+ All Categories
Home > Documents > The pre-mRNA Splicing Machinery of Trypanosomes: Co mplex ... · 25.06.2010  · Both types of...

The pre-mRNA Splicing Machinery of Trypanosomes: Co mplex ... · 25.06.2010  · Both types of...

Date post: 19-Oct-2020
Category:
Upload: others
View: 1 times
Download: 0 times
Share this document with a friend
37
1 The pre-mRNA Splicing Machinery of Trypanosomes: Complex or Simplified? Arthur Günzl Department of Genetics and Developmental Biology and Department of Molecular, Microbial and Structural Biology, University of Connecticut Health Center, 263 Farmington Avenue, Farmington, CT 06030-3301, USA Dr. Arthur Günzl Department of Genetics and Developmental Biology University of Connecticut Health Center 263 Farmington Avenue Farmington, CT 06030-3301 USA Phone (860) 679-8878 Fax (860) 679-8345 E-mail [email protected] Short Title: pre-mRNA splicing in trypanosomes Copyright © 2010, American Society for Microbiology and/or the Listed Authors/Institutions. All Rights Reserved. Eukaryotic Cell doi:10.1128/EC.00113-10 EC Accepts, published online ahead of print on 25 June 2010 on March 29, 2021 by guest http://ec.asm.org/ Downloaded from
Transcript
  • 1

    The pre-mRNA Splicing Machinery of Trypanosomes: Complex or Simplified?

    Arthur Günzl

    Department of Genetics and Developmental Biology and Department of Molecular, Microbial and

    Structural Biology, University of Connecticut Health Center, 263 Farmington Avenue, Farmington, CT

    06030-3301, USA

    Dr. Arthur Günzl

    Department of Genetics and Developmental Biology

    University of Connecticut Health Center

    263 Farmington Avenue

    Farmington, CT 06030-3301

    USA

    Phone (860) 679-8878

    Fax (860) 679-8345

    E-mail [email protected]

    Short Title: pre-mRNA splicing in trypanosomes

    Copyright © 2010, American Society for Microbiology and/or the Listed Authors/Institutions. All Rights Reserved.Eukaryotic Cell doi:10.1128/EC.00113-10 EC Accepts, published online ahead of print on 25 June 2010

    on March 29, 2021 by guest

    http://ec.asm.org/

    Dow

    nloaded from

    http://ec.asm.org/

  • 2

    ABSTRACT

    Trypanosomatids are early-diverged, protistan parasites of which Trypanosoma brucei,

    Trypanosoma cruzi, and several species of Leishmania cause severe, often lethal diseases in humans.

    To better combat these parasites, their molecular biology has been a research focus for more than

    three decades and the discovery of spliced leader (SL) trans splicing in T. brucei established a key

    difference between parasites and hosts. In SL trans splicing, the capped 5' terminal region of the

    small nuclear SL RNA is fused onto the 5' end of each mRNA. This process, in conjunction with

    polyadenylation, generates individual mRNAs from polycistronic precursors and creates functional

    mRNA by providing the cap structure. The reaction is a two step transesterification process

    analogous to intron removal by cis splicing which, in trypanosomatids, is confined to very few pre-

    mRNAs. Both types of pre-mRNA splicing are carried out by the spliceosome consisting of five U-

    rich small nuclear (sn)RNAs and, in humans, of up to ~170 different proteins. While

    trypanosomatids possess a full set of spliceosomal U snRNAs, only few splicing factors were

    identified by standard genome annotation because trypanosomatid amino acid sequences are

    among the most divergent in the eukaryotic kingdom. This review focuses on recent progress made

    in the characterization of the splicing factor repertoire in T. brucei which was achieved by tandem

    affinity purification of splicing complexes, by systematic analysis of proteins containing RNA

    recognition motifs, and by mining the genome database. In addition, recent findings about

    functional differences between trypanosome and human pre-mRNA splicing factors are discussed.

    on March 29, 2021 by guest

    http://ec.asm.org/

    Dow

    nloaded from

    http://ec.asm.org/

  • 3

    Trypanosomatids are protistan parasites infecting hosts as diverse as mammals, insects and plants. In

    humans, vector-borne Trypanosoma brucei, Trypanosoma cruzi, and Leishmania spp. cause lethal

    diseases and the strong impact of these parasites on global health has spurred investigations of the

    molecular processes in these organisms early on. One of the first key discoveries in regard to gene

    expression was spliced leader (SL) trans splicing which was eventually found to be an essential

    maturation step for all nuclear pre-mRNA in trypanosomatids.

    The initial discoveries of SL trans splicing were made in T. brucei and until now this organism has

    remained the preferred trypanosomatid organism for spliceosomal studies. T. brucei is an extracellular

    parasite which evades the mammalian immune system by antigenic variation of its variant surface

    glycoprotein (VSG) coat. VSG expression has therefore been a research focus and it was on VSG mRNAs

    that the 5' terminal region was first discovered to contain a leader sequence which was not encoded in the

    VSG gene (11, 87). Further analysis showed that the 39 nt-long leader was derived from the 5' terminus of

    a separate, small nuclear RNA, which has been termed SL RNA or mini-exon-derived RNA (13, 35, 57).

    Discovery of a Y structure intermediate, which corresponds to the cis splicing intron-exon-lariat structure

    (Fig. 1), strongly indicated that the SL transfer functions analogously to intron removal entailing the same

    two transesterification reactions (59, 80). This notion was confirmed by the demonstration that destruction

    of spliceosomal Uridine-rich small nuclear RNAs (U snRNAs) blocked SL transfer (85).

    SL trans splicing is not restricted to VSG mRNA but is an essential maturation step for all

    trypanosomatid mRNAs. In trypanosomatid genomes, coding genes are tandemly arranged in large

    polygenic clusters which are transcribed in a polycistronic fashion (8). Trans splicing and polyadenylation

    lead to precursor cleavages up- and downstream of a coding region, respectively, and therefore are

    mechanistically required to process individual mRNAs from polycistronic pre-mRNA. Moreover, the SL

    carries an m7G cap and the first four nucleotides of its sequence are methylated; some of these

    on March 29, 2021 by guest

    http://ec.asm.org/

    Dow

    nloaded from

    http://ec.asm.org/

  • 4

    methylations are unique to trypanosomes and the unusual 5' terminal structure has been termed cap 4 (5).

    Since cap 4 is transferred onto mRNA 5' ends as part of the SL, trans splicing represents a post-

    transcriptional mode of mRNA capping and, therefore, is essential in the formation of functional mRNA.

    Since all trypanosomatid mRNAs are trans spliced and trypanosomatid genes typically do not harbor

    introns, it was long thought that these organisms use RNA splicing exclusively for SL transfer and

    accordingly, trypanosome-specific deviations of splicing factors were hypothesized to be trans splicing-

    specific. It therefore came as a surprise when the T. brucei PAP gene (Access. No. Tb927.3.3160)

    encoding Poly(A) polymerase was shown to harbor a single intron that was removed by conventional cis

    splicing (49). The search for further introns revealed only one more gene in T. brucei (Tb927.8.1510),

    encoding a putative RNA helicase (8), whose pre-mRNA was shown to be cis spliced (31). Interestingly,

    a recent characterization of the T. brucei transcriptome by RNA-seq strongly indicates that there are no

    other introns disrupting protein coding genes (76).

    SL trans splicing is a more widespread phenomenon in eukaryotes and after its initial discovery in

    trypanosomes, it was found to occur in a variety of organisms including euglenids (81), nematodes (36),

    trematodes (71), and even lower chordates such as the sea squirt (89). However, there is no indication that

    this particular mode of trans splicing occurs in hosts of trypanosomatid parasites, e.g. vertebrates or

    arthropods (20), and therefore it can be regarded as a parasite-specific process. This specificity and the

    ubiquitous requirement of SL trans splicing for mRNA maturation has made this process an attractive

    research focus. Long-term aims have been to find out how the trypanosome splicing machinery differs

    from its human counterpart, to identify factors or factor domains which are specifically required for the

    trans splicing process, and to analyze whether these features can be inactivated in a parasite-specific

    manner. The challenge of this research is that the splicing machinery, termed spliceosome, is a huge,

    dynamic complex composed of structural RNAs and proteins that is difficult to characterize.

    on March 29, 2021 by guest

    http://ec.asm.org/

    Dow

    nloaded from

    http://ec.asm.org/

  • 5

    The spliceosome consists of the U1, U2, U4, U5, and U6 snRNAs and, in the human system, of up to

    170 spliceosome-associated protein factors (92). Trypanosomatids possess all five spliceosomal U

    snRNAs which are typically somewhat smaller and deviate in several aspects from their human

    counterparts. In contrast, until recently, our knowledge of spliceosomal protein factors in trypanosomatids

    was very limited. A main reason for this lack of knowledge comes from the fact that amino acid

    sequences of trypanosomatid proteins have diverged dramatically from their human and yeast orthologs

    and thus, only a few splicing factors were identified by standard annotation of the completed TriTryp

    genomes (30). In the past years, however, major progress has been made in the identification of

    spliceosomal proteins and the characterization of U small nuclear ribonucleoprotein particles (U snRNPs)

    in trypanosomes. Three factors have contributed to this success: firstly, the unrestricted access to the

    sequenced and annotated TriTryp genome databases (8) at GeneDB (http://www.genedb.org/) and,

    recently, also at TriTrypDB (http://tritrypdb.org/; ref. 3), secondly, the systematic analysis of RNA

    binding proteins harboring an RNA recognition motif (RRM; ref. 17), and thirdly, tandem affinity

    purification of splicing complexes combined with mass spectrometric identification of co-purified

    proteins (1, 64). A current list of identified splicing factors is presented in Table 1.

    In an excellent previous review on trypanosomatid RNA splicing, Liang et al. described the

    discoveries and functional characterizations of the trypanosome spliceosomal U snRNAs and early

    characterizations of the corresponding snRNPs (40). This review omits a general discussion of the U

    snRNAs and instead focuses on proteins involved in splicing.

    U snRNPs and the spliceosome in higher eukaryotes

    Our mechanistic insight of RNA splicing and our biochemical and structural knowledge of snRNPs,

    splicing factors and the spliceosome stem predominantly from work in the human and yeast systems.

    on March 29, 2021 by guest

    http://ec.asm.org/

    Dow

    nloaded from

    http://ec.asm.org/

  • 6

    Unfortunately, there is no uniform nomenclature for the protein factors in the two systems and typically,

    there are two distinct names for orthologous factors (listed in ref. 32). In this review, the default is the

    denotation from the human system.

    The main building blocks of the spliceosome are the U snRNPs whose biogenesis requires several

    distinct assembly steps. First, all spliceosomal U snRNAs, except U6, are exported to the cytoplasm

    where they bind a set of seven common proteins, known as the Sm proteins B, D1, D2, D3, E, F, and G.

    These proteins form a heteromeric ring around a conserved Sm binding site that resides in a single-

    stranded region in the 3' terminal domain of the U snRNA. This RNA-protein interaction is typically very

    stable and thus, the U snRNA/Sm complex is referred to as the core snRNP. Core snRNP assembly takes

    place in the cytoplasm and is linked to U snRNA cap hypermethylation which in turn co-determines the

    re-import of the core snRNP into the nucleus. The U6 snRNA does not have a cytoplasmic phase and, in

    the nucleus, binds a different complex of seven Sm-like (LSm) proteins termed LSm2-8. Back in the

    nucleus, the core snRNPs bind various snRNP-specific proteins, and overall there are approximately 45

    different proteins in the human system directly interacting with the spliceosomal U snRNAs (92). As

    follows, most of the spliceosome-associated proteins are considered to be non-snRNP proteins.

    The RNA sequence determinants for the splicing reaction comprise the 5' and 3' splice sites (SS), and

    a branch point (BP) sequence upstream of the 3'SS. In addition, a polypyrimidine tract is typically present

    between BP and 3'SS (Fig. 1). Importantly, the spliceosome is assembled step-by-step on the pre-mRNA

    and before and during splicing, it undergoes highly dynamic changes in which both RNA and protein

    compositions are altered (92 and refs. therein). In brief, the U1 snRNP first recognizes the 5'SS, the

    protein factor SF1 the branch point, and the heterodimeric U2 auxiliary factor (U2AF) both branch point

    and 3'SS. Subsequently, the U2 snRNP is recruited and the U2 snRNA base-pairs with the BP sequence

    displacing SF1, a process which is mediated by the U2-associated protein complexes SF3a and SF3b. At

    on March 29, 2021 by guest

    http://ec.asm.org/

    Dow

    nloaded from

    http://ec.asm.org/

  • 7

    this stage, the factor assembly is called pre-spliceosome or complex A. Subsequently, the U4/U6 snRNP

    and the U5 snRNP enter the spliceosome in the form of the U4/U6.U5 tri-snRNP which results in the pre-

    catalytic complex B. Although in this complex all snRNPs are on board, the spliceosome undergoes major

    rearrangements for activation (complex B') including the discard of U1 and U4 snRNPs. After the first

    transesterification, the spliceosome is transformed into complex C, and following the second splicing step

    it is disassembled. The snRNPs are then recycled for new rounds of splicing. The different spliceosomal

    complexes have been purified and biochemically characterized in the yeast and human systems. Besides

    the described snRNP changes, these complexes are associated with distinct sets of proteins (Fig. 2;

    reviewed in refs. 32, 92).

    Is the spliceosome different for SL trans splicing? Interestingly, as first shown for the nematode

    Caenorhabditis elegans, the SL RNA splicing substrate itself is assembled into a core snRNP binding the

    Sm proteins (12). This finding led to the hypothesis that the SL RNP activates its own splice site and

    trans splicing does not require the U1 snRNP (82, 88). Indeed, in vitro studies in the parasitic nematode

    Ascaris lumbricoides showed that the destruction of U1 snRNA affected only cis but not trans splicing

    (28). Moreover, in the same system, two specific SL RNP proteins were identified and termed SL175 and

    SL30 according to their molecular masses (18). Protein-protein interaction experiments suggested that

    these proteins bridge the SL RNP, and thus the 5'SS of the SL RNA, via SF1 to the BP, a function which

    in cis splicing is mediated by the U1-specific FBP11/Prp40p (human/yeast nomenclature) subunit (18).

    SL175 and SL30 are indispensable for SL trans splicing but they have no function in cis splicing or in an

    SL-independent mode of trans splicing (18) which has also been described in the human system

    (reviewed in ref. 23). While these factors and their interactions are potential anti-parasitic targets, the

    amino acid sequences of these proteins are not conserved, and putative orthologs have not been identified

    outside of nematodes.

    on March 29, 2021 by guest

    http://ec.asm.org/

    Dow

    nloaded from

    http://ec.asm.org/

  • 8

    Trypanosome Sm and LSm proteins, and Sm core variation in U2 and U4 snRNPs

    Sm and LSm proteins are small proteins with a molecular mass typically of ~10-20 kDa that share a

    highly conserved bipartite Sm motif. The corresponding Sm fold characteristically consists of an N-

    terminal helix and a strongly bent, anti-parallel beta-sheet of five strands. While antibodies directed

    against the Sm domain of human proteins recognize Sm proteins in a wide range of organisms, they did

    not cross-react with trypanosome proteins (56, 62, 63). Hence, it required affinity purification of U

    snRNPs and protein analysis to show that trypanosome U snRNAs and the SL RNA bind a set of common

    proteins (63). The identity of five of these proteins was revealed in the classic way: U snRNPs were

    affinity-purified, amino acid sequence information from common proteins was obtained by protein micro-

    sequencing, and the respective genes were cloned with the help of degenerate primers and PCR (66). In

    the same study, the missing SmB and SmD3 orthologs, however, could already be identified by mining

    the growing T. brucei genome data base (66). Later, this was the exclusive route to identify the orthologs

    of LSm2-8 (42). However, while the Sm motifs were readily identifiable in all these proteins, the

    remaining amino acid sequences exhibited limited similarity to their putative orthologs in other

    eukaryotes and therefore needed functional verification. In case of the Sm proteins, SmG was shown to

    complement an SmG-deficient yeast strain (66) whereas the others exhibited protein-protein interactions

    which were consistent with the known arrangement in the ring structure (64, 66). For the LSm proteins,

    only LSm8 and LSm3 were functionally analyzed at first, and all others identified by sequence similarity

    alone (42). This approach backfired because LSm2 and LSm5 ended up to be very interesting Sm proteins

    (see below) but not LSm proteins. Eventually, a second study clarified the trypanosome LSm repertoire

    and identified new LSm2 and LSm5 orthologs, and provided strong evidence through functional studies

    that the correct set of LSm proteins was identified (83). Formation of core snRNPs stabilizes the U

    snRNAs and expression silencing of a single Sm or LSm protein leads to a loss of the cognate U snRNA.

    on March 29, 2021 by guest

    http://ec.asm.org/

    Dow

    nloaded from

    http://ec.asm.org/

  • 9

    Accordingly, conditional RNA interference experiments targeting each of the seven LSm proteins resulted

    in a specific loss of U6 snRNA confirming the new identifications (1, 42, 83).

    In initial studies of trypanosome snRNPs, it was found that the trypanosome U2 core snRNP, in

    contrast to its human counterpart and other trypanosome U snRNPs, completely disassembled in a cesium

    chloride density gradient (15) and in high salt buffers (26). While this instability to salt was originally

    attributed to a deviating U2 Sm binding site, which in T. brucei contains an unusual central guanosine

    residue, it was found only recently that the different core includes the Sm complex as well. U2 snRNP

    purification revealed two U2-specific proteins with apparent sizes of 15 and 16.5 kDa that contained the

    bipartite Sm motif (93). This was odd because the whole Sm repertoire had already been characterized

    and, moreover, Sm15K, at that time, had been identified as LSm5. However, a comprehensive tagging

    and co-immunoprecipitation analysis clarified the issue and showed that Sm15K and Sm16.5k are

    paralogs of SmB and SmD3, respectively; they replace these proteins specifically in the U2 Sm core and

    do not bind other U snRNAs. Furthermore, snRNP reconstitution assays with recombinant Sm proteins

    and synthetic RNAs demonstrated that the guanosine residue of the U2 Sm binding site is the recognition

    determinant of the U2-specific Sm core complex (93). In an independent study, the identification of the

    two U2-specific Sm paralogs was confirmed and the previously mis-annotated LSm2 was shown to be a

    second SmD3 paralog that replaces SmD3 in the U4 snRNP (84). Importantly, the study by Tkacz et al.

    (84) provided an in vivo analysis demonstrating that RNAi-mediated expression silencing of the specific

    Sm paralogs reduced the abundance of only the corresponding U snRNA. The U4-specific Sm core was

    subsequently characterized at the biochemical level verifying the U4-association of the SmD3 paralog

    (31). Unfortunately, the studies on Sm core variation established different nomenclatures and Sm15K is

    also referred to as specific spliceosomal Sm2-1 protein (SSm2-1), Sm16.5K as SSm2-2, and the U4-

    specific protein as SSm4.

    on March 29, 2021 by guest

    http://ec.asm.org/

    Dow

    nloaded from

    http://ec.asm.org/

  • 10

    What is the significance of Sm core variation? It has been speculated that it assists U2 and U4 snRNP

    assembly (84, 93) and indeed, this has recently been demonstrated for the U2 snRNP. Earlier it was found

    that stable protein binding to the 3' terminal region of U2 snRNA, which included the Sm binding site,

    was dependent on residues in the 3' terminal loop IV sequence. This suggested that the U2 core snRNP

    was not formed by Sm protein binding alone but required cooperative binding of Sm and loop IV-binding

    proteins (26). This model was recently verified by the demonstration that the U2-specific Sm15k/16.5K

    doublet interacts with the U2 snRNP protein U2A' which in turn interacts with the loop IV-binding

    protein U2B'' (70). Only this ternary complex efficiently and specifically interacts with the 3'-terminal U2

    snRNA region. In the human system, U2A' is separated from the Sm core by stem-loop III. Since this

    structure is completely missing in trypanosome U2 snRNA, it is likely that the Sm15K/16.5K - U2A'

    interaction occurs through an essential, parasite-specific protein-protein interface that compensates for the

    lack of this RNA structure.

    Furthermore, it was suggested that Sm core variation may facilitate snRNP function in the splicing

    process (93). For example, human and yeast U2 snRNAs share a conserved motif which is

    complementary to the BP sequence. Conversely, in trypanosomes there is no conserved BP sequence and

    typically no complementarity between BP and U2 snRNA sequences (46). Possibly, the U2-specific Sm

    paralogs undergo specific protein-protein interactions positioning the U2 snRNP at the BP in the absence

    of sequence complementarity. A third speculation stated that the different Sm cores may be connected to

    different U snRNA cap structures (84). In vertebrate and yeast systems, the U6 snRNA carries a γ-

    monomethyl phosphate cap whereas U1, U2, U4 and U5 snRNAs obtain co-transcriptionally an m7G cap

    which is further methylated to a 2,7,7 trimethylguanosine (m3G) cap after the formation of the core

    snRNP. It was shown that the binding of the Sm proteins to the U snRNAs is a pre-requisite for the

    recruitment of the enzyme trimethylguanosine synthase 1 (55, 68). In trypanosomes, U1, U2, U4 and U6

    on March 29, 2021 by guest

    http://ec.asm.org/

    Dow

    nloaded from

    http://ec.asm.org/

  • 11

    snRNAs have the same caps as their yeast and human orthologs (19, 58, 65) whereas SL RNA has cap 4

    (5) and U5 snRNA lacks a cap (21, 96). Theoretically, Sm core variation could facilitate the recruitment

    of different cap-modifying enzymes into the core RNP but there is no correlation between the type of cap

    and the type of Sm core. For example, U1, U2, and U4 snRNAs bind different Sm complexes but share

    the same m3G cap. Moreover, it was demonstrated experimentally for the T. brucei U2 snRNA that cap

    trimethylation does not depend on the presence of the Sm binding site nor on formation of the core RNP,

    thus excluding the possibility that the U2-specific Sm core is involved in cap hypermethylation (25).

    SMN-mediated assembly of canonical Sm cores

    When Wang et al. (93) reconstituted core snRNPs with recombinantly expressed Sm proteins, they

    detected specific binding of the canonical and U2-specific Sm cores to their cognate Sm binding sites only

    with short RNA fragments whereas full-length U snRNAs did not discriminate between the two Sm

    complexes. This suggested that other activities in the cell confer specificity of Sm core binding. A

    candidate for such an activity was the SMN (survival motor neuron) complex, which in the human system

    was shown to act as a catalyst for core snRNP formation (reviewed in refs. 34, 60). The human SMN

    complex consists of the SMN protein and seven additional subunits, termed Gemin2-8, and it binds the

    SmD1/D2-E/F/G and SmD3/B sub-complexes in an open ring formation (14). This interaction then leads

    to U snRNA binding, ring closure, and dissociation of the SMN complex. While standard annotation of

    the TriTryp genomes did not identify SMN or Gemin homologs, tandem affinity purification of the T.

    brucei SmB protein (see below) led to the identification of highly divergent orthologs of SMN and

    Gemin2 (64). No other Gemin orthologs were found and it is possible that they do not exist in

    trypanosomes because lower eukaryotes in general have a strongly reduced Gemin repertoire and, in

    Drosophila melanogaster, the SMN/Gemin2 complex was sufficient to mediate core RNP assembly in

    vitro (37). In trypanosomes, in vitro core snRNP assembly experiments functioned efficiently in the

    on March 29, 2021 by guest

    http://ec.asm.org/

    Dow

    nloaded from

    http://ec.asm.org/

  • 12

    absence of SMN but when the factor was added it exhibited a striking discriminatory role: in its presence

    the canonical Sm core was efficiently loaded onto its cognate U5 snRNA but not onto U4 and U2

    snRNAs nor onto a U5 snRNA with a mutated Sm site (64). In contrast, the SMN complex had no effect

    on the binding of the U2-specific Sm core. These findings suggested that the SMN complex specifically

    bound the canonical SmD3/B subcomplex and directly interacted with SmB because this protein, in

    contrast to SmD3, is replaced in both the U2 and U4-specific Sm cores. This was indeed the case. SMN

    purification co-isolated only the SmB and SmD3 proteins and not their paralogs, and pull-down assays

    with recombinant, tagged SMN proteins identified a direct interaction with SmB and the N-terminal part

    of SMN (64). The latter finding, again, is highly significant because it identified an important, potentially

    parasite-specific protein-protein interaction: human SMN utilizes an internal Tudor domain and C-

    terminal regions to interact with dimethylated arginines in the RG-rich C-termini of Sm proteins (74)

    whereas in trypanosomes neither Tudor domain nor RG-rich domains are present in SMN and Sm

    proteins, respectively (64). Another striking difference to the human system was found in regard to SMN

    localization. In the human system, core snRNP assembly takes place in the cytoplasm and, accordingly,

    human SMN is primarily localized in this compartment. Conversely, trypanosome SMN was found almost

    exclusively in the nucleus suggesting that U snRNP assembly in this organism is nuclear, a finding which

    is consistent with localizations of SL RNA and U2 snRNA by fluorescence in situ hybridization (9, 84).

    In summary, it appears that despite its small size, the trypanosome SMN complex is mechanistically

    complex entailing both chaperone and specificity functions in core snRNP assembly.

    If the SMN complex only chaperones the assembly of the canonical Sm core, how are then the U2 and

    U4-specific Sm cores put together? One possibility is that they require a different, yet to be determined

    assembly chaperone. Alternatively, the specific interactions of the U2 Sm paralogs SmK15/SmK16.5 with

    U2A'/B'' may facilitate correct assembly of the U2-specific Sm complex onto the U2 Sm binding site. On

    the other hand, U4 core snRNP formation appears to be independent of snRNP-specific proteins because

    on March 29, 2021 by guest

    http://ec.asm.org/

    Dow

    nloaded from

    http://ec.asm.org/

  • 13

    in core RNP reconstitution assays, SSm4 alone determined efficient and specific assembly of the U4-

    specific Sm complex onto the U4 snRNA (31).

    Tandem affinity purification of splicing complexes in T. brucei

    Until recently, only two snRNP-specific proteins had been studied in trypanosomes, namely the

    orthologs of human U2A' (originally termed U2-40K; ref. 16) and the U5-specific PRP8 (45). While the

    latter was identified by sequence homology, U2A' was co-purified with the U2 snRNA at high stringency

    U snRNP purifications which typically left only the core structures intact (63). For a more comprehensive

    biochemical characterization of U snRNPs and/or of the spliceosome it was therefore essential to purify

    the RNA-protein complexes at conditions of lower stringency. A method well-suited for this purpose is

    tandem affinity purification (TAP) which is based on expressing a known protein factor fused to a

    composite TAP tag. TAP comprises two consecutive high affinity chromatography steps which are carried

    out at nearly physiological conditions. Since the advent of this technology (72), the TAP tag and the TAP

    method have been modified in various ways to accommodate different systems, extracts or protein

    complexes (27). For the purification of nuclear protein complexes in trypanosomes, the PTP modification

    of TAP has proven to be very useful (27, 73). One of the first applications of the PTP tag was the

    purification of the trypanosome U1 snRNP. A first characterization of this snRNP had revealed a protein

    with sequence homology to the human U1-70K protein (65). And indeed, PTP-tagging and purification of

    T. brucei U1-70K-PTP resulted in the specific co-purification of the U1 snRNA (67). The protein profile

    of the purification comprised the Sm proteins, the tagged protein, two additional proteins of major

    abundance and several proteins of minor abundance. The proteins of the two major bands were identified

    by mass spectrometry and found to be annotated as conserved hypotheticals meaning that they were

    conserved among trypanosomatids but had no obvious similarity to proteins of other eukaryotes.

    However, when the kinetoplastid sequences of one of the new proteins was compared to those of known

    on March 29, 2021 by guest

    http://ec.asm.org/

    Dow

    nloaded from

    http://ec.asm.org/

  • 14

    U1-specific proteins of model organisms, it was identified as the ortholog of human U1C (67). In contrast,

    the second protein, termed U1-24K, could not be meaningfully aligned to known U1 proteins and

    therefore likely represents a trypanosome-specific U1 snRNP subunit.

    Since this initial tandem affinity purification of a snRNP was successful, more comprehensive

    proteomic analyses of trypanosomal splicing complexes were carried out by PTP tagging and purification

    of the common proteins SmD1 (1) and SmB (64). Overall, mass spectrometry identified 53 proteins in

    these purifications and the majority of the proteins co-purified in both studies (Table 1). Moreover, with

    the exception of three LSm proteins and two non-snRNP proteins, all known trypanosomal snRNP

    proteins were identified in these proteomic analyses confirming the high significance of the proteomic

    data sets. Consequently, bioinformatic analyses of the amino acid sequences of un-annotated proteins

    revealed several new orthologs of known splicing factors (Table 1) and for LSm2 (U6), U1A, PRP4 (U4),

    and U5-40K, the bioinformatic identifications were confirmed by co-immunoprecipitation experiments

    which showed that these proteins were bound to their predicted snRNAs (1).

    While these proteomic analyses increased the number of spliceosomal protein orthologs in

    trypanosomes considerably and identified potentially novel splicing factors (see below), the number of

    proteins that co-purified with SmD1 or SmB are ~3 fold lower than the protein count in human

    spliceosomes. Proteomics of yeast spliceosomal complexes revealed about 90 proteins (22) which is

    lower than the count in the human system but still about twofold higher than the identified trypanosome

    repertoire. One possible interpretation of this finding is that the splicing machinery of early-diverged

    trypanosomatids is simplified. However, this is unlikely because the vast majority of newly identified

    proteins are snRNP proteins, and non-snRNP proteins are strongly underrepresented (Fig. 2). In fact,

    trypanosome orthologs have been identified for nearly all known bona fide snRNP proteins indicating that

    a trypanosome spliceosome comprises additional non-snRNP proteins possibly in comparable numbers to

    on March 29, 2021 by guest

    http://ec.asm.org/

    Dow

    nloaded from

    http://ec.asm.org/

  • 15

    those in yeast and humans. Hence, the question arises why only a few non-snRNP proteins co-purified

    with SmB and SmD1. Both proteomics studies were carried out according to the standard PTP protocol

    which includes the extract preparation procedure (38, 73). Since these extracts contain an estimated

    overall salt concentration of 250-300 mM, it is likely that the spliceosome did not withstand the extract

    preparation procedure. Accordingly, a sucrose gradient sedimentation analysis of SmD1-PTP-purified

    material showed that the U snRNPs did not co-sediment as part of a larger complex and complexes with S

    values greater than 20 were not detected (1). In contrast, spliceosomal 45S complexes were characterized

    by a combination of glycerol gradient sedimentation and native gel electrophoresis in extracts of lower

    salt concentration (41). It is therefore likely that modifying the extract preparation procedure will result in

    formation of higher order spliceosomal complexes which possibly can be isolated by tandem affinity

    purification and characterized by mass spectrometry in the future.

    As discussed above, in both proteomic studies, the highly divergent SMN and Gemin2 orthologs co-

    purified. To better understand the trypanosome SMN complex, Palfi et al. (64) PTP-tagged and tandem

    affinity-purified both proteins, and identified co-purified proteins by mass spectrometry. While no other

    Gemin proteins were detected, which supports the idea that a SMN/Gemin2 complex is sufficient for

    chaperone function, surprisingly, all coatomer subunits co-purified. While the coatomer complex

    functions in vesicular transport between Golgi and endoplasmic reticulum (48), the significance of the

    coatomer-SMN/Gemin2 interaction is not understood. Possibly, the trypanosome SMN/Gemin2 complex

    has a cytoplasmic function independent of snRNP core assembly (64) or there is a cytoplasmic component

    of the core snRNP assembly process which is vesicular and has not been detected yet.

    Analysis of proteins carrying an RRM

    Besides by tandem affinity purification, trypanosome splicing factors have been identified through a

    focus on RRM-containing proteins. Since trypanosomatid protein coding genes are typically arranged in

    on March 29, 2021 by guest

    http://ec.asm.org/

    Dow

    nloaded from

    http://ec.asm.org/

  • 16

    long tandem gene arrays which are transcribed polycistronically, differential gene expression is typically

    regulated post-transcriptionally, for example at the level of RNA stability. Many proteins which affect

    RNA stability bind to mRNAs directly by virtue of an RRM motif and thus, RRM-containing proteins

    have become a research focus in gene expression studies in both T. brucei and Trypanosoma cruzi (17).

    Since the spliceosome comprises several RRM proteins, their identification came as a benefit from the

    attempt to determine the role of RRMs in gene expression regulation.

    One RRM protein that was identified as a splicing factor was a subunit of the U2-associated SF3b

    complex. SF3a and SF3b are two essential multi-subunit splicing factors that interact with the U2 snRNP

    after its recognition of the BP. The trypanosome protein was identified as the ortholog of human SF3b49

    and accordingly, expression silencing of the corresponding gene was lethal and affected RNA splicing in

    T. brucei (51). Moreover, TAP-tagging and purification of the protein, using the original TAP method, led

    to the complete characterization of the trypanosome SF3b complex. Orthologs of all seven human SF3b

    subunits were identified including the RRM protein SF3b14, often referred to as p14 (51). The SF3a

    complex appears to be also present in trypanosomatids because a putative homolog of the SF3a60 subunit

    was annotated in the genome database (Table 1).

    Other RRM proteins that were found to be splicing factors comprise the snRNP protein U1A (1) and

    the U2 auxiliary factor components U2AF65 and U2AF35 (90, 91). In addition, RRM-containing serine-

    argine-rich (SR) proteins have been identified. SR proteins comprise a phylogenetically conserved protein

    family and, as has been shown in other systems, play significant roles in constitutive and alternative

    splicing of pre-mRNA (reviewed in ref. 43). SR proteins contain one or two N-terminal RRMs and a C-

    terminal RS domain, rich of arginine-serine dipeptides. The first such protein discovered in trypanosomes

    was termed RRM1 (52). While RRM1 was shown to be encoded by an essential gene and located in the

    nucleus, its specific function has not been determined yet (53). A second SR protein, termed

    on March 29, 2021 by guest

    http://ec.asm.org/

    Dow

    nloaded from

    http://ec.asm.org/

  • 17

    trypanosomal SR-rich protein 1 (TSR1), was localized to the nucleus, was shown to bind to the

    heterologous human U2AF complex and, in a yeast three hybrid system, it appeared to interact with the

    SL RNA (29). While these findings led to the speculation that TSR1 may facilitate recognition of the SL

    RNA by the trans spliceosome (29), a functional characterization of TSR1 strongly indicated that the

    factor has an essential role in cis splicing but not in SL trans splicing (Christian Tschudi, Yale University,

    personal communication). This result is in accordance with a previous study of the T. cruzi ortholog TcSR

    which showed that TcSR was functional in cis splicing in a heterologous system (69). Finally, RRM

    protein analysis in trypanosomes revealed two homologs (PTB1 and PTB2) of the mammalian

    polypyrimidine tract binding protein. While mammalian PTB did not co-purify with spliceosomal

    complexes and has several non-splicing functions, it negatively affects the splicing process presumably by

    binding to the polypyrimidine tract near the 3'SS thereby interfering with U2AF65 function (78).

    Functional characterization of trypanosome PTB1 and PTB2 did not reveal a repressor function of these

    proteins in splicing. In contrast, a detailed study provided very strong evidence that both proteins are

    essential for trans splicing of pre-mRNAs that contain C-rich polypyrimidine tracts (79). In addition,

    expression silencing of PTB1, but not of PTB2, affected cis splicing indicating that both proteins have

    distinct activating functions in trypanosome RNA splicing (79).

    Bioinformatic identification of trypanosome splicing factors

    A third route to identify RNA splicing factors has been data mining. Some of the splicing factors in

    trypanosomes are conserved enough to be identified by in silico analysis alone. For example, it was

    straightforward to identify the missing orthologs of the human snRNP proteins Snu13 and U5-200K for

    this study (Table 1). Similarly, the important CDC5 subunit of the PRP19 complex, which is an essential

    component of the active spliceosome, was readily identifiable in the trypanosome genome database (Table

    1). Two splicing factors which had previously been identified bioinformatically are PRP43 and PRP31

    on March 29, 2021 by guest

    http://ec.asm.org/

    Dow

    nloaded from

    http://ec.asm.org/

  • 18

    (41). PRP43 is a conserved spliceosomal helicase with essential functions in intron lariat release from the

    spliceosome (54) and in spliceosome disassembly (2) whereas PRP31 is a factor of the U4/U6.U5 tri-

    snRNP being important for tri-snRNP formation and assembly into the spliceosome (50). The

    trypanosome PRP31 and PRP43 appear to be functionally equivalent because both proteins were shown to

    be essential for both cis and SL trans splicing, and PRP31 was specifically associated with the

    trypanosome U4/U6.U5 tri-snRNP (41). Nevertheless, the bioinformatics route of identifying

    trypanosome splicing proteins has not been exploited extensively and it is very likely that a systematic

    approach will reveal additional [putative] orthologs of non-snRNP proteins.

    Overall, our knowledge of the spliceosomal protein repertoire of trypanosomes has greatly increased

    in the past years. While the set of snRNP proteins appears to be nearly complete, most of the non-snRNP

    factors have probably not been identified yet. However, the identification of individual components of

    spliceosomal protein complexes such as PRP19 and SF3a (Fig. 2) indicate that these complexes are

    present and that they can be further analyzed. For example, in yeast more than twenty splicing proteins

    were identified by tandem affinity purification of the CDC5 ortholog Cef1p (reviewed in ref. 32). The

    identification of CDC5 in this study will enable a comparable analysis in trypanosomes.

    Another important aspect of the newly identified trypanosome splicing factors is that they strongly

    indicate that trypanosomes form a spliceosome that possesses the same basic components and undergoes

    the same dynamic rearrangements as its human and yeast counterparts. It should be kept in mind that with

    the exception of a 45S spliceosome detection by native gel electrophoresis (41) there is so far no

    biochemical evidence yet that trypanosomes do form complexes that correspond to the well-characterized

    spliceosome E, A, B, or C complexes in the yeast and human systems. On the other hand, a comparison of

    human proteins that enter the spliceosome at these defined stages and of the known trypanosome

    on March 29, 2021 by guest

    http://ec.asm.org/

    Dow

    nloaded from

    http://ec.asm.org/

  • 19

    repertoire shows that for each spliceosome transition characteristic trypanosome orthologs have been

    identified (Fig. 2).

    Trypanosome-specific aspects of the spliceosome

    Despite the rapidly increasing number of identified trypanosomal RNA splicing factors, only a few

    functional protein characterizations have been published thus far. Nevertheless, several trypanosome-

    specific characteristics of the splicing machinery have already been identified. As discussed above, Sm

    core variation, including the trypanosome-specific interaction between SmB and SMN, as well as the

    particular architecture of the U2 RNP core, involving potentially unique interactions between

    Sm15K/16.5K and U2A', are trypanosome-specific U snRNP features. Other notable differences, shown

    in the T. cruzi system, include the demonstration that the U2AF subunits U2AF35 and U2AF65 exhibit

    weak or no interaction (91), that instead U2AF65 forms a stable complex with the BP binding protein SF1

    (91), and that, within the U2-related SF3b complex, the protein interface between the SF3b155 and

    SF3b14 subunits appears to be larger and more complex than in the human system (4).

    Another interesting trypanosome splicing factor is U5-Cwc21. This protein shares a highly conserved

    N-terminus with the human SRm300/SRRM2 protein and yeast Cwc21p (complexed with Cef1p protein

    21). Co-immunoprecipitation analysis showed that the trypanosome protein is predominantly associated

    with U5 snRNA, and expression silencing of U5-Cwc21 was lethal and affected both cis and trans

    splicing (1). In contrast, yeast Cwc21p and human SRm300 have redundant, non-essential roles in RNA

    splicing because CWC21 is a non-essential gene and SRm300 can be immunodepleted from extract

    without affecting splicing efficiency in vitro (10). Moreover, while yeast Cwc21p does interact with the

    U5-protein PRP8 (24), it is predominantly associated with U2 snRNA and not with U5 snRNA (33).

    on March 29, 2021 by guest

    http://ec.asm.org/

    Dow

    nloaded from

    http://ec.asm.org/

  • 20

    These findings therefore strongly indicate that trypanosome U5-Cwc21 has an essential function in RNA

    splicing that is unique to trypanosomes.

    A further peculiarity of trypanosome splicing factors is the expression level of U1 snRNP

    components. In the nematode system, the U1 snRNP exclusively functions in cis splicing and not in SL

    trans splicing (28). If the trypanosome U1 snRNP functions analogously, it would be required only for the

    removal of a single intron from two different pre-mRNAs. However, the U1-specific proteins U1-70K,

    U1-24K, and U1C are among the most abundant proteins that co-purified with SmD1 (1). This

    discrepancy between intron number and U1 snRNP expression level suggests that the trypanosome U1

    snRNP has functions beyond intron removal. There is evidence that the trypanosome 45S spliceosome

    contains both SL and U1 snRNA and it was suggested that there may only be one kind of spliceosome for

    both cis and trans splicing (41). If this is true, the U1 snRNP may be essential for spliceosome integrity or

    it may have a yet undetected, trypanosome-specific function in trans splicing. Alternatively, the

    trypanosome U1 snRNP, as its human counterpart, may function beyond intron removal in transcription

    initiation and/or elongation (reviewed in ref. 6).

    Finally, there seems to be a difference of SL RNP recruitment to the BP between nematode and

    trypanosome systems. While the nematode SL RNP apparently docks on SF1 via a protein bridge (18),

    immunoprecipitation of trypanosome SF1 at low stringency conditions did not co-precipitate SL RNA

    (D.L. Ambrósio & A. Günzl, unpublished results). It is therefore likely that other proteins and protein-

    protein interactions than in the nematode system mediate the recruitment of the trypanosome SL RNP.

    Possibly, U5 and U6 snRNPs play a role in this process because U5 and U6 snRNAs were convincingly

    shown to interact with the 5'SS of the SL RNA (94, 97). If SL RNP recruitment to pre-mRNA requires

    trans splicing-specific factors as in the nematode system, potential candidates of such proteins are listed

    in Table 1; there are currently eight proteins which co-purified with trypanosomal splicing complexes but

    on March 29, 2021 by guest

    http://ec.asm.org/

    Dow

    nloaded from

    http://ec.asm.org/

  • 21

    could not be annotated convincingly. Three of these proteins may be the orthologs of cyclophylins and of

    the PRP19-related factor Cwc15 although the sequence similarities are very weak. The remaining five

    proteins are novel in sequence because they do not exhibit any sequence similarity to non-trypanosomatid

    proteins.

    Perspectives

    The spliceosome is one of the most complex molecular machineries in the cell and it is a great

    challenge to functionally characterize this dynamic RNP-protein machinery. In the past years, major

    progress has been made in the biochemical and structural analysis of the human spliceosome (47, 92). If

    corresponding studies can be carried out in trypanosomes, it will be possible to determine in detail

    essential differences between trypanosome and human spliceosomes. While such differences may be the

    consequence of evolutionary divergence or may represent SL trans splicing-specific requirements, they

    are potential anti-parasitic drug targets. This notion is not remote since the spliceosome has been validated

    as a drug target, for example for anticancer treatment (reviewed in ref. 86). Although the undertaking of

    comprehensively analyzing the trypanosome spliceosome appears overwhelming, the prospects are

    nevertheless good because all necessary tools are in place. As shown in figure 2, there are now several

    new spliceosomal proteins which can serve as baits in tandem affinity purification to broadly characterize

    the trypanosome splicing factor repertoire. The conditional RNAi-based expression silencing system in T.

    brucei (95) in combination with established RT-PCR and primer extension assays for the analysis of trans

    and cis splicing defects provides an in vivo platform for determining splicing functions of individual

    proteins. Moreover, a homologous in vitro trans splicing system was recently established in T. brucei

    which will allow the functional dissection of important splicing factors (75). Finally, the recent

    demonstration that the tandem affinity-purified trypanosome transcription factor complex TFIIH was

    on March 29, 2021 by guest

    http://ec.asm.org/

    Dow

    nloaded from

    http://ec.asm.org/

  • 22

    sufficiently intact and pure to determine its molecular structure by macromolecular electron microscopy

    (39) strongly indicates that similar structures can be obtained from tandem affinity-purified splicing

    complexes.

    And there is another potentially exciting perspective. While introns and alternative splicing greatly

    enhance the protein repertoire in higher eukaryotes (recently reviewed in ref. 61), the functional role of

    introns in lower eukaryotes is not well understood. Trypanosomes appear to have reduced their intron

    repertoire to only two (76). Why did they not eliminate these two introns as well? The fact that the

    insertion site of the PAP intron is conserved in trypanosomatids argues that the intron has a specific and

    essential function which was retained throughout trypanosomatid evolution. Since it should be

    straightforward to test the outcome of [conditionally] deleting these intron sequences in the trypanosome

    genome, it may be possible to determine the specific function of these introns and understand the

    functional significance of cis splicing in these early-diverged organisms.

    ACKNOWLEGMENTS

    I am thankful to Christian Tschudi (Yale University) for communicating unpublished data and to Tu

    N. Nguyen and Daniela L. Ambrósio for critical reading of the manuscript. This work was supported by

    National Institutes of Health R01 grants AI059377 and AI073300 to A.G.

    on March 29, 2021 by guest

    http://ec.asm.org/

    Dow

    nloaded from

    http://ec.asm.org/

  • 23

    REFERENCES

    1. Ambrósio D.L., J. H. Lee, A. K. Panigrahi, T. N. Nguyen, R. M. Cicarelli, and A. Günzl. 2009.

    Spliceosomal proteomics in Trypanosoma brucei reveal new RNA splicing factors. Eukaryot. Cell

    8:990-1000.

    2. Arenas, J. E. and J. N. Abelson. 1997. Prp43: An RNA helicase-like factor involved in

    spliceosome disassembly. Proc. Natl. Acad. Sci. USA 94:11798-11802.

    3. Aslett, M., C. Aurrecoechea, M. Berriman, J. Brestelli, B. P. Brunk, M. Carrington, D. P.

    Depledge, S. Fischer, B. Gajria, X. Gao, M. J. Gardner, A. Gingle, G. Grant, O. S. Harb, M.

    Heiges, C. Hertz-Fowler, R. Houston, F. Innamorato, J. Iodice, J. C. Kissinger, E. Kraemer,

    W. Li, F. J. Logan, J. A. Miller, S. Mitra, P. J. Myler, V. Nayak, C. Pennington, I. Phan, D. F.

    Pinney, G. Ramasamy, M. B. Rogers, D. S. Roos, C. Ross, D. Sivam, D. F. Smith, G.

    Srinivasamoorthy, C. J. Stoeckert, Jr., S. Subramanian, R. Thibodeau, A. Tivey, C. Treatman,

    G. Velarde, and H. Wang. 2010. TriTrypDB: a functional genomic resource for the

    Trypanosomatidae. Nucleic Acids Res. 38:D457-D462.

    4. Avila, M. L., N. Bercovich, G. Westergaard, M. J. Levin, and M. P. Vazquez. 2007. Mapping of

    the protein-binding interface between splicing factors SF3b155 and p14 of Trypanosoma cruzi.

    Biochem. Biophys. Res. Commun. 364:26-32.

    5. Bangs, J. D., P. F. Crain, T. Hashizume, J. A. McCloskey, and J. C. Boothroyd. 1992. Mass

    spectrometry of mRNA cap 4 from trypanosomatids reveals two novel nucleosides. J. Biol. Chem.

    267:9805-9815.

    6. Barrandon, C., B. Spiluttini, and O. Bensaude. 2008. Non-coding RNAs regulating the

    transcriptional machinery. Biol. Cell 100:83-95.

    7. Bercovich, N., M. J. Levin, C. Clayton, and M. P. Vazquez. 2009. Identification of core

    components of the exon junction complex in trypanosomes. Mol. Biochem. Parasitol. 166:190-193.

    8. Berriman, M., E. Ghedin, C. Hertz-Fowler, G. Blandin, H. Renauld, D. C. Bartholomeu, N. J.

    Lennard, E. Caler, N. E. Hamlin, B. Haas, U. Bohme, L. Hannick, M. A. Aslett, J. Shallom, L.

    Marcello, L. Hou, B. Wickstead, U. C. Alsmark, C. Arrowsmith, R. J. Atkin, A. J. Barron, F.

    Bringaud, K. Brooks, M. Carrington, I. Cherevach, T. J. Chillingworth, C. Churcher, L. N.

    Clark, C. H. Corton, A. Cronin, R. M. Davies, J. Doggett, A. Djikeng, T. Feldblyum, M. C.

    on March 29, 2021 by guest

    http://ec.asm.org/

    Dow

    nloaded from

    http://ec.asm.org/

  • 24

    Field, A. Fraser, I. Goodhead, Z. Hance, D. Harper, B. R. Harris, H. Hauser, J. Hostetler, A.

    Ivens, K. Jagels, D. Johnson, J. Johnson, K. Jones, A. X. Kerhornou, H. Koo, N. Larke, S.

    Landfear, C. Larkin, V. Leech, A. Line, A. Lord, A. MacLeod, P. J. Mooney, S. Moule, D. M.

    Martin, G. W. Morgan, K. Mungall, H. Norbertczak, D. Ormond, G. Pai, C. S. Peacock, J.

    Peterson, M. A. Quail, E. Rabbinowitsch, M. A. Rajandream, C. Reitter, S. L. Salzberg, M.

    Sanders, S. Schobel, S. Sharp, M. Simmonds, A. J. Simpson, L. Tallon, C. M. Turner, A. Tait,

    A. R. Tivey, S. Van Aken, D. Walker, D. Wanless, S. Wang, B. White, O. White, S. Whitehead,

    J. Woodward, J. Wortman, M. D. Adams, T. M. Embley, K. Gull, E. Ullu, J. D. Barry, A. H.

    Fairlamb, F. Opperdoes, B. G. Barrell, J. E. Donelson, N. Hall, C. M. Fraser, S. E. Melville,

    and N. M. El Sayed. 2005. The Genome of the African Trypanosome Trypanosoma brucei. Science

    309:416-422.

    9. Biton, M., M. Mandelboim, G. Arvatz, and S. Michaeli. 2006. RNAi interference of XPO1 and

    Sm genes and their effect on the spliced leader RNA in Trypanosoma brucei. Mol. Biochem.

    Parasitol. 150:132-143.

    10. Blencowe, B. J., G. Bauren, A. G. Eldridge, R. Issner, J. A. Nickerson, E. Rosonina, and P. A.

    Sharp. 2000. The SRm160/300 splicing coactivator subunits. RNA 6:111-120.

    11. Boothroyd, J. C. and G. A. Cross. 1982. Transcripts coding for variant surface glycoproteins of

    Trypanosoma brucei have a short, identical exon at their 5' end. Gene 20:281-289.

    12. Bruzik, J. P., K. Van Doren, D. Hirsh, and J. A. Steitz. 1988. Trans splicing involves a novel

    form of small nuclear ribonucleoprotein particles. Nature 335:559-562.

    13. Campbell, D. A., D. A. Thornton, and J. C. Boothroyd. 1984. Apparent discontinuous

    transcription of Trypanosoma brucei variant surface antigen genes. Nature 311:350-355.

    14. Chari, A., M. M. Golas, M. Klingenhager, N. Neuenkirchen, B. Sander, C. Englbrecht, A.

    Sickmann, H. Stark, and U. Fischer. 2008. An assembly chaperone collaborates with the SMN

    complex to generate spliceosomal SnRNPs. Cell 135:497-509.

    15. Cross, M., A. Günzl, Z. Palfi, and A. Bindereif. 1991. Analysis of small nuclear

    ribonucleoproteins (RNPs) in Trypanosoma brucei: structural organization and protein components

    of the spliced leader RNP. Mol. Cell. Biol. 11:5516-5526.

    on March 29, 2021 by guest

    http://ec.asm.org/

    Dow

    nloaded from

    http://ec.asm.org/

  • 25

    16. Cross, M., B. Wieland, Z. Palfi, A. Günzl, U. Röthlisberger, H. W. Lahm, and A. Bindereif.

    1993. The trans-spliceosomal U2 snRNP protein 40K of Trypanosoma brucei: cloning and analysis

    of functional domains reveals homology to a mammalian snRNP protein. EMBO J. 12:1239-1248.

    17. De Gaudenzi, J., A. C. Frasch, and C. Clayton. 2005. RNA-binding domain proteins in

    Kinetoplastids: a comparative analysis. Eukaryot. Cell 4:2106-2114.

    18. Denker, J. A., D. M. Zuckerman, P. A. Maroney, and T. W. Nilsen. 2002. New components of

    the spliced leader RNP required for nematode trans-splicing. Nature 417:667-670.

    19. Djikeng, A., L. Ferreira, M. D'Angelo, P. Dolezal, T. Lamb, S. Murta, V. Triggs, S. Ulbert, A.

    Villarino, S. Renzi, E. Ullu, and C. Tschudi. 2001. Characterization of a candidate Trypanosoma

    brucei U1 small nuclear RNA gene. Mol. Biochem. Parasitol. 113:109-115.

    20. Douris, V., M. J. Telford, and M. Averof. 2010. Evidence for multiple independent origins of

    trans-splicing in Metazoa. Mol. Biol. Evol. 27:684-93.

    21. Dungan, J. M., K. P. Watkins, and N. Agabian. 1996. Evidence for the presence of a small U5-

    like RNA in active trans- spliceosomes of Trypanosoma brucei. EMBO J. 15 :4016-4029.

    22. Fabrizio, P., J. Dannenberg, P. Dube, B. Kastner, H. Stark, H. Urlaub, and R. Lührmann.

    2009. The evolutionarily conserved core design of the catalytic activation step of the yeast

    spliceosome. Mol. Cell 36:593-608.

    23. Gingeras, T. R. 2009. Implications of chimaeric non-co-linear transcripts. Nature 461:206-211.

    24. Grainger, R. J., J. D. Barrass, A. Jacquier, J. C. Rain, and J. D. Beggs. 2009. Physical and

    genetic interactions of yeast Cwc21p, an ortholog of human SRm300/SRRM2, suggest a role at the

    catalytic center of the spliceosome. RNA 15:2161-2173.

    25. Günzl, A., A. Bindereif, E. Ullu, and C. Tschudi. 2000. Determinants for cap trimethylation of the

    U2 small nuclear RNA are not conserved between Trypanosoma brucei and higher eukaryotic

    organisms. Nucleic Acids Res. 28:3702-3709.

    26. Günzl, A., M. Cross, Z. Palfi, and A. Bindereif. 1993. Assembly of the U2 small nuclear

    ribonucleoprotein from Trypanosoma brucei. A mutational analysis. J. Biol. Chem. 268:13336-

    13343.

    on March 29, 2021 by guest

    http://ec.asm.org/

    Dow

    nloaded from

    http://ec.asm.org/

  • 26

    27. Günzl, A. and B. Schimanski . 2009. Tandem affinity purification of proteins. Curr. Protoc. Protein

    Sci. Chapter 19:Unit 19.19.

    28. Hannon, G. J., P. A. Maroney, and T. W. Nilsen. 1991. U small nuclear ribonucleoprotein

    requirements for nematode cis- and trans-splicing in vitro. J. Biol. Chem. 266 :22792-22795.

    29. Ismaili, N., D. Perez-Morga, P. Walsh, A. Mayeda, A. Pays, P. Tebabi, A. R. Krainer, and E.

    Pays. 1999. Characterization of a SR protein from Trypanosoma brucei with homology to RNA-

    binding cis-splicing proteins. Mol. Biochem. Parasitol. 102:103-115.

    30. Ivens, A. C., C. S. Peacock, E. A. Worthey, L. Murphy, G. Aggarwal, M. Berriman, E. Sisk, M.

    A. Rajandream, E. Adlem, R. Aert, A. Anupama, Z. Apostolou, P. Attipoe, N. Bason, C.

    Bauser, A. Beck, S. M. Beverley, G. Bianchettin, K. Borzym, G. Bothe, C. V. Bruschi, M.

    Collins, E. Cadag, L. Ciarloni, C. Clayton, R. M. Coulson, A. Cronin, A. K. Cruz, R. M.

    Davies, J. De Gaudenzi, D. E. Dobson, A. Duesterhoeft, G. Fazelina, N. Fosker, A. C. Frasch,

    A. Fraser, M. Fuchs, C. Gabel, A. Goble, A. Goffeau, D. Harris, C. Hertz-Fowler, H. Hilbert,

    D. Horn, Y. Huang, S. Klages, A. Knights, M. Kube, N. Larke, L. Litvin, A. Lord, T. Louie, M.

    Marra, D. Masuy, K. Matthews, S. Michaeli, J. C. Mottram, S. Muller-Auer, H. Munden, S.

    Nelson, H. Norbertczak, K. Oliver, S. O'neil, M. Pentony, T. M. Pohl, C. Price, B. Purnelle, M.

    A. Quail, E. Rabbinowitsch, R. Reinhardt, M. Rieger, J. Rinta, J. Robben, L. Robertson, J. C.

    Ruiz, S. Rutter, D. Saunders, M. Schafer, J. Schein, D. C. Schwartz, K. Seeger, A. Seyler, S.

    Sharp, H. Shin, D. Sivam, R. Squares, S. Squares, V. Tosato, C. Vogt, G. Volckaert, R.

    Wambutt, T. Warren, H. Wedler, J. Woodward, S. Zhou, W. Zimmermann, D. F. Smith, J. M.

    Blackwell, K. D. Stuart, B. Barrell, and P. J. Myler. 2005. The Genome of the Kinetoplastid

    Parasite, Leishmania major. Science 309:436-442.

    31. Jaé, N., P. Wang, T. Gu, M. Huhn, Z. Palfi, H. Urlaub, and A. Bindereif. 2010. Essential role of

    a trypanosome U4-specific Sm core protein in small nuclear ribonucleoprotein assembly and

    splicing. Eukaryot. Cell 9:379-386.

    32. Jurica, M. S. and M. J. Moore. 2003. Pre-mRNA splicing: awash in a sea of proteins. Mol. Cell

    12:5-14.

    33. Khanna, M., H. Van Bakel, X. Tang, J. A. Calarco, T. Babak, G. Guo, A. Emili, J. F.

    Greenblatt, T. R. Hughes, N. J. Krogan, and B. J. Blencowe. 2009. A systematic characterization

    of Cwc21, the yeast ortholog of the human spliceosomal protein SRm300. RNA 15:2174-2185.

    on March 29, 2021 by guest

    http://ec.asm.org/

    Dow

    nloaded from

    http://ec.asm.org/

  • 27

    34. Kolb, S. J., D. J. Battle, and G. Dreyfuss. 2007. Molecular functions of the SMN complex. J.

    Child Neurol. 22:990-994.

    35. Kooter, J. M., T. de Lange, and P. Borst. 1984. Discontinuous synthesis of mRNA in

    trypanosomes. EMBO J. 3:2387-2392.

    36. Krause, M. and D. Hirsh. 1987. A trans-spliced leader sequence on actin mRNA in C. elegans.

    Cell 49:753-761.

    37. Kroiss, M., J. Schultz, J. Wiesner, A. Chari, A. Sickmann, and U. Fischer. 2008. Evolution of

    an RNP assembly system: a minimal SMN complex facilitates formation of U snRNPs in

    Drosophila melanogaster. Proc. Natl. Acad. Sci. USA 105:10045-10050.

    38. Laufer, G., G. Schaaf, S. Bollgönn, and A. Günzl. 1999. In vitro analysis of alpha-amanitin-

    resistant transcription from the rRNA, procyclic acidic repetitive protein, and variant surface

    glycoprotein gene promoters in Trypanosoma brucei. Mol. Cell. Biol. 19:5466-5473.

    39. Lee, J. H., H. S. Jung, and A. Günzl. 2009. Transcriptionally active TFIIH of the early-diverged

    eukaryote Trypanosoma brucei harbors two novel core subunits but not a cyclin-activating kinase

    complex. Nucleic Acids Res. 37:3811-3820.

    40. Liang, X. H., A. Haritan, S. Uliel, and S. Michaeli. 2003. trans and cis Splicing in

    Trypanosomatids: Mechanism, Factors, and Regulation. Eukaryot. Cell 2:830-840.

    41. Liang, X. H., Q. Liu, L. Liu, C. Tschudi, and S. Michaeli. 2006. Analysis of spliceosomal

    complexes in Trypanosoma brucei and silencing of two splicing factors Prp31 and Prp43. Mol.

    Biochem. Parasitol. 145:29-39.

    42. Liu, Q., X. H. Liang, S. Uliel, M. Belahcen, R. Unger, and S. Michaeli. 2004. Identification and

    functional characterization of LSm proteins in Trypanosoma brucei. J. Biol. Chem. 279:18210-

    18219.

    43. Long, J. C. and J. F. Caceres. 2009. The SR protein family of splicing factors: master regulators of

    gene expression. Biochem. J. 417:15-27.

    44. Lücke, S., K. Jurchott, L. H. Hung, and A. Bindereif. 2005. mRNA splicing in Trypanosoma

    brucei: branch-point mapping reveals differences from the canonical U2 snRNA-mediated

    recognition. Mol. Biochem. Parasitol. 142:248-251.

    on March 29, 2021 by guest

    http://ec.asm.org/

    Dow

    nloaded from

    http://ec.asm.org/

  • 28

    45. Lücke, S., T. Klöckner, Z. Palfi, M. Boshart, and A. Bindereif. 1997. Trans mRNA splicing in

    trypanosomes: cloning and analysis of a PRP8-homologous gene from Trypanosoma brucei

    provides evidence for a U5-analogous RNP. EMBO J. 16:4433-4440.

    46. Lücke, S., K. Jurchott, L. H. Hung, and A. Bindereif. 2005. mRNA splicing in Trypanosoma

    brucei: branch-point mapping reveals differences from the canonical U2 snRNA-mediated

    recognition. Mol. Biochem. Parasitol. 142:248-251.

    47. Lührmann, R. and H. Stark. 2009. Structural mapping of spliceosomes by electron microscopy.

    Curr. Opin. Struct. Biol. 19:96-102.

    48. Maier, A. G., H. Webb, M. Ding, M. Bremser, M. Carrington, and C. Clayton. 2001. The

    coatomer of Trypanosoma brucei. Mol. Biochem. Parasitol. 115:55-61.

    49. Mair, G., H. Shi, H. Li, A. Djikeng, H. O. Aviles, J. R. Bishop, F. H. Falcone, C. Gavrilescu, J.

    L. Montgomery, M. I. Santori, L. S. Stern, Z. Wang, E. Ullu, and C. Tschudi. 2000. A new

    twist in trypanosome RNA metabolism: cis-splicing of pre-mRNA. RNA 6:163-169.

    50. Makarova, O. V., E. M. Makarov, S. Liu, H. P. Vornlocher, and R. Lührmann. 2002. Protein

    61K, encoded by a gene (PRPF31) linked to autosomal dominant retinitis pigmentosa, is required

    for U4/U6*U5 tri-snRNP formation and pre-mRNA splicing. EMBO J. 21:1148-1157.

    51. Manful, T., M. Cristodero, and C. Clayton. 2009. DRBD1 is the Trypanosoma brucei homologue

    of the spliceosome-associated protein 49. Mol. Biochem. Parasitol. 166:186-189.

    52. Manger, I. D. and J. C. Boothroyd. 1998. Identification of a nuclear protein in Trypanosoma

    brucei with homology to RNA-binding proteins from cis-splicing systems. Mol. Biochem. Parasitol.

    97:1-11.

    53. Manger, I. D. and J. C. Boothroyd. 2001. Targeted disruption of an essential RNA-binding protein

    perturbs cell division in Trypanosoma brucei. Mol. Biochem. Parasitol. 116:239-245.

    54. Martin, A., S. Schneider, and B. Schwer. 2002. Prp43 is an essential RNA-dependent ATPase

    required for release of lariat-intron from the spliceosome. J. Biol. Chem. 277:17743-17750.

    55. Mattaj, I. W. 1986. Cap trimethylation of U snRNA is cytoplasmic and dependent on U snRNP

    protein binding. Cell 46:905-911.

    on March 29, 2021 by guest

    http://ec.asm.org/

    Dow

    nloaded from

    http://ec.asm.org/

  • 29

    56. Michaeli, S., T. G. Roberts, K. P. Watkins, and N. Agabian. 1990. Isolation of distinct small

    ribonucleoprotein particles containing the spliced leader and U2 RNAs of Trypanosoma brucei. J.

    Biol. Chem. 265:10582-10588.

    57. Milhausen, M., R. G. Nelson, S. Sather, M. Selkirk, and N. Agabian. 1984. Identification of a

    small RNA containing the trypanosome spliced leader: a donor of shared 5' sequences of

    trypanosomatid mRNAs? Cell 38:721-729.

    58. Mottram, J., K. L. Perry, P. M. Lizardi, R. Lührmann, N. Agabian, and R. G. Nelson. 1989.

    Isolation and sequence of four small nuclear U RNA genes of Trypanosoma brucei subsp. brucei:

    identification of the U2, U4, and U6 RNA analogs. Mol. Cell. Biol. 9:1212-1223.

    59. Murphy, W. J., K. P. Watkins, and N. Agabian. 1986. Identification of a novel Y branch structure

    as an intermediate in trypanosome mRNA processing: evidence for trans splicing. Cell 47:517-525.

    60. Neuenkirchen, N., A. Chari, and U. Fischer. 2008. Deciphering the assembly pathway of Sm-

    class U snRNPs. FEBS Lett. 582:1997-2003.

    61. Nilsen, T. W. and B. R. Graveley. 2010. Expansion of the eukaryotic proteome by alternative

    splicing. Nature 463:457-463.

    62. Palfi, Z. and A. Bindereif. 1992. Immunological characterization and intracellular localization of

    trans-spliceosomal small nuclear ribonucleoproteins in Trypanosoma brucei. J. Biol. Chem.

    267:20159-20163.

    63. Palfi, Z., A. Günzl, M. Cross, and A. Bindereif. 1991. Affinity purification of Trypanosoma

    brucei small nuclear ribonucleoproteins reveals common and specific protein components. Proc.

    Natl. Acad. Sci. USA 88:9097-9101.

    64. Palfi, Z., N. Jaé, C. Preußer, K. H. Kaminska, J. M. Bujnicki, J. H. Lee, A. Günzl, C.

    Kambach, H. Urlaub, and A. Bindereif. 2009. SMN-assisted assembly of snRNP-specific Sm

    cores in trypanosomes. Genes Dev. 23:1650-1664.

    65. Palfi, Z., W. S. Lane, and A. Bindereif. 2002. Biochemical and functional characterization of the

    cis-spliceosomal U1 small nuclear RNP from Trypanosoma brucei. Mol. Biochem. Parasitol.

    121:233-243.

    66. Palfi, Z., S. Lücke, H. W. Lahm, W. S. Lane, V. Kruft, E. Bragado-Nilsson, B. Seraphin, and

    A. Bindereif. 2000. The spliceosomal snRNP core complex of Trypanosoma brucei: cloning and

    on March 29, 2021 by guest

    http://ec.asm.org/

    Dow

    nloaded from

    http://ec.asm.org/

  • 30

    functional analysis reveals seven Sm protein constituents. Proc. Natl. Acad. Sci. USA 97:8967-

    8972.

    67. Palfi, Z., B. Schimanski, A. Günzl, S. Lücke, and A. Bindereif. 2005. U1 small nuclear RNP

    from Trypanosoma brucei: a minimal U1 snRNA with unusual protein components. Nucleic Acids

    Res. 33:2493-2503.

    68. Plessel, G., U. Fischer, and R. Lührmann. 1994. m3G cap hypermethylation of U1 small nuclear

    ribonucleoprotein (snRNP) in vitro: evidence that the U1 small nuclear RNA- (guanosine-N2)-

    methyltransferase is a non-snRNP cytoplasmic protein that requires a binding site on the Sm core

    domain. Mol. Cell. Biol. 14:4160-4172.

    69. Portal, D., J. M. Espinosa, G. S. Lobo, S. Kadener, C. A. Pereira, M. M. De La, Z. Tang, R. J.

    Lin, A. R. Kornblihtt, F. E. Baralle, M. M. Flawia, and H. N. Torres. 2003. An early ancestor in

    the evolution of splicing: a Trypanosoma cruzi serine-arginine-rich protein (TcSR) is functional in

    cis-splicing. Mol. Biochem. Parasitol. 127:37-46.

    70. Preußer, C., Z. Palfi, and A. Bindereif. 2009. Special Sm core complex functions in assembly of

    the U2 small nuclear ribonucleoprotein of Trypanosoma brucei. Eukaryot. Cell 8:1228-1234.

    71. Rajkovic, A., R. E. Davis, J. N. Simonsen, and F. M. Rottman. 1990. A spliced leader is present

    on a subset of mRNAs from the human parasite Schistosoma mansoni. Proc. Natl. Acad. Sci. USA

    87:8879-8883.

    72. Rigaut, G., A. Shevchenko, B. Rutz, M. Wilm, M. Mann, and B. Seraphin. 1999. A generic

    protein purification method for protein complex characterization and proteome exploration. Nat.

    Biotechnol. 17:1030-1032.

    73. Schimanski, B., T. N. Nguyen, and A. Günzl. 2005. Highly efficient tandem affinity purification

    of trypanosome protein complexes based on a novel epitope combination. Eukaryot. Cell 4:1942-

    1950.

    74. Selenko, P., R. Sprangers, G. Stier, D. Buhler, U. Fischer, and M. Sattler. 2001. SMN tudor

    domain structure and its interaction with the Sm proteins. Nat. Struct. Biol. 8:27-31.

    75. Shaked, H., C. Wachtel, P. Tulinski, N. H. Yahia, O. Barda, E. Darzynkiewicz, T. W. Nilsen,

    and S. Michaeli. 2010. Establishment of an in vitro trans-splicing system in Trypanosoma brucei

    that requires endogenous spliced leader RNA. Nucleic Acids Res., Epub PMID: 20159996.

    on March 29, 2021 by guest

    http://ec.asm.org/

    Dow

    nloaded from

    http://ec.asm.org/

  • 31

    76. Siegel, T. N., D. R. Hekstra, X. Wang, S. Dewell, and G. A. Cross. 2010. Genome-wide analysis

    of mRNA abundance in two life-cycle stages of Trypanosoma brucei and identification of splicing

    and polyadenylation sites. Nucleic Acids Res. Epub PMID: 20385579.

    77. Siegel, T. N., K. S. Tan, and G. A. Cross. 2005. Systematic study of sequence motifs for RNA

    trans splicing in Trypanosoma brucei. Mol. Cell. Biol. 25:9586-9594.

    78. Spellman, R. and C. W. Smith. 2006. Novel modes of splicing repression by PTB. Trends

    Biochem. Sci. 31:73-76.

    79. Stern, M. Z., S. K. Gupta, M. Salmon-Divon, T. Haham, O. Barda, S. Levi, C. Wachtel, T. W.

    Nilsen, and S. Michaeli. 2009. Multiple roles for polypyrimidine tract binding (PTB) proteins in

    trypanosome RNA metabolism. RNA 15:648-665.

    80. Sutton, R. E. and J. C. Boothroyd. 1986. Evidence for trans splicing in trypanosomes. Cell

    47:527-535.

    81. Tessier, L. H., M. Keller, R. L. Chan, R. Fournier, J. H. Weil, and P. Imbault. 1991. Short

    leader sequences may be transferred from small RNAs to pre- mature mRNAs by trans-splicing in

    Euglena. EMBO J. 10:2621-2625.

    82. Thomas, J. D., R. C. Conrad, and T. Blumenthal. 1988. The C. elegans trans-spliced leader RNA

    is bound to Sm and has a trimethylguanosine cap. Cell 54:533-539.

    83. Tkacz, I. D., S. Cohen, M. Salmon-Divon, and S. Michaeli. 2008. Identification of the heptameric

    LSm complex that binds U6 snRNA in Trypanosoma brucei. Mol. Biochem. Parasitol. 160:22-31.

    84. Tkacz, I. D., Y. Lustig, M. Z. Stern, M. Biton, M. Salmon-Divon, A. Das, V. Bellofatto, and S.

    Michaeli. 2007. Identification of novel snRNA-specific Sm proteins that bind selectively to U2 and

    U4 snRNAs in Trypanosoma brucei. RNA 13:30-43.

    85. Tschudi, C. and E. Ullu. 1990. Destruction of U2, U4, or U6 small nuclear RNA blocks trans

    splicing in trypanosome cells. Cell 61:459-466.

    86. van Alphen, R. J., E. A. Wiemer, H. Burger, and F. A. Eskens. 2009. The spliceosome as target

    for anticancer treatment. Br. J. Cancer 100:228-232.

    on March 29, 2021 by guest

    http://ec.asm.org/

    Dow

    nloaded from

    http://ec.asm.org/

  • 32

    87. Van der Ploeg, L. H., A. Y. Liu, P. A. Michels, T. D. De Lange, P. Borst, H. K. Majumder, H.

    Weber, G. H. Veeneman, and J. Van Boom. 1982. RNA splicing is required to make the

    messenger RNA for a variant surface antigen in trypanosomes. Nucleic Acids Res. 10:3591-3604.

    88. Van Doren, K. and D. Hirsh. 1988. Trans-spliced leader RNA exists as small nuclear

    ribonucleoprotein particles in Caenorhabditis elegans. Nature 335:556-559.

    89. Vandenberghe, A. E., T. H. Meedel, and K. E. Hastings. 2001. mRNA 5'-leader trans-splicing in

    the chordates. Genes Dev. 15:294-303.

    90. Vazquez, M., C. Atorrasagasti, N. Bercovich, R. Volcovich, and M. J. Levin. 2003. Unique

    features of the Trypanosoma cruzi U2AF35 splicing factor. Mol. Biochem. Parasitol. 128:77-81.

    91. Vazquez, M. P., D. Mualem, N. Bercovich, M. Z. Stern, B. Nyambega, O. Barda, D. Nasiga, S.

    K. Gupta, S. Michaeli, and M. J. Levin. 2009. Functional characterization and protein-protein

    interactions of trypanosome splicing factors U2AF35, U2AF65 and SF1. Mol. Biochem. Parasitol.

    164:137-146.

    92. Wahl, M. C., C. L. Will, and R. Lührmann. 2009. The spliceosome: design principles of a

    dynamic RNP machine. Cell 136:701-718.

    93. Wang, P., Z. Palfi, C. Preußer, S. Lücke, W. S. Lane, C. Kambach, and A. Bindereif. 2006. Sm

    core variation in spliceosomal small nuclear ribonucleoproteins from Trypanosoma brucei. EMBO

    J. 25:4513-4523.

    94. Watkins, K. P., J. M. Dungan, and N. Agabian. 1994. Identification of a small RNA that interacts

    with the 5' splice site of the Trypanosoma brucei spliced leader RNA in vivo. Cell T6:171-182.

    95. Wirtz, E., S. Leal, C. Ochatt, and G. A. M. Cross. 1999. A tightly regulated inducible expression

    system for conditional gene knock-outs and dominant-negative genetics in Trypanosoma brucei.

    Mol. Biochem. Parasitol. 99:89-101.

    96. Xu Yx, H. Ben Shlomo, and S. Michaeli. 1997. The U5 RNA of trypanosomes deviates from the

    canonical U5 RNA: the Leptomonas collosoma U5 RNA and its coding gene. Proc. Natl. Acad. Sci.

    USA 94:8473-8478

    97. Xu, Y., L. Liu, and S. Michaeli. 2000. Functional analyses of positions across the 5' splice site of

    the trypanosomatid spliced leader RNA. Implications for base-pair interaction with U5 and U6

    snRNAs. J. Biol. Chem. 275:27883-27892.

    on March 29, 2021 by guest

    http://ec.asm.org/

    Dow

    nloaded from

    http://ec.asm.org/

  • 33

    Figure Legends

    FIG. 1. Schematic of the mammalian cis splicing and the trypanosome SL trans splicing reactions.

    Upstream exon and spliced leader are drawn as grey rectangles, and downstream exon and the

    trypanosome gene are drawn as black rectangles. 5' and 3' splice sites (SSs) are represented by small open

    boxes, branch points (BPs) by closed circles, polypyrimidine tracts by small striped boxes, and the cap 4

    structure of the spliced leader as an oval. Conserved sequences are provided below the drawing with

    invariant residues underlined. While in mammalian systems, 5'SSs, BPs and 3'SSs exhibit partly

    conserved sequences (R, purine; Y, pyrimidine, N, any base), there is no obvious sequence conservation

    at trypanosome BPs (44) and 3'SSs, although it was shown for the latter that an AC dinucleotide (*)

    preceding the AG residues drastically reduces splicing efficiency unless a compensatory AG dinucleotide

    is present within the 5' untranslated region (77). It appears that the importance of the polypyrimidine tract

    becomes more important when consensus sequences are lacking. Yeast has highly conserved splice site

    and BP sequences and some yeast introns function without a polypyrimidine tract (not shown). The partly

    conserved sequences in mammals require a small polypyrimidine tract in the range of 10 to 12 residues

    (Y10-12) whereas in trypanosomes the polypyrimidine tract is large, an essential sequence determinant for

    efficient splicing, and it starts typically right downstream of the BP (44, 77). After the first

    transesterification reaction, cis splicing results in a lariat intron structure whereas a Y-structure

    intermediate is formed in the SL trans splicing process. After the second transesterification, these intronic

    structures are debranched (not shown) and rapidly degraded.

    FIG. 2. Comparison of known spliceosomal factors of humans and trypanosomes. Schematic drawing of

    spliceosomal complexes during a splicing reaction as described in the mammalian and yeast systems. For

    each complex, proteins are listed that enter the spliceosome at the outlined stage (slightly modified human

    protein repertoire according to ref. (92). Please note that only incoming proteins are listed and proteins

    on March 29, 2021 by guest

    http://ec.asm.org/

    Dow

    nloaded from

    http://ec.asm.org/

  • 34

    leaving the spliceosome in the transitions are not recognized. Bold, blue lettering indicates proteins for

    which orthologs have been found in trypanosomes whereas red lettering specifies trypanosome-specific

    factors. 1, Highly divergent, putative cyclophilin orthologs have been co-purified with SmD1 and SmB1

    (Table 1). 2, U5-100K is a DExD/H-box helicase and it is unclear whether one of the putative

    trypanosome DExD/H-box helicases (Table 1) represents a U5-100K ortholog. 3, U5-Cwc21 is possibly

    the ortholog of human SRM300 but seems to have a trypanosome-specific function (see text). 4, The

    trypanosome exon junction complex has recently been characterized (7) but its specific function in RNA

    splicing or metabolism remains unclear.

    on March 29, 2021 by guest

    http://ec.asm.org/

    Dow

    nloaded from

    http://ec.asm.org/

  • Table 1: Spliceosomal proteins of Trypanosoma brucei

    ANNOTATION 1 ACCESSION NO 2 Mr (kDa) TAP 3 REF./E VALUE 4 ANNOTATION 1 ACCESSION NO 2 Mr (kDa) TAP 3 REF./E VALUE 4

    Sm/LSm proteins PRP19 complex SmB Tb927.2.4540 12.3 1,2,3,4 (66) PRP19 Tb927.2.5240 54.3 1,3,4 E = 3e-42 SmD1 Tb927.7.3120 11.7 1,2 (66) * CDC5 Tb927.5.2060 80.1 - E = 1e-30 SmD2 Tb927.2.5850 12.5 1,2 (66) CRN Tb927.10.9660 87.7 1 E = 7e-41 SmD3 Tb927.4.890 12.4 1,2,3,4 (66) SYF1 Tb927.5.1340 92.2 1 E = 7e-23 SmE Tb927.6.2700 9.6 1,2 (66) ISY1 Tb927.8.1930 31.7 1 ISY1 domain E = 6e-7 SmF Tb09.211.1695 8.4 1,2 (66) KIAA1604/Cwc22 Tb11.01.2520 66.82 2 E = 2e-49. SmG Tb11.01.5915 8.9 1,2 (66)

    SSm2-1/Sm15K Tb927.6.4340 12.8 1,2 (84, 93) un-annotated proteins that co-purified in spliceosomal complexes 5 SSm2-2/Sm16.5K Tb927.10.4950 14.7 1,2 (84,93) cons. hypo. Tb927.8.6280 27.1 1,2 putative cyclophilin (64) SSm4 .Tb927.7.6380 23.2 1,2 (84) cons. hypo. Tb927.8.2090 21.6 2 putative cyclophilin (64)

    LSm2 Tb927.8.5180 13.2 1,2 (1, 83) cons. hypo. Tb927.10.11950 22.4 2 putative Cwc15 (64) LSm3 Tb927.7.7380 10.1 - (42) cons. hypo. Tb927.8.4790 26.0 1 novel LSm4 Tb11.01.5535 14.2 1,2 (42) cons. hypo. Tb927.2.3400 42.0 1 novel LSm5 not assigned 6 12.0 - (83) cons. hypo. Tb927.7.1890 31.0 1 novel LSm6 Tb09.160.2150 9.1 - (42) cons. hypo. Tb927.5.2910 20.0 1 novel LSm7 Tb927.5.4030 10.2 1,2 (42) cons. hypo. Tb11.02.0465 12.1 1 novel LSm8 Tb927.3.1780 14.0 1,2 (42) annotated proteins w/o known splicing function that co-purified in spliceosomal complexes

    SMN/Gemin2 and associated proteins eEF-1α Tb927.10.2100 49.1 2 SMN Tb11.01.6640 17.0 1,2,3,4 (64) HSP70 Tb11.01.3110 75.4 1,2,3 Gemin2 Tb927.10.5640 55.4 1,2,3,4 (64) Importin α Tb927.6.2640 58.0 2 Coatomer α Tb927.4.450 132.0 3,4 (48) La protein Tb927.10.2370 37.7 1,2,3 Coatomer β Tb927.1.2570 110.0 3,4 (48) NORF1 Tb927.5.2140 93.3 1 Coatomer β' Tb927.2.6050 93.9 3,4 (48) PABP1 Tb09.211.2150 62.1 1,2 Coatomer γ Tb11.01.3740 97.5 3 (48) TRYP1 Tb09.160.4250/80 22.4 1 Coatomer δ Tb927.8.5250 57.3 3,4 (48) Coatomer ε Tb11.01.6530 34.8 3,4 (48) miscellaneous splicing factors Coatomer ζ Tb927.10.4270 20.5 3 (48) U2AF35 Tb927.10.3200 29.1 - (90) U2AF65 Tb927.10.3500 96.6 - (91)

    U1 proteins SF1 Tb927.10.9400 31.6 - (91) U1-70K Tb927.8.4830 31.7 1,2 (67) PRP17 Tb927.3.1930 52.8 2 E = 4e-59 U1A Tb927.10.8280/8300 18.0 1 (1) PRP31 Tb927.10.10700 39.7 - (41)

    U1-24K Tb927.3.1090 24.2 1,2 (67) PRP43 Tb927.5.1150 82.9 - (41) U1C Tb927.10.2120 21.7 1,2 (67) PTB1 Tb09.211.0560 37.0 - (79) PTB2 Tb11.01.5690 54.7 - (79)

    U2 proteins TSR1 Tb927.8.900 37.5 - SR-like protein (29) U2A' (U2-40K) Tb927.10.2120 36.5 1,2 (16) RRM1 Tb927.2.4710 50.0 - SR-like protein (52) U2B'' Tb927.3.3480 13.6 1,2 (70) SR protein Tb09.160.5020 17.6 - TriTrypDB, E = 3e-06 SF3a60 Tb927.6.3160 61.5 - TritrypDB, E = 1e-13 SF3b(SAP)155 Tb11.01.3690 122.0 - (4, 51) putative spliceosomal DExD/H-box helicases SF3b(SAP)145 Tb927.6.2000 52.5 - (51) Tb927.6.4600; Tb927.6.4600; Tb927.10.5280; Tb927.10.7280; Tb927.10.9130; Tb11.02.3460; SF3b(SAP)130 Tb927.7.6980 195.0 1 (51) Tb927.7.7300; Tb11.02.1930 SF3b(SAP)49 Tb927.3.5280 29.8 - (51) SF3b(SAP)14b Tb927.10.7390 12.7 - E = 6e-12 (51) putative spliceosomal peptidyl-prolyl cis/trans isomerases SF3b(SAP)10 Tb09.211.2205 10.4 - SF3b10 domain, E= 4e-7 (51) >20 candidates SF3b14 (p14) Tb927.10.7470 13.3 - (4, 51) U4 proteins 1 Annotation according to the human system. PRP3 Tb09.160.2900 63.2 1,2 PRP3 domain, E = 2e-42 2 Accession numbers of the TriTrypDB data base (http://www.tritryp.org/). PRP4 Tb927.10.960 65.5 1,2 (1) 3 Protein was co-tandem affinity-purified with SmD1 (1), SmB (2), SMN (3), or Gemin2 (4).

    * Snu13 Tb09.160.3670 13.6 - E = 5e-34 4 Protein sequences of identifications without experimental support were compared to the human genome and E values determined by NCBI BLAST. REF., references.

    U5 proteins 5 E-values lower than 1e-05 were considered to be not significant. PRP8 Tb09.211.2420 277.0 1,2,4 (45) 6 The gene of LSm5 has not been recognized as a protein coding gene yet.

    * U5-200K Tb927.5.2290 249.3 1 E ≈ 0

    U5-102K Tb11.01.7330 111.0 1 E = 1e-5 Mr, molecular mass. U5-116K Tb11.01.7080 105.5 1,2 E = 8e-100 Asterisks mark genes which have been annotated in this study. U5-40K Tb11.01.2940 35.0 1,2 (1) Trypanosome-specific proteins are shaded in gray. U5-15K Tb927.8.2560 17.7 1 E = 4e-26

    U5-Cwc21 Tb09.160.2110 16.2 1,2 (1)

    on March 29, 2021 by guest

    http://ec.asm.org/

    Dow

    nloaded from

    http://ec.asm.org/

  • on March 29, 2021 by guest

    http://ec.asm.org/

    Dow

    nloaded from

    http://ec.asm.org/

  • on March 29, 2021 by guest

    http://ec.asm.org/

    Dow

    nloaded from

    http://ec.asm.org/

Recommended