-protein interactions.
RNA binding activity of PUF60
Like U2AF65, PUF60 was purified from nuclear extracts based on its ability to
associate with poly[U] RNA at high salt concentrations. We subsequently tested the RNA
binding activity of bacterially expressed His 6 PUF60 and found that this protein bound a
pyrimidine-tract RNA of 23 nucleotides with high affinity. This affinity, 130 nM, was
comparable to that of U2AF65, 300 nM, for the same RNA. PUF60 RNA binding to
pyrimidine-tract RNAs of 14, 11 and 7 nucleotides was also tested. PUF60 requires
pyrimidine-tract RNAs of greater than 11 nucleotides for high affinity binding. As the RNAs
tested for binding were short oligonucleotides it is not known what effect, if any, flanking RNA
would have on PUF60's affinity for these short RNAs. U2AF65 has been reported to contact
the branch sequence via its N-terminal SR domain. Interestingly, PUF60 does not have an SR
domain and so may not be able to make similar contacts.
The RNA binding specificity of PUF60 for the pyrimidine tract was tested by competing
PUF60 with RNA homopolymers and heteropolymers. Only polymers containing uridine could
successfully compete for pyrimidine-tract binding. Notably poly(C) could not compete PUF60
binding although both poly(C,U) and poly(U) could. This result suggests that PUF60, like
U2AF65 and unlike PTB, prefers binding uridine-rich sequences, although both appear to
tolerate the presence of cytosine (Singh et aL, 1995). This sequence specificity mimics the
consensus sequence of the pyrimidine tract, which is composed predominantly of uridine
(Roscigno et al., 1993).
111
PUF60 is a U2AF65 homologue
PUF60 is distantly similar to U2AF65; however this homology extends across the entire
length of these two proteins with the exception of the very N-terminal domain. Both proteins
have a relatively non-conserved N-terminal domain, two central RRM domains and a C-terminal
RRM-like PUMP domain. Comparison of the N-terminal domains is informative in that only
PUF60, among these proteins, lacks SR dipeptide repeats. DPUF68, however, has five SRdipeptide repeats in this region. This is reminiscent of the variable number of SR dipeptide
repeats observed between invertebrates and vertebrates in the large subunit of U2AF65 (Kanaar
et al., 1993). Phylogenetic analysis of these proteins and their metazoan and yeast homologues
showed that PUF60 and U2AF65 were approximately equally related to the yeast protein
Mud2p. Mud2p, while not required for viability, does play a central role in 3' splice site
recognition. MUD2 interacts genetically with Ul snRNA and is present in the commitment
complex. Mud2p and U2AF65 are both known to interact with the branch site interacting
protein SF1/BBP (Berglund et al., 1998). PUF60 has also been shown to weakly interact with
SFl/BBP (chapter 2). Mud2 is also known to interact with the splicing factor Prpl 1 (which
adds to the spliceosome coincidentally with U2 snRNP; Abovich et al., 1994), suggesting that
Mud2p plays a role in bringing U2 snRNP to the branch sequence. Analysis of the complete
yeast genome database demonstrates that there are no other PUF60 or U2AF65 homologues. It
is interesting to speculate that PUF60 and U2AF65 may interact with SF1/BBP at different
points of spliceosome assembly. Alternatively, PUF60 may function on a discrete set of introns
and may play a regulatory role. We do not favor this model as PUF60 and U2AF65 show
similar binding affinities and specificities and PUF60 has been shown to be required for
efficient splicing of each intron tested (chapter 2).
The PUMP domain is a subset of the RRM domain family
The highest degree of conservation between PUF60, U2AF65, and Mud2p is found in
the C-terminal PUMP domain. This domain had previously been identified as an RRM-like
domain but has also been recognized as being divergent from the RRM domain sequence
(Birney et al., 1993). The PUF60, U2AF65 and Mud2p C-terminal domains were used to
search the protein database to identify other possible homologues. Several other domains that
previously had been identified as RRM domains, but are more closely related to the PUMP
domain subfamily, were identified. The small subunit of U2AF35 and its homologue
U2AFbpl/Urp, in contrast, had not previously been identified as containing an RRM
homology, but showed significant homology to the PUMP domain.
112
Additional PUMP domain homologues can be found among several proteins that have
previously been identified as RRM homologues: kis/P-CIP2, a kinase that binds the
cytoplasmic tail of the transmembrane protein PAM; Tat-SF1, which binds to the HIV tat-TAR
complex; D 111, an Arabidopsis protein implicated in DNA-damage repair; CUS2, a splicing
factor in S. cerevisiae;and UAP2, which interacts with S. pombe U2AF large subunit. Less
clearly homologous are domains found in PRP24, a splicing factor, Not4, which has been
shown to regulate basal and activated transcription, Nrdl a regulator of pre-mRNA abundance
in yeast (Steinmetz and Brow, 1996), Nop4 and RNA12 which are important for rRNA
maturation, and Ylfl which encodes a GTP binding protein and was identified as a polymersase
III mutant suppressor. The function of these domains is unknown, but it is interesting to note
that all known examples of PUMP domains are in some way associated with RNA metabolism
in the nucleus except for P-CIP2 which may be an accessory factor in Golgi shuttling.
Because the RRM domain is indicative of RNA binding activity, we tested whether the
PUMP domain of PUF60 contributed to RNA binding activity. Surprisingly, deletion of the Cterminal half of this domain had no detectable effect on the RNA binding activity of PUF60,
strongly arguing that the PUMP domain of PUF60, in contrast to that of U2AF65, does not
contribute to the RNA binding activity of the protein. In contrast to the PUMP domain of
PUF60, the PUMP domain of U2AF65 does contribute to RNA binding as deletions of either
RRM domain or the PUMP domain severely effected RNA-binding activity. The PUMP
domain of U2AF65 has recently been shown to interact with SFl/BBP, suggesting that it may
play multiple roles in 3' splice site recognition.
RRM domains are generally found to be RNA binding domains, but exceptions to this
rule have been identified. Perhaps the best known example of an RRM that has no known RNA
binding activity is the second RRM domain of U1A. While UIA is known to bind RNA, it is
the first RRM domain of this protein that is sufficient for the known RNA-binding activities of
this protein (Scherly et al., 1990). We have identified an family of RRM domains that form a
distinct subset of the RRM domain class, the PUMP family. The PUMP domain of PUF60 is
unlikely to have RNA binding activity and is a protein-protein interaction domain. This appears
to be a general feature of the PUMP domain class. Since the second RRM of U1A does not
bind RNA, it was of interest to determine ifU1A is a PUMP domain; surprisingly, it is not
(data not shown). This raises the question of whether RRM domains can generally be assumed
to be RNA-binding domains.
113
The PUMP domain is a protein-protein interaction domain
The PUMP domain was shown by deletion analysis to be responsible for the unusual
SDS-resistant dimerization of PUF60. The fact that dimerization is stable to the addition of
SDS was unexpected but is not without precedent. Bacteriophage P22 tailspike
Endorhanosidase is a protein that is resistant to SDS denaturation (Goldenberg et al., 1982).
Like the RRM domain, and presumably the PUMP domain, the tailspike protein consists
predominantly of B strands (Steinbacher et al., 1994). B-strand containing proteins have been
shown to be remarkably resistant to denaturants. Examples of this are found in amyloid plaques
and Prion protein which are insoluble under most conditions and are composed predominantly
of B sheets (Meyer et al., 1986; Pan et al., 1993).
Denaturation of protein by SDS is believed to occur by disruption of the proteins'
hydrophobic core, and it is interesting to speculate that the affinity of the PUF60 PUMP domain
dimerization may have only limited contribution by a hydrophobic pocket. In support of this
idea it has recently been shown that a B sheet can form stable, non-aggregating structures in the
absence of a hydrophobic core (Pham et al., 1998).
PUMP domain interactions:
other proteins
It is interesting to note that the region of U2AF35 and Urp that are implicated in binding
U2AF65 contain a PUMP domain (Tronchere et al., 1997; Wentz and Potashkin, 1996; Zhang
et al., 1992). This domain has previously been described as the H2 homologous region of Urp
and U2AF35. The heterodimerization reaction between U2AF65 and U2AF35 is known to be
very stable. U2AF35 remains bound to U2AF65 in the presence of at least 2 M KCl and coelutes from poly[U] Sepharose with U2AF65 in 2M guanidine. This heterodimerization
reaction is different from the homodimerization reaction of PUF60 in that the U2AF65 PUMP
domain is not involved. Instead, a small region of U2AF65, N-terminal to the first RRM, is
required for this interaction. This heterodimerization interaction is reminiscent of the
heterodimerization between the short cytoplasmic domain of PAM (Eipper et al., 1993), the PCIP2 ligand, and the PUMP domain protein P-CIP2 (Alam et al., 1996). Deletion analysis of
the C-terminal domain of PAM implicates a 35 amino acid region as the P-CIP2 binding site
(Alam et al., 1996). Comparison of the region of the large subunit of U2AF from vertebrates,
Drosophilaand the fission yeast S. pombe required for small subunit binding together with the
C-terminal domain of PAM, required for the interaction of the PUMP domain of P-CIP2 with
PAM, suggests that the PUMP domains of the small subunits of U2AF and of P-CIP2 bind the
peptide motif glycine, phenylalanine or tyrosine, glutamate or aspartate, X, valine or leucine,
114
serine or threonine (G, F/Y, E/D, X, V/I, T/S). It will be of interest to determine if this is the
PUMP domain interaction peptide of these proteins and if this motif is more generally found to
be a PUMP domain interaction motif.
115
METHODS AND MATERIALS:
Identification of ESTs, sequencing, and alignments
The human EST clone 33065 (accession numbers R43914 and R18804) was sequenced
by oligo directed automated dideoxy sequencing (ABI) at MIT Biopolymers. The Drosophila
clone was identified by a BLAST search of the DrosophilaEST database and clone CK001 12
was sequenced. Management of the sequencing project was by DNA* (Lasergene). Murine
ESTs sequences from the following accession numbers were aligned to generate the murine
PUF60 sequence: AA139823, W33907, W29734, AA086867, AA068510, R75215,
AA062267, W29345, AA023679, 11166178, AA002661, AA109731, W98367, W78451,
AA073303, W14352, W64149, AA075822, AA184449, AA105954, W44199, AA003590,
W34769, W82745, W65772, W65505, AA175197, AA182190, and R75214. Alignments of
the PUF60, U2AF65, and Mud2p protein sequence was by Clustal W. Identification of new
PUMP domains was achieved by identifying blocks of similarity in the C-terminal domains of
PUF60, U2AF65, and Mud2p using BLOCKMAKER. The resulting blocks were used to
search the non-redundant database using the MAST program. Alignment of the PUMP domains
and the compiled sequences of the RRM domain (PFAM) was by Clustal W. The PUMP and
PFAM seed RRM dendrogram was displayed by TreeViewPPC (Page, 1996).
Expression and purification of His 6PUF60 and His6PUF60AC
pET15-His 6PUF60 and pET15-His 6PUF60AC were transfected into BL21 [DE3] cells
and grown to mid-log phase where they were induced with 1 mM IPTG. Induced cells were
harvested after 7 hours by centrifugation. Cells were lysed in QBA (6 M Guanidine HC1, 10
mM 2-mercaptoethanol, 100 mM NaPO 4, 10 mM Tris, pH 8.0) and sonicated to shear the
DNA. Lysate was spun 25,000g for 15 minutes and the pellet was discarded. Lysate was
loaded onto a Ni-NTA Agarose (Qiagen) gel according to the manufacturers instructions,
washed with QBB (8.0 M Urea, 10 mM 2-mercaptoethanol, 100 mM NaPO 4 , 10 mM Tris, pH
6.3 with HC1) and QBC (QBB at pH 4.5). The protein was eluted in QBE (QBB at pH 3.7)
and the pH was brought to 7.5 with the addition of Tris base. Eluted protein was refolded by
applying to fresh Ni-NTA Agarose, washing the column with a linear gradient of 6 M Urea to 1
M Urea in QR (500 mM KC1, 20% glycerol, 20 mM HEPES, 10 mM 2-mercaptoethanol, pH
7.9) and eluted with 250 mM imidazole, 50 mM EDTA in 1 M Urea QR. Protein is dialyzed
against 100 mM KC1, 20 mM HEPES (pH 7.9) 0.2 mM EDTA, 20% glycerol, 40 mM DTT
and stored frozen. Yield on refolding was 10 and 18% respectively, 4.5 and 4.7 mg was
116
obtained from 1 liter of cultured cells. The purified, refolded protein was polished on Mono Q
Sepharose (Pharmacia) on a linear gradient of 350 mM KCl to 500 mM KCl in 20 mM Tris pH
7.5 (at room temperature), 0.2 mM EDTA and 10% glycerol. Peak fractions were dialyzed
against 0.1 M KC1, 20 mM HEPES, 0.2 mM EDTA, 0.05% Np40, 20% glycerol, pH7.9,
frozen on liquid nitrogen and stored at -800 C.
Translation in vitro
Plasmids for translation in vitro were pCITE 4 with the following fragments of PUF60
cloned. Translation of PUF60 and the deletion series was in the coupled transcription and
translation system, TnT (Promega), and was labeled by incorporation of 35S-Methionine
Dimerization assay
All samples were made 1 x with SDS-electrophoresis buffer (1% SDS, 62.5 mM TrisCl
pH 6.8, 10% glycerol and 180 mM 2-mercaptoethanol) and boiled for six minutes before
resolution on BioRad 4-15% ReadyGels. For figure 3B the proteins were diluted serially,
quantitation of this and similar gels gave results similar to that shown in figure 3D. For figure
3D two quantities of His 6PUF60 were used and the concentration was varied by changing the
volume. This allowed better quantitation of the small amount of dimer formed at low
concentrations. Protein concentration was determined by Bradford Assay (BioRad). Gels were
stained with SYPRO Red and scanned with a Storm 860 (Molecular Dynamics). Binding
curves were generated by determining the fraction of dimer and monomer at each concentration
and plotting fraction of dimer and monomer as a function of protein concentration. Data was fit
to the Hill equation using Kaleidagraph: 0 = 1/(I+(Kd /[A]) n where 0 is fraction of dimer or
monomer, Kd is the apparent dissociation constant, [A] is the protein concentration, and n is the
Hill coefficient.
RNA binding assay
RNA binding were carried out in 83 mM KC1, 60 gtM EDTA, 3 mM MgC 2, 26 mM
HEPES pH 7.9, 2.5 mM dithiothreitol, 6% glycerol and 10 tg/ml BSA (New England
Biolabs). Binding reactions were run out on an 8% (60:1) polyacrylamide gel in 50 mM Tris,
50 mM glycine, 3 mM MgAcetate at 10 V/cm. Dried gels were exposed to Phosphorimager
plates (Molecular Dynamics), and binding was quantitated by determining the volume of each
band, corrected for background. Fraction bound at each concentration was determined for three
identical experiments. Protein concentration was determined by Bradford Assay (BioRad).
117
Binding curves were generated by determining the fraction bound at each concentration and
plotting the fraction bound as a function of protein concentration. Data was fit to the Hill
equation using Kaleidagraph as for the protein dimerization assay.
118
REFERENCES
Abovich, N., Liao, X. C., and Rosbash, M. (1994). The yeast MUD2 protein: an interaction
with PRP 11 defines a bridge between commitment complexes and U2 snRNP addition. Genes
Dev 8, 843-54.
Alam, M. R., Caldwell, B. D., Johnson, R. C., Darlington, D. N., Mains, R. E., and Eipper,
B. A. (1996). Novel proteins that interact with the COOH-terminal cytosolic routing
determinants of an integral membrane peptide-processing enzyme. J Biol Chem 271, 28636-40.
Avis, J. M., Allain, F. H., Howe, P. W., Varani, G., Nagai, K., and Neuhaus, D. (1996).
Solution structure of the N-terminal RNP domain of U1A protein: the role of C-terminal
residues in structure stability and RNA binding. J Mol Biol 257, 398-411.
Bailey, T. L., and Gribskov, M. (1998). Combining evidence using p-values: application to
sequence homology searches. Bioinformatics 14, 48-54.
Bailey, T. L., and Gribskov, M. (1997). Score distributions for simultaneous matching to
multiple motifs. J Comput Biol 4, 45-59.
Berglund, J. A., Abovich, N., and Rosbash, M. (1998). A cooperative interaction between
U2AF65 and mBBP/SF1 facilitates branchpoint region recognition. Genes Dev 12, 858-67.
Birney, E., Kumar, S., and Krainer, A. R. (1993). Analysis of the RNA-recognition motif and
RS and RGG domains: conservation in metazoan pre-mRNA splicing factors. Nucleic Acids
Res 21, 5803-16.
Creighton, T. E. (1984). Proteins: Structures and Molecular Principles (New York: W. H.
Freeman and Company).
Eipper, B. A., Milgram, S. L., Husten, E. J., Yun, H. Y., and Mains, R. E. (1993).
Peptidylglycine alpha-amidating monooxygenase: a multifunctional protein with catalytic,
processing, and routing domains. Protein Sci 2, 489-97.
Goldenberg, D. P., Berget, P. B., and King, J. (1982). Maturation of the tail spike
endorhamnosidase of Salmonella phage P22. J Biol Chem 257, 7864-71.
Henikoff, S., Henikoff, J. G., Alford, W. J., and Pietrokovski, S. (1995). Automated
construction and graphical presentation of protein blocks from unaligned sequences. Gene 163,
GC 17-26.
Hoffman, D. W., Query, C. C., Golden, B. L., White, S. W., and Keene, J. D. (1991).
RNA-binding domain of the A protein component of the Ul small nuclear ribonucleoprotein
119
analyzed by NMR spectroscopy is structurally similar to ribosomal proteins. Proc Natl Acad Sci
U S A 88, 2495-9.
Imai, H., Chan, E. K., Kiyosawa, K., Fu, X. D., and Tan, E. M. (1993). Novel nuclear
autoantigen with splicing factor motifs identified with antibody from hepatocellular carcinoma. J
Clin Invest 92, 2419-26.
Kanaar, R., Roche, S. E., Beall, E. L., Green, M. R., and Rio, D. C. (1993). The conserved
pre-mRNA splicing factor U2AF from Drosophila:requirement for viability. Science 262, 56973
Kenan, D. J., Query, C. C., and Keene, J. D. (1991). RNA recognition: towards identifying
determinants of specificity. Trends Biochem Sci 16, 214-20.
MacMillan, A. M., McCaw, P. S., Crispino, J. D., and Sharp, P. A. (1997). SC35-mediated
reconstitution of splicing in U2AF-depleted nuclear extract. Proc Natl Acad Sci U S A 94, 1336.
Meyer, R. K., McKinley, M. P., Bowman, K. A., Braunfeld, M. B., Barry, R. A., and
Prusiner, S. B. (1986). Separation and properties of cellular and scrapie prion proteins. Proc
Natl Acad Sci U S A 83, 2310-4.
Nagai, K., Oubridge, C., Ito, N., Avis, J., and Evans, P. (1995). The RNP domain: a
sequence-specific RNA-binding domain involved in processing and transport of RNA. Trends
Biochem Sci 20, 235-40.
Nagai, K., Oubridge, C., Ito, N., Jessen, T. H., Avis, J., and Evans, P. (1995). Crystal
structure of the U1A spliceosomal protein complexed with its cognate RNA hairpin. Nucleic
Acids Symp Ser, 1-2.
Nagai, K., Oubridge, C., Jessen, T. H., Li, J., and Evans, P. R. (1990). Crystal structure of
the RNA-binding domain of the Ul small nuclear ribonucleoprotein A. Nature 348, 515-520.
Oubridge, C., Ito, N., Evans, P. R., Teo, C. H., and Nagai, K. (1994). Crystal structure at
1.92 A resolution of the RNA-binding domain of the U1A spliceosomal protein complexed with
an RNA hairpin. Nature 372, 432-8.
Page, R. D. M. (1996). TREEVIEW: An application to display phylogenetic trees on personal
computers. Computer Applications in the Biosciences 12, 357-358.
Pan, K. M., Baldwin, M., Nguyen, J., Gasset, M., Serban, A., Groth, D., Mehlhorn, I.,
Huang, Z., Fletterick, R. J., Cohen, F. E., and et al. (1993). Conversion of alpha-helices into
beta-sheets features in the formation of the scrapie prion proteins. Proc Natl Acad Sci U S A 90,
10962-6.
120
Pham, T. N., Koide, A., and Koide, S. (1998). A stable single-layer beta-sheet without a
hydrophobic core. Nat Struct Biol 5, 115-9.
Roscigno, R. F., Weiner, M., and Garcia-Blanco, M. A. (1993). A mutational analysis of the
polypyrimidine tract of introns. Effects of sequence differences in pyrimidine tracts on splicing.
J Biol Chem 268, 11222-9.
Scherly, D., Dathan, N. A., Boelens, W., van Venrooij, W. J., and Mattaj, I. W. (1990). The
U2B" RNP motif as a site of protein-protein interaction. EMBO J 9, 3675-81.
Singh, R., Valcarcel, J., and Green, M. R. (1995). Distinct binding specificities and functions
of higher eukaryotic polypyrimidine tract-binding proteins. Science 268, 1173-6
Sonnhammer, E. L., Eddy, S. R., Birney, E., Bateman, A., and Durbin, R. (1998). PFAM:
multiple sequence alignments and HMM-profiles of protein domains. Nucleic Acids Res 26,
320-2.
Steinbacher, S., Seckler, R., Miller, S., Steipe, B., Huber, R., and Reinemer, P. (1994).
Crystal structure of P22 tailspike protein: interdigitated subunits in a thermostable trimer.
Science 265, 383-6.
Steinmetz, E. J., and Brow, D. A. (1996). Repression of gene expression by an exogenous
sequence element acting in concert with a heterogeneous nuclear ribonucleoprotein-like protein,
Nrdl, and the putative helicase Senl. Mol Cell Biol 16, 6993-7003.
Tang, J., and Rosbash, M. (1996). Characterization of yeast Ul snRNP A protein:
identification of the N- terminal RNA binding domain (RBD) binding site and evidence that the
C- terminal RBD functions in splicing. RNA 2, 1058-70
Thompson, J. D., Higgins, D. G., and Gibson, T. J. (1994). CLUSTAL W: improving the
sensitivity of progressive multiple sequence alignment through sequence weighting, positionspecific gap penalties and weight matrix choice. Nucleic Acids Res 22, 4673-80.
Tronchere, H., Wang, J., and Fu, X. D. (1997). A protein related to splicing factor U2AF35
that interacts with U2AF65 and SR proteins in splicing of pre-mRNA. Nature 388, 397-400.
Wells, S. E., Neville, M., Haynes, M., Wang, J., Igel, H., and Ares, M., Jr. (1996). CUSl,
a suppressor of cold-sensitive U2 snRNA mutations, is a novel yeast splicing factor
homologous to human SAP 145. Genes Dev 10, 220-32.
Wentz, H. K., and Potashkin, J. (1996). The small subunit of the splicing factor U2AF is
conserved in fission yeast. Nucleic Acids Res 24, 1849-54.
Zhang, M., Zamore, P. D., Carmo, F. M., Lamond, A. I., and Green, M. R. (1992). Cloning
and intracellular localization of the U2 small nuclear ribonucleoprotein auxiliary factor small
subunit. Proc Natl Acad Sci U S A 89, 8769-73.
121
FIGURE LEGENDS
Figure 1. Sequence of PUF60 and DPUF68 and comparison of PUF60,
U2AF65 and DPUF68.
A. Alignment of HPUF60 and DPUF68, with RRM and PUMP domain alignments of
U2AF65 and Mud2p. The middle line shows identity (*) and similarity (:) between HPUF60
and DPUF68, underlined amino acids sequence are peptides identified by amino acid
sequencing.
B, C, and D. The two RRM domains and PUMP domain of HPUF60, DPUF68, Mud2p and
HU2AF65 were aligned separately and residues that are absolutely conserved between the four
proteins are shown in white on a black background while residues conserved between three of
the proteins are shown with gray background. The locations of the RNP 1 and RNP2 motifs is
indicated for each RRM and the PUMP domain.
122
A
HPUF60
MAT---ATIALQVNGQQGGGSEPAAAAAVVAAG --- DKWKP--POGTDSIK ---*
*:
:
:
:
**:
: *:
:
*
:*
*
*
MENGQ
47
*
::
:*
DPUF68
MGSNDRASRSPRSDDQREISDMPATKRTRSDSGKSTDSKIPYLSQPLYDLKQTGDVKFGP
60
HPUF60
ST-AAKLG ----- LPPLTPEOOEALOKAKKYAMEOSIKSVLVKQTIAHQQQQLTNLQMAA
102
*
**
:*
**
*:
**::
: ************
**:******:***
DPUF68
GTRSALLGLLGGALPKLSSEQHDLVSKAKKYAMEQSIKMVLMKQTLAHQQQQLA ------
114
HPUF60
VTMGFGDPLSPLQSMAAQRQRALAIMCRVYVGSIYYELGEDTIRQAFAPFGPIKSIDMSW
162
DPUF68
----------- TQRTQVQRQQALALMCRVYVGSISFELKEDTIRVAFTPFGPIKSINMSW
163
HPUF60
DSVTMKHKGFAFVEYEVPEAAOLALEQMNSVMLGGRNIKVGRPSNIGQAQPIIDQLAEEA
222
DPUF68
DPITQKHKGFAFVEYEIPEGAQLALEQMNGALMGGRNIKVGRPSNMPQAQQVIDEVQEEA
223
HPUF60
RAFNRIYVASVHODLSDDDIKSVFEAFGKIKSCTLARDPTTGKHKGYGFIEYEKAOSSOD
282
*
***:.**********
::*********
*
*
**:
*********
::
: *:
DPUF68
KSFNRIYVASIHPDLSEEDIKSVFEAFGPILYCKLAQGTSLHTHKGYGFIEYANKQAMDE
283
HPUF60
AVSSMNLFDLGGOYLRVGKAVTPPMPLLTPATPGGLPPAAAVAAAAATAKITAQEAVAGA
342
**
******W***
**t******
*
*.*
**
*************
*
:***
DPUF68
AIASMNLFDLGGQLLRVGRSITPPNALACPTTNSTMPTAAAVAAAAATAKIQALDAVASN
343
HPUF60
AVLG-TLGTP ----------- G-LVS----------PALTLAQPLGTLPQAVMAAQAP-
377
AtW*
:
**
*
.**
***.
*
*
**
*:
*
.*
DPUF68
AVLGLSQNTPVMAAGAVVTKVGAMPVVSAATSAAALHPALAQAAP-ALLPPGIFQAPTPV
402
HPUF60
----- GVITG------ VTPARPP--------IPVTIPSVGV-VNPILAS---- PPTLG--
411
**
*
:*
**
*:
:*:*:
***
: *
*
:*:
*
DPUF68
APSLLGVPAGLQXLQAVVPTLPPPALLATPTLPMTVGGVGVGLVPTVATLAGAEASKGAA
462
HPUF60
------------------ LLE--PKK-EKEEEELFPESERP---EMLSEOEHMSISGSSA
447
*
*
*
: *
**::***
:*****
*
**
DPUF68
AAAALSAAANNAAVTAANLSENIKKAHEKQQEELQKKLMDEGDVQTLQQQENMSIKGQSA
522
HPUF60
RHMVMQKLLRKQESTVMVLRNMVDPKDIDDDLEGEVTEECGKFGAVNRVIIYQEKQGEEE
507
*::***:*:*
:*
*::*****
*:*:*:
*:
*:
***
***:*
****::***
*:*
DPUF68
RQLVMQRLMRPVDSRVIILRNMVGPEDVDETLQEEIQEECSKFGTVSRVIIFNEKQTENE
582
HPUF60
D---AEIIVKIFVEFSIASETHKAIOALNGRWFAGRKVVAEVYDOERFDNSDLSA
559
DPUF68
DDDEAEIIVKIFVEFSAGAEAMRGKEALDGRFFGGRRVVAELYDQGIFDQGDLSG
DPUF68
DDDEAEIIVK****IFVEFSAGAEAMR***
*
GKEALDGRFFGGRRVVAEL**:**YDQGIF*
******QGDLSG***
**
***
637
B
RNP2
111%
RNP1
-%9%
HPUF60
DPUF68
HU2AF65
MUD2P
C
RNP2
HPUF60 1
DPUF68 ]
U2AF65 I
MUD2P
D
D
HPUF 60
DPUF68
HU2AF65
MUD2P
10
ITEORRMDMKI
I.
RLFIT
30
40
LPFGP
S DM.WDS~TMKKG....
rPFGP KS
.WDP TQ KG ....
AQMRGG
TQ PGNP LAVQINQDKN
KTESEDFK
.NFY GEGIPD ....
60
70
80
90
100
EQMNSNRNG-GSNIaGAQPI DQLAEEA
:VP..EAQ
.EQMNBRN
SNMP QQVDM.QEEA
IP..E
Q
S
P...
AFDGDY.PLP
LS...VDETTQ
QLDHnFCRGT
RSFF4NKTFD WRNDY
ISQICST
RNP1
nA
30
40
fKIKSCT
PTTGKH
:A
LY C KQSLHTH
P&rAF N
TGLS
KCS.SNTNN
EFTKC
20
30
70
80
90
GQYPPMPTPATP
PPINACPTTN
D GQL
G GMQDK
VGAK TLVSP.
SLID
PYK
40
50
NR IIIDYEIEKQGEEED....
KPNDG..
RNP1
60
EJ
DLE
T ....
LQE
....
TSRIFEEKQTENEDDD.EEIl
EYEIVDVRDIS
KSIEDPRPVDGVEVPG .... CG..
KT... LKYSI
DTK CPGVDYRLNFENLSGIC
T ...........
so
90
100
110
ELY QGI
Q
G..
lFW...
HR
D
TKYC PIE
MMMLS
ED
T MTYI
Figure 2.
The PUMP domain is a distinct subset of the RRM domain family.
A. Alignment of the PUMP domain. The top panel is an alignment of the best matches to the
PUMP family consensus. The PUMP consensus and RRM consensus is shown at the bottom
of the figure. The sequence of the first RRM of U1A is shown on the bottom line.
B. A dendrogram showing that the PUMP family represents a distinct subset of the RRM
domain family. Alignment of 40 RRM domains with the PUMP domains. The RRM seed
database from PFAM was aligned with the PUMP family sequences and displayed as a
dendrogram. The PUMP family is boxed and is consistently found to form a distinct group
from the set of RRMs in the alignment.
125
A
RNP
HPUF60
DPUF68
ATD111
SPUAP2P
TAT-SF1
HU2AF65
DU2AF50
CeU2AF
SpU2AF
HU2AF35
DU2AF38
SPU2AF23
HURP
ScMud2p
SCCUS2P
YMC7_CA32
YHS7_YE159
NRD1_YE341
LU15_HU233
YAX9_SC137
NOP4_YE149
RN12_YE200
YG3Q_ YE22
EECG ----OPKDIDD----- DL
TL
QEECS -----GPEDVDE--GECA----PGQVDD----EL D
-LEELDKTP--ELI
KDDITEEAE- FHPMDFEDD---PL
REDLRVECS-ECS----5PEELLDDEE-YEEI
rPDELRDEEE-YEDI
KEECT----1EDELKRDDE-YEEI
ECS----S. TQFS-----rGDEIMDVQE-YEDI
6DVEMQEH---YDEF
TEMEE--CED----PDEEMQEH---YDNF
CEFS-----QRELAEQ---FD
LEYSEEE---TYQQ
EDVLPEFK-KY------PLDLKDET--FITE
LEGCE-----S, NDDIND-------I
SS-------PEDVFDN------KQ
EN-------
LGVNQE------- N
K---------
LNMKEW ------- D
S---------
PHTVVD ------- S
F
KPVTTNG-------S R--------I
FG--------PWSCRDP------VK
S FR-------GPALTEE-------E
S
PEWNQD-------I
1
--- IASW K
QEKQGE--EED-----AEII
EKQTE--NEDDDE--AEII'
EITEPN-------- FPVHEA
-------KE-------PD
--------DR------HPD
RPVDG---------VEVPGC
RPIEG---------VEVPGC
RPYED---------LPVPGV
RSIGTR-------- NSGLGT
DNLGD------------HLV
DNLGD------------HLV
DNVGD------------HLV
CNLEP------------HLR
R-R--
3------ NNE
RPE
---
--DPE
--- SVF
--- SVL
--- DLPS----GS-----AT
---N-------------SR
IKDKQTQQ ---------NR
-S
--- DLS
--- KIS
--- SF
S----------ETG----DL
-- RKRD-----GK-----LC
---
ITK
Yi
1(
I
VDGTT
R-R-R--
R---- DIR
--- REE
--- N
--- YEE
--- SEE
--- TL
-::NN
-SP
--- TTQ
--- SRH
QPGVDYR- -- LNFENLASGA
----------- PN------ K
FD--------FLR-----SF
----------
SDLSAGDLSGKNELAPQKSGKHT
QVEETSR
oDPDS RRDFW"DPDK RREF-DVDK
QF----
DQER
IDQGI
DEEK
DGK
R--
TAA-----NN
ND------PR-----TG
C-ir
GEC
3PVTD
CCRQ
3PVTD RECCRQ
R-R--
N
A
Q1
A
K
SPVTD
k-iR---D--
Q
Q.
I
is.
-YLTS
EACCRQ
.'PVTR
ICGL
DEDD
MMEATQ
jGDEN SSTSDKN
ANSQML
kQRI
SY--kSDES
QHGY3PRDC
LVLSDG
AKSA
NHPK PNAEHL
DYKKA
AVQKN
,NIRR LVSNFF
BPNY
LRETAN
iI-I--
PUMP consensus
RRM consensus
RU1AHU12
NE-KIKKDE--LKKS
-- P1-RNP 2
loopl
FS---------
--- al---
loop 2
RS---------LK-----MR
loop 3
--
---
EVS
P3 -- loop4----t2-----
RNP
1
-PZRPQkKTDS
loop 5
--
)4-
IAKMKG
~,
PUB1 YEAST/163-234
PABP DROME/4-75
PES4 YEAST/93-164
EWS
fus/tls-H.s.
0.1
Figure 3.
HPUF60 forms SDS-resistant dimers.
A. Translation in vitro of the full length PUF60 yields a predominant band of 60 kD and a band
of 130 kD (lane 1); an N-terminal truncation of HPUF60 yields a 55 kD band and a 120 kD
band (lane 3); co-translation of both yields the 60 and 55 kD products as well as products of
120, 125 and 130 kD (lane 2).
B. Bacterially expressed His6PUF60 also forms SDS-resistant dimers. Serial two-fold
dilutions of the protein (lanes 1-6) showed that monomeric His 6PUF60 could be chased into
dimeric form at high concentrations. A small C-terminal deletion of PUF60, His6PUF60AC,
prepared identically to His6PUF60, does not form SDS-resistant dimers (lanes 9 and 10).
C. Plot of the fraction PUF60 in monomer and dimer forms upon boiling in SDS. In order to
more accurately measure the concentration of dimer at low protein concentrations for this
experiment two different quantities of protein were used and the volume was varied to vary the
concentration over the range shown. Similar results are obtained by using the method shown in
A and B.
D. The domain responsible for SDS-resistant dimerization was mapped by expression of
PUF60 fragments, as indicated.
128
I
_
_
_
__
A
PUF60
+
PUF60AN
+
+
DIMERS
MONOMERS
PUF60
PUF60AN
4
RRM1 I RRM2
PUMP
RRM2
PUMP
RRMF
B
His 6 PUF60
His 6 PUF60
'
,'
O*
O*
'
r*
--:':::::
::::::::::::::::L::::i::~
:::X::
:.::.:i::
:~:;:::i::::::
j_
4,
O*
His 6 PUF60 dimers
7
3
4
5
6
150 kD
100 kD
75 kD
:::
:;:l
.;:::::,:u
:
::::::::::'::::::
::::::
::":':':
His 6 PUF60
2
r*k
:::::::::
::
-::::: : :::::
:-~
::"':":':::'::::
1
His 6 PUF60AC
8
50 kD
9
10
C
0.8 -i
1.8
0.6 -
0.4 -
0.2 -
0 I
I
I
110 -
210-6
I
I
310 -s
410-6
Concentration of HisPUF60 (M)
y = m2/(1+(1 .5e-6/MO)^m1)
Error
Value
ml
1.5
0.1
m2
0.97
0.025
Chisq
0.0057
NA
R
1
NA
5 10 -6
1l
+I
I
Ir
I
!l03 3
+
!ot 3
+
!1oo 3
I
''
I r
I
!lO 3
I
Lu U
Izwuti
I
dwnd
I
Figure 4.
RNA binding activity of His 6PUF60.
A. Binding of His 6PUF60 to pyrimidine-tract RNAs of different lengths was tested.
Pyrimidine-tract RNAs of various lengths were incubated with a titration of PUF60 protein, and
complexes formed were resolved by native gel electrophoresis. Complexes formed on the
longer and not the shorter RNAs.
B. Specificity ofPUF60 RNA binding was determined by competing His 6PUF60 binding with
cold competitor RNA homo- and heteropolymers. The specificity of PUF60 binding was tested
by binding PUF60 to pyr23 in the presence of varying concentrations of homo- and
heteropolymeric RNAs. Added competitor RNA is x 0.1 ng.
C. Comparison of the binding of His 6PUF60 with His 6PUF60AC for three independent
experiments. A plot of three replicates of a PUF60 binding reaction to pyr23.
133
A
His6PUF60
o0p
0
pyr23
0o
0
pyrl4
pyr11
pyr7
! ',i
WOW
'i!
...........
BOUND
FREE
1 2 3
4 5 6
7 8 9101112
13 14 15 16 17 18
19 20 21 22 23 24
:-:-::
+50000
+5000
+500
"
0
'a
%z
)
C
C
"
0
>
0
0
S50000
+0 +0
+50000
+5000
+500
+50
+5
, 50000
+0
+500
+50
+5
S50000
+0
+ 50000
' 50000
+0
+50000
g
+5000
+500
..!iii:::ri
...
......
+50
+5
0
C
"
' 50000
C
"
0
+0
+50000
+5000
+500
+50
+5
+50000
+5000
+500
+50
+5
S50000
+0
0
mj (0.1 ng)
C COMPETITOR
-o
H
C
1
0.8
0.6
0.4
0.2 -
0
10-8
10-6
10-7
Concentration (M)
10-s
CHAPTER 4:
SC35 MEDIATED RECONSTITUTION OF
SPLICING IN U2AF-DEPLETED NUCLEAR EXTRACT
Andrew M. MacMillan, Patrick S. McCaw, John D. Crispino, and Phillip A. Sharp*
Center for Cancer Research and Department of Biology
Massachusetts Institute of Technology
Cambridge, Massachusetts
02139-4307 USA
This chapter was published originally in the Proceedings of the National Academy of
Sciences, volume 94, pp. 133-6.
137
ABSTRACT
Assembly of the mammalian spliceosome is known to proceed in an ordered
fashion through several discrete complexes, but the mechanism of this assembly process
may not be universal. In an early step, pre-mRNAs are committed to the splicing pathway
through association with U 1 snRNP and non-snRNP splicing factors including U2AF and
members of the SR protein family. As a means of studying the steps of spliceosome
assembly, we have prepared HeLa nuclear extracts specifically depleted of the splicing
factor U2AF. Surprisingly, the SR protein SC35 can functionally substitute for U2AF 65 in
the reconstitution of pre-mRNA splicing in U2AF-depleted extracts. This reconstitution is
substrate-specific and is reminiscent of the SC35 mediated reconstitution of splicing in
extracts depleted of Ul snRNP. However, SC35 reconstitution of splicing in U2AFdepleted extracts is dependent on the presence of functional Ul snRNP. These
observations suggest that there are at least three distinguishable mechanisms for the binding
of U2 snRNP to the pre-mRNA including U2AF dependent and independent pathways.
138
INTRODUCTION
Pre-mRNA splicing occurs via two sequential transesterification reactions in a 60S
complex known as the spliceosome which assembles on the pre-mRNA substrate in an
ordered fashion through several discrete complexes (E, A, B, C; ref. 1). The spliceosome
includes the small nuclear ribonucleoprotein (snRNP) particles U1, U2, U4/6, and U5, as
well as associated splicing factors (2-4). Commitment of a pre-mRNA substrate to
assembly of the spliceosome involves the ATP-independent formation of the E (early) or
commitment complex (5-8). This complex, in the mammalian system, contains U1 snRNP
as well as non-snRNP protein factors including the U2 auxiliary factor, U2AF, and
members of the SR protein family (8).
SR proteins contain extensive serine/arginine (SR) repeats and a subset contain
RNA recognition motifs (RRM); the predominant members of the family are conserved
from Drosophilato humans. Many SR proteins are important splicing factors which
function in both constitutive and alternative RNA splicing (9). SR proteins containing
RRMs display modest affinity and sequence specificity in their association with RNA and
probably bind cooperatively in association with other factors including other SR proteins
(9). SR family members associate with pre-mRNAs early in spliceosomal assembly (10,
11) and this association may persist through the chemistry of splicing (12). It has been
suggested that SR proteins are required for specific transitions during the course of
spliceosomal assembly such as the progression from A to B complex (13). The 35 kD SR
protein SC35 has been reported to stimulate E complex formation (8) and has been shown
to be associated with a complex formed at the 3' end of the intron at early stages in
spliceosome assembly (14).
Conserved sequence elements at the 5' and 3' splice sites and in the branch
sequence and pyrimidine tract of the pre-mRNA direct formation of the spliceosome (3, 4).
In particular, recognition of the pyrimidine tract is required early in the formation of
commitment complexes. A number ofpolypeptides have been reported to bind specifically
to the pyrimidine tract including the U2 auxiliary factor, U2AF (15-18).
The splicing factor U2AF was first identified as an activity required for the stable
association of U2 snRNP with the pre-mRNA branch site in the formation of the A
complex (15). U2AF is a heterodimer consisting of both a large (65 kD) and a small (35
kD) subunit (18). The large subunit, U2AF 65 , is an essential splicing factor containing an
N-terminal SR domain and three C-terminal RRM domains. U2AF 65 binds with avidity to
139
the pyrimidine tract while U2AF35 is in turn tightly associated with the larger sub-unit
through protein-protein interactions (17, 18).
The interactions between the various components of the commitment complex and
their precise role in progression through spliceosomal assembly remain to be determined.
U2AF has been detected in affinity selected commitment (E) complexes isolated by gel
filtration (8). Although U2AF is clearly important for the transition from E to A complex,
it'has not been found to be stably associated with either the A or B/C complexes (19). In
the commitment complex, U2AF is probably bound to the pyrimidine tract while Ul
snRNP is associated with the 5' splice site. Far Western analysis has suggested that the
SR protein SC35 is associated with both the U1 snRNP associated factor U1-70K and
U2AF through interactions with the small sub-unit U2AF 35 (20). Thus, SC35 may
function as a bridge across the 5' and 3' splice sites.
In order to study the mechanism of spliceosomal assembly, we have prepared HeLa
nuclear extracts depleted of the splicing factor U2AF and reconstituted splicing of premRNA substrates by the combination of both column fractions and purified recombinant
splicing proteins. Interestingly, the SR protein SC35 can functionally substitute for
U2AF 65 in this reconstitution in a manner which is both substrate specific and dependent
on the presence of functional Ul snRNP.
140
MATERIALS AND METHODS
RNA Transcription
The PIP85.A RNA pre-mRNA substrate was transcribed from plasmid pPIP85.A
(22). The PIPPG pre-mRNA substrate is a chimera consisting of the 5' portion of
PIP85.A and the 3' portion of 13-globin (23). It was transcribed from a template
constructed by ligating the PCR product representing P-globin sequences between +252
and +386 (which include the last 92 nucleotides of the intron and the complete 3' exon) to
pPIP85.A digested with Xba I and Hind III.
Nuclear Extracts
Nuclear extracts were prepared from HeLa cells as described by Dignam et al. (24).
Extracts depleted of poly[U]-binding proteins including U2AF65 (P.S. McCaw & P.A.
Sharp, chapter 2) were prepared by dialyzing nuclear extract directly into 1 M KCl/buffer D
(20 mM Hepes, pH 7.9, 20% glycerol, 0.2 mM EDTA, 0.05% NP-40, 0.5 mM DTT).
The resulting extract was passed over a poly[U]-Sepharose 4B column (Pharmacia) at 0.1
ml/min and subsequently dialyzed against 0.1 M KCl/buffer D. After washing this column
with 1 M KCl/buffer D, the column was eluted with buffer D containing 2 M KCl and the
eluate was dialyzed against 0.1 M KC1. Combination of the poly[U]-depleted nuclear
extract with the 2M KCl eluate gave an extract, AU2AF NE, specifically depleted of U2AF
activity. The extent of U2AF depletion in this extract was assayed by Western analysis
with a-U2AF65 antibody.
U2AF and SC35 Preparation
Recombinant His6-tagged U2AF 6 5 was prepared from an insoluble E. coli lysate
and was further purified by a 60% ammonium sulfate precipitation (P.S. McCaw & P.A.
Sharp, chapter 2).
SC35 was purified as described elsewhere (10) from baculovirus infected Hi5
insect cells. The protein recovered from phenyl-Sepharose chromatography was treated
with micrococcal nuclease, concentrated by precipitation with 20 mM MgCl 2 , and then
resuspended in buffer D.
141
Splicing Assays
Splicing reactions (25 tl) were performed under standard conditions (25) using
20% HeLa nuclear extract, incubated at 30"C, and resolved on 20% denaturing
polyacrylamide gels. Reactions containing U2AF-depleted extract, AU2AF NE, were
supplemented with 500 ng of recombinant U2AF 65 and/or 300 ng recombinant SC35. For
the Ul blocking experiments, mock or U2AF-depleted nuclear extract was incubated in the
presence of 7 pM (-15 fold excess over Ul snRNP; 26) a-U1 2'-OMe oligonucleotide
(27) for 15 min. at 30"C followed by addition of substrate RNA and SR protein and
incubation under splicing conditions for 60 min.
RESULTS
U2AF 65 and SC35 Mediated Reconstitution of Splicing in U2AF-Depleted
Extracts
In order to examine the mechanism of action of the splicing factor U2AF, HeLa
nuclear extracts were depleted of this activity. HeLa nuclear extracts depleted of poly-[U]
binding factors and supplemented with a 2M KCI column fraction (P.S. McCaw & P.A.
Sharp, chapter 2) were efficiently depleted of the splicing factor U2AF 65 (400 fold by
Western analysis; Fig. IB and data not shown). Splicing of pre-mRNA substrates
including PIPPG could be restored to the U2AF-depleted extract by either the addition of a
3M KCl column fraction containing U2AF 65 or recombinant U2AF 65 (Fig. lA and data not
shown).
Combinations of U2AF-depleted extract, purified SR proteins, and recombinant
U2AF 65 were tested for reconstitution of splicing. Surprisingly, addition of SR proteins to
nuclear extracts depleted of U2AF65 reconstituted splicing of the PIPPG pre-mRNA (data
not shown). To examine this effect more closely, reconstitution reactions were carried out
with specific SR proteins purified from baculovirus infected insect cells. Addition of
recombinant SC35, purified from insect cells, restored splicing of the PIPPG pre-mRNA in
U2AF-depleted extracts (Fig. lA). This effect was substrate specific since splicing of the
PIP85.A pre-mRNA in depleted extract could not be reconstituted with the addition of
142
SC35 although addition of recombinant U2AF 65 resulted in reconstitution of PIP85.A
splicing at the levels observed with the PIPPG pre-mRNA substrate (Fig. 2). The SC35
reconstitution activity was not due to the presence of the insect homologue of U2AF; insect
U2AF 50 was detected by Western analysis in crude insect cell lysates but not in samples of
purified SC35 (Fig. 1C).
Factor Dependent Splicing in U1-Blocked Extracts
SR proteins, specifically SC35, have been shown to reconstitute splicing in a
substrate specific manner in reactions in which Ul snRNP has been either depleted by
affinity selection (28, 23) or blocked by the pre-incubation of extract with an antisense-U1
oligonucleotide (26). Because of this observation and because Ul snRNP is a component
of the commitment complex, we examined the role of Ul snRNP in the reconstitution of
U2AF-depleted extracts.
U 1 snRNP was inactivated in both mock and U2AF-depleted extracts by preincubation of the extracts with a 15-fold excess (over endogenous U1 snRNP; 26) of an
antisense-U1 2'-OMe oligonucleotide. This blocking of Ul snRNP severely decreased the
splicing of PIPPG pre-mRNA (Fig. 3; lanes 2 and 5). In accordance with previous
observations (26), an excess of SC35 restored splicing activity in Ul blocked reactions in
the presence of U2AF (Fig. 3; lanes 3 and 6). More interestingly, SC35 could only
restore splicing activity in U2AF-depleted reactions in the presence of functional Ul
snRNP (Fig. 3; compare lanes 7 and 8).
We have shown (Fig. 1) that SC35 functionally substitutes for U2AF 65 in a U2AFdepleted extract (Fig 3, lane 7). However, when U2AF-depleted extract was pre-incubated
with an antisense-U I oligonucleotide in the absence of added U2AF 65 , addition of
recombinant SC35 alone did not restore splicing of the PIPiG pre-mRNA (Fig. 3, lane 8).
Thus, SC35 can only functionally substitute for U2AF in the presence of Ul snRNP.
These results distinguish a U2AF independent pathway from a Ul snRNP independent
pathway (Fig. 4; ref. 28)
DISCUSSION
HeLa nuclear extracts depleted of the splicing factor U2AF were reconstituted for
splicing of pre-mRNA substrates by the addition of purified recombinant SC35.
143
Furthermore, the SR protein SC35 restored splicing in a U2AF-depleted extract in a
manner that was both substrate specific and U snRNP dependent. This suggests that
U2AF 65 is not an essential factor in either spliceosome assembly or RNA splicing in the
presence of high concentrations of SC35; it is likely that the enhanced concentrations of
SR domains provided by the addition of SC35 complement the U2AF deficiency.
The pyrimidine tract binding protein U2AF 65 has been shown previously to be
required for the formation of the first stable complex formed between the pre-mRNA and
U2 snRNP, the A complex (15). However, addition of purified SR proteins and more
particularly recombinant SC35 can functionally substitute for U2AF 65 in the reconstitution
of splicing in U2AF-depleted HeLa nuclear extracts. This complementation required
addition of SR protein to approximately a 10-fold excess over levels present in a typical
extract. Complementation by SC35 was substrate specific: the PIPPG pre-mRNA was
efficiently spliced in an SC35 reconstituted extract while the PIP85.A pre-mRNA was not.
The basis for this specificity is unclear, but it is intriguing in that it mirrors the specificity of
the SC35 mediated reconstitution of splicing in extracts depleted of Ul snRNP (28, 29).
There are no obvious specific SC35 binding sites (30) in the PIPPG substrate, but the
specific interaction of SR proteins with the pre-mRNA substrate may only occur in the
context of complex protein-RNA and RNA-RNA interactions (9).
Several lines of evidence suggest that the requirement of U2AF for in vitro splicing
may not be stringent. First, it does not appear that U2AF is absolutely required for
spliceosome assembly. Green and coworkers have shown that either the SR domain of
U2AF 65 fused to a heterologous RNA binding domain or heterologous SR domains fused
to the U2AF 65 RNA binding domains can restore U2AF function in a depleted extract (31,
32). These results suggest that the function of U2AF is to position an SR domain in the
vicinity of the branch region/pyrimidine tract. Second, U2AF does not appear to be present
in catalytically active spliceosomes isolated by gel filtration and thus the chemistry of
splicing probably occurs in the absence of U2AF (19). Finally, the reported
Saccharomyces cerevisiae homolog of U2AF, MUD2, is not a required splicing factor
indicating that its function in U2 snRNPopre-mRNA association is either not essential or
redundant (33). Thus, although in the mammalian system U2AF is highly conserved, it is
possible that there are a variety of mechanisms or factors responsible for directing U2
snRNP to the branch region of the pre-mRNA.
144
The mechanism of the SC35 complementation for U2AF 65 deficiency is not clear.
Members of the SR protein family have been proposed to recruit U2AF to the branch site
via exon enhancers (34) and it is possible that trace amounts of U2AF in the depleted
extract were recruited to a commitment complex by excess SC35. However, this seems
unlikely for several reasons. First, U2AF cannot be detected in either the depleted extract
or in the purified SC35 which complements the reactions - the upper limit of
contamination is 1/400th of endogenous levels of U2AF (Fig. 1 and data not shown). The
U2AF-depleted extracts contained at least 10 fold less U2AF than that required for
restoration of splicing (as determined by adding mock depleted extract to depleted extract;
data not shown). Second, a recruitment mechanism would suggest that excess SC35
should reconstitute a U2AF-depleted extract even when Ul activity was blocked in
accordance with the observed SC35 reconstitution of U1 snRNP blocked (26) or depleted
extracts (28, 29). This was not the case: pre-incubation of U2AF-depleted extract with
antisense-U1 oligonucleotide effectively blocked the SC35 reconstitution (Fig. 3; see
below).
It is likely that SC35 plays several roles in the reconstitution reactions including
substitution for the activity of U2AF (Fig. 4). The mechanism of U2AF activity in
recruitment of U2 snRNP to the branch region is not well understood but clearly involves
recognition of the N terminal SR region (18). While most SR domains are believed to be
involved in protein-protein interactions, it has been suggested that the SR domain of U2AF
functions as an RNA annealing activity (35). Most probably, SC35 complements the
U2AF deficiency in depleted extracts by providing a surrogate SR domain required for
critical interactions in the course of spliceosome assembly.
High levels of SR proteins, including SC35, promote the splicing of the PIP3G
substrate in the absence of Ul snRNP (23). Under these conditions, the SR proteins
facilitate the formation of the U2 snRNP containing A complex independent of Ul snRNA
and the association of U6 snRNA with the substrate RNA can become rate-limiting (29).
The U1 snRNP-bypass reaction does not occur with PIP85.A pre-mRNA. The observed
substrate specificity is identical to that in the SC35-mediated U2AF bypass reaction. This
intriguing observation might have reflected a common pathway in which the Ul snRNP
independent reaction was also U2AF independent. However, this was not the case:
interference with the activity of Ul snRNP inactivated splicing in U2AF-depleted extracts,
even in the presence of high concentrations of SC35. The common substrate specificities
in the two reconstitutions perhaps reflect the sequence specificity of SC35-pre-mRNA
interaction and not common splicing pathways. Thus, there are at least three different
145
pathways to the formation of an active spliceosome: the conventional pathway requiring
both UI snRNP and U2AF 65 ; a second pathway, which is independent of U1 snRNP but
dependent on U2AF; and a third pathway which is independent of U2AF but dependent
upon Ul snRNP (Fig. 4). Any one of these mechanisms could be operative for a particular
intron under specific conditions in vivo. However, since the splicing of a typical intron
requires both Ul snRNP and U2AF, both of these entities probably act at a common step
in stabilizing the interaction of U2 snRNP with the pre-mRNA.
ACKNOWLEDGMENTS
We thank M. Green for the generous gift of U2AF 65 antibody, R. Kanaar and D.
Rio and for the generous gift of DrosophilaU2AF 50 antibody, and B. Blencowe for the
generous gift of anti-U1 oligonucleotide. We also thank L. Lim, J. Pomerantz, and C.
Query for their critical reading of the manuscript; M. Siafaca for her ever-present
assistance; M. Beddall and R. Issner for indispensable technical support. This work was
supported by United States Public Health Service MERIT award R37-GM34277 and grant
RO1-AI32486 from the National Institutes of Health to P.A.S. and partially by a Cancer
Center Support (core) grant P30-CA14051 from the National Cancer Institute. A.M.M.
was supported by the Medical Research Council of Canada.
146
REFERENCES
1.
Konarska, M.M. & Sharp, P.A. (1986) Cell 46, 845-855.
2.
Guthrie, C. (1991) Science 253, 157-163.
Moore, M.J., Query, C.C., & Sharp, P.A. (1993) in The RNA World, eds.
3.
Gesteland, R.F. & Atkins, J.F. (Cold Spring Harbor, New York) pp. 303-357.
4.
Krnimer, A. (1995) in Pre-mRNA Processing,ed. Lamond, A.I. (R.G. Landes,
Austin) pp. 35-64.
5.
Ruby, S.R. & Abelson, J. (1988) Science 242, 1028-1035.
6.
Seraphin, B. & Rosbash, M. (1989) Cell 59, 349-358.
7.
Michaud, S. & Reed, R. (1991) Genes Dev. 5, 2534-2546.
8.
Staknis, D. & Reed, R. (1994) Mol. Cell Biol. 14, 7670-7682.
9.
Fu, X.-D. (1996) RNA 1, 663-680.
10.
Fu, X.-D. (1993) Nature 365, 82-85.
11.
MacMillan, A.M., Query, C.C., Allerson, C.R., Chen, S., Verdine, G.L., &
Sharp, P.A. (1994) Genes Dev. 8, 3008-3020.
12.
Blencowe, B.J.; Nickerson, J.A., Issner, R., Penman, S., & Sharp, P.A. (1994)
J. Cell Biol. 127, 583-607.
13.
Roscigno, R.F. & Garcia-Blanco, M.A. (1995) RNA 1, 692-706.
14.
Fu, X.-D. & Maniatis, T. (1992) Proc. Natl. Acad. Sci. USA 89, 1725-1729.
15.
Ruskin, B., Zamore, P.D., Green, M.R. (1988) Cell 52, 207-219.
16.
Zamore, P.D. & Green, M.R. (1989) Proc. Natl. Acad. Sci. USA 86, 9243-9247.
17.
Zamore, P.D. & Green, M.R. (1991) EMBO J. 10, 207-214.
18.
Zamore, P.D., Patton, J.G., & Green, M.R. (1992) Nature 355, 609-614.
147
19.
Bennett, M., Michaud, S., Kingston, J., & Reed, R. (1992) Genes Dev. 6, 19862000.
20.
Wu, J.Y. & Maniatis, T. (1993) Cell 75, 1061-1070.
21.
Robberson, B.L., Cote, G.J., & Berget, S.M. (1990) Mol. Cell. Biol. 10, 84-94.
22.
Moore, M.J. & Sharp, P.A. (1992) Science 256, 992-997.
23.
Crispino, J.D., Mermoud, J.E., Lamond, A.I., & Sharp, P.A. (1996) RNA 2,
664-673.
24.
Dignam, J.D., Lebovitz, R.M., & Roeder, R.D. (1983) Nucleic Acids Res. 11,
1475-1489.
25.
Grabowski, P.J., Padgett, R.A., & Sharp, P.A. (1984) Cell 37, 415-427.
26.
Tarn, W.-Y. & Steitz, J.A. (1994) Genes Dev. 8, 2704-2717.
27.
Barabino, S.M.L., Blencowe, B.J., Ryder, U., Sproat, B.S., & Lamond, A.I.
(1990)Cell 63, 293-302.
28.
Crispino, J.C., Blencowe, B.J., & Sharp, P.A. (1994) Science 265, 1866-1869.
29.
Crispino, J.D. & Sharp, P.A. (1995) Genes Dev. 9, 2314-2323.
30.
Tacke, R. & Manley, J.L. (1995) EMBO J. 14, 3540-3551.
31.
175.
Valdcrcel, J., Singh, R., Zamore, P.D., & Green, M.R. (1993) Nature 362, 171-
32.
Valcarcel, J., Gaur, R.J., Singh, R., & Green, M.R. (1996) Science 273, 17061709.
33.
Abovich, N., Liao X.C., & Rosbash, M. (1994) Genes Dev. 8, 843-854.
34.
Wang, Z., Hoffinann, H.M., Grabowski, P.J. (1995) RNA 1, 21-35.
35.
Lee, C.G., Zamore, P.D., Green, M.R., & Hurwitz, J. (1993) J. Biol. Chem.
268, 13,472-13,478.
148
FIGURE LEGENDS
Figure 1. SC35 functionally substitutes for U2AF 6 5 in the reconstitution
of pre-mRNA splicing in U2AF-depleted extracts.
(A) Splicing of PIPOG pre-mRNA in mock depleted extract (lane 1); U2AF-depleted
extract (lane 2); and U2AF65 reconstituted (lane 3) and SC35 reconstituted (lane 4) U2AFdepleted extract.
(B) Western analysis with anti-U2AF 65 antibody of mock depleted (lane 1) and U2AFdepleted (lane 2) extract.
(C) Western analysis of: (left) crude lysate of SC35 overexpression with a-p50 (antiDrosophilaU2AF 50 antibody) and anti-SC35 antibody (Mab 104); (right) purified SC35
with a-p50 and anti-SC35 antibody (Mab 104).
149
__~__
A
NE
IU2AF NE
SIVS-E2
IWS
E1-IVS-E2
~\C~
94
E1-E2
U2AF 6 5 -
1
234
2
lysate
purified SC35
az-p50 Mabl04
a-p50 Mabl04
H
p50 --sC35
-
4-SC35
Figure 2. SC35 reconstitution of splicing in U2AF-depleted reactions is
substrate specific.
Splicing of PIP3G pre-mRNA in mock (lane 1), U2AF-depleted (lane 2), U2AF 65
reconstituted (lane 3) and SC35 reconstituted (lane 4) extracts. Splicing of PIP85.A in
mock (lane 5), U2AF-depleted (lane 6), U2AF 6 5 reconstituted (lane 7) and SC35
reconstituted (lane 8) extracts.
151
PIPBG
NE
U2AF 6 5
SC35
AU2AF
NE
-
-
PIP85.A
NE AU2AF
NE
-E-
IVS-E2
IVS
E1-IVS-E2
El-E2
12
345678
Figure 3. SC35 reconstitutes pre-mRNA splicing in U2AF-depleted
extracts dependent on the presence of functional U1 snRNP.
Splicing of PIPI3G pre-mRNA in: nuclear extract (lane 1), nuclear extract pre-blocked with
a-U1 oligonucleotide (lane 2), nuclear extract pre-blocked with a-U1 oligonucleotide and
supplemented with SC35 (lane 3); U2AF-depleted extract supplemented with U2AF 65
(lane 4), U2AF-depleted extract supplemented with U2AF 65 and pre-blocked with a-U1
oligonucleotide (lane 5), U2AF-depleted extract supplemented with U2AF 65 , pre-blocked
with a-U1 oligonucleotide, and supplemented with SC35 (lane 6); U2AF-depleted extract
supplemented with SC35 (lane 7), U2AF-depleted extract blocked with a-U1
oligonucleotide and supplemented with SC35 (lane 8).
153
NE
65
U2AF
t-Ul
SC35
- - - + +
- - +
AU2AF NE
+ + +
- ++
- - +
- +
+ +
VS-E2
E1-IVS-E2
123
456
78
Figure 4.
Three distinct pathways resulting in spliceosome assembly.
Under typical splicing conditions, both U2AF and Ul snRNP are required for spliceosome
assembly with a network of stabilizing interactions between U2AF, U1 snRNP, and SR
proteins (B; 20). In a substrate specific manner, excess SC35 reconstitutes splicing in
U2AF-depleted reactions in a Ul snRNP dependent pathway (A) and in U1 snRNPdepleted reactions in a U2AF dependent pathway (C).
155
(C)
U2AF independent
U2
snRNP
U2AF dependent
Ut snRNP dependent
U2
U1 snRNP independent
U2
snRNP
snRNP
S3snRNP
A
A Complex
CHAPTER 5:
A MINIMAL SPLICEOSOMAL A COMPLEX
RECOGNIZES BRANCH SITE AND POLYPYRIMIDINE
TRACTS
Charles C. Query, Patrick S. McCaw, and Phillip A. Sharp*
Center for Cancer Research and Department of Biology
Massachusetts Institute of Technology
Cambridge, MA 02139-4307
This chapter was originally published in the Journal of Molecular and Cellular Biology,
vol. 17, pp. 2944-53.
157
ABSTRACT
The association of U2 snRNP with the pre-mRNA branch region is a critical step in
the assembly of spliceosomal complexes. We describe an assembly process that reveals
both minimal requirements for formation of a U2 snRNP-substrate RNA complex, here
designated A., and specific interactions with the branch-site adenosine. The substrate is a
minimal RNA oligonucleotide, containing only a branch sequence and polypyrimidine tract.
Interactions at the branch-site adenosine and requirements for polypyrimidine tract-binding
proteins for An are the same as those of authentic pre-spliceosome complex A.
Surprisingly, A formation does not require Ul snRNP or ATP, suggesting that these
factors are not necessary for stable binding of U2 snRNP per se, but rather for accessibility
of components on longer RNA substrates. Further, there is an ATP-dependent activity that
releases or destabilizes U2 snRNP from branch sequences. The simplicity of the An
complex will facilitate a detailed understanding of the assembly of pre-spliceosomes.
158
INTRODUCTION
The removal of introns from precursors to messenger RNA molecules (pre-mRNA)
is catalyzed by the spliceosome, a dynamic 50-60S complex composed of small nuclear
RNAs (snRNAs) U1, U2, U5, and U4/6, as well as protein components (for review, see
34, 39, 43, 46). Such intron excision proceeds by way of two sequential
transesterification reactions. The spliceosome assembles de novo on each substrate premRNA and several distinct intermediates in an assembly pathway can be observed in vitro.
The E (early) or commitment complex contains Ul snRNP and non-snRNP protein factors
(28, 42, 58). Complex A is generated by the stable binding of U2 snRNP to the branch
region of the pre-mRNA; a larger complex, B, is formed by association of U4/5/6 trisnRNP with complex A. Complex C follows B after significant rearrangements and
contains splicing intermediates (29, 30, 43).
The branch region contains the nucleophile for the first chemical step of splicing,
and its recognition is required early in splicing complex assembly. U2 snRNP binds the
pre-mRNA, in part, through U2 snRNA*branch region base-pairing (48, 69, 73), and the
first step nucleophile is selected, in part, by virtue of being bulged from this duplex (51).
Early branch site recognition in yeast requires U1 snRNP and a non-snRNP splicing
factor, a component of which may be MUD2 (2, 55, 59). In mammals factors SF3a,
SF3b (both of which join 12S U2 to form 17S U2 snRNP), SF1, U2AF 65 , U2AF35 , Ul
snRNP and members of a family of proteins containing arginine-serine dipeptide repeats
(SR proteins; for review see 23, 40, 66) are important for the stable association of U2
snRNP with the pre-mRNA (3, 6, 7, 9, 10, 33, 74). U2AF 65 binds specifically to
polypyrimidine tracts in early complexes (24, 42, 56, 70). Another factor, PUF-2
(poly[U]-binding factor-2), which contains two more polypyrimidine-binding proteins, a
p54 SR protein (14, 71) and a p130, is also important for efficient complex A formation
(41). Some of the components of SF3 have been shown to cross-link to the pre-mRNA
upstream of the branch region and are suggested to tether or stabilize U2 snRNP binding to
the pre-mRNA (13, 25). Within the branch region, but not at the adenosine, two proteins,
BPS7 2 and BPS 70 , have been cross-linked in E and A complexes, respectively (16, 52).
At the branch-site adenosine itself, three proteins have been detected in complex A within
15 A: p14, p35, and p150 (38); one of these, p14, can be directly photo-cross-linked to
the branch-site adenosine (52).
159
ATP is required at numerous points during the splicing process and probably for
multiple distinct functions, although it is not involved directly in either of the two
transesterification reactions (45). Phosphorylation and dephosphorylation of SR proteins
are believed to occur, as well as structural rearrangements of the snRNAs (reviewed in 23,
46, 65). The earliest detected requirement for ATP is during the transition from E complex
to complex A, when U2 snRNP joins the pre-mRNA (e.g., 15, 29, 32, 37, 42, 50).
Although this has been generally interpreted to indicate that U2 snRNP binding requires
ATP, the exact mechanism is unclear. By analogy to known systems operative in the
ribosome for the fidelity of translation, there have been many suggested steps of proofreading during the splicing process (11). The yeast protein PRP 16 may be part of a proofreading/discard pathway that examines the branched nucleotide after chemical step one, as
mutant prpl16 alleles increase the rate of progression to the second step of splicing of certain
non-adenine branches (12, 17).
In the present study, we have determined the minimal substrate requirements for
formation of complexes containing U2 snRNP. A variety of criteria indicates that this
minimal complex, A ,, represents an accurate model for interactions with many factors
influencing assembly of pre-spliceosome complex A. A~i
n formation is a more sensitive
system, as it is more affected by subtle modifications than is complex A. Surprisingly,
formation of Amin does not require ATP; but, the complex is subject to an ATP-dependent
dissociation, which may reflect a fidelity mechanism normally operative at the time of prespliceosome assembly.
MATERIALS AND METHODS
RNA transcription and synthesis of substrates.
pPIP85.B is a modification of pPIP85.A (44) that has only one adenosine in the
branch region and encodes the following 234-nucleotide sequence:
5 '-GGGCGAAUUCGAGCUCACUCUCUUCCGCAUCGCUGUCUGCGAGGUACCCUACCAG GU
GAGUAUGGAUCCCUCUAAAAGCGGGCAUGACUUCUAGAGUAGUCCAGGGUUUCCGAGGGUU
UCCGUCGACGAUGUCAGCUCGUCUCGAGGGUGCUGACUGGCUUCUUCUCUCUUUUUC
CCUCAG GUCCUACACAACAUACUGCAGGACAAACUCUUCGCGGUCUCUGCAUGCAAGCU 3 '. Arrows indicate the 5' and 3' splice sites, and the underlined A indicates the branch
site. The bold sequence represents RNA(146-179), or BS-PPT RNA. Transcription of
this full-length pre-mRNA and of other RNAs were performed under standard conditions
(see 51).
160
Two-way RNA ligation reactions and gel purification of products were performed
as described previously (44, 51). Briefly, oligo-ribonucleotides containing a branch
sequence and polypyrimidine tract [BS-PPT RNA: RNA(146-179)] were prepared by
joining a branch region decamer [RNA(146-155): 5'-GGGUGCUGAC-3'] and a 5'-32p phosphorylated polypyrimidine tract [RNA(156-179): 5'UGGCUUCUUCUCUCUUUUUCCCUC-3'] using T4 DNA ligase (USB) and a
bridging oligonucleotide [cDNA(169-136): 5'-GAGAGAAGAAGCCAGTCAGCACCCTCGAGACGAG-3']. PPT-BS RNA [RNA(156-179, 145-155)] was prepared by
joining RNA(156-179) and 5'- 32 p-phosphorylated branch region decamer [RNA(146155)] using cDNA (5'-GTCAGCACCCGAGGGAAAAAGAGAGAAGAAGCC-3').
Ligation products were purified on 15% polyacrylamide (29:1), 8 Murea gels run in lx
TBE (89 mM Tris-borate, 2 mM EDTA). All-RNA and 2,6-diaminopurine-containing
branch region decamers were prepared by chemical synthesis as described (62). 2'-H
substituted branch region decamers and polypyrimidine tract-containing RNA(156-179)
were prepared by chemical synthesis on an ExpediteTM 8909 oligonucleotide synthesizer
(by M.J. Moore) and purified similarly. Branch region decamer containing a convertible
adenosine for cross-linking experiments was described in (38).
Formation and native gel analysis of splicing complexes.
To form splicing complexes, RNAs were incubated under standard splicing
conditions (26) using nuclear extracts as described below; or, for ATP-depleted reactions,
ATP and creatine phosphate were omitted from the mixes, which were preincubated for 15
min at 30 0 C to deplete endogenous ATP and, in some cases, then adjusted to 10 mM
EDTA. RNAs were then added and incubated at 300 C for the times indicated. Reactions
were adjusted to 0.5 mg/ml heparin and separated by electrophoresis in 50 mM Tris-glycine
through non-denaturing 4% (80:1) polyacrylamide gels.
Nuclear extracts and purification of splicing factors.
Nuclear extracts were prepared from HeLa cells as described (21). Extracts
depleted of individual snRNPs were generous gifts from John Crispino, were prepared as
described (6, 8), and were characterized in (18). Extracts depleted of poly[U]-binding
proteins and U2AF 65 were prepared by dialyzing nuclear extract directly into 1 M
KCl/buffer D [20 mM Hepes (pH 7.9), 20% glycerol, 0.2 mM EDTA, 0.05% NP-40, 0.5
mM DTT]. The resulting extract was passed over a poly[U]-Sepharose 4B column
(Pharmacia) at 0.1 ml/min and subsequently dialyzed against 0.1 MKCl/buffer D. After
161
washing this column with 1 MKCl/buffer D, the column was eluted with buffer D
containing 2 MKCI; the eluate was dialyzed against 0.1 M KCI to obtain the "2 M KC1
fraction", or PUF-2, which contains the poly[U]-binding proteins p54 and p130 (41).
Mock-depleted extract was prepared in parallel to depleted extract by dialyzing nuclear
extract into 1 MKCl/buffer D and subsequently against 0.1 MKCl/buffer D. Recombinant
His6-tagged U2AF 65 was prepared from a 60% ammonium sulfate precipitate of a soluble
E. coli lysate. This was loaded onto a Ni2+-NTA-agarose column (Qiagen), eluted with
250 mM imidazole/buffer D, and dialyzed into 0.1 MKCl/buffer D.
Photo-cross-linking
assays.
High specific activity substrate (107 c.p.m./reaction) containing an N6-ethylthiolmodified adenosine was reduced by treatment with 5 mi dithiothreitol (DTT) in 20 mM
NaHCO3 at 30"C for 1 hr and then derivatized by reaction with 20 mM benzophenone
maleimide (Molecular Probes) in 50% dimethyl formamide at room temperature for 1 hr
(38). Reactions were extracted with phenol/chloroform and chloroform and then ethanol
precipitated. The RNA was incubated in HeLa nuclear extract as above except that RNasin
was omitted, and was adjusted to 0.5 mg/ml heparin prior to UV irradiation with a 302-nm
lamp (0.12 W/cm 2 at 1 cm; Ultraviolet Products) for 20 min on ice.
Alternatively, cross-linking of 2,6-diaminopurine-containing RNA was performed
on ice by irradiation with a 254-nm lamp (0.12 W/cm 2 at 1 cm; Ultraviolet Products) for 60
min. After either photo-cross-linking technique, reactions were separated on 4% (80:1)
native polyacrylamide gels (29) and frozen; the individual complexes, visualized by
autoradiography, were excised. These were incubated with 0.32 mg RNase A/ml of gel in
125 mM Tris-HCI (pH 6.8) at 37 0 C overnight, then incubated with SDS loading buffer at
37 0C for 2 hours and 65 0 C for 5 min, and placed directly onto the stacking gel of a
disassembled SDS 16% (200:1) polyacrylamide gel, which was reassembled and
electrophoresed in 0.25 MTris (pH 8.3), 0.192 M glycine, 0.1 % SDS.
RESULTS
A short oligonucleotide can form complexes with U2 snRNP.
U2 snRNP complexes form on full-length pre-mRNAs (complex A) as well as on
3' half RNAs that lack a 5' splice site (A3' complexes; 29). These RNAs contain a
number of elements, illustrated in Figure lA, upper, believed to contribute to complex A
162
formation and stability. 5' to the branch site is a region that interacts with SF3a and SF3b
components, which is believed to stabilize complex A (25). Surrounding the branch site is
a region of U2 snRNA complementarity important for efficient complex formation (48, 69,
72, 73). The polypyrimidine tract interacts with several factors, including U2AF 65 and
PUF-2 (41, 56, 70), and exon enhancer sequences or downstream 5' splice sites interact
with SR and other proteins or Ul snRNP to promote U2AF 65 binding and complex
formation (e.g., 27, 36, 61, 67). In addition, binding of U1 snRNP and other factors to
the 5' splice site probably stimulates complexes; and, in a role that is not understood, Ul
snRNP is also required for complex A formation independently of 5' splice site interaction
(6). To establish minimal requirements for this process, shorter RNAs, made by deleting
from both ends of a 234-nucleotide model pre-mRNA, PIP85.B RNA (which contains a
well-defined branch site with only one adenosine in the region; 51), were tested for
formation of A-like U2 snRNP complexes (data not shown). The shortest RNA efficiently
forming a complex that co-migrated with complex A on native gels was RNA(146-179), a
34-nucleotide RNA containing only a branch sequence and polypyrimidine tract (BS-PPT
RNA; Figures lA, lower, and lB). This complex is designated Amin, since it represents an
A-like complex on a minimal substrate.
This RNA substrate notably lacks several elements discussed above that
presumably contribute to efficient complex A formation. It does not contain the region
thought to be the binding site for SF3a and SF3b (25). Nor does it contain any sequence
3' to the 3' splice site that could act as an exon enhancer element. In addition, it does not
contain the 3' splice site AG: comparison of RNAs either containing or deleted of the 3'
splice site AG or containing a mutated 3' splice site region did not show detectable
differences in complex formation (data not shown). As indicated in Figure IB and
discussed in depth below, formation of Amin does not require ATP. In the analysis of
truncated pre-mRNAs, RNAs containing additional sequences 3' to the BS-PPT region
also formed A-like complexes in the absence of ATP (e.g., Figure 4C, lane 9); but, RNAs
containing additional sequences 5' to the BS-PPT region did not [RNA(1-234), RNA(64179), and RNA(104-179); Figure IB, lane 2, and data not shown].
Northern blot analysis of the A, complex separated by native gel electrophoresis
showed no detectable U1, U4, U5, or U6 snRNA in the complex; however, free 17S U2
snRNP (in the absence of BS-PPT RNA) migrates close to Amin in this gel system, making
evaluation of the U2 snRNA content of Amin indeterminate (data not shown). To verify that
contained U2 snRNP, the snRNA composition was analyzed by Northern blot after
streptavidin-agarose affinity selection using BS-PPT RNA containing 3'-terminal biotin
Amin
163
[RNA(146-179, bio); Figure IC]. The A. complex was highly enriched for U2 snRNA
(lane 2) as compared to all five snRNAs in spliceosomes formed on full-length pre-mRNA
(lane 3). A small amount of U4, U5, and U6 snRNAs was selected (<5-10% of the level
of U2 snRNA relative to full-length pre-mRNA); this may relate to larger, as yet
uncharacterized, complexes sometimes observed after long incubations (e.g., Figure 3B,
lanes 6 and 7, or Figure 4B, lane 7).
Ul snRNA was also selected using biotin-tagged BS-PPT RNA; this was not
unexpected since formation of complex A on full-length pre-mRNA, as well as complex
A3' on 3' partial RNA substrates, is dependent on both U 1 and U2 snRNPs (6, 58).
However, since the Ul snRNP association was not stoichiometric with U2 snRNP, the
snRNP requirements for A. n complex formation were tested in extracts depleted of various
snRNPs (6, 8). These extracts alone did not form mature spliceosomes on pre-mRNA,
but when mixed they complement for spliceosome formation and for splicing (data not
shown; for an analysis of these specific extracts, see 18). In particular, the extracts
depleted of either Ul or U2 snRNP did not form complex A on pre-mRNA (see Figure 1 in
ref 20). As expected, extracts depleted of U2 snRNP did not form An n complex (Figure
lD, lane 3), and extracts depleted of U4/6 snRNP formed complexes just as well as mockdepleted extract (cf. lane 4 to 1). Surprisingly, however, U 1-depleted extracts also formed
An n complexes efficiently (cf. lane 2 to 1); thus, the binding of U2 snRNP to the branch
region per se does not require Ul snRNP.
Both branch sequence and polypyrimidine tract are required.
BS-PPT RNA contains two sequence elements - a branch sequence (i.e., U2
complementarity region; 5'-UGCUGAC-3', where the underlined A represents the branchsite adenosine) and a polypyrimidine tract (5'-CUUCUUCUCUCUUUUUCCCUC-3')
(Figure 1A, lower). To investigate the individual contributions of each of these elements,
we tested RNAs containing mutations in each element (Figure 2). RNAs containing a
mutated branch sequence (5'-...UGCUGAC...-3' -- 5'-...GUCGUAC...-3') did not
form Amin complex (Figure 2A, lane 3). Similarly, RNAs in which the polypyrimidine tract
was replaced by 5'-...GACGGACAUGCAAUGCAACUC-3' did not form Amin complex
(lane 2). Furthermore, RNAs containing shorter polypyrimidine tracts did not form
complexes with U2 snRNP as efficiently. For example, removal of 7 or 14 pyrimidines
from the 3' end [RNA(146-172) and RNA(146-165), respectively] or an internal deletion
[RNA(146-155, 169-179)] in the polypyrimidine tract resulted in significantly less
164
complex (data not shown). These data suggest that both sequence elements make specific
contributions to An complex formation, as expected for an analog of complex A.
Both elements, the branch sequence and the polypyrimidine tract, were required in
cis. As expected from the mutations tested above, neither sequence alone formed A-like
complexes (Figure 2B, lanes 1 and 6). When added in trans they also could not form
complexes: labeled branch sequence RNA mixed with unlabeled polypyrimidine-tract RNA
did not form detectable complexes (Figure 2B, lanes 2-5); similarly, unlabeled branch
sequence RNA mixed with labeled polypyrimidine-tract RNA also did not form detectable
complexes (lanes 7-10). We next tested the ability of each of the two RNAs to compete
with BS-PPT RNA in complex formation. Although neither branch sequence RNA nor
polypyrimidine-tract RNA formed a stable complex alone, polypyrimidine-tract RNA did
compete with BS-PPT RNA (lanes 11-15). Branch sequence RNA competed with BSPPT RNA only at the highest concentrations tested (1 gM; lanes 16-20). Therefore,
although each element may interact with required factors independently at high
concentrations, both elements are required in cis to form a stable complex. Furthermore,
factors recognizing the polypyrimidine tract are either more limiting, required earlier in the
binding process, or more critical than factors recognizing the branch sequence.
In addition, Amn complex will form only on RNAs in which the branch sequence
and polypyrimidine tract elements are in the correct orientation. When the polypyrimidine
tract was placed 5' of the branch sequence [PPT-BS RNA: RNA(156-179, 145-155)], no
A-like complexes were detected (Figure 2C). Thus, interactions between factors binding to
these two elements are sensitive to their relative positions, and the branch sequence must be
5' of the polypyrimidine tract in order to form correct interactions in making A n.
Similarities of Amin to complex A containing U2 snRNP.
In addition to the above, several lines of evidence suggest that ,in reflects many
aspects of authentic complex A. For example, the ionic strength dependence of A,. n
complex formation corresponds to that required for splicing (Figure 3A). When assayed
across a series of KC1 concentrations, the optimum was 60 mM, as is found for splicing
conditions (reviewed in 47). No complexes were observed at high ionic strength (>200
mM), which was previously found to stabilize the formation of pseudospliceosomes (31).
It should be noted that these high ionic strengths would be expected to stabilize simple
duplexes, so destabilization of the An complex suggests that the latter is not simply due to
RNA-RNA base-pairing. This is also supported by several other lines of evidence. When
RNA-RNA pairing was enhanced by making the branch sequence perfectly complementary
165
to U2 snRNA (5'-...UGCUGC...-3'), Amin complexes were reduced 96% (52); this
contrasts with the stable binding of oligonucleotides for tagging or depletion that is via a
much longer sequence complementarity to U2 snRNP (5, 35). Also, unlike the stabilizing
effect of 2'-O-methyl sugars on simple hybridization, 2'-O-methylation across the branch
region of BS-PPT RNA abrogated formation of A,, n complexes (data not shown). Finally,
when the melting temperatures of several RNA-RNA duplexes were measured in the
absence of proteins, a branch sequence-U2 RNA duplex was not stable under these
conditions (see 51). In contrast, after formation, Amin complex was stable to chase with
excess cold competitor for greater than 4 hours at 300 C (in the absence of ATP, see below;
Figure 3B, lanes 1-7). If added first, this level of competitor completely saturated the A n
complex-forming components (lanes 8-14), demonstrating that the maintenance of
complexes in lanes 1-7 was not due to release and reformation. These data, together with
the requirement for both branch and polypyrimidine tract sequences, argues strongly that
in
complex is not based principally on base-pairing interactions.
The factor requirements for Amin are similar to those for complex A. Assembly of
U2 complexes on full-length pre-mRNA requires the presence of U2AF 65 (56, 70) and is
strongly stimulated by the presence of a factor PUF-2, which elutes from a poly[U] column
at 2 MKC1. This factor contains primarily two polypyrimidine tract-binding proteins, p54
and p130 (41). Extracts depleted of these factors did not support Amin formation (Figure
3C, lane 2), whereas a mock-treated extract did form Amin (lane 1). Addition of the PUF-2
fraction alone did not significantly restore activity (lane 3), and addition of recombinant
U2AF 65 restored only a low level of activity (lane 4). Addition of both PUF-2 and
U2AF65 restored the ability to form Amin complex (lane 5), in keeping with the requirement
of these protein factors for efficient formation of complex A and for splicing (41).
Previously, three proteins - p14, p35, and p150 - were photo-cross-linked to the
branch site as components of both complexes A and A3', using a linker and photo-active
agent that could sample distances up to 15 A (38). The same benzophenone photo-reagent
was placed site-specifically on the branch-site adenosine ofBS-PPT RNA (Figure 3D).
This modified RNA was incubated to form A. complex and UV irradiated; the complexes
were separated on a native gel, the A n complexes excised and digested with RNase A, and
the proteins cross-linked to the labeled RNA fragment analyzed on an SDS gel. The same
three molecular weight proteins, p14, p35, and p150, were labeled within Amin as were
observed within full complex A. Using direct UV irradiation, one of these three proteins,
the p14, cross-linked directly to the branch-site nucleotide in An n complex as it does in
complex A (Figure 3E and 52). The other protein detected in this assay, p70, is cross166
linked at another site within the branch region (see 52). and likely corresponds to BPS 70
previously observed to cross-link in complex A (16, 52). Thus, Ami n contains similar
components and similar interactions proximal to the branch-site adenosine as those detected
in authentic complex A.
Amin complex formation is ATP independent and undergoes an ATPdependent dissociation.
Formation of complex A on full-length pre-mRNA, as well as A3' complexes on 3'
partial RNAs, requires ATP (29). Surprisingly, as was suggested in Figures IB and 2,
assembly of A. n complexes does not require ATP. A complex formed more efficiently
(see below) in the absence of ATP (i.e., in extracts depleted of ATP; see materials and
methods) compared to levels observed in the presence of ATP (Figure 4A, cf. lanes 8-14
to 1-7). As expected, A-type complexes did not form on full-length pre-mRNA in the
absence of ATP (cf. lanes 24-25 to 22-23). Also, Ain complex formation was even more
efficient, or stabilized, in the presence of EDTA (lanes 15-21); this increase may be due to
many effects, including stabilization of the RNA from degradation or chelation of Mg 2+
from trace levels of contaminating ATP (data not shown). Other studies have suggested
that the presence of EDTA does not inhibit the formation of functional splicing complexes
(1, 15). Furthermore, A n complexes, but not A or A3' complexes, form at 40 C, albeit
with slower kinetics than at 30 0 C, also suggesting that ATP hydrolysis is not required (data
not shown).
The increase and subsequent decrease in A.i complexes in the presence of ATP
(Figure 4A, lanes 1-7; and Figure 4D, curve a) suggests that two distinct processes are at
work: both formation and dissociation. The increased level of A. complexes observed in
the absence of ATP (Figure 4A, lanes 8-14 or 15-21; 4D, cf. curves b to a) suggested that
the dissociation process was ATP-dependent. To test whether this represented an active
process, complexes were formed in the absence of ATP, challenged with excess cold
competitor BS-PPT RNA, and re-incubated either with or without the addition of ATP.
During this re-incubation, A, complexes were dissociated in the presence of ATP, but not
in the absence of ATP (Figure 4B upper, cf.lanes 8-13 to 2-7). This was not due to
degradation of the RNA, which remained at similar levels throughout the incubations
(Figure 4B lower, cf. lanes 8-13 to 2-7). The dissociation of complexes required both
magnesium cation and hydrolyzable nucleotide-triphosphate. For example, AMP-PcP,
AMP-cPP, or AMP-PnP could not replace ATP, although other NTPs or dNTPs could
(data not shown). Thus, A.. complex is a substrate for an NTP-dependent dissociation
167
activity that results in rapid disassembly of U2 snRNP-containing complexes (Figure 4D,
curve c). The level of complexes formed in the presence of ATP (curve a) is probably the
sum of the two processes of complex formation without ATP (curve b) and of dissociation
using ATP (curve c), indicating a dynamic assembly and disassembly of U2 snRNP
complexes.
To test whether the presence of additional sequences would alter the susceptibility
of the complex to the dissociation activity, the stability of ,n complexes was compared to
complexes formed on RNA additionally containing a 3' splice site and 3' exon [RNA(146234); Figure 4C]. Although this RNA ostensibly is similar to 3' half RNAs used to form
A3' complex, it does not contain sequences 5' to the branch region and forms complexes in
the absence of ATP, making this comparison possible. As before, preformed An n complex
dissociated rapidly when challenged with ATP and competitor BS-PPT RNA (lanes 2-8)
compared to no chase (lane 1) or chase without added ATP (lane 2). In contrast,
complexes containing the RNA with additional 3' sequences were relatively stable to this
challenge with ATP and competitor RNA(146-234) (lanes 11-16) compared to no chase
(lane 9) or chase without ATP (lane 10). Thus, the presence of additional 3' sequences
stabilizes
Amin
complex from disassembly in the presence of ATP.
Effects of 2'-H substitutions.
Formation of the A n complex is exquisitely sensitive to branch-site modifications.
In contrast, formation of complexes at the branch site of full-length pre-mRNAs is only
minimally affected by branch site modifications (51, 52). For example, a double
2'-deoxynucleotide (2'-H) substitution at the branch-site adenosine and immediately 5' to it
(5'-UGCUGHAHC -3'; where the superscripted letters indicate the 2' moiety) only slightly
reduced U2 snRNP complex formation on a full-length pre-mRNA; rather, there was an
accumulation of later complexes unable to undergo the first chemical step of splicing (51).
In contrast, the same double substitution in BS-PPT RNA resulted in a 97% decrease in
A complex formation relative to the all-ribose RNA (Figure 4, cf. lanes 8-14 to 1-7;
Table 1).
To test whether the large effect of the double 2'-H substitutions at the branch site
and adjacent nucleotide were specific to these positions, a similar double 2'-H substitution
was prepared three and four residues 5' to the branch site ( lanes 15-21). This resulted in a
70% decrease in the level of A complexes, significant but much less than the effect at the
two positions above. Single 2'-deoxynucleotide substitutions at the branch site or at the
immediately 5' residue resulted in approximately 40% and 20% decreases in A.i n complex
168
formation, respectively (Table 1). These effects are comparable to that of a 2'-H placed
four nucleotides 5'-distal to the branch site, which decreased complex formation by
approximately 12%. The modest effects of single substitutions compared to the dramatic
effect of two 2'-H substitutions are consistent with either position contributing an important
contact (see Discussion). The strong effect of double substitutions at the branch site and 5'
to it (97% decrease) is not due just to cumulative effects of individual substitutions, as two
separated 2'-H substitutions resulted in only a 40% decrease (which is roughly cumulative
of the individual effects) and the two adjacent substitutions discussed above inhibited only
70%. Nor are one or two 2'-H substitutions likely to alter the conformation of the branch
sequence-U2 helix (4, 22, 49). Thus, the simplified Amin system revealed an important
2'-OH contact at the branch site.
DISCUSSION
Interactions of the pre-mRNA branch site with U2 snRNP have been studied using
a minimal RNA sequence containing only the branch region and polypyrimidine tract. The
Amin complex formed under these conditions is an accurate reflection of many interactions in
the generation of complex A, as both are critically dependent upon the sequences of the
branch region and polypyrimidine tract and both require U2AF 65 and PUF-2 factors.
Further, the adenine base and 2'-OH constituents of the branch site are important for
formation of APin. Finally, the protein-RNA contacts around the branch site in Amin are
identical to those of complex A, as shown by two photo-cross-linking methods. These
characteristics indicate that the engagement of Amin complex components with the branch
site is the same as within complex A. Surprisingly, formation of Amin complex does not
require ATP or U1 snRNP, indicating that these factors are not necessary for stable
association of U2 snRNP with a branch sequence per se. In the absence of ATP, Ain-type
complexes do not form with RNAs containing sequences upstream of the branch site,
suggesting that accessibility of the branch site in these RNAs might be ATP-dependent.
Finally, the association of U2 snRNP with the branch region on a minimal substrate is
dynamic - rapid ATP-dependent turnover indicates the presence of an active mechanism
that releases or destabilizes U2 snRNP from branch sequences.
A more sensitive system -
2'-OH and adenine interactions.
The dramatic effects of 2'-deoxynucleotide and branch-site base substitutions on the
formation of Amin demonstrate that this complex is more sensitive to subtle atomic changes
than is pre-spliceosomal complex A. If multiple interactions contribute to overall stability
169
of complex A, then the absence of some of these interactions should result in complex
formation being more critically dependent upon the remaining ones. In the case of the
branch region, multiple weak interactions almost certainly contribute to the formation and
stability of complex A (reviewed in 54). When some of these are absent - e.g.,
interactions at the 5' splice site, at the "U2 anchoring site" 5' to the branch region, and 3' to
the polypyrimidine tract at the 3' splice site and exon enhancer sequences (e.g., 25, 36,
53, 60, 63, 66, 68, 74) - recognition of the branch site and polypyrimidine tract becomes
more important. This increased sensitivity to the precise nature of chemical groups at the
branch site has revealed 2'-OH and adenine contacts in Amin. The large effect of double
2'-H substitutions at the branch and the 5' adjacent sites, unlike single substitutions that
have modest effects, indicates that contacts with these two positions are critical and, in
some way, cooperative. This may relate to the alternative bulging of these two positions
described previously (51) which would allow either 2'-OH present to fill a similar
position; or, interaction with either 2'-OH might be adequate for formation of a stable
complex.
Recognition of the adenine base at the branch site is critical for Ar n formation and
previously was shown to contribute to complex A stability (52). Relative to complex A,
Ami has enhanced dependency upon an exocyclic C6-NH 2 group at the branch site, which
contributes a significant positive effect; a C2-NH 2 group has a significant negative effect
and a C6-oxo/N 1-H of guanine is strongly inhibitory. Thus, even at the time of initial U2
snRNP addition, and without any ATP-dependent transitions, there are specific contacts
both with the adenine base and with 2'-OH groups in the ribose-phosphate backbone in the
branch region. Interestingly, recognition of these specific contacts strongly correlates with
the direct cross-linking of a p14 protein. This protein is thus a good candidate for the
component of complex A that directly recognizes the branch-site adenosine.
Amin complex forms independently of U1 snRNP and ATP.
Formation of complexes in nuclear extracts depleted for individual snRNPs
demonstrated that U2 snRNP, but neither Ul nor U4/6 snRNPs, is required for Amin
formation. The Ul snRNP independence of Amin is surprising, since this snRNP is
generally required for assembly of complexes on full-length pre-mRNAs (6). However,
this requirement is not absolute, since both complex formation and splicing can sometimes
occur in the absence of Ul snRNP (18-20, 64). Such Ul snRNP-independent splicing
has been observed for a subset of pre-mRNAs exemplified by fushi tarazu (ftz) and with
certain substrates in the presence of elevated concentrations of SR proteins; other substrates
170
do not show Ul -independent activity under any condition tested. The conditions reported
here for An formation do not have elevated levels of SR proteins and the BS-PPT RNA is
derived from a pre-mRNA that does not exhibit U -independent splicing even with added
SR proteins (18). Since Am readily forms on the isolated branch sequencepolypyrimidine tract in the absence of Ul snRNP, the general requirement of Ul snRNP
for complex A must be due to sequences external to these elements.
The mechanism requiring ATP during the formation of complex A on pre-mRNA is
not known. The independence of A. formation from this ATP requirement implies that
ATP is not needed for U2 binding per se. This is consistent with previous results that
suggested that U2 addition could occur without ATP in the background of a weakened U15' splice site interaction (37). The A. complex does not form on RNA substrates with
sequences 5' of the branch site; formation of complex A or A3' on these substrates requires
ATP. This suggests that there may be an ATP-dependent step required for exposure of the
longer substrate RNA for the binding to U2 snRNP. For example, a helicase-type activity
might unfold the substrate RNA for the subsequent binding of U2 snRNP; alternatively, a
conformational change in U2 snRNP or another complex A factor might be necessary to
allow interaction with sequences 5' to the branch region.
In the presence of ATP, the binding of U2 snRNP in complex A is probably
stabilized by interactions with both Ul snRNP and SR proteins in a dynamic equilibrium.
The strength of interactions with Ul snRNP and SR proteins summed with recognition of
the branch region and polypyrimidine tract would determine the level of complex A. If the
other interactions were weak, then the determinants for stable formation of A
a
consensus branch region and extended polypyrimidine tract - would be critical. Thus, the
sequence requirements of A. probably reflect those of complex A at introns containing
other weak splice site elements. That formation of A is not as dependent upon the above
dynamic processes is likely due to the simplicity of the short consensus substrate RNA. At
the moment, it is conjectured that U2AF 65 and the PUF-2 complex of proteins bind the
short substrate RNA since these proteins tightly bind poly[U] tracts and are required for
complex formation. The other components that subsequently bind the substrate are the 17S
U2 snRNP complex and perhaps other factors (Figure 6). The simplicity of the Ain
complex will facilitate a full analysis of the assembly of pre-spliceosomes.
An active mechanism of U2 snRNP removal.
The A.
complex rapidly dissociates in the presence of ATP. This represents an
active mechanism that removes or destabilizes U2 snRNP from branch sequences. There
171
are several points in the spliceosome cycle at which such a mechanism may be required.
This process might reflect the pathway of U2 snRNP removal from excised lariat introns,
although this seems unlikely as both the chemical nature of the RNA substrate (no branched
nucleotide) and the snRNP complement (U2 vs. U2/6/5) are different. More likely, it
could represent a destabilization of complex A interactions that normally occurs during
formation of spliceosomes; this destabilization process could be a weakening of interactions
needed to progress beyond complex A, or, alternatively, this dynamic process could act as
a proof-reading step. We propose that the ATP-dependent step may test the fidelity or total
stability of the U2 snRNP-RNA complex. If the U2 snRNP complex is not stabilized by
interactions with components recognizing other splicing signals, such as Ul snRNP and
SR proteins bound to nearby sequences, then U2 snRNP would dissociate (Figure 6,
dashed arrow). This mechanism would preclude stable formation of an A-type complex on
sequences fortuitously resembling branch sequences and polypyrimidine tracts (see 57)
found within introns and exons of nuclear precursor mRNAs. The biochemical assay
demonstrated here will allow characterization of this active mechanism and its role in the
splicing process.
ACKNOWLEDGMENTS
We are grateful to J. Crispino for generous sharing of reagents and to M. Moore for
synthesis of oligo-ribonucleotides. We thank D. Bartel, B. Blencowe, J. Crispino, L.
Lim, M. Moore and R. Pulak for their critical review of this manuscript, M. Siafaca for her
patience and ever-present assistance, and the members of the Sharp lab for their continued
interest and support. C.C.Q. is supported by Leukemia Society of America postdoctoral
fellowship (3075-94). This work was supported by United States Public Health Service
grant ROl-GM34277 and ROl-AI32486 from the National Institutes of Health to P.A.S.
and partially by a Cancer Center Support (core) grant P30-CA14051 from the National
Cancer Institute.
REFERENCES
1.
Abmayr, S. M., R. Reed, and T. Maniatis. 1988. Identification of a
functional mammalian spliceosome containing unspliced pre-mRNA. Proc. Natl. Acad.
Sci. USA 85: 7216-7220.
2.
Abovich, N., X. C. Liao, and M. Rosbash. 1994. The yeast
MUD2 protein: an interaction with PRP 11 defines a bridge between commitment
complexes and U2 snRNP addition. Genes & Dev. 8: 843-854.
172
3.
Arning, S., P. Griiter, G. Bilbe, and A. Krimer. 1996.
Mammalian splicing factor SF1 is encoded by variant cDNAs and binds to RNA. RNA 2:
794-810.
4.
Ban, C., B. Ramakrishnan, and M. Sundaralingam. 1994. A
single 2'-hydroxyl group converts B-DNA to A-DNA. Crystal structure of the DNA-RNA
chimeric decamer duplex d(CCGGC)r(G)d(CCGG) with a novel intermolecular G*C basepaired quadruplet. J. Mol. Biol. 236: 275-285.
5.
Barabino, S. M., B. S. Sproat, U. Ryder, B. J. Blencowe,
and A. I. Lamond. 1989. Mapping U2 snRNP-pre-mRNA interactions using
biotinylated oligonucleotides made of 2'-OMe RNA. EMBO J. 8: 4171-4178.
6.
Barabino, S. M. L., B. J. Blencowe, U. Ryder, B. S. Sproat,
and A. I. Lamond. 1990. Targeted snRNP depletion reveals an additional role for
mammalian Ul snRNP in spliceosome assembly. Cell 63: 293-302.
7.
Behrens, S.-E., F. Galisson, P. Legrain, and R. Liihrmann.
1993. Evidence that the 60-kDa protein of 17S U2 small nuclear ribonucleoprotein is
immunologically and functionally related to the yeast PRP9 splicing factor and is required
for the efficient formation of prespliceosomes. Proc. Natl. Acad. Sci. USA 90: 82298233.
8.
Blencowe, B. J., and S. M. L. Barabino. 1995. Antisense affinity
depletion of RNP particles: application to spliceosomal snRNPs. In M. J. Tymms (ed.),
Methods in Molecular Biology, Vol. 37: In Vitro Transcription and Translation Protocols.
Humana Press Inc.: Totowa, N.J. p. 67-76.
Brosi, R., K. Groining, S.-E. Behrens, R. Liihrmann, and A.
9.
Kraimer. 1993. Interaction of mammalian splicing factor SF3a with U2 snRNP and
relation of its 60-kD subunit to yeast PRP9. Science 262: 102-105.
10.
Brosi, R., H.-P. Hauri, and A. Krimer. 1993. Separation of
splicing factor SF3 into two components and purification of SF3a activity. J. Biol. Chem.
268: 17640-17646.
Burgess, S. M., and C. Guthrie. 1993. Beat the clock: paradigms in
the maintenance of biological fidelity. Trends Biochem. Sci. 18: 381-384.
11.
Burgess, S. M., and C. Guthrie. 1993. A mechanism to enhance
mRNA splicing fidelity: the RNA-dependent ATPase Prpl6 governs usage of a discard
pathway for aberrant lariat intermediates. Cell 73: 1377-1391.
12.
173
13.
Champion-Arnaud, P., and R. Reed. 1994. The prespliceosome
components SAP 49 and SAP 145 interact in a complex implicated in tethering U2 snRNP
to the branch site. Genes & Dev. 8: 1974-1983.
14.
Chaudhary, N., C. McMahon, and G. Blobel. 1991. Primary
structure of a human arginine-rich nuclear protein that colocalizes with spliceosome
components. Proc. Natl. Acad. Sci. USA 88: 8189-8193.
15.
Cheng, S. C., and J. Abelson. 1987. Spliceosome assembly in yeast.
Genes & Dev. 1: 1014-1027.
16.
Chiara, M. D., O. Gozani, M. Bennett, P. Champion-Arnaud,
L. Palandjian, and R. Reed. 1996. Identification of proteins that interact with exon
sequences, splice sites, and the branchpoint sequence during each stage of spliceosome
assembly. Mol. Cell. Biol. 16: 3317-3326.
17.
Couto, J. R., J. Tamm, R. Parker, and C. Guthrie. 1987. A
trans-actingsuppressor restores splicing of a yeast intron with a branch point mutation.
Genes & Dev. 1: 445-455.
18.
Crispino, J. D., B. J. Blencowe, and P. A. Sharp. 1994.
Complementation by SR proteins of pre-mRNA splicing reactions depleted of Ul snRNP.
Science 265: 1866-1869.
19.
Crispino, J. D., J. E. Mermoud, A. I. Lamond, and P. A.
Sharp. 1996. Cis-actingelements distinct from the 5' splice site promote Ul-independent
pre-mRNA splicing. RNA 2: 664-673.
20.
Crispino, J. D., and P. A. Sharp. 1995. A U6 snRNA:pre-mRNA
interaction can be rate-limiting for Ul-independent splicing. Genes & Dev. 9: 2314-2323.
21.
Dignam, J. D., R. M. Lebovitz, and R. D. Roeder. 1983.
Accurate transcription initiation by RNA polymerase II in a soluble extract from isolated
mammalian nuclei. Nucleic Acids Res. 11: 1475-1489.
22.
Egli, M., N. Usman, and A. Rich. 1993. Conformational influence
of the ribose 2'-hydroxyl group: crystal structures of DNA-RNA chimeric duplexes.
Biochemistry 32: 3221-3237.
Fu, X. D. 1995. The superfamily of arginine/serine-rich splicing factors.
23.
RNA 1: 663-680.
174
24.
Gaur, R. K., J. Valcaircel, and M. R. Green. 1995. Sequential
recognition of the pre-mRNA branch point by U2AF 65 and a novel spliceosome-associated
28-kDa protein. RNA 1: 407-417.
25.
Gozani, O., R. Feld, and R. Reed. 1996. Evidence that sequence-
independent binding of highly conserved U2 snRNP proteins upstream of the branch site is
required for assembly of spliceosomal complex A. Genes & Dev. 10: 233-243.
Grabowski, P. J., R. A. Padgett, and P. A. Sharp. 1984.
26.
Messenger RNA splicing in vitro: An excised intervening sequence and a potential
intermediate. Cell 37: 415-427.
Hoffman, B. E., and P. J. Grabowski. 1992. Ul snRNP targets an
essential splicing factor, U2AF65, to the 3' splice site by a network of interactions
spanning the exon. Genes & Dev. 6: 2554-2568.
27.
Jamison, S. F., and M. A. Garcia-Blanco. 1992. An ATP28.
independent U2 small nuclear ribonucleoprotein particle/precursor mRNA complex requires
both splice sites and the polypyrimidine tract. Proc. Natl. Acad. Sci., USA 89: 54825486.
Konarska, M. M., and P. A. Sharp. 1986. Electrophoretic separation
of complexes involved in the splicing of precursors to mRNAs. Cell 46: 845-855.
29.
Konarska, M. M., and P. A. Sharp. 1987. Interactions between
30.
small nuclear ribonucleoprotein particles in formation of spliceosomes. Cell 49: 763-774.
31.
Konarska, M. M., and P. A. Sharp. 1988. Association of U2, U4,
U5 and U6 small nuclear ribonucleoproteins in a spliceosome-type complex in absence of
precursor RNA. Proc. Natl. Acad. Sci. USA 85: 5459-5462.
Krimer, A. 1988. Presplicing complex formation requires two proteins
32.
and U2 snRNP. Genes & Dev. 2: 1155-1167.
Krimer, A. 1995. The biochemistry of pre-mRNA splicing. In A. I.
33.
Lamond (ed.), Pre-mRNA Processing. R. G. Landes Company: Austin, TX. p. 35-64.
Krimer, A. 1996. The structure and function of proteins involved in
mammalian pre-mRNA splicing. Ann. Rev. Biochem. 65: 367-409.
34.
Lamond, A. I., B. Sproat, U. Ryder, and J. Hamm. 1989.
Probing the structure and function of U2 snRNP with antisense oligonucleotides made of
35.
2'-OMe RNA. Cell 58: 383-390.
175
Lavigueur, A., H. LaBranche, A. R. Kornblihtt, and B.
36.
Chabot. 1993. A splicing enhancer in the human fibronectin alternate ED1 exon interacts
with SR proteins and stimulates U2 snRNP binding. Genes & Dev. 7: 2405-2417.
Liao, X. C., H. V. Colot, Y. Wang, and M. Rosbash. 1992.
Requirements for U2 snRNP addition to yeast pre-mRNA. Nucleic Acids Res. 20: 42374245.
37.
MacMillan, A. M., C. C. Query, C. R. Allerson, S. Chen, G.
L. Verdine, and P. A. Sharp. 1994. Dynamic association of proteins with the premRNA branch region. Genes & Dev. 8: 3008-3020.
38.
Madhani, H. D., and C. Guthrie. 1994. Dynamic RNA-RNA
interactions in the spliceosome. Annu. Rev. Genet. 28: 1-26.
39.
40.
Manley, J. L., and R. Tacke. 1996. SR proteins and splicing control.
Genes & Dev. 10: 1569-1579.
41.
McCaw, P. S., and P. A. Sharp. In preparation.
42.
Michaud, S., and R. Reed. 1991. An ATP-independent complex
commits pre-mRNA to the mammalian spliceosome assembly pathway. Genes & Dev. 5:
2534-2546.
Moore, M. J., C. C. Query, and P. A. Sharp. 1993. Splicing of
precursors to mRNA by the spliceosome. In R. Gesteland and J. Atkins (ed.), The RNA
World. Cold Spring Harbor Laboratory Press: New York. p. 303-357.
43.
Moore, M. J., and P. A. Sharp. 1992. Site-specific modification of
pre-mRNA: the 2' hydroxyl groups at the splice sites. Science 256: 992-997.
44.
Moore, M. J., and P. A. Sharp. 1993. The stereochemistry of pre45.
mRNA splicing: evidence for two active sites in the spliceosome. Nature 365: 364-368.
Nilsen, T. W. 1994. RNA-RNA interactions in the spliceosome:
unraveling the ties that bind. Cell 78: 1-4.
46.
Padgett, R. A., P. J. Grabowski, M. M. Konarska, S. Seiler,
and P. A. Sharp. 1986. Splicing of messenger RNA precursors. Ann. Rev. Biochem.
55: 1119-1150.
47.
Parker, R., P. G. Siliciano, and C. Guthrie. 1987. Recognition of
the TACTAAC box during mRNA splicing in yeast involves base pairing to the U2-like
snRNA. Cell 49: 229-39.
48.
176
49.
Portmann, S., S. Grimm, C. Workman, N. Usman, and M.
Egli. 1996. Crystal structures of an A-form duplex with single-adenosine bulges and a
conformational basis for site-specific RNA self-cleavage. Chemistry & Biology 3: 173184.
50.
Pruzan, R., H. Furneaux, P. Lassota, G. Y. Hong, and J.
Hurwitz. 1990. Assemblage of the prespliceosome complex with separated fractions
isolated from HeLa cells. J. Biol. Chem. 265: 2804-2813.
Query, C. C., M. J. Moore, and P. A. Sharp. 1994. Branch
51.
nucleophile selection in pre-mRNA splicing: evidence for the bulged duplex model. Genes
& Dev. 8: 587-597.
52.
Query, C. C., S. A. Strobel, and P. A. Sharp. 1996. Three
recognition events at the branch site adenine. EMBO J. 15: 1392-1402.
Ramchatesingh, J., A. M. Zahler, K. M. Neugebauer, M. B.
53.
Roth, and T. A. Cooper. 1995. A subset of SR proteins activates splicing of the
cardiac troponin T alternative exon by direct interactions with an exonic enhancer. Mol.
Cell. Biol. 15: 4898-4907.
54.
Reed, R. 1996. Initial splice-site recognition and pairing during pre-
mRNA splicing. Curr. Opin. Genet. Dev. 6: 215-220.
55.
Ruby, S. W., and J. Abelson. 1988. An early hierarchic role of U1
small nuclear ribonucleoprotein in spliceosome assembly. Science 242: 1028-35.
56.
Ruskin, B., P. D. Zamore, and M. R. Green. 1988. A factor,
U2AF, is required for U2 snRNP binding and splicing complex assembly. Cell 52: 207219.
57.
Senapathy, P., M. B. Shapiro, and N. L. Harris. 1990. Splice
junctions, branch point sites, and exons: sequence statistics, identification, and applications
to genome project. Methods Enzymol. 183: 252-278.
58.
Sraphin, B., and M. Rosbash. 1989. Identification of functional U1
snRNP-pre-mRNA complexes committed to spliceosome assembly and splicing. Cell 59:
349-358.
59.
S6raphin, B., and M. Rosbash. 1991. The yeast branchpoint
sequence is not required for the formation of a stable Ul snRNP pre-mRNA complex and
is recognized in the absence of U2 snRNA. EMBO J. 10: 1209-1216.
177
60.
Staknis, D., and R. Reed. 1994. Direct interactions between premRNA and six U2 small nuclear ribonucleoproteins during spliceosome assembly. Mol.
Cell. Biol. 14: 2994-3005.
Staknis, D., and R. Reed. 1994. SR proteins promote the first specific
recognition of pre-mRNA and are present together with the Ul small nuclear
ribonucleoprotein particle in a general splicing enhancer complex. Mol. Cell. Biol. 14:
7670-7682.
61.
62.
Strobel, S. A., T. R. Cech, N. Usman, and L. Beigelman.
1994. The 2,6-diaminopurine riboside*5-methylisocytidine wobble base pair: an
isoenergetic substitution for the study of G*U pairs in RNA. Biochemistry 33: 1382413835.
Sun, Q., R. K. Hampson, A. T. Krainer, and F. M. Rottman.
1993. General splicing factor SF2/ASF promotes alternative splicing by binding to an
exonic splicing enhancer. Genes & Dev. 7: 2598-2608.
63.
Tarn, W. Y., and J. A. Steitz. 1994. SR proteins can compensate for
64.
the loss of Ul snRNP functions in vitro. Genes & Dev. 8: 2704-2717.
65.
Umen, J. G., and C. Guthrie. 1995. The second catalytic step of premRNA splicing. RNA 1: 869-885.
Valcircel, J., and M. R. Green. 1996. The SR protein family:
pleiotropic functions in pre-mRNA splicing. Trends Biol. Sci. 21: 296-301.
66.
Wang, Z., H. M. Hoffmann, and P. J. Grabowski. 1995.
Intrinsic U2AF binding is modulated by exon enhancer signals in parallel with changes in
67.
splicing activity. RNA 1: 21-35.
Watakabe, A., K. Tanaka, and Y. Shimura. 1993. The role of exon
68.
sequences in splice site selection. Genes & Dev. 7: 407-418.
Wu, J., and J. Manley. 1989. Mammalian pre-mRNA branch site
69.
selection by U2 snRNP involves base pairing. Genes & Dev. 3: 1553-1561.
Zamore, P. D., J. G. Patton, and M. R. Green. 1992. Cloning
and domain structure of the mammalian splicing factor U2AF. Nature 355: 609-614.
70.
Zhang, W.-J., and J. Y. Wu. 1996. Functional properties of p54, a
71.
novel SR protein active in constitutive and alternative splicing. Mol. Cell. Biol. 16: 54005408.
178
Zhuang, Y., A. M. Goldstein, and A. M. Weiner. 1989.
72.
UACUAAC is the preferred branch site for mammalian mRNA splicing. Proc. Natl. Acad.
Sci. USA 86: 2752-2756.
Zhuang, Y., and A. M. Weiner. 1989. A compensatory base change
in human U2 snRNA can suppress a branch site mutation. Genes & Dev. 3: 1545-1552.
73.
Zuo, P., and T. Maniatis. 1996. The splicing factor U2AF 3 5 mediates
critical protein-protein interactions in constitutive and enhancer-dependent splicing. Genes
& Dev. 10: 1356-1368.
74.
179
LEGENDS TO FIGURES
FIGURE 1.
BS-PPT RNA forms an A-like complex with U2 snRNP.
(A) Schematic comparison of RNAs that form complexes A (upper) and A.
(lower). Regions that promote formation of complex A on pre-mRNA are bracketed. SS,
splice site; BS, branch site adenosine; PPT, polypyrimidine tract; Ul compl., region
complementary to Ul snRNA; SF3, binding site for SF3a and SF3b components; U2
compl., region complementary to U2 snRNA; exon seq., exon sequences that typically
include enhancer elements, SR protein binding sites, and/or downstream 5' splice sites
(none of which specifically are known to exist in this 234 nt pre-mRNA).
(B) Co-migration. BS-PPT RNA [RNA(146-179)] (lane 1) or full-length
PIP85.B pre-mRNA (lanes 2 and 3) were incubated in nuclear extract at 300 C for 20 min,
adjusted to 0.5 mg/ml heparin, and separated on a native 4% polyacrylamide gel. A., a
minimal U2 snRNP complex; H, nonspecific complexes.
(C) snRNA composition. Biotinylated RNAs were incubated in nuclear extract at
300 C for 20 min, bound to streptavidin-agarose beads, and washed. Bound complexes
were digested with protease, eluted, separated on a 10% polyacrylamide (19:1) gel,
transferred to Nytran, and probed with antisense RNA probes for Ul, U2, U4, US and U6
snRNAs (30). Lane 1, beads alone; lane 2, biotinylated BS-PPT RNA [RNA(146-179,
bio)]; lane 3, full-length PIP85.B pre-mRNA with biotin incorporated at random positions.
(D) Dependence on snRNPs. BS-PPT RNA was incubated in mock-depleted
extract (lane 1) or extracts depleted of Ul snRNPs (lane 2), U2 snRNPs (lane 3), or U4/6
snRNPs (lane 4), and analyzed as in (A).
180
5' SS
Full-length pre-mRNA
(234 nt)
BS
IU11--1GU-AG
I PPT 1W
UA
EI
U1 compl.
BS-PPT RNA
(34 nt)
0
3' SS
SF3
exon seq.
GGGUGCUGACUGGCUUCUUCUCUCUUUUUCCCUC
U2 compl.
polypyrimidine tract
<
z
-Z
/
C-o
m ~E
ATPAmin-
-
An- B/C
-A
i
-H
H1
D
nuclear
extract
E s
Z
C
Amin-
4l 1
cD E
ul
e,""
-U2
-Ul
-U4
-U5
-U6
H1234
1
2
3
FIGURE 2. Both branch sequence and polypyrimidine tract are required in
cis.
(A) RNA(146-179) with wild-type branch sequence and polypyrimidine tract (BSPPT RNA, lane 1), with a mutated polypyrimidine tract
(5'-...CUUCUUCUCUCUUUUUCCCUC-3' -5'-...GACGGACAUGCAAUGCAACUC-3', lane 2), or with scrambled branch sequence
(5'-...UGCUGAC...-3' -,
5'-...GUCGUAC...-3', lane 3) were incubated in nuclear
extract at 30 0 C for 20 min and analyzed as in Figure IB.
(B) Labeled branch sequence RNA (5'-GGGUGCUGAC-3', lanes 1-5), labeled
polypyrimidine-tract RNA (5'-UGGCUUCUUCUCUCUUUUUCCCUC-3', lanes 6-10),
or labeled BS-PPT RNA (lanes 11-20) were incubated in nuclear extract at 30 0 C for 20
min in the presence of 0 tM (lanes 1, 6, 11, 16), 0.001 gM (lanes 2, 7, 12, 17), 0.01 tM
(lanes 3, 8, 13, 18), 0.1 giM (lanes 4, 9, 14, 19), or 1 LM (lanes 5, 10, 15, 20) cold
competitor RNA. Competitor RNAs were either polypyrimidine-tract RNA (lanes 1-5,
11-15) or branch sequence RNA (lanes 6-10, 16-20). Reactions were adjusted to 0.5
mg/ml heparin and analyzed as above.
(C) BS-PPT RNA [RNA(146-179)] (lane 1) or PPT-BS RNA [RNA(156-179,
146-155)] (lane 2) were incubated in nuclear extract at 300 C for 20 min and analyzed as
above.
182
B
A
labeled RNA -
BS
r-
PPT
BS-PPT
Amin
cold RNA -
Amin -
~ ~
Amin-
Amin-
Hf
H-1
1 2 3 4 5
123
6 7 8 9 10
11 12 13 14 15
16 17 18 18 20
1 2
FIGURE 3.
Characteristics of
Ain
complex.
(A) Dependence on ionic strength. BS-PPT RNA was incubated in extracts
adjusted to the KCl concentration indicated (mM), and adjusted to 0.5 mg/ml heparin, and
separated on a native 4% polyacrylamide gel.
(B) Stability. BS-PPT RNA was incubated in nuclear extracts depleted of ATP for
20 min to form A., complexes, then challenged with 1 nmol/ml cold competitor
RNA(146-179), reincubated for the time course indicated, analyzed as above (lanes 1-7).
Alternatively, the cold competitor BS-PPT RNA was incubated first for 20 min, and
labeled BS-PPT RNA was then added and reincubated for the times indicated (lanes 8-14).
(C) Dependence on U2AF 65 and PUF-2 (poly[U]-binding factor-2). BS-PPT
RNA was incubated in mock-depleted extract (lane 1) or extract depleted of poly[U]binding proteins (lane 2); or poly[U]-depleted extract supplemented with a PUF-2 fraction
(lane 3), supplemented with recombinant U2AF 65 (lane 4), or supplemented with both
recombinant U2AF 65 and the PUF-2 fraction (lane 5) and analyzed as in (A) above.
(D) Proteins that cross-link in An,. complexes using a 15 A probe. BS-PPT RNA
modified at the N6 position of the branch-site adenosine to contain benzophenone (38)
was incubated to form A. complexes, UV irradiated at 302 nm and separated on a native
polyacrylamide gel. Complexes were isolated and treated with RNase, and the proteins
subsequently separated on a 16% (200:1) polyacrylamide-SDS gel.
(E) Proteins that cross-link in A. complexes within 2 A of the branch site. BSPPT RNA modified at the branch site to contain 2,6-diaminopurine (52) was incubated to
form A. complexes, UV irradiated at 254 nm, and analyzed as in (D) above.
184
[KCI] -
20 60 100
20250300
I
Amin-
chase with
cold compet.
cold
compet. first
time (hours) - '0.25 .5 1 1.5 2 4
0 .25 .5 1 1.5 2 4
500
of chase
Amin -
Io
H-
H- t
1 2 3 4 5 6 7
1 23456789
E
Dap
Benzophenone
r nuclear _
extract
Amin-
H
Amin
200 I
.
46-
r ,---p35
30 -
I
14-
1
45
10069-
46-
123
1
H Amin
I
S-- p150
20010069-
8 9 1011 121314
- -p14
30--
-
14-
1
2
p1 4
FIGURE 4.
Independence of Amin complex formation on the presence of
ATP and dissociation of Amin complex in the presence of ATP.
(A) Time course of complex assembly for BS-PPT RNA (lanes 1-21) or for fulllength PIP85.B pre-mRNA (lanes 22-27) in the presence of ATP (lanes 1-7, 22-23),
absence of ATP (lanes 8-14, 24-25), or absence of ATP and presence of EDTA (lanes 1521, 26-27). RNAs were incubated for the times indicated as described in Materials and
methods, adjusted to 0.5 mg/ml heparin, and separated on a native 4% polyacrylamide gel.
A n, a minimal U2 snRNP complex; B/C, spliceosomal complexes B and C containing
U2/4/5/6 snRNPs and pre-mRNA; A, pre-spliceosomal complex A containing U2 snRNP
and pre-mRNA; H, nonspecific complexes; *, a faster migrating complex observed at low
levels in the presence of ATP.
(B) upper. BS-PPT RNA was incubated in nuclear extract depleted of ATP for
20 min to form A n complex (lane 1), then chased with 1 nmol/ml cold competitor RNA
and reincubated for the time courses shown either in the absence (lanes 2-7) or presence of
ATP (lanes 8-13). Reactions then were adjusted to 0.5 mg/ml heparin and loaded onto a
native 4% polyacrylamide gel. lower. The RNA in samples from the above reactions was
analyzed on a 15% (19:1) 8 Murea gel.
(C) BS-PPT RNA (lanes 1-8) or BS-PPT-3'Exon RNA [RNA(146-234); lanes
9-16] were incubated in nuclear extract depleted of ATP for 20 min to form Amn or An
like complexes, respectively. Cold competitor RNA was added [cold BS-PPT RNA for
lanes 1-8, or cold RNA(146-234) for lanes 9-16] and reactions were reincubated at 300 C
in the presence of ATP for the time courses indicated (lanes 3-8 and 11-16) or in the
absence of ATP for 60 min (lanes 2 and 10). Lanes 1 and 9, no reincubation. Reactions
were then analyzed as above.
186
FIGURE 4. (continued)
(D) Graph of kinetics of formation and dissociation of A n complexes. For
formation, complexes were assembled on BS-PPT RNA in the presence of ATP [--0-curve a; as in (A) lanes 1-7] or in the absence of ATP [--- curve b; as in (A) lanes 1521]. For dissociation, Ai n complexes were first assembled by incubation for 30 min in
nuclear extract depleted of ATP, and then reincubated in the presence of ATP [---curve c; as in (B) lanes 8-13]. Relative complex formation was determined as the fraction
of A . complex relative to the input RNA. Polyacrylamide gels were quantitated using a
Molecular Dynamics PhosphorImager and ImageQuant software version 3.22.
187
BS-PPT RNA
time(min) -
Amin -
+ATP
t o In)-.oo
soo
1 2 5 10
-ATP
I 012510--
20 30' '0
.1
pre-mRNA
-ATP/+EDTA
1 2 5 10 20 30
01261O0 1 2 5 10
2
30
-
+
time (min)
of chase
-1+
is
0 ~0200 20'
(
Amin
-ATP
+ATP
0 2 5 102060
0 2 5 102060
""aI1
00
2 3 4 5 6 7
8 9 10
11 1213
-B/C
$A
1
time(min) 1 2 3 4 5 5 7
8 91011121314
15161718192021
ofchase
222324252627
-ATP
+ATP
0 2 5 1020 o
0 2 5 10 20 o
1 2 3 4 5 6 7
8 910111213
BS-PPT RNA --
RNA -
BS-PPT
BS-PPT-3'Exon
+ATP
time (min)
of chase
P 60
2 5 10 2060
I
i
Amin
+ATP
-ATP
+ATP
2 5 102060
8
D
dki
Sa (+ATP
i'in
0
10
20
30
30
50
70
time (min)
123456
s
78
9 101112 13141516
Formation
D issociat ion
90
FIGURE 5. Time course of complex assembly for RNA oligonucleotides
containing 2'-H substitutions.
BS-PPT RNAs containing an all-ribose branch sequence (lanes 1-7), two
2'-deoxynucleotides at the branch site and immediately 5'-adjacent (UGCUGHAHC ; lanes
8-14), or two 2'-deoxynucleotides 5'-distal to the branch position (UGHCHUGAC ; lanes
15-21) were incubated for the times indicated as described in Materials and methods,
adjusted to 0.5 mg/ml heparin, and loaded onto a native 4% polyacrylamide gel. See Table
I for quantitations of effects of these and other substitutions.
189
all-ribo
double deoxy
double deoxy
-
-UGCUGAC -
-UGCUGAc
0 1 2 5 102030
0 1 2 5 102030
0 1 2 5 102030
-UGCUGAc
time (min) -
-
S.
Amin -
I
H1 2 3 4 5 6 7
8 9 10 11 12 13 14
15 16 17 18 19 20 21
FIGURE 6.
Summary of formation of complexes A (left) and Amin (right).
Schematic comparison of assembly of complexes A and At. At top of the panels
are shown diagrams of full-length pre-mRNA and the minimal BS-PPT RNA substrates.
Both complexes require branch sequence and polypyrimidine-tract RNA elements, U2
snRNP, and protein factors U2AF 65 and PUF-2. Complex A requires U1 snRNP and
ATP, which are not necessary for An. formation. In both complexes A and An., three
proteins are detected within 15 A of the branch-site adenosine - p14, p35, and p150; the
pl14 cross-links directly to the adenosine in both complexes. In the presence of ATP, U2
snRNP is released from A. complexes (dashed arrow). Other components not shown,
such as SR proteins (23, 40, 66) and SF1 (3, 31), are important for complex A formation
in other systems and may also be required for A. n. The arrangement of proteins is
illustrative only, and not meant to imply a known spatial order. SS, splice site; BS, branch
site adenosine; PPT, polypyrimidine tract; PUF-2, poly[U]-binding factor-2; Ul, U1
snRNP; U2, U2 snRNP; CC/E, commitment or early complex; A, pre-spliceosomal
complex A containing U2 snRNP and pre-mRNA; A., a minimal substrate RNA-U2
snRNP complex.
191
M
5' SS
BS
1
1
3'ISS
BS
PPT
GU
AM
Full-length pre-mRNA
(234 nt)
1 PPT
AAG
-A"
BS-PPT RNA
(34 nt)
CC/E
-A
VIP
AT P
\ATP
/
Amin
TABLES
TABLE 1. Relative yields for Am,i complex formation of modified
substrates.
Modification
Branch
Relative Am
sequence a
complex formationb
all-ribose
UGCUGAC
1.0
2'-deoxyribose
UGCUGAHC
0.62
2'-deoxyribose
UGCUGHAC
0.80
2'-deoxyribose
UGHCUGAC
0.88
double
2'-deoxyribose
UGCUGHAHC
0.03
double
2'-deoxyribose
UGHCHUGAC
0.30
double
UGHCUGHAC
0.59
2'-deoxyribose
193
aSite-specific 2'-deoxyribose modifications at or near to the branch site are indicated
by the superscripted letters.
bRelative complex formation was determined as the fraction of A. complex formed
during a time course relative to the input RNA and normalized to the respective all-ribosecontaining RNA. Polyacrylamide gels were quantitated using a Molecular Dynamics
PhosphorImager and ImageQuant software version 3.22. For each band in every lane an
individual background value was determined from the area in the same lane immediately
above or below that band.
194
SPECULATIVE APPENDIX:
A PROPOSED INTERACTION
BETWEEN THE BRANCH ADENOSINE AND THE U5
LOOP NUCLEOTIDE URIDINE 4,
A MECHANISM TO
JUXTAPOSE THE SUBSTRATES OF FIRST STEP OF premRNA SPLICING
Patrick Schonleber McCaw and Phillip A. Sharp
195
While much is known about the constituents and secondary structural elements that
are found in the catalytic spliceosome, many questions remain about what is the chemical
mechanism of splicing. It is not yet clear, for example, whether the splicing reaction is
catalyzed by RNA or by protein. The mechanism by which the first step and second step
substrates are positioned is also not known. A model for the structure of the active site has
been proposed (Steitz, 1992), but it remains controversial. What is clear is that RNA
secondary structural elements are important and conserved features of the spliceosome
(Madhani and Guthrie, 1994; Moore et al., 1993). These known secondary structural
features do not suggest a mechanism for the juxtaposition of the 2' hydroxyl nucleophile of
the branch adenosine with the phosphodiester bond of the 5' splice site (Figure 1, reprinted
from (Moore et al., 1993)). Juxtaposition of the two substrates of the first step (the 2'
hydroxyl of the branch adenosine and the phosphodiester bond of the 5' splice site) and the
two substrates of the second step (the 3' hydroxyl of the 5' terminal exonic nucleotide and
the phosphodiester bond of the 3' splice site) will be an essential feature of any successful
model for the structure of the catalytic core of the spliceosome. This speculative appendix
will briefly describe a model that juxtaposes the branch adenosine 2' hydroxyl nucleophile
and the phosphodiester bond of the 5' splice site. Before the model is described a different
representation of the secondary structural elements than the one shown in figure 1 will be
described.
A NEW REPRESENTATION OF THE SPLICEOSOMAL SECONDARY STRUCTURE
A rearrangement of the secondary structure diagram of the group I ribozyme,
proved to be a useful heuristic tool in understanding the structure of the group I ribozyme
(Cate et al., 1996; Cech et al., 1994). I have arranged the standard secondary structure of
the spliceosome (Madhani and Guthrie, 1994; Moore et al., 1993) in a way that mimics the
method used for the group I ribozyme (Figure 2). The RNAs are indicated as vertical lines,
the pre-mRNA is shown as a thick line and the snRNAs are shown as thin lines. Base
pairs are indicated by horizontal lines. The rearrangement organizes the known helices in
three columns. On the left is shown the 5' splice site helices with U5 snRNA (U5:5'ss)
(Cortes et al., 1993; Newman and Norman, 1992) and U6 snRNA (Lesser and Guthrie,
1993; Sawa and Shimura, 1992). In the center is shown the helices formed between U6
snRNA and U2 snRNA, both at the top (Madhani and Guthrie, 1992) and at the bottom
(Sun and Manley, 1995) of the page. On the right is shown the U2 snRNA helix with the
196
branch sequence (U2:BS). The branch adenosine is shown flipped out of the U2:BS helix.
The branch adenosine is thought not to be base-paired to U2 snRNA for either chemical
step (Query et al., 1994). The 2' hydroxyl and the phosphodiester bond of the 5' splice
site are indicated. The position of the introns present in U6 genes are indicated by arrows
(Tani and Ohshima, 1989). These introns are suggested to be the result of a trans-splicing
reaction into a U6 molecule that was subsequently incorporated into the genome and so are
thought to lie near to the active center of the catalytic spliceosome. The interaction between
U2 snRNA A25 and U6 snRNA G52 deduced from genetic co-variation (Madhani and
Guthrie, 1992) is indicated by the gray line. Crosslinking detected between the pre-mRNA
terminal nucleotide of the 5' exon and U5 snRNA is indicated by gray lines (Sontheimer
and Steitz, 1993; Wyatt et al., 1992). Nucleotides that are important in yeast for the first
step of splicing are in gray boxes and U2 (Fabrizio and Abelson, 1990). The principal
advantage to this secondary structural representation is that residues important for the first
step of splicing are seen to cluster. This is even more apparent when the proposed
interaction of Madhani and Guthrie is taken into account. This interaction would fold the
top part of the U2:U6 helix back over the page and bring the boxed residues shown in
figure 2 at the top of the page close to the center of the page where both the branch
adenosine and the 5' splice site are found.
This new diagrammatic arrangement of the secondary structural elements does not
itself solve the juxtaposition problem; the branch adenosine 2' hydroxyl is shown on the far
right of the diagram, while the 5' splice site phosphodiester bond is shown on the left hand
side of the diagram.
DESCRIPTION OF THE MODEL
The proposed model for the secondary structure of the catalytic spliceosome
requires that the branch adenosine (Ab) stacks between the last nucleotide of the exon (G-1)
and the first nucleotide of the intron (G,). That is it stacks into to the 5'ss:U5 helix. Based
on the genetic evidence and crosslinking evidence, I propose that the branch adenosine
base-pairs to Uridine 4 of the U5 snRNA loop. Base-pairing of the branch adenosine and
Uridine 4 displaces the G-_:Uridine 4 base-pair, forcing G_1 to base-pair with Uridine 5. This
interaction is shown in figure 3 and the Ab:Uridine 4 base pair is indicated in bold. This
base-pair interaction juxtaposes the 2' hydroxyl and the splice site phosphodiester bond in a
way that is consistent with the known stereochemistry of splicing (Maschloff and Padgett,
1992; Moore and Sharp, 1993) and aligns the 2' hydroxyl nucleophile for attack of the
phosphodiester bond of the 5' splice site, discussed in greater detail below. It should be
197
noted that the U5 loop contains a series of four uridines (Uridine4 to Uridine7) and in some
of the mutations of the U5 loop, discussed below, the register of the base-pairing scheme is
not conserved. In these cases, the branch adenosine is proposed to base-pair either to
Uridine 5 or to Uridine 6. Evidence that supports the 5'ss:U5 interaction in the context of the
Ab:Uridine4 base-pair will be discussed. The experiments performed to date do not
directly test the model, however the evidence is consistent with the model. I will conclude
with a brief description of experiments that may be done to test the model.
The branch adenosine is remarkably conserved. It is perhaps the best conserved
nucleotide in the vertebrate pre-mRNA, but surprisingly, it is not known to be base-paired
to any of the snRNAs at any point in spliceosome assembly or catalysis. The branch
adenosine conservation may be due to a required protein interaction; alternatively, a branch
adenosine functional group could play a direct role in catalysis or the branch adenosine
could base-pair to another nucleotide. It seems unlikely that the branch adenosine base is
directly involved in catalysis as many of the substituents of the base can be changed and the
first step of splicing can occur (Query et al., 1996). Most other highly conserved
nucleotides of the spliceosome have been shown to base pair with other nucleotides, for
example the 5' splice site sequences base pair to U 1 snRNA and to U6 and U5 snRNA,
conserved nucleotides of U2 and U6 snRNAs are known to basepair (Madhani and
Guthrie, 1994). A role for branch nucleotide-protein interaction cannot be ruled out, and in
fact, the branch nucleotide is in contact with several proteins during assembly and probably
in the active spliceosome (MacMillan et al., 1994; Query et al., 1996). Notably, both
guanosine and 2-aminopurine can act as the branch nucleophile, both should be able to base
pair with Uridine4.
STRUCTURAL CONSIDERATIONS
Unstacking an unpaired nucleotide from a helix is thermodynamically unfavorable
due in large part to the loss of base-stacking interactions. How might the cost of
unstacking this base effect the model? It is not known what the energetic cost is of moving
an unpaired nucleotide from one helix into another. For the branch adenosine unstacking
from the U2-pre-mRNA helix and base-pairing with Uridine4 while stacking into the
5'ss:U5 helix, the thermodynamics might be favorable as two hydrogen bonds would be
gained upon base-pairing with Uridine4 and none would be lost by unstacking from the U2
helix. Moving the phosphodiester backbones of the two helices close enough to allow this
interaction will be energetically unfavorable; however, this interaction must occur whether
198
the branch adenosine is base-paired with Uridine 4 or not, as the 2' hydroxyl and its
adjacent 3' and 5' phosphates must approach the phosphodiester bond of the 5' splice site.
A model for a branch adenosine that is bulged from the bs:U2 helix is provided by
Portmann et al. who solved the crystal structure of duplex RNA-DNA containing an
unpaired adenosine (Portmann et al., 1996). In this structure, the helix from which the
adenosine is bulged stacks normally, the adenosine is bulged from the helix and is stacked
on a bulged adenosine of an adjacent helix. Although the crystal structure provides no
measure of the thermodynamic cost of unstacking, it suggests that bulged adenosines can
be removed from the helix. Furthermore, the presence of a single base on which to stack,
and not a helix may be sufficient to stabilize such a structure. The base-stacking interaction
observed was believed to occur in solution as well as in the crystal (Portmann et al., 1996).
This result argues that the branch adenosine can be unstacked and bulged, it could then
stack into an adjacent helix. Portmann et al. also point out that the 2' hydroxyl is exposed
upon unstacking and bulging, possibly facilitating its role as a nucleophile.
Why would the branch adenosine interact with Uridine4? In order for the branch
adenosine to base pair with Uridine 4, the 5' exonic terminal nucleotide must shift register
one nucleotide in the 3' direction on U5 snRNA. This means that G_1, the exonic
nucleotide, will base pair with Uridine 5 of U5 snRNA stretching the phosphodiester bond
at the 5' splice site. If the branch adenosine enters the 5' ss:U5 helix from the major
groove, the 2' hydroxyl is brought into apposition with the phosphodiester bond stretched
across this base. The 2' hydroxyl nucleophile is arranged in this structure in a way that
facilitates direct, in-line attack of the phosphodiester bond of the 5' splice site stretched
across the branch adenosine basepair position. This stretched phosphodiester is itself not
unprecedented, as intercalating DNA dyes should stretch the helix by an equivalent
distance. Further support can be found in the crystal structure of a four stranded DNA
molecule in which the two DNA helices are coaxial and the bases of each helix are
intercalated with the bases of the other helix (Gehring et al., 1993).
GENETIC EXPERIMENTS THAT ARE CONSISTENT WITH THE MODEL
Is a base-pair between G_- and Uridine, predicted or allowed by the known
interactions of this base and U5 snRNA as determined by genetic and biochemical
experiments? There is genetic evidence that the branch adenosine is base-paired with
Uridine 4 in both yeast and mammalian systems, though the evidence is indirect. I will
discuss the evidence presented by Newman and Norman (Newman and Norman, 1992). In
these experiments Newman and Norman use an activated cryptic 5' splice site to identify
199
the nucleotides of the U5 snRNA loop that are important in mediating splice site
determination. U5 snRNA molecules that have a randomized U5 loop were selected and
the sequence of the loops was determined (shown in figure 5). Seven loop sequences were
identified; the consensus sequence of these loops is 5'
GCNNUAUYC 3 '. Position 6, an
invariant A (underlined), is interpreted to be sufficient for activating the cryptic 5' splice
site used in these experiments, as Adenosine 6 would base-pair with the unusual exonic base
U_-. The adjacent invariant position, Uridine 8, base-pairs with the 5' exon position A-2.
Uridine5 is described as basepairing with G1 of the 5' splice site sequence. Why is
Uridine5 an invariant uridine and not cytosine in every U5 loop sequenced, if position 5
interacts with G, and only G, in these reactions? The branch adenosine model predicts that
position 5 must base-pair to both G, and the branch adenosine and so must be a uridine and
cannot be a cytosine as uridine and not cytosine can base pair to both the branch adenosine
and G1 . Newman and Norman test mutants of the U5 loop sequence against mutations
made in the 5' exon sequences of the cryptic splice site. Each of the splicing phenotypes of
these mutations is consistent with a branch adenosine-Uridine 4 or Uridine5 base-pairing
interaction. None of the mutations made directly tests the proposed model.
Similar experiments have been performed by Cortes et al. in mammalian cells
(Cortes et al., 1993). In this experiment mutations of the U5 loop were made and the
cryptic 5' splice site that was activated by the mutant U5 in the presence of a mutant 5'
splice site was identified and sequenced. In only one case, does the addition of the branch
adenosine to the 5'ss:U5 helix decrease the number of hydrogen bonds in the helix;
splicing to this cryptic 5' splice site is very weak. In the other cases, addition of the branch
adenosine increases or does not change the total number of hydrogen bonds in this helix.
In some cases where the U5 loop Uridine 4 was mutant Uridine 6 was the base-pairing
partner of the branch adenosine, more often it was Uridine5 .
Deletion of the U5 loop does not prevent splicing (O'Keefe et al., 1996). This
unexpected result would seem to contradict the proposed model. However, the fact that a
reaction is robust to the deletion of a well established interaction; here, the interaction of the
5' splice site sequence with the U5 loop, should not be interpreted to mean that the
interaction does not occur or that it is unimportant. There are two examples of redundant
function in splicing already described; first, the interaction of U1 snRNA with the 5' splice
site is known to be redundant in splicing (Crispino et al., 1994) and, second, the
interaction of U2AF with the pyrimidine tract is known to be redundant as well (chapter 4).
The 5'ss:U5 helix has been demonstrated both genetically and biochemically, described
below, in both vertebrates and in yeast. This conserved interaction is likely to occur,
200
though it may not be absolutely required, other factors, such as the protein p200/PRP8,
may substitute for this interaction to align the 5'ss:U5 helix. The 5'ss:U5 helix, present
during catalysis, is known not to inhibit splicing (Sontheimer and Steitz, 1993).
Sontheimer et al. demonstrate that the terminal nucleotide of the exon can be crosslinked to
the U5 loop early in the splicing reaction and that this crosslink does not inhibit the splicing
reaction.
BIOCHEMICAL EVIDENCE
Site-specific crosslinking experiments also support a base-pairing interaction
between the U5 loop and a branch adenosine. In the first experiment Wyatt et al. use a
thio-uridine at the -2 position of the exon, tU_2, they observe a strong crosslink to Uridine6
of the U5 loop and weaker crosslinks to adjacent residues of the U5 loop (Wyatt et al.,
1992). Again demonstrating the close proximity of the U5 loop and the 5' splice site
sequence. These experiments were extended by positioning the thio-uridine at positions -1
and +2 of the 5' splice site and -1 of the 3' exon (Sontheimer and Steitz, 1993).
Crosslinks of the 5' splice site -1 position were mapped to two positions in the U5 loop
Uridine4 and Uridine 5. This is consistent with the model in that it is expected that prior to
the branch adenosine-Uridine 4 base-pairing that G_1 will base-pair to Uridine4 and
subsequently be displaced to Uridine 5. Based on this model, we would expect that
crosslinks formed between tU_, and Uridine 5 will splice significantly more efficiently than
crosslinks formed between tU_1 and Uridine4, as crosslinks to Uridine4 should inhibit
branch adenosine intercalation with the 5' splice site sequence.
The stereochemistry of the first and second steps of splicing has been determined
(Maschloff and Padgett, 1993; Moore and Sharp, 1993). Is the model consistent with the
stereochemistry of the first step? It is known that the Rp phosporotioate diastereomer and
not the Sp diastereomer inhibits splicing. This has been interpreted to mean that the Rp
diastereomer may be bound to Mg +2 (Moore and Sharp, 1993). A plastic model built to
represent the U5 loop-5' splice site branch adenosine helix demonstrated that the Spoxygen is directed in to the center of the 5 'ss:U5 helix and is unlikely to be bound by a
metal due to steric constraints. In contrast, the Rp-oxygen faces out toward the U2
snRNA-branch sequence helix where it is entirely possible that it interacts with a metal.
The model is shown in figure 4. To distinguish the Rp from the Sp oxygens in the figure,
the Sp oxygen is a small white ball and the Rp oxygen is indicated with an arrow.
201
EXPERIMENTS THAT TEST THE MODEL
Perhaps the easiest and most direct test of model is to repeat the experiments of
Newman and Norman described above (Newman and Norman, 1992), using a mutant
branch residue. It is known that a G at the branch position will complete the first step of
splicing in vertebrates (Query et al., 1995), if this is true for yeast as well, then it might be
possible demonstrate a compensatory mutagenesis between position 4 or 5 of the U5 loop
and the branch position. It may also be possible to use a 4-thiouridine derivitized U5 loop
to demonstrate the proposed base-pairing interaction, while tU favors RNA-RNA
crosslinks in regions of non-Watson-Crick interaction, A:tU crosslinks have been observed
(Sontheimer and Steitz, 1993).
ACKNOWLEDGMENTS:
This speculation is the result of many long, productive conversations with Charles
Query and Andrew MacMillan. They have been great teachers. I am forever grateful for
their patience and kind criticism. While any credit due this speculation belongs equally to
them, its inevitable shortcomings are a reflection of the author, and not the teachers. The
errors in reasoning and judgment are solely my own.
202
REFERENCES
Cate, J. H., Gooding, A. R., Podell, E., Zhou, K., Golden, B. L., Kundrot, C. E.,
Cech, T. R., and Doudna, J. A. (1996). Crystal structure of a group I ribozyme domain:
principles of RNA packing. Science 273, 1678-85.
Cech, T. R., Damberger, S. H., and Gutell, R. R. (1994). Representation of the
secondary and tertiary structure of group I introns. Nat Struct Biol 1, 273-80.
Cortes, J. J., Sontheimer, E. J., Seiwert, S. D., and Steitz, J. A. (1993). Mutations in the
conserved loop of human U5 snRNA generate use of novel cryptic 5' splice sites in vivo.
EMBO J 12, 5181-9.
Crispino, J. D., Blencowe, B. J., and Sharp, P. A. (1994). Complementation by SR
proteins of pre-mRNA splicing reactions depleted of Ul snRNP. Science 265, 1866-9.
Fabrizio, P., and Abelson, J. (1990). Two domains of yeast U6 small nuclear RNA
required for both steps of nuclear precursor messenger RNA splicing. Science 250, 404409.
Gehring, K., Leroy, J. L., and Gueron, M. (1993). A tetrameric DNA structure with
protonated cytosine.cytosine base pairs. Nature 363, 561-5.
Lesser, C. F., and Guthrie, C. (1993). Mutations in U6 snRNA that alter splice site
specificity: implications for the active site. Science 262, 1982-8.
MacMillan, A. M., Query, C. C., Allerson, C. R., Chen, S., Verdine, G. L., and Sharp,
P. A. (1994). Dynamic association of proteins with the pre-mRNA branch region. Genes
& Dev. 8, 3008-3020.
Madhani, H. D., and Guthrie, C. (1994). Dynamic RNA-RNA interactions in the
spliceosome. Annu Rev Genet 28, 1-26.
Madhani, H. D., and Guthrie, C. (1992). A novel base-pairing interaction between U2 and
U6 snRNAs suggests a mechanism for catalytic activation of the spliceosome. Cell 71,
803-817.
Maschloff, K. L., and Padgett, R. A. (1992). Phosphorothioate substitution identifies
phosphate groups important for pre-mRNA splicing. Nucleic Acids Res. 20, 1949-1957.
203
Maschloff, K. L., and Padgett, R. A. (1993). The stereochemical course of the first step of
pre-mRNA splicing. Nucleic Acids Res. 21, 5456-5462.
Moore, M. J., Query, C. C., and Sharp, P. A. (1993). Splicing of precursors to mRNA
by the spliceosome. In The RNA World, R. Gesteland and J. Atkins, eds. (New York:
Cold Spring Harbor Laboratory Press), pp. 303-357.
Moore, M. J., and Sharp, P. A. (1993). Evidence for two active sites in the spliceosome
provided by stereochemistry of pre-mRNA splicing. Nature 365, 364-8.
Newman, A., and Norman, C. (1992). U5 snRNA interacts with exon sequences at 5' and
3' splice sites. Cell 68, 743-754.
O'Keefe, R. T., Norman, C., and Newman, A. J. (1996). The invariant U5 snRNA loop
1 sequence is dispensable for the first catalytic step of pre-mRNA splicing in yeast. Cell
86, 679-89.
Portmann, S., Grimm, S., Workman, C., Usman, N., and Egli, M. (1996). Crystal
structures of an A-form duplex with single-adenosine bulges and a conformational basis for
site-specific RNA self-cleavage. Chemistry & Biology 3, 173-184.
Query, C. C., Moore, M. J., and Sharp, P. A. (1994). Branch nucleophile selection in
pre-mRNA splicing: evidence for the bulged duplex model. Genes & Dev. 8, 587-597.
Query, C. C., Strobel, S. A., and Sharp, P. A. (1995). The branch site adenosine is
recognized differently for the two steps of pre-mRNA splicing. Nucleic Acids Symp Ser,
224-5.
Query, C. C., Strobel, S. A., and Sharp, P. A. (1996). Three recognition events at the
branch-site adenine. EMBO J 15, 1392-402.
Sawa, H., and Shimura, Y. (1992). Association of U6 snRNA with the 5'-splice site
region of pre-mRNA in the spliceosome. Genes & Dev. 6, 244-254.
Sontheimer, E. J., and Steitz, J. A. (1993). The U5 and U6 small nuclear RNAs as active
site components of the spliceosome . Science 262, 1989-96.
Steitz, J. A. (1992). Splicing takes a holliday. Science 257, 888-9.
Sun, J. S., and Manley, J. L. (1995). A novel U2-U6 snRNA structure is necessary for
mammalian mRNA splicing. Genes Dev 9, 843-54.
204
Tani, T., and Ohshima, Y. (1989). The gene for the U6 small nuclear RNA in fission yeast
has an intron. Nature 337, 87-90.
Wyatt, J. R., Sontheimer, E. J., and Steitz, J. A. (1992). Site-specific cross-linking of
mammalian U5 snRNP to the 5' splice site before the first step of pre-mRNA splicing.
Genes Dev 6, 2542-53.
205
FIGURE LEGENDS
Figure 1.
Known base-pairing interactions of U1, U2, U5, and U6
The conventionally drawn base pairing interactions of the pre-mRNA and U 1, U2, U5 and
U6 are shown. Reprinted from Moore et al. (1993).
206
caaagagAuuuaUUucgUUUU>p
GuuUCUcuaag cA
U2
G
Figure 2.
The rearranged base-pairing interactions.
The basepairing interactions between the pre-mRNA and Ul, U2, U5 and U6 are shown.
The U2:U6 helices extend beyond the top and bottom of the page. The U6 stem-loop, in
figure 1, is not shown, but begins at the top of the page.
208
U25'
C*
1..
U6 3'
I I
00000
I
sss9la~8ossp~ssss~Bs~
intron 3'
I
I
I
4
rQ
I
Vo
oCo
1
I
v
0*
I II" " I"
1
p
I
XN1I
00
00
U5 5'
I I I I I l
U5 3'
I
"D
.I.iL..T
.
0O
|I/I
SO
@
00
U2 3'
D
D
I I I I I I
U6 5'
Figure 3. The proposed base-pairing interaction between the branch
adenosine and Uridine4.
The branch adenosine is shown basepairing with Uridine 4 of the U5 stem loop. Note that
the exonic G (G,) has been displaced one nucleotide 3' on the U5 loop to Uridine 5.
210
U2 5
l~~lI
I I
I
QUD~ 0
U6 31
IIl
intron 3'
i
fzj 0
D~~0D~D
U
C)
00
0
-0
z
U5 5
ii i I I
LD ID I I I
U5 3'
D
-,qq
I
U2 3
u 0 0
Figure 4.
Photographs of the model of the proposed structure.
A. The 5' splice site G-l/G1 U5 loop helix with the intercalated branch adenosine (Ab) is
shown, view is in to the major groove. The pre-mRNA:U2 snRNA helix is shown to the
left. The boxed region is shown in greater detail in B.
B. Close-up view of the 5' splice site phosphodiester bond and the branch adenosine
ribose sugar. The Rp oxygen is indicated, the Sp oxygen is facing out of the page.
212
p
H,OIZ.
. . -----
Figure 5. Proposed base-pairing interactions of the selected U5 snRNA
molecules in the Experiment of Newman and Norman and selected 5'splice
site sequences of Cortes et al.
Selected U5 loop sequeces are shown in the top half of the figure, the consensus of the
selected sequences is indicated and the sequence of the 5' splice site and stacked branch
adenosine is shown. Adapted from Newman and Norman (1992), figure 2. Below the
dashed line is the 5' splice site selections with mutant and wild type U5 snRNA of Cortes
et al.. Branch adenosine is in outline font, exon sequences are underlined and proposed
base pairs are indicated by a dash, these base pairs include G:U pairs. In the Cortes
experiment the mutant U5 bases are indicated as plain rather than bold text.
214
pre-mRNA
3
GAUGAUAUC
5
CONSESUS
5
GCNNUAUYC
3
GC
G C
GC
G C
GC
G C
GC
selected
U5
loops
5
snRNA
5,
GGUAUCC
ACUAUUC
UAUAUCC
CGUAUUC
UAUAUUC
CUUAUCC
CCUAUCC
re-mRN A
GUGAGGACU
5
snRNA
GCCUUUUAC
NA
UUGAAGUGG
snR NA
GCCUUUUAC
r e -mR
5
r e -mR
NA
'
GUGAGAAUC
5
NA
S
GCCUUUUAC
snR
r e -mRNA
5
UCG
snRNA
AAUCG
GCCUUUUAC
re-mRNA
UUGAGACGG
snRNA
GCCUUUUAC
5
re-m RNA
5
sn
RNA
3
GCCUUUUAC
3
5 '
GGUG
CAC
GCCGUUUAC
215
pre-mRNA
U5
snRNA
pre-mRNA
U5
snRNA
pre-mRNA
I I I I
snRNA
I
5 '
GCCUGUUAC
3 '
3
GAGUGAGUG
5 '
'
5
GCCAUUUA
3 '
GAGUGAGUG
5 '
GCC
I
U5
5
GUGACACU
3
C
3
'
5
'
3
'
x I-I
U AUUAC
216
AFTERWORD
A mechanistic understanding of the process by which splice site sequence
information and context information is read by the spliceosome, requires an understanding
of the proteins and spliceosomal complexes that form on the pre-mRNA and an
understanding of the basic cell biology of splicing. The identification of a new pyrimidine
tract binding factor, PUF, that is required for the efficient splicing of pre-mRNA in vitro
suggests that the process of pyrimidine tract recognition is more complicated than has been
anticipated. There are four outstanding issues to be resolved with regard to the PUF factor
and the PUF protein PUF60. 1. Is PUF60 and/or p54 required for PUF activity? Is some
other component of the PUF fraction required? 2. What is the function of the PUF
activity? At what step in spliceosome assembly does it act? 3. What cytological structure
is PUF60 associated with? Are the discrete nuclear bodies observed to stain with PUF60,
PML bodies, coiled bodies or some unidentified nuclear body? Is the staining that we
observe representative of all or just a subset of the PUF60 in the cell? 4. Is the SDSresistant dimerization representative of a native state interaction? Is the proposed PUMP
domain interaction motif G, F/Y, E/D, X, V/I, T/S responsible for the interaction of
the U2AF35 PUMP domain with U2AF65?
PUF60: a splicing factor?
The principal question that needs to be answered is, is PUF60 a splicing factor? A
related question is, is the PUF60/p54 complex responsible for the PUF activity that was
purified in chapter 2?
Without an antiserum that immunoprecipitates PUF60 or a reconstituting expressed
PUF60/p54 fraction, it will be difficult to determine if PUF60 is a splicing factor. These
two problems have been major stumbling blocks to progress. One approach that is being
taken to solve this problem is to affinity tag the PUF60 protein and express this protein in
mammalian cells. This experiment is being pursued both here and at the University of
Sherbrooke by Pascal Bouffard and Gilles Boire and a collaboration has been established to
share reagents. We hope to show, using this method, that the expressed-tagged protein
restores splicing activity to NEAU. A large-scale immunoprecipitation (IP) with the weakly
immunoprecipitating anti p54 antibody can also be attempted, and the eluate of such an IP
could be tested for reconstitution of NEAU activity. This has not previously been
attempted due to the limiting quantity of this antibody (the gift of Nilabh Chaudhary).
217
Experiments that address functional issues
It will be interesting to determine what proteins associate with PUF60 and p54.
Under what conditions do PUF60 and SF1/BBP interact. Current experiments show only
a weak interaction, but this interaction may be stronger under some conditions. It will be
important to determine, both on functional grounds and on pedagogical grounds what
cofactors or conditions enhance this activity.
It will be of interest to determine what snRNAs and splicing protein components are
present on pre-mRNAs that are incubated in NEAU in the presence of PUF or U2AF or
both proteins. This experiment may most easily be done using tagged PUF and U2AF.
Complexes formed can then be compared directly for snRNA content and protein content.
A similar experiment was performed using GstU2AF several years ago that demonstrated
an association with both U2 and Ul snRNPs dependent on the presence of the 3' half premRNA (P. S. M., Anna Gil and P. A. S., unpublished). A similar approach was taken in
which pre-mRNAs associated with SRm160 were examined for proteins that crosslinked to
the pyrimidine tract. This experiment may be revisited to determine a time course of the
association of PUF60 and p54 with the pre-mRNA .
More straightforward and short term experiments include direct
immunoprecipitation of tagged PUF60 and determination of what proteins associate with
PUF60 in the cell using a panel of antibodies directed against the various nuclear proteins
including the splicing factors SRml60, SFl/BBP, SF3a components, U2AF65 and 35,
Urp, PTB, Sm, Ro and p54.
To demonstrate that the interaction between PUF60 and p54 is conserved in
evolution, an immunoprecipitation using the weakly immunoprecipitable p54 antiserum will
be attempted from insect cells and the pellet will be tested for the presence of DPUF68
using the PUF60 antiserum. It is expected that due to the high degree of conservation the
anitserum directed against the human proteins will cross react with the Drosophilaproteins
The human autoimmune antigen Ro, of unknown function, immunoprecipitates
PUF60 (Pascal Bouffard and Gilles Boire, personal communication). Does Ro
immunoprecipitate the PUF splicing activity as well? Does PUF60 associated Ro associate
with the pre-mRNA? Ro has been shown to immunoprecipitate PUF60 and it should be
determined if the Ro IP pellet contains PUF activity. It is possible that Ro
immunoprecipitates will have p54 as well, and this should be tested. As Ro's function is
unknown, any associated proteins that are suggestive of function is of interest.
218
p54 translated in vitro and added to NEAU in the presence or absence of U2AF or
in vitro translated PUF60 caused degradation of the pre-mRNA (data not shown).
Unprogrammed and control programmed lysate did not have this effect. The mechanism of
degradation is not known, but it is phenomenologically interesting.
PUF60 immunolocalization: is the native protein detectable?
PUF60, detected in situ by immunofluorescence experiments, does not localize to
the expected nuclear structures. The p54 protein, shown to co-purify with and associate
with PUF60, localizes to the speckle structures as do most splicing factors. The lack of
PUF60 staining of this structure and the unexpectedly faint staining that is observed
suggests that the PUF60 epitopes are masked in situ, as they are in vitro. To determine if
this is the case fixed cells will be treated with denaturing agents, SDS and heat, and the
cells will be stained for PUF60. We expect to see increased staining under these conditions
This experiment is being actively pursued. Tagged PUF60 will also be transfected into
cells and the localization of the expressed protein determined by using anti-tag antibodies.
Is PUF60 a dimer in the native state?
PUMP domain interaction motif?
Is the PUMP domain bound to the
One easy way to test this hypothesis is to transfect cells with tagged PUF60 and
determine if this tagged protein associates with untagged endogenous protein by
immunoprecipitation.
Native state dimerization can also be tested using native gel electrophoresis,
crosslinking, analytical ultracentrifugation and gel filtration chromatography.
It will also be of interest to see if the Drosophilahomolog of PUF60, DPUF68, is
an SDS-resistant dimer.
The U2AF35 PUMP domain may interact with a short peptide motif of U2AF65.
This may be tested by mutagenesis and far western blotting as performed by Zhang et al.
1992.
219
Download