Han 2010

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 14

www.biochemj.

org
Biochem. J. (2010) 430, 379–392 (Printed in Great Britain) doi:10.1042/BJ20100396 379

REVIEW ARTICLE
Functional diversity of the hnRNPs: past, present and perspectives
Siew Ping HAN, Yue Hang TANG and Ross SMITH
School of Chemistry and Molecular Biosciences, University of Queensland, Building 76, Cooper Road, Brisbane, Queensland 4072, Australia

The hnRNPs (heterogeneous nuclear ribonucleoproteins) are increasingly incompatible with current knowledge about their

Biochemical Journal
RNA-binding proteins with important roles in multiple aspects structural and functional divergence. Hence, we review the current
of nucleic acid metabolism, including the packaging of nascent literature to examine hnRNP diversity, and discuss how this
transcripts, alternative splicing and translational regulation. impacts upon approaches to the classification of RNA-binding
Although they share some general characteristics, they vary proteins in general.
greatly in terms of their domain composition and functional
properties. Although the traditional grouping of the hnRNPs as a Key words: functional divergence, heterogeneous nuclear
collection of proteins provided a practical framework, which has ribonucleoprotein (hnRNP), RNA-binding protein, structural
guided much of the research on them, this approach is becoming divergence.

INTRODUCTION In the present review, we describe the overall features of the


hnRNPs that have previously formed the basis for their description
The hnRNPs [heterogeneous nuclear RNPs (ribonucleoproteins)] as a structurally and functionally distinct group of proteins. We
are a set of primarily nuclear proteins that bind to nascent then examine our current knowledge of the properties of sub-
transcripts produced by RNA polymerase II, and which do not groups of structurally similar hnRNPs, highlighting the degree of
stably associate with other RNA–protein complexes [1]. The divergence within the hnRNPs. Finally, we discuss the extent of
basis for this definition can be found in the history of the overlap between the hnRNPs and other RBPs, and the implications
experimental procedures used to isolate the hnRNPs. Originally, for how we define and understand the hnRNPs.
sucrose density gradients were used to separate soluble RNA–
protein complexes, leading to the identification of the 40S
core particle, which comprises the hnRNPs A/B and C, and STRUCTURAL FEATURES OF THE hnRNPs
associated RNAs [2]. Furthermore, the use of UV cross-linking to
reliably isolate RBPs (RNA-binding proteins) from intact cells The hnRNPs are modular proteins that usually consist of
increased specificity and the range of hnRNPs detected [3]. multiple domains connected by linker regions of varying length
However, the definitive list of hnRNPs was only generated by (Figure 1). The most prevalent domain amongst the hnRNPs,
immunopurification with monoclonal antibodies against hnRNP the RRM (RNA recognition motif) [5], is characterized by a
C, in combination with two-dimensional gel electrophoresis [4], β 1 -α 1 -β 2 -β 3 -α 2 -β 4 structure and the presence of two highly
which yielded a collection of 20 proteins that were named hnRNPs degenerate RNP consensus sequences, RNP-1 and RNP-2 [6].
A–U (Figure 1). The production of monoclonal antibodies It is one of the most common protein motifs in the proteome,
against specific hnRNPs provided essential tools for the isolation and is estimated to be present in approx. 0.5–1 % of gene
of individual hnRNPs and further characterization of their products [7]. The RRM contacts RNA using the RNP-1 and
sequences and structures. In addition, immunostaining using RNP-2 consensus sequences, which are present on the β 3 and
the same antibodies revealed that they generally had a diffuse β 1 strands respectively. This involves primarily hydrophobic
nucleoplasmic distribution that differed from the speckled patterns interactions between four conserved aromatic protein side chains
of snRNPs (small nuclear ribonucleoproteins) or SR (serine– and two bases, resulting in the binding of RNA to the β-sheet
arginine) proteins. surface. The small set of interactions and exposed binding
Thus the description of the defining features of hnRNPs has platform allow the RRM to bind single-stranded nucleic acids
evolved to incorporate new sequence, structural and functional of variable length, including ssDNA (single-stranded DNA), in a
data. With the advent of genomic databases and bioinformatics non-sequence-specific manner. At the same time, the external β 2
tools, another approach has more recently joined the fold, and the and β 4 strands, linker regions, and C- and N-termini can enhance
hnRNP group of proteins has expanded as paralogues to individual binding affinity for specific sequences or types of nucleic acids,
members were identified based on sequence similarities. As the with binding affinities often in the nanomolar range (reviewed in
inclusion of these later members has been unsystematic and non- [8]). Thus RRMs have the capacity to participate in both general
uniform, this review will mostly be limited to the ‘standard’ list of and specific interactions with nucleic acids.
hnRNPs as set out in Figure 1. Also, hnRNPs N, S and T are poorly It should be noted that not all hnRNPs contain canonical
characterized, apart from their identification as components of RRMs. hnRNPs E/K bind RNA via the hnRNP KH (K homology)
hnRNP complexes, and thus will not be discussed further. domain, which is structurally distinct from the RRM. KH domains

Abbreviations used: aRRM, atypical RNA recognition motif; A2RE, A2-response element; C/EBPα, CCAAT/enhancer-binding protein-α; ELAV, embryonic
lethal abnormal vision; hnRNP, heterogeneous nuclear ribonucleoprotein; IRES, internal ribosome entry site; KH domain, K homology domain; miRNA,
microRNA; NLS, nuclear localization signal; PCBP, poly(C)-binding protein; PTB, polypyrimidine tract-binding protein; PyT, polypyrimidine tract; qRRM,
quasi-RNA recognition motif; RBD, RNA-binding domain; RBP, RNA-binding protein; RNP, ribonucleoprotein; RRM, RNA recognition motif; SMN, survival
motor neuron; snRNP, small nuclear ribonucleoprotein; SR protein, serine–arginine protein; ssDNA, single-stranded DNA; 3 UTR, 3 -untranslated region.
1
To whom correspondence should be addressed (email ross.s@uq.edu.au).


c The Authors Journal compilation 
c 2010 Biochemical Society
380 S. P. Han, Y. H. Tang and R. Smith

Figure 1 hnRNP structures


‘A’ represents the hnRNP A/B proteins (A0, A1, A2/B1 and A3) that are structurally similar. Gly-Rich, glycine-rich.

participate in a range of biological processes through interactions they frequently undergo post-translational modifications, such as
with RNA or ssDNA. The type I KH fold, which is commonly phosphorylation of serine and threonine residues, and methylation
found in eukaryotic proteins, has a β 1 -α 1 -α 2 -β 2 -β  -α  structure of arginine residues (reviewed in [1] and [13]).
that binds nucleic acids in a cleft between the β-sheet and the Another key characteristic of the hnRNPs is that they undergo
α-helices. Unlike the RRM, the KH domain can only nucleocytoplasmic shuttling [14]. This was originally proposed as
accommodate four bases within this narrow groove, primarily a means of transporting mRNA out of the nucleus, and is also es-
recognises nucleic acids by hydrogen bonding and electrostatic sential for the numerous cytoplasmic functions in which hnRNPs
interactions, and binds nucleic acids with micromolar affinity participate. There are differences between the mechanisms by
(reviewed in [9]). hnRNPs F/H contain qRRMs (quasi-RRMs), which hnRNPs move across the nuclear envelope: hnRNPs A1 and
which contain an extra β 3  loop compared with classical RRMs I do not contain classical NLSs (nuclear localization signals), and
and lack the RNP consensus sequences. Consequently, they have their movement is coupled to transcription, whereas the shuttling
a completely different mode of RNA recognition from the RRM of hnRNP K, which contains an NLS, is transcription-independent
[10]. Similarly, there are five strands in the β-sheet of RRM II and [15]. Shuttling hnRNPs undergo homomeric and heteromeric
RRM III of hnRNP I {PTB [PyT (polypyrimidine tract)-binding interactions with each other, thus adding another layer of
protein]}, and all of its RRMs lack canonical RNP motifs [11]. In complexity to their functional properties [16]. Although hetero-
the present review, we will refer to these and other non-standard karyon assays revealed that hnRNPs C and U did not exit the
RRMs (such as the RRM homologues of SR proteins) as atypical nucleus [14], these proteins have subsequently been detected in
(aRRMs). In addition, hnRNP U binds RNA via the glycine-rich cytoplasmic RNA granules [17], and hnRNP C localizes to the
domain [12]. Thus the domain composition of the hnRNPs is cytoplasm under certain cellular conditions, such as viral infection
highly diverse, and importantly, no domain is shared across all of [18].
the members within this protein group. Although these structural and shuttling properties are common
The modularity of the hnRNPs creates structural variation in amongst the hnRNPs, they are not unique to this group of proteins.
terms of domain combinations and arrangements, which in turn As such, they are insufficient for distinguishing hnRNPs from
generates functional diversity. As a result, the hnRNPs are highly other RBPs, such as the SR proteins. Structurally, SR proteins,
versatile proteins that can participate in a wide range of biological similar to hnRNPs, are modular proteins that usually contain
functions (Table 1). one or more RBDs in combination with a characteristic auxillary
domain that is rich in arginine and serine residues [19].
GENERAL CHARACTERISTICS AND FUNCTIONS Functionally, SR proteins are well-known antagonistic partners
with hnRNP A1 in splicing, and are also involved in other
When the hnRNPs were originally described, their presence post-splicing activities, such as mRNA export and translational
in the same complex suggested a corresponding commonality regulation [19,20]. In addition, the nucleocytoplasmic shuttling
of structure and function. Indeed, there are several structural of both hnRNPs and SR proteins is coupled to transcription [21].
features that are shared among the hnRNPs. Firstly, with the These similarities in characteristics between hnRNPs and SR
exception of hnRNP U, all hnRNPs contain RBDs (RNA-binding proteins may reflect trends that are common to proteins involved
domains), often in tandem. Secondly, many of them contain in mRNA metabolism.
RGG boxes, which are repeats of Arg-Gly-Gly tripeptides
with interspersed aromatic amino acids. Thirdly, in addition
to RBDs, hnRNPs tend to contain auxiliary domains with Nucleic acid metabolism
distinctive amino acid compositions, such as glycine-rich, acidic mRNA metabolism can be partitioned into nuclear (transcription,
or proline-rich domains. In addition, alternative splice variants splicing, 5 capping and polyadenylation) and cytoplasmic
have been described for the majority of the hnRNPs. Moreover, (transport, translation and degradation) components, and the


c The Authors Journal compilation 
c 2010 Biochemical Society
hnRNP diversity 381

Table 1 hnRNP functions


For clarity, functions have been categorized based on the predominant nature of the hnRNP interaction, but it should be noted that these categories are not mutually exclusive.

hnRNP DNA interactions Reference(s) RNA interactions Reference(s) Protein interactions Reference(s)

A/B/D Telomere maintenance [57–62,158,159] Splicing [42,45–55,145,160,161]


Transcription [74,162–164] Viral unspliced mRNA transport [165]
DNA replication [73] miRNA processing [69]
mRNA stability [166–170]
Translational regulation [71,171–173]
mRNA editing [174]
mRNA trafficking [35,40,63–65,67,175]
mRNA packaging [38]
C1–2 Chromatin remodelling [176] Splicing [55]
Transcription [177] mRNA retention [1]
mRNA packaging [38]
Translational regulation [82–84]
Telomere biogenesis [61]
E/K Transcription [93,95,96,101,178] Splicing [55,93,97,101,179] Signal transduction and integration [104]
Chromatin remodelling [101] Translational regulation [68,89,90,92–94,99–101]
Telomere biogenesis [61,101] mRNA stability [86–89,94,98,101]
F/H mRNA stability [108] Splicing [110,127]
Splicing [10,56,110,111,138]

G DNA repair [132] Splicing [55,129,130,180]


Tumour suppressor [132]
Transcription factor [131,132]
I/L Transcription [181] Splicing [55,111,112,115–118,122–124,
127,182,183]
mRNA export [184]
Translation regulation [120,125,126,185]
mRNA stability [119,121,125,185]
Polyadenylation [184]
M Splicing [55] Antigen receptor (only for the M4 isoform) [33]
Heat-shock response [31] Thyroglobulin receptor (only for the M4 isoform) [33]
P Transcription [140] Splicing [55,141] Cell spreading and stress response [37]
Genome stability [186] Transcription [141]

Q/R Splicing [146,187]


RNA replication [188]
mRNA stability [189]
mRNA trafficking [36,190]
U Chromatin organization [191] RNA binding [192] Transcription [192,193]
DNA binding [192]

nucleocytoplasmic shuttling of hnRNPs provides an essential bind, in vitro, to a wide variety of RNA and ssDNA sequences
avenue for the coupling of the two compartments. Consequently, in a manner resistant to salt or heparin treatment [1]. However,
the hnRNPs participate in virtually every step of this process more stringent assays using salt concentrations of up to 2 M NaCl
(Figure 2), and also in other aspects of nucleic acid processing, revealed distinct binding affinities, and distinct binding motifs
such as telomere maintenance, chromatin remodelling and DNA were identified using SELEX (systematic evolution of ligands
repair. There is considerable overlap between the functions of by exponential enrichment) [24,25]. Subsequently, more specific
different hnRNPs, particularly between paralogues, but they are binding sequences were identified for each of the hnRNPs using
not functionally redundant. Many of the biological functions different experimental setups in a range of functional contexts
ascribed to the hnRNPs involve dynamic and co-operative (Table 2). The conclusions drawn from such assays are highly de-
interactions within large numbers of proteins, and it is likely pendent on the experimental conditions utilized, as protein–RNA
that the different binding affinities of the hnRNPs target them to interactions are highly dynamic, and RNA secondary structure is
distinct nucleic acid and protein targets within these assemblages. greatly influenced by environmental factors, such as the presence
For instance, the range and specificity of alternative splicing of ions [26,27]. The methods used to study these interactions,
reactions differs between hnRNPs, and different sets of hnRNPs especially in earlier approaches, may not be representative of
assemble on each mRNA as they undergo transport and maturation actual intracellular conditions. In addition, techniques based on
[22,23]. Thus hnRNPs have both generalised roles as RNA the crude separation of RNA–protein complexes by size or density
packaging proteins, and specialised roles that are dependent on cannot distinguish between sets of proteins bound to different
specific RNA–protein or protein–protein interactions. mRNAs. Moreover, although the hnRNPs, in particular hnRNPs
A/B and C, are among the most highly expressed proteins in
the cell, with levels comparable with those of the histones [1],
RNA binding their distribution is not uniform within the cell. Thus local protein
The dual nature of the hnRNPs as both generalists and specialists concentrations, as well as those of accessory binding partners,
is perhaps most evident in their RNA-binding properties. hnRNPs may influence the nature of their binding.


c The Authors Journal compilation 
c 2010 Biochemical Society
382 S. P. Han, Y. H. Tang and R. Smith

Figure 2 Nuclear and cytoplasmic functions of hnRNPs at multiple stages of mRNA metabolism
(1) Packaging of nascent RNA transcripts. (2) Regulation of constitutive and alternative splicing. (3) Assembly of A2RE-containing mRNAs into RNA granules for trafficking, followed by localized
translation of trafficked mRNAs at distal sites. (4) Regulation of mRNA stability. (5) Activation or silencing of mRNA translation. Note that not all hnRNPs participate at each of these stages. Please
refer to Table 1 for specific functions of each hnRNP. An animation of this Figure is available at http://www.BiochemJ.org/bj/430/0379/bj4300379add.htm.

Intracellular localization interesting to see if different patterns are observed in other species
The hnRNPs generally have a diffuse nucleoplasmic distribution and cell types.
but are excluded from nucleoli. In addition, hnRNP L is localized
to discrete non-nucleolar foci associated with chromosomes [28].
hnRNP I is also found within nuclear foci, but each nucleus STRUCTURAL AND FUNCTIONAL DIVERGENCE OF THE hnRNPs
usually only contains a single perinucleolar structure [29]. The The coverage of hnRNPs in the current literature is highly
hnRNPs F/H and M exhibit a speckled pattern similar to, but uneven, with detailed and extensive research into the more popular
distinct from, the distribution of snRNPs [30,31], whereas the hnRNPs, and almost no information available about the rest. The
intranuclear localization of hnRNP G and its paralogues appear following section reviews our current understanding of the better-
to overlap with that of SR proteins [32]. known sets of hnRNPs, and highlights some of their functional
Although hnRNPs are mostly expressed in the nucleus, there are and structural correlations with each other.
a few exceptions to the rule. One of the isoforms of hnRNP M is a
membrane-bound receptor [33], whereas hnRNP Q (SYNCRIP)
is predominantly cytoplasmic [34]. Additionally, the limited
localization of certain hnRNPs to the cytoplasm can also be hnRNPs A/B
of functional importance. For instance, hnRNP A2/B1 and Q The hnRNPs A/B comprise four paralogues: A1, A2/B1, A3
are present in mRNA granules and regulate mRNA trafficking and A0. With the exception of hnRNP A0, for which there is
[35,36], whereas hnRNP P (FUS) is localized to cytoplasmic little functional data in the current literature, the hnRNPs A/B
stress granules and participates in the cellular stress response [37]. are known to be involved in many aspects of RNA processing.
It should be noted that the characterization of hnRNP localization hnRNPs A/B, in combination with hnRNP C, package nascent
patterns has mostly been conducted in HeLa cells, which are transcripts in a non-sequence-specific manner [38]. At the same
human epithelial (cervical carcinoma) cells, and it would be time, the hnRNPs A/B also have distinct and specific preferences


c The Authors Journal compilation 
c 2010 Biochemical Society
hnRNP diversity 383

Table 2 hnRNP-binding sequences and the functional contexts in which II), neurogranin and Arc [67]. In addition, hnRNP A2/B1 recruits
they were identified hnRNP E1 to RNA granules, which inhibits translation of
trafficked mRNAs until they reach their intended destination [68].
hnRNP Binding sequence Functional context Reference(s) The recently discovered, and highly unexpected, role of hnRNP
A1 in miRNA (microRNA) regulation highlights the astounding
A1 UAGAGU, UAGGGU, UAGRG(A/U), Alternative splicing [41–43] functional diversity of this protein. hnRNP A1 binds human
UAGAGA, UAGG, GGGU, UGGGGU
AU-rich mRNA stability [166]
pri-miR-18a and is required for efficient processing of the
A2/B1 GCCAAGGAGCC mRNA trafficking [66] miRNA precursor by Drosha [69]. In addition, it accumulates
TTTAGGG Telomere maintenance [59] in cytoplasmic stress granules, in stress-activated cells, and is
A3 GCCAAGGAGCC mRNA trafficking [40] required for recovery from stress [70]. Cellular stress can also
G-rich, GGCAG repeats, TTAGGG Telomere maintenance [62,159,194] induce the cytoplasmic redistribution of hnRNP A1, whereupon
repeats it acts as an IRES (internal ribosome entry site) trans-acting factor
C U-rich Translation [83]
D UUAGG Alternative splicing [194]
[71]. Besides this, hnRNP A1 can disrupt steroid hormone signal
AUUUA mRNA stability [195] transduction by suppressing the activity of vitamin D receptor,
TTAGGG repeats Telomere maintenance [194,196] thus contributing to vitamin D resistance [72]. Furthermore,
E UUAGG Alternative splicing [194] hnRNP A1 binds directly to and stimulates the nuclease activity
C-rich mRNA stability and translation [197] of FEN-1 (flap endonuclease-1), thus increasing the efficiency
TTAGGG repeats Telomere maintenance [194] of Okazaki fragment maturation during DNA replication [73].
CU-rich elements Translational silencing [198]
F/H G-tract, GGGA Alternative splicing [10,43,109]
Rounding off this remarkable but non-exhaustive list is the role
G-rich Translational regulation [108] of hnRNP A1 in regulating APOE (apolipoprotein E) promoter
G CC(A/C)-rich, AAGU Alternative splicing [130,199] activity, which is important in the development of coronary heart
I UCUUC, UCUU, CUCUCU, Alternative splicing [200–203] disease and Alzheimer’s disease [74].
CUUCUCUCU
K dCdT elements Transcription [204]
L CA repeats RNA stability [121]
U AT-rich Chromatin organization [191]
hnRNP C and Raly
hnRNP C is the founding member of the hnRNPs, and was
also the first hnRNP implicated in splicing [75]. There are two
human spliceoforms, C1 and C2, which differ by 13 residues
for RNA sequences [1,23,39–43]. This seemingly contradictory [76]. Interestingly, mouse hnRNP C has four spliceoforms, thus
behaviour may depend upon the effective local concentration of suggesting that species-specific splicing may be common among
the hnRNPs A/B, as they are among the most abundant proteins the hnRNPs [77]. hnRNP C contains an N-terminal RRM, a C-
in the cell and are often present in molar excess over their high- terminal auxiliary domain that is rich in acidic residues [78],
affinity binding targets [44]. and a leucine zipper motif that promotes oligomerization [79,80].
hnRNPs A1 and A2/B1 have been well-characterized as C13 C2 tetramers bind RNA in a non-sequence-specific manner
splicing repressors that promote distal splice site selection, and to form an hnRNP C–RNA complex, which is the obligate first
regulate the alternative splicing of many mRNAs, including those event in the assembly of the 40S core particle [38]. From 230
for β-globin, NMDA (N-methyl-D-aspartate) receptor, HIV-1, to 240 nucleotides of RNA loop around each C13 C2 tetramer,
Src, survival of motor neuron, follicle-stimulating hormone β with the RNA binding to exposed RNPs on the surface of the
subunit, neurofibromatosis and BRCA1 (breast cancer early-onset tetramer. Surprisingly, mouse cells that do not express detectable
1) [45–54] (see also [55] for a review). However, they apparently levels of hnRNP C (due to insertion of a provirus into the gene)
can also promote exon inclusion [56], which demonstrates the are still viable, do not exhibit any aberrations in gene expression,
context-dependency of splice regulation. Interestingly, A1 also and can differentiate in vitro, albeit more slowly than wild-type
modulates alternative splicing of its own pre-mRNA by binding cells [77]. This implies that other proteins can compensate for
multiple intronic splicing silencer elements that flank alternative hnRNP C in the biologically indispensable process of mRNA
exon 7b [41,42]. Efficient splicing requires the presence of biogenesis. This kind of functional redundancy is not uncommon
multiple hnRNP A1 recognition motifs, which suggests that among the hnRNPs and other RBPs. However, hnRNP C-deficient
hnRNP A1 proteins may act co-operatively to suppress exon mice do not survive embryonic development, presumably because
inclusion, perhaps by looping out the pre-mRNA such that internal their cells cannot meet the heightened metabolic demands during
5 splice sites are repressed [41,43]. this period of rapid growth [77]. Similarly, mice that do not
In addition, hnRNPs A1, A2/B1 and A3 bind to both single- express Raly (hnRNP associated with lethal yellow), which shares
stranded telomere DNA and telomerase RNA, and may act
as molecular adapters that regulate the interactions between a high level of sequence similarity with hnRNP C, do not survive
telomerase and telomeres [57–60]. Also, the hnRNPs A/B, embryonic development [81].
in concert with other hnRNPs and telomere-binding factors, Besides its role in the packaging of nascent transcripts, hnRNP
may influence the higher order structure of the telomere and C also increases mRNA translation by binding the IRES of
protect it from degradation or regulate access to telomerase, thus mRNAs such as those encoding c-Myc, Unr (upstream of N-Ras)
maintaining genome stability [61,62]. and c-sis [82–84]. Although hnRNP C is generally thought of as
hnRNP A2/B1 is well-characterized as an mRNA trafficking having wholly nuclear functions, its binding to IRESs occurs in the
trans-acting factor in both oligodendrocytes and neurons [35,63]. cytoplasm, and this cytoplasmic localization is usually triggered
It recognises the A2RE (A2-response element), a 21-nucleotide by entry into mitosis [82–84].
cis-acting element present in myelin basic protein mRNA [64,65].
hnRNP A2/B1 is necessary and sufficient for the process of
hnRNPs E/K
localization of myelin basic protein mRNA [66], as well as
other mRNAs containing A2RE-like sequences, such as those The hnRNPs E/K, which include hnRNPs E1, E2 and K, are
encoding CaMKII (Ca2+ /calmodulin-dependent protein kinase markedly different from the other hnRNPs in that they contain


c The Authors Journal compilation 
c 2010 Biochemical Society
384 S. P. Han, Y. H. Tang and R. Smith

three KH domains instead of RRMs or RRM-like domains. Owing to a typical RRM β-platform. Rather surprisingly, qRRMs are
to their binding affinities, hnRNPs E1 and E2 are often known deleted from three and severely truncated in one of the six known
as PCBPs [poly(C)-binding proteins] 1 and 2, and, based on their splice variants of hnRNP H3 [106], which implies that these
sequences, they should logically be grouped together with closely- spliceoforms do not directly participate in RNA metabolism.
related paralogues PCBPs 3 and 4 in the same protein family [85]. Although the hnRNPs F/H are highly similar in sequence,
It should be noted, though, that PCBPs 3 and 4 are cytoplasmic, structure and binding preferences, they are differentially regulated
and are not classified as hnRNPs. by treatment with phorbol esters, which reduce expression of
Although the hnRNPs E1 and E2 are highly expressed within hnRNP F but not H1 or H2 [105]. They are also differentially
cell nuclei, most work has centred on their cytoplasmic roles in expressed in both normal and cancerous tissues [107], antagonize
regulating mRNA stability and translation. They are assembled each other in the regulation of polyadenylation of mRNAs
into complexes that stabilize mRNAs via interactions with PABP [108], and have different binding specificities for gene regulatory
[poly(A)-binding protein] [86]. Their mRNA targets include elements [109]. Notably, hnRNP F is predominantly cytoplasmic,
transcripts for C/EBPα (CCAAT/enhancer-binding protein-α), whereas hnRNPs H1 and H2 are mostly nuclear [107]. This feature
which regulates granulocyte differentiation [87], and androgen of overlapping but distinct functionality between paralogues is
receptor, which may play a role in carcinogenesis [88]. In addition, common amongst the hnRNPs.
hnRNPs E1 and E2 are IRES-binding factors that enhance The hnRNPs F/H are perhaps best known for their role in
translation of target mRNAs [89,90]. It has been suggested that regulation of alternative splicing, where interactions between
binding of hnRNP E1 unfolds the secondary structure of the multiple hnRNPs bound to target sites causes looping out
mRNA to allow access to ribosomes [90]. In contrast, hnRNP E1 of intervening introns [56] (the same mechanism has also
inhibits translation of trafficked mRNAs in transit within RNA been proposed for the hnRNPs A/B [41,43]). In addition,
granules [68], and silences lipoxygenase mRNA by binding to hnRNP F regulates polyadenylation site choice in the alternative
its 3 UTR (3 -untranslated region) in a complex with hnRNP K splicing of immunoglobulin heavy chain pre-mRNA by blocking
[91,92]. The role of hnRNP E1 in integrating manifold aspects of recruitment of cleavage stimulation factor, thereby promoting the
gene expression and co-ordinating them with signal transduction differentiation of memory cells into B-cells [110]. As the ratio of
pathways is exemplified in its regulation of the transcription, hnRNP F to hnRNPs H1 and H2 is decreased in memory cells
splicing and translation of p21-activated kinase 1 in response compared with B-cells, and these proteins have been shown to
to mitogens [93]. Likewise, hnRNP E2 is involved in multiple form heteromers, hnRNPs H1 and H2 may sequester hnRNP F
steps in the poliovirus replication cycle, including regulation of and prevent it from promoting differentiation [110]. In addition,
the stability, translation and replication of polioviral mRNA [94]. these proteins are present together with hnRNP I in a neuronal
Similarly, hnRNP K has a wide range of nuclear and splicing enhancer complex that assembles on c-src mRNA [111].
cytoplasmic functions, including mRNA silencing [91,92],
transcription [95,96], splicing [97], and regulation of mRNA
stability [98] and translation [99,100]. Importantly, hnRNP K hnRNP I (PTB)/hnRNP L/nPTB
regulates expression of translation initiation factor 4E and Myc, hnRNP I is more commonly known as PTB because it regulates
which have many downstream targets and global effects on splicing by binding to the PyT at the branch point upstream of
cellular physiology [96]. The functional versatility of hnRNP exons [112]. It contains four RRMs, as does the highly similar
K arises from its modular structure. Its three KH domains hnRNP L [29]. There are conflicting reports about the involvement
can interact with both RNA and ssDNA, and it also contains of each RRM to RNA binding. Oh et al. [113] proposed that only
a K interactive region that can recruit a variety of proteins, RRMs 3 and 4 contributed to RNA binding, and that RRM 2
including kinases and factors regulating transcription, splicing promoted oligomerization. In contrast, Simpson et al. [11] found
and translation [101]. In this way, hnRNP K acts as a that hnRNP I exists as a monomer, and all its RRMs bound
‘docking platform’ to co-ordinate nucleic acid metabolism and RNA. The latter result was supported by Vitali et al. [114], who
mediate cross-talk between signalling pathways [101]. Its role in established that RRMs 1 and 2 act independently of each other and
integrating cellular signals is important in several key pathways, therefore have substantial substrate flexibility, whereas RRMs 3
including the p53 transcriptional response to DNA damage [102] and 4 participate in unusually rigid domain–domain interactions
and the mitochondrial response to insulin [103]. Thus hnRNP K to create an RNA-binding platform that induces formation of RNA
is at the centre of a vast interaction network, with 114 binding loops.
partners having been identified via affinity purification coupled The mechanism by which hnRNP I represses splicing has
with MS [104]. been examined by Sharma et al. [115] in a particularly elegant
illustration of splicing dynamics. Splicing requires a shift from
exon definition, during which splicing and other accessory factors
hnRNPs F/H
mark out exon boundaries, to the assembly of an intron-defined
The hnRNPs F/H include hnRNPs F, H1 (H), H2 (H ) and H3 spliceosome, which catalyses the splicing reaction. This involves
(2H9). Unlike most hnRNPs, which can bind both RNA and a number of transitions through a defined series of spliceosomal
ssDNA independently of sequence, the hnRNPs F/H appear to complexes of varying protein compositions. hnRNP I inhibits
be specific for poly(G) tracts [30]. Their RBDs differ from those the interaction of splicing factors bound to exons upstream and
in most other hnRNPs, in that the residues that contact RNA in downstream of branch points, thus causing splicing to be arrested
standard RRMs are not conserved [30]. Thus the RBDs of the at the pre-spliceosomal A complex. In addition, hnRNP I represses
hnRNPs F/H have been described as qRRMs [105]. Three qRRMs exon inclusion by competing with splicing factor U2AF65 for
are present in each protein, of which qRRM 3 assumes the binding to the PyT [116], and may also act by looping out
canonical RRM fold, whereas qRRMs 1 and 2 are structurally exons flanked by PyTs [117]. Importantly, hnRNP I can induce
distinct from, and have a RNA-binding surface that is more either exon inclusion or skipping, depending on the position of its
exposed, than that of standard RRMs [10]. This may be required binding site relative to the splice site [118].
due to the stability of G-rich secondary structures, which would The pyrimidine-rich sequences to which hnRNP I preferentially
tend not to adopt the extended configuration necessary for binding binds are common in other mRNA regulatory elements that


c The Authors Journal compilation 
c 2010 Biochemical Society
hnRNP diversity 385

control mRNA stability and translation. Stimulation of pancreatic telomere biogenesis, and deviations in telomere length can
β-cells triggers the translocation of hnRNP I to the cytosol, where lead to tumorigenesis [61]. Defects in mRNA splicing are also
it stabilizes mRNAs encoding proteins that form insulin secretory common in cancer development, and changes in the activity of
granules, thus promoting insulin secretion [119]. Besides this, splicing factors, such as hnRNPs A1 and F/H, could lead to
during apoptosis, hnRNP I binds the IRESs of a subset of mRNAs, mis-splicing of their mRNA targets, which include regulators
thereby rendering them resistant to translational inhibition [120]. of cell proliferation such as c-Src and CD44 [50,111,137,138].
hnRNP L preferentially binds to CA repeats [121]. It regulates Similarly, the transcriptional and translational regulatory targets
the inducible exon skipping in CD45 mRNA in response to of hnRNPs E/K include c-Myc, the androgen receptor, eIF4E
T-cell activation, possibly via the stabilization of snRNP binding (eukaryotic translation initiation factor 4E), p53 and C/EBPα
[122,123]. hnRNP L also regulates the alternative splicing of [87,88,96,100,102,139], for which changes in expression levels
other genes via additional mechanisms, including intron retention, could lead to excessive cell proliferation. In addition, hnRNP
suppression of multiple exons and alternative poly(A) site G reduced tumorigenicity in oral carcinoma cells by upregulating
selection [124]. Both hnRNP I and L interact with the 3 UTR thioredoxin-interacting protein, a tumour suppressor [131]. On the
of inducible nitric oxide synthase mRNA, and may regulate its other hand, hnRNP M4 interacts with carcinoembryonic antigen,
stability [125]. Furthermore, both hnRNP L and PTB may co- a cell membrane receptor that may contribute to metastasis in
operatively regulate translation of Cat-1 mRNA during amino colorectal cancer [33]. Finally, chromosomal translocations
acid starvation [126]. in multiple types of cancer, including leukaemias and sarcomas,
nPTB is a truncated paralogue of hnRNP I that is expressed in create chimaeric oncoproteins in which FUS (hnRNP P) is fused
vertebrate neuronal cells, and both hnRNP I and nPTB assemble to transcription factors [140–143].
into common splicing complexes (along with the hnRNPs F/H) Recently, mutations in the FUS gene itself have been linked
[111,127]. However, as nPTB has higher binding affinity, but to familial amyotrophic lateral sclerosis, revealing a novel role
lower splicing activity, than hnRNP I, the expression of nPTB for FUS in neurodegeneration [144]. Another neurodegenerative
reduces splicing repression by hnRNP I [127]. Thus differential disease associated with the hnRNPs is spinal muscular atrophy,
expression of closely-related proteins with divergent functional which is caused by loss of the SMN1 (survival motor neuron
properties results in species- and tissue-specific regulation of RNA 1) gene. SMN2, a nearly identical gene copy, undergoes exon
metabolism. skipping to produce truncated and functionally deficient SMN
protein. As this exon skipping is regulated by hnRNPs A1, G and
Q/R, they could potentially be therapeutic targets for this common
hnRNP G/GT and RBMY
genetic disease [129,145,146]. Furthermore, hnRNP A2/B1 has
hnRNP G (also known as RBMX, which refers to the location of its been implicated in Alzheimer’s disease, and expression of its
gene on the X chromosome), is unique in that it is to date the only isoforms is altered in the hippocampi of patients at different stages
hnRNP shown to be glycosylated [128]. It contains an N-terminal of the disease [147].
RRM and a C-terminal auxiliary domain that is rich in glycine Although many of the hnRNPs play significant roles in the
and basic residues [128]. Depending on the mRNA substrate, development of cancer and neurodegeneration, it is important
hnRNP G can act synergistically with or antagonistically to to note that different hnRNPs contribute to disease progression
Tra2β, a splicing factor, to regulate inclusion of alternative via distinct mechanisms, and exert their effects via pathways
exons [129,130]. In addition, hnRNP G promotes the expression involving different sets of genes. Furthermore, these pathways in-
of tumour suppressor Txnip, and protects DNA double-strand volve multiple RBPs, thus placing the hnRNPs within an
breaks from further degradation, thus reducing tumorigenicity interconnected network of functionally related proteins.
and maintaining genomic stability [131,132].
RBMY is the Y chromosome counterpart of hnRNP G, whereas
hnRNP GT is thought to have originated from retrotransposition
OVERLAPPING FEATURES OF hnRNPs AND OTHER RBPs
of hnRNP G [32,133]. Unlike hnRNP G, which is ubiquitously
expressed, RBMY and hnRNP GT are primarily expressed in The hnRNPs have many features in common with other RBPs,
the testis [133,134]. All three proteins have been shown to such as the SR proteins and ELAV (embryonic lethal abnormal
interact with SR proteins and Tra2β, and to inhibit splicing by vision)-like proteins. Structurally, they comprise multiple RBDs,
the latter [32]. RBMY contains four sets of SRGY-rich repeats in particular the RRM, and often also contain auxiliary domains
(SRGY boxes), which are characteristic of the RS (arginine– (Figure 4). Functionally, they have multiple and overlapping
serine) domains of SR proteins, and localizes to intranuclear roles in the regulation of alternative splicing, mRNA stability,
concentrations of SR proteins known as SR speckles [32]. hnRNP mRNA export and other aspects of post-transcriptional processing
G also contains one SRGY box, which suggests that it may [148,149]. In addition, they are ubiquitously expressed, and most
also have properties that resemble those of the SR proteins. It is shuttle between the nucleus and cytoplasm [13,21,150,151].
therefore tempting to speculate that if hnRNP G had not already Although the hnRNPs, SR proteins and ELAV-like proteins
been described as an hnRNP, it and its paralogues might have have many similarities at both the structural and functional
been classified as SR proteins instead. level, they have generally been described and studied as separate
protein families. The basis for this has more to do with the
history of the field, rather than phylogenetic, structural or
CLINICAL SIGNIFICANCE OF THE hnRNPs
functional evidence. The uncertainty over the delineation of
Given their central roles in the regulation of gene expression, it what distinguishes hnRNPs from other RBPs has prompted
is not surprising that the hnRNPs have been linked to numerous repeated calls for a redefinition of hnRNPs [1,44,55]. The original
diseases. Many of the hnRNPs have been directly or indirectly definition of hnRNPs was mainly operational and was based on the
implicated in cancer development, and aberrant expression of experimental methods available at the time. Although this scheme
hnRNPs is characteristic of various types of cancer [135,136]. has provided a useful conceptual framework for studying RNA–
Dysregulation of hnRNP function contributes to carcinogenesis protein interactions, it is difficult to reconcile with our current
in multiple ways (Figure 3). Many hnRNPs are involved in knowledge of RBP diversity, function and evolution, and has


c The Authors Journal compilation 
c 2010 Biochemical Society
386 S. P. Han, Y. H. Tang and R. Smith

Figure 3 hnRNPs can contribute to carcinogenesis


Note that pathways (1)–(3) can involve multiple hnRNPs, whereas pathways (4) and (5) are specific for hnRNP M4 and Fus (hnRNP P2) respectively. (1) Deviations in telomere biogenesis result
in genome instability. (2) Defects in splicing regulation result in mis-splicing of transcripts involved in cellular proliferation and differentiation, which could alter cell-cycle regulation. (3) Changes
in gene expression at the transcriptional or translational level can up- or down-regulate expression of oncogenes or tumour suppressors. (4) hnRNP M4 interacts with cell membrane receptors to
trigger signalling pathways that promote metastasis. (5) Chromosome translocations involving FUS create chimaeric proteins that act as oncoproteins. Please see text for further details.

Figure 4 Modular structure of selected hnRNPs and other RBPs


Glu-Rich, glycine-rich; Arg/Ser Rich, ariginine- and serine-rich.


c The Authors Journal compilation 
c 2010 Biochemical Society
hnRNP diversity 387

a limited basis in terms of structural, functional or expression features of cis-acting elements that facilitate their association
characteristics. with trans-acting factors, and consequently the molecular basis
However, the description of hnRNPs as a family has slipped into for recognition, we need to determine their three-dimensional
common usage, thus implying that they have a common ancestry structures. For example, mutational analysis has revealed partial
and shared functional properties (although it should be noted that clues to how the A2RE interacts with hnRNP A2/B1, but in
most of the long-standing research groups in this field are usually the absence of complete structural data, our knowledge falls
careful not to describe them as such). This has resulted in cases short of explaining their binding at the atomic level. In contrast,
in which experimental findings based on a subset of hnRNPs are X-ray crystallographic studies have shown that binding of hnRNP
extrapolated to all members of the group, which may be confusing A1 with the telomere repeat, which forms a single-stranded G-
and potentially misleading. In addition, the identification of novel rich overhang, involves telomeric repeat bases interacting with
hnRNPs based on structural similarity to a subset of hnRNPs hydrophobic residues that are on the β1 and β3 strands of the
has led to a growing list of members, but the rules for inclusion RRM module [57]. The identification of specific nucleotides
remain ambiguous and inconsistent [55]. As existing approaches and residues critical for the telomere–hnRNP A1 interaction
do not seem to adequately address the structural, functional and will provide valuable information about RNA–protein binding
evolutionary features of the RBPs, it is important to be aware of in general, and facilitate future experimental design.
these limitations when interpreting the current literature. Bioinformatics approaches have been useful in expanding the
library of hnRNP paralogues and isoforms. This has paved the
way for further studies of their three-dimensional structures in
order to better understand the functions and regulation of these
CONCLUSIONS AND PERSPECTIVES
biologically important, multi-functional proteins. The greatest
The hnRNP proteins were initially viewed as prime constituents need is to identify and characterize their alternatively spliced
of 40S heterogeneous nuclear ribonucleoprotein particles, which isoforms, at the sequence, structural and functional level. To
bind to and stabilize nascent pre-mRNA. However, more recent complete the picture of molecular recognition, complementary
findings have shown that the functions of these proteins extend information is also needed for the cis-acting elements. The
far beyond the packaging of nascent RNA, and they are now three-dimensional structures of these cis-elements are complex:
viewed as a highly divergent group of proteins with roles in many progress has been made in predicting nucleotide sequences that
important aspects of nucleic acid metabolism. will bind specified trans-acting factors, but the details of cis-
Functional variation within the hnRNPs can arise from element association with proteins remain largely obscure.
expression of multiple paralogues and alternatively-spliced In light of the functional complexity of the hnRNPs, the
isoforms. However, paralogues and isoforms are often ascribed continued utility of the existing definition of the hnRNPs is
parallel functions, often in the absence of extant data. For example, questionable, given the lack of commonality within the hnRNPs
hnRNPs A1 and A2/B1 have some common functions, with and their lack of distinction from other RBPs. The prevalence
both being implicated in telomere maintenance and regulation of isoforms adds to the complexity of the nomenclature, which
of cell proliferation. However, hnRNP A2/B1 is important in requires resolution. Ideally, the classification of hnRNPs and other
cytoplasmic RNA trafficking, whereas there is no evidence for RBPs should be based on current knowledge of the structural and
the involvement of hnRNP A1 in this process. Likewise, there is functional properties of these proteins, and accurately describe
functional discrimination between hnRNP A2 isoforms: although relationships based on objective and consistent measures. This
the majority of the hnRNP A2 isoforms are spatially restricted could incorporate approaches based on their evolutionary histories
to the nucleus, we have recently shown that only one minor and homology, or domain-centric approaches that reflect the
isoform, A2b, is associated with the formation of RNA granules highly modular nature of these proteins. We envisage that a more
that undergo cytoplasmic trafficking [152]. Thus it will be logically coherent and robust approach to the classification of
important to determine if other hnRNPs have paralogue- or hnRNPs and other RBPs will facilitate a global understanding
isoform-specific locations and functions. of the functional diversity of these key transcriptome regulators.
To better understand the functional roles of the hnRNPs, further
studies of their three-dimensional structures are required, and Note added in proof (received 23 July 2010)
more effort needs to be invested in attempts to crystallize RBPs
and their complexes with nucleic acids. In particular, we need There has been a recent classification of SR proteins as described
to address the question of whether the protein, nucleic acid or by Manley and Krainer [205].
both adopt a modified structure in the protein–RNA complex.
The three-dimensional structures of several modules of hnRNPs
have been solved to high resolution [10,57,114,153–155]. The
GRD (glycine-rich domain), which is a common domain in ACKNOWLEDGMENTS
the hnRNPs and other RBPs, introduces two complications for
crystallographic studies. First, it lowers protein solubility and We thank Dr Sam Lukowski for helpful feedback.
therefore impedes ready crystallization. Secondly, this unusual
amino acid sequence is predicted to form flexible loops (omega FUNDING
loops) [156] that are pinned together on a backbone and would
This work was supported in part by a grant from the Cancer Council of Queensland to R.S.
not be conducive to crystal formation. For these reasons, to date,
most of the solved hnRNP structures are of individual or tandem
domains, and none have been of intact proteins. REFERENCES
Besides protein structures, RNA structures would provide 1 Dreyfuss, G., Matunis, M. J., Pinolroma, S. and Burd, C. G. (1993) hnRNP proteins and
further information about the nature of RNA binding to the the biogenesis of messenger-RNA. Annu. Rev. Biochem. 62, 289–321
hnRNPs and other RBPs. Tiedge [157] has predicted that 2 Beyer, A. L., Christensen, M. E., Walker, B. W. and Lestourgeon, W. M. (1977)
many RNA elements form three-dimensional spatial codes, Identification and characterization of packaging proteins of core 40S hnRNP particles.
described as kink–turn motifs. Thus, in order to understand the Cell 11, 127–138


c The Authors Journal compilation 
c 2010 Biochemical Society
388 S. P. Han, Y. H. Tang and R. Smith

3 van Eekelen, C. A., Riemen, T. and Van Venrooij, W. J. (1981) Specificity in the 31 Gattoni, R., Mahe, D., Mahl, P., Fischer, N., Mattei, M. G., Stevenin, J. and Fuchs, J. P.
interaction of heterogeneous nuclear RNA and messenger RNA with proteins as revealed (1996) The human hnRNP-M proteins: structure and relation with early heat
by in-vivo cross linking. FEBS Lett. 130, 223–226 shock-induced splicing arrest and chromosome mapping. Nucleic Acids Res. 24,
4 Pinol-Roma, S., Choi, Y. D., Matunis, M. J. and Dreyfuss, G. (1988) Immunopurification 2535–2542
of heterogeneous nuclear ribonucleoprotein-particles reveals an assortment of 32 Venables, J. P., Elliott, D. J., Makarova, O. V., Makarov, E. M., Cooke, H. J. and Eperon,
RNA-binding proteins. Genes Dev. 2, 215–227 I. C. (2000) RBMY, a probable human spermatogenesis factor, and other hnRNP G
5 Hoffman, D. W., Query, C. C., Golden, B. L., White, S. W. and Keene, J. D. (1991) proteins interact with Tra2 beta and affect splicing. Human Mol. Gen. 9, 685–694
RNA-binding domain of the A-protein component of the U1 small nuclear 33 Bajenova, O., Stolper, E., Gapon, S., Sundina, N., Zimmer, R. and Thomas, P. (2003)
ribonucleoprotein analyzed by NMR-spectroscopy is structurally similar to Surface expression of heterogeneous nuclear RNA binding protein M4 on Kupffer cell
ribosomal-proteins. Proc. Natl. Acad. Sci. U.S.A. 88, 2495–2499 relates to its function as a carcinoembryonic antigen receptor. Exp. Cell Res. 291,
6 Birney, E., Kumar, S. and Krainer, A. R. (1993) Analysis of the RNA-recognition motif and 228–241
RS and RGG domains - conservation in metazoan pre-messenger-RNA splicing factors. 34 Mizutani, A., Fukuda, M., Ibata, K., Shiraishi, Y. and Mikoshiba, K. (2000) SYNCRIP, a
Nucleic Acids Res. 21, 5803–5816 cytoplasmic counterpart of heterogeneous nuclear ribonucleoprotein R, interacts with
7 Lunde, B. M., Moore, C. and Varani, G. (2007) RNA-binding proteins: modular design ubiquitous synaptotagmin isoforms. J. Biol. Chem. 275, 9823–9831
for efficient function. Nat. Rev. Mol. Cell Biol. 8, 479–490 35 Shan, J. G., Munro, T. P., Barbarese, E., Carson, J. H. and Smith, R. (2003) A molecular
mechanism for mRNA trafficking in neuronal dendrites. J. Neurosci. 23, 8859–8866
8 Maris, C., Dominguez, C. and Allain, F. H.T. (2005) The RNA recognition motif, a plastic
36 Bannai, H., Fukatsu, K., Mizutani, A., Natsume, T., Iemura, S., Ikegami, T., Inouie, T. and
RNA-binding platform to regulate post-transcriptional gene expression. FEBS J. 272,
Mikoshiba, K. (2004) An RNA-interacting protein, SYNCRIP (heterogeneous nuclear
2118–2131
ribonuclear protein Q1/NSAP1) is a component of mRNA granule transported with
9 Valverde, R., Edwards, L. and Regan, L. (2008) Structure and function of KH domains.
inositol 1,4,5-trisphosphate receptor type 1 mRNA in neuronal dendrites. J. Biol. Chem.
FEBS J. 275, 2712–2726
279, 53427–53434
10 Dominguez, C. and Allain, F. H. T. (2006) NMR structure of the three quasi RNA
37 Andersson, M. K., Stahlberg, A., Arvidsson, Y., Olofsson, A., Semb, H., Stenman, G.,
recognition motifs (qRRMs) of human hnRNP F and interaction studies with Bcl-x
Nilsson, O. and Aman, P. (2008) The multifunctional FUS, EWS and TAF15
G-tract RNA: a novel mode of RNA recognition. Nucleic Acids Res. 34, 3634–3645 proto-oncoproteins show cell type-specific expression patterns and involvement in cell
11 Simpson, P. J., Monie, T. P., Szendroi, A., Davydova, N., Tyzack, J. K., Conte, M. R., spreading and stress response. BMC Cell Biol. 9, 37
Read, C. M., Cary, P. D., Svergun, D. I., Konarev, P. V. et al. (2004) Structure and RNA 38 Huang, M., Rech, J. E., Northington, S. J., Flicker, P. F., Mayeda, A., Krainer, A. R. and
interactions of the N-terminal RRM domains of PTB. Structure 12, 1631–1643 Lestourgeon, W. M. (1994) The C-protein tetramer binds 230 to 240 nucleotides of
12 Kiledjian, M. and Dreyfuss, G. (1992) Primary structure and binding-activity of the premessenger RNA and nucleates the assembly of 40S heterogeneous nuclear
hnRNP U-protein: binding RNA through RGG box. EMBO J. 11, 2655–2664 ribonucleoprotein-particles. Mol. Cell. Biol. 14, 518–533
13 Gorlach, M., Burd, C. G., Portman, D. S. and Dreyfuss, G. (1993) The hnRNP proteins. 39 Shan, J. G., Moran-Jones, K., Munro, T. P., Kidd, G. J., Winzor, D. J., Hoek, K. S. and
Mol. Biol. Rep. 18, 73–78 Smith, B. (2000) Binding of an RNA trafficking response element to heterogeneous
14 Pinol-Roma, S. and Dreyfuss, G. (1992) Shuttling of pre-mRNA binding proteins nuclear ribonucleoproteins A1 and A2. J. Biol. Chem. 275, 38286–38295
between nucleus and cytoplasm. Nature 355, 730–732 40 Ma, A. S. W., Moran-Jones, K., Shan, J., Munro, T. P., Snee, M. J., Hoek, K. S. and
15 Gama-Carvalho, M. and Carmo-Fonseca, M. (2001) The rules and roles of Smith, R. (2002) Heterogeneous nuclear ribonucleoprotein A3, a novel RNA trafficking
nucleocytoplasmic shuttling proteins. FEBS Lett. 498, 157–163 response element-binding protein. J. Biol. Chem. 277, 18010–18020
16 Kim, J. H., Hahm, B., Kim, Y. K., Choi, M. Y. and Jang, S. K. (2000) Protein-protein 41 Chabot, B., Blanchette, M., Lapierre, I. and LaBranche, H. (1997) An intron element
interaction among hnRNPs shuttling between nucleus and cytoplasm. J. Mol. Biol. 298, modulating 5 splice site selection in the hnRNP A1 pre-mRNA interacts with hnRNP A1.
395–405 Mol. Cell. Biol. 17, 1776–1786
17 Kanai, Y., Dohmae, N. and Hirokawa, N. (2004) Kinesin transports RNA: isolation and 42 Hutchison, S., LeBel, C., Blanchette, M. and Chabot, B. (2002) Distinct sets of adjacent
characterization of an RNA-transporting granule. Neuron 43, 513–525 heterogeneous nuclear ribonucleoprotein (hnRNP) A1/A2 binding sites control 5 splice
18 Kneller, E. L. P., Connor, J. H. and Lyles, D. S. (2009) hnRNPs relocalize to the site selection in the hnRNP A1 mRNA precursor. J. Biol. Chem. 277, 29745–29752
cytoplasm following infection with vesicular stomatitis virus. J. Virol. 83, 770–780 43 Han, K., Yeo, G., An, P., Burge, C. B. and Grabowski, P. J. (2005) A combinatorial code
19 Long, J. C. and Caceres, J. F. (2009) The SR protein family of splicing factors: master for splicing silencing: UAGG and GGGG motifs. PLoS Biol. 3, 843–860
regulators of gene expression. Biochem. J. 417, 15–27 44 Dreyfuss, G., Kim, V. N. and Kataoka, N. (2002) Messenger-RNA-binding proteins and
20 Graveley, B. R. (2000) Sorting out the complexity of SR protein functions. RNA 6, the messages they carry. Nat. Rev. Mol. Cell Biol. 3, 195–205
1197–1211 45 Mayeda, A. and Krainer, A. R. (1992) Regulation of alternative pre-messenger-RNA
21 Caceres, J. F., Screaton, G. R. and Krainer, A. R. (1998) A specific subset of SR proteins splicing by hnRNP-A1 and splicing factor-SF2. Cell 68, 365–375
shuttles continuously between the nucleus and the cytoplasm. Genes Dev. 12, 55–66 46 Mayeda, A., Munroe, S. H., Caceres, J. F. and Krainer, A. R. (1994) Function of conserved
22 Mili, S., Shu, H. J., Zhao, Y. M. and Pinol-Roma, S. (2001) Distinct RNP complexes of domains of hnRNP A1 and other hnRNP A/B proteins. EMBO J. 13, 5483–5495
shuttling hnRNP proteins with pre-mRNA and mRNA: candidate intermediates in 47 Tingley, W. G., Roche, K. W., Thompson, A. K. and Huganir, R. L. (1993) Regulation of
NMDA receptor phosphorylation by alternative splicing of the C-terminal domain.
formation and export of mRNA. Mol. Cell. Biol. 21, 7307–7319
Nature 364, 70–73
23 Venables, J. P., Koh, C. S., Froehlich, U., Lapointe, E., Couture, S., Inkel, L., Bramard,
48 Bilodeau, P. S., Domsic, J. K., Mayeda, A., Krainer, A. R. and Stoltzfus, C. M. (2001)
A., Paquet, E. R., Watier, V., Durand, M. et al. (2008) Multiple and specific mRNA
RNA splicing at human immunodeficiency virus type 1 3 splice site A2 is regulated by
processing targets for the major human hnRNP proteins. Mol. Cell. Biol. 28, 6033–6043
binding of hnRNP A/B proteins to an exonic splicing silencer element. J. Virol. 75,
24 Swanson, M. S. and Dreyfuss, G. (1988) Classification and purification of proteins of
8487–8497
heterogeneous nuclear ribonucleoprotein-particles by RNA-binding specificities. Mol.
49 Domsic, J. K., Wang, Y. B., Mayeda, A., Krainer, A. R. and Stoltzfus, C. M. (2003) Human
Cell. Biol. 8, 2237–2241
immunodeficiency virus type 1 hnRNP A/B-Dependent exonic splicing silencer ESSV
25 Tuerk, C. and Gold, L. (1990) Systematic evolution of ligands by exponential antagonizes binding of U2AF65 to viral polypyrimidine tracts. Mol. Cell. Biol. 23,
enrichment: RNA ligands to bacteriophage-T4 DNA-polymerase. Science 249, 505–510 8762–8772
26 Chu, V. B. and Herschlag, D. (2008) Unwinding RNA’s secrets: advances in the biology, 50 Rooke, N., Markovtsov, V., Cagavi, E. and Black, D. L. (2003) Roles for SR proteins and
physics, and modeling of complex RNAs. Curr. Opin. Struct. Biol. 18, 305–314 hnRNP A1 in the regulation of c-src exon N1. Mol. Cell. Biol. 23, 1874–1884
27 Draper, D. E. (2004) A guide to ions and RNA structure. RNA 10, 335–343 51 Singh, N. N., Androphy, E. J. and Singh, R. N. (2004) An extended inhibitory context
28 Pinol-Roma, S., Swanson, M. S., Gall, J. G. and Dreyfuss, G. (1989) A novel causes skipping of exon 7 of SMN2 in spinal muscular atrophy. Biochem. Biophys. Res.
heterogeneous nuclear RNP protein with a unique distribution on nascent transcripts. Commun. 315, 381–388
J. Cell Biol. 109, 2575–2587 52 Feng, J., Lawson, M. A. and Melamed, P. (2008) A proteomic comparison of immature
29 Ghetti, A., Pinolroma, S., Michael, W. M., Morandi, C. and Dreyfuss, G. (1992) hnRNP-I, and mature mouse gonadotrophs reveals novel differentially expressed nuclear proteins
the polypyrimidine tract-binding protein: distinct nuclear-localization and association that regulate gonadotropin gene transcription and RNA splicing. Biol. Reprod. 79,
with hnRNAs. Nucleic Acids Res. 20, 3671–3678 546–561
30 Matunis, M. J., Xing, J. and Dreyfuss, G. (1994) The hnRNP F-protein: unique primary 53 Skoko, N., Baralle, M., Buratti, E. and Baralle, F. E. (2008) The pathological splicing
structure, nucleic acid-binding properties, and subcellular-localization. Nucleic Acids mutation c.6792C>G in NF1 exon 37 causes a change of tenancy between antagonistic
Res. 22, 1059–1067 splicing factors. FEBS Lett. 582, 2231–2236


c The Authors Journal compilation 
c 2010 Biochemical Society
hnRNP diversity 389

54 Goina, E., Skoko, N. and Pagani, F. (2008) Binding of DAZAP1 and hnRNPA1/A2 to an 78 Swanson, M. S., Nakagawa, T. Y., Levan, K. and Dreyfuss, G. (1987) Primary structure of
exonic splicing silencer in a natural BRCA1 exon 18 mutant. Mol. Cell. Biol. 28, human nuclear ribonucleoprotein particle C-proteins: conservation of sequence and
3850–3860 domain-structures in heterogeneous nuclear-RNA, messenger-RNA, and
55 Martinez-Contreras, R., Cloutier, P., Shkreta, L., Fisette, J. F., Revil, T. and Chabot, B. pre-ribosomal-RNA-binding proteins. Mol. Cell. Biol. 7, 1731–1739
(2007) hnRNP proteins and splicing control. Adv. Exp. Med. Biol. 623, 123–147 79 Shahied, L., Braswell, E. H., LeStourgeon, W. M. and Krezel, A. M. (2001) An antiparallel
56 Martinez-Contreras, R., Fisette, J. F., Nasim, F. H., Madden, R., Cordeau, M. and Chabot, four-helix bundle orients the high-affinity RNA binding sites in hnRNP C: a mechanism
B. (2006) Intronic binding sites for hnRNP A/B and hnRNP F/H proteins stimulate for RNA chaperonin activity. J. Mol. Biol. 305, 817–828
pre-mRNA splicing. PLoS Biol. 4, 172–185 80 Whitson, S. R., LeStourgeon, W. M. and Krezel, A. M. (2005) Solution structure of the
57 Ding, J. Z., Hayashi, M. K., Zhang, Y., Manche, L., Krainer, A. R. and Xu, R. M. (1999) symmetric coiled coil tetramer formed by the oligomerization domain of hnRNP C:
Crystal structure of the two-RRM domain of hnRNP A1 (UP1) complexed with implications for biological function. J. Mol. Biol. 350, 319–337
single-stranded telomeric DNA. Genes Dev. 13, 1102–1115 81 Michaud, E. J., Bultman, S. J., Stubbs, L. J. and Woychik, R. P. (1993) The embryonic
58 LaBranche, H., Dupuis, S., Ben-David, Y., Bani, M. R., Wellinger, R. J. and Chabot, B. lethality of homozygous lethal yellow mice (AY /AY ) is associated with the disruption of a
(1998) Telomere elongation by hnRNP A1 and a derivative that interacts with telomeric novel RNA-binding protein. Genes Dev. 7, 1203–1213
repeats and telomerase. Nat. Gen. 19, 199–202 82 Sella, O., Gerlitz, G., Le, S. Y. and Elroy-Stein, O. (1999) Differentiation-induced internal
59 Moran-Jones, K., Wayman, L., Kennedy, D. D., Reddel, R. R., Sara, S., Snee, M. J. and translation of c-sis mRNA: analysis of the cis elements and their differentiation-linked
Smith, R. (2005) hnRNP A2, a potential ssDNA/RNA molecular adapter at the telomere. binding to the hnRNP C protein. Mol. Cell. Biol. 19, 5429–5440
Nucleic Acids Res. 33, 486–496 83 Kim, J. H., Paek, K. Y., Choi, K. B., Kim, T. D., Hahm, B. S., Kim, K. T. and Jang, S. K.
60 Tanaka, E., Fukuda, H., Nakashima, K., Tsuchiya, N., Seimiya, H. and Nakagama, H. (2003) Heterogeneous nuclear ribonucleoprotein C modulates translation of c-myc
(2007) hnRNP A3 binds to and protects mammalian telomeric repeats in vitro . Biochem. mRNA in a cell cycle phase-dependent manner. Mol. Cell. Biol. 23, 708–720
Biophys. Res. Commun. 358, 608–614 84 Schepens, B., Tinton, S. A., Bruynooghe, Y., Parthoens, E., Haegman, M., Beyaert, R.
61 Ford, L. P., Wright, W. E. and Shay, J. W. (2002) A model for heterogeneous nuclear and Cornelis, S. (2007) A role for hnRNP C1/C2 and Unr in internal initiation of
ribonucleoproteins in telomere and telomerase regulation. Oncogene 21, 580–583 translation during mitosis. EMBO J. 26, 158–169
62 Nakagama, H., Higuchi, K., Tanaka, E., Tsuchiya, N., Nakashima, K., Katahira, M. and 85 Makeyev, A. V. and Liebhaber, S. A. (2002) The poly(C)-binding proteins: a multiplicity
Fukuda, H. (2006) Molecular mechanisms for maintenance of G-rich short tandem of functions and a search for mechanisms. RNA 8, 265–278
repeats capable of adopting G4 DNA structures. Mut. Res. Fund. Mol. Mech. Mutagen. 86 Wang, Z. R., Day, N., Trifillis, P. and Kiledjian, M. (1999) An mRNA stability complex
598, 120–131 functions with poly(A)-binding protein to stabilize mRNA in vitro . Mol. Cell. Biol. 19,
63 Hoek, K. S., Kidd, G. J., Carson, J. H. and Smith, R. (1998) hnRNP A2 selectively binds 4552–4560
the cytoplasmic transport sequence of myelin basic protein mRNA. Biochemistry 37, 87 Perrotti, D., Cesi, V., Trotta, R., Guerzoni, C., Santilli, G., Campbell, K., Iervolino, A.,
7021–7029 Condorelli, F., Gambacorti-Passerini, C., Caligiuri, M. A. and Calabretta, B. (2002)
64 Ainger, K., Avossa, D., Diana, A. S., Barry, C., Barbarese, E. and Carson, J. H. (1997) BCR-ABL suppresses C/EBP alpha expression through inhibitory action of hnRNP E2.
Transport and localization elements in myelin basic protein mRNA. J. Cell Biol. 138, Nat. Genet. 30, 48–58
1077–1087 88 Yeap, B. B., Voon, D. C., Vivian, J. P., McCulloch, R. K., Thomson, A. M., Giles, K. M.,
65 Carson, J. H., Worboys, K., Ainger, K. and Barbarese, E. (1997) Translocation of myelin Czyzyk-Krzeska, M. F., Furneaux, H., Wilce, M. C. J., Wilce, J. A. and Leedman, P. J.
basic protein mRNA in oligodendrocytes requires microtubules and kinesin. Cell Motil. (2002) Novel binding of HuR and poly(C)-binding protein to a conserved UC-rich motif
Cytoskel. 38, 318–328 within the 3 -untranslated region of the androgen receptor messenger RNA. J. Biol.
66 Munro, T. P., Magee, R. J., Kidd, G. J., Carson, J. H., Barbarese, E., Smith, L. M. and Chem. 277, 27183–27192
Smith, R. (1999) Mutational analysis of a heterogeneous nuclear ribonucleoprotein A2 89 Ostareck-Lederer, A., Ostareck, D. H. and Hentze, M. W. (1998) Cytoplasmic regulatory
response element for RNA trafficking. J. Biol. Chem. 274, 34389–34395 functions of the KH-domain proteins hnRNPs K and E1/E2. Trends Biochem. Sci. 23,
67 Gao, Y., Tatavarty, V., Korza, G., Levin, M. K. and Carson, J. H. (2008) Multiplexed 409–411
dendritic targeting of alpha-calcium calmodulin-dependent protein kinase II, 90 Pickering, B. M., Mitchell, S. A., Spriggs, K. A., Stoneley, M. and Willis, A. E. (2004)
neurogranin, and activity-regulated cytoskeleton-associated protein RNAs by the A2 Bag-1 internal ribosome entry segment activity is promoted by structural changes
pathway. Mol. Biol. Cell 19, 2311–2327 mediated by Poly(rC) binding protein 1 and recruitment of polypyrimidine tract binding
68 Kosturko, L. D., Maggipinto, M. J., Korza, G., Lee, J. W., Carson, J. H. and Barbarese, E. protein 1. Mol. Cell. Biol. 24, 5595–5605
(2006) Heterogeneous nuclear ribonucleoprotein (hnRNP) E1 binds to hnRNP A2 and 91 Ostareck, D. H., Ostareck-Lederer, A., Wilm, M., Thiele, B. J., Mann, M. and
inhibits translation of A2 response element mRNAs. Mol. Biol. Cell 17, 3521–3533 Hentze, M. W. (1997) mRNA silencing in erythroid differentiation:
69 Guil, S. and Caceres, J. F. (2007) The multifunctional RNA-binding protein hnRNP A1 is hnRNP K and hnRNP E1 regulate 15-lipoxygenase translation from the 3 end. Cell 89,
required for processing of miR-18a. Nat. Struct. Mol. Biol. 14, 591–596 597–606
70 Guil, S., Long, J. C. and Caceres, J. F. (2006) hnRNP A1 relocalization to the stress 92 Ostareck, D. H., Ostareck-Lederer, A., Shatsky, I. N. and Hentze, M. W. (2001)
granules reflects a role in the stress response. Mol. Cell. Biol. 26, 5744–5758 Lipoxygenase mRNA silencing in erythroid differentiation: the 3 UTR regulatory complex
71 Cammas, A., Pileur, F., Bonnal, S., Lewis, S. M., Leveque, N., Holcik, M. and Vagner, S. controls 60S ribosomal subunit joining. Cell 104, 281–290
(2007) Cytoplasmic relocalization of heterogeneous nuclear ribonucleoprotein A1 93 Meng, Q., Rayala, S. K., Gururaj, A. E., Talukder, A. H., O’Malley, B. W. and Kumar, R.
controls translation initiation of specific mRNAs. Mol. Biol. Cell 18, 5048–5059 (2007) Signaling-dependent and coordinated regulation of transcription, splicing, and
72 Chen, H., Hewison, M., Hu, B. and Adams, J. S. (2003) Heterogeneous nuclear translation resides in a single coregulator, PCBP1. Proc. Natl. Acad. Sci. U.S.A. 104,
ribonucleoprotein (hnRNP) binding to hormone response elements: A cause of vitamin 5866–5871
D resistance. Proc. Natl. Acad. Sci. U.S.A. 100, 6109–6114 94 Spear, A., Sharma, N. and Flanegan, J. B. (2008) Protein-RNA tethering: the role of
73 Chai, Q., Zheng, L., Zhou, M., Turchi, J. J. and Shen, B. H. (2003) Interaction and poly(C) binding protein 2 in poliovirus RNA replication. Virology 374, 280–291
stimulation of human FEN-1 nuclease activities by heterogeneous nuclear 95 Stains, J. P., Lecanda, F., Towler, D. A. and Civitelli, R. (2005) Heterogeneous nuclear
ribonucleoprotein A1 in alpha-segment processing during Okazaki fragment maturation. ribonucleoprotein K represses transcription from a cytosine/thymidine-rich element in
Biochemistry 42, 15045–15052 the osteocalcin promoter. Biochem. J. 385, 613–623
74 Campillos, M., Lamas, J. R. N., Garcia, M. A., Bullido, M. J., Valdivieso, F. and Vazquez, 96 Lynch, M., Chen, L., Ravitz, M. J., Mehtani, S., Korenblat, K., Pazin, M. J. and Schmidt,
J. (2003) Specific interaction of heterogeneous nuclear ribonucleoprotein A1 with the E. V. (2005) hnRNP K binds a core polypyrimidine element in the eukaryotic translation
-219T allelic form modulates APOE promoter activity. Nucleic Acids Res. 31, 3063–3070 initiation factor 4E (eIF4E) promoter, and its regulation of eIF4E contributes to neoplastic
75 Choi, Y. D., Grabowski, P. J., Sharp, P. A. and Dreyfuss, G. (1986) Heterogeneous transformation. Mol. Cell. Biol. 25, 6436–6453
nuclear ribonucleoproteins: role in RNA splicing. Science 231, 1534–1539 97 Expert-Bezancon, A., Le Caer, J. P. and Marie, J. (2002) Heterogeneous nuclear
76 Merrill, B. M., Barnett, S. F., Lestourgeon, W. M. and Williams, K. R. (1989) Primary ribonucleoprotein (hnRNP) K is a component of an intronic splicing enhancer complex
structure differences between protein-C1 and protein-C2 of HeLa 40S nuclear that activates the splicing of the alternative exon 6A from chicken beta-tropomyosin
ribonucleoprotein-particles. Nucleic Acids Res. 17, 8441–8449 pre-mRNA. J. Biol. Chem. 277, 16614–16623
77 Williamson, D. J., Banik-Maiti, S., DeGregori, J. and Ruley, H. E. (2000) hnRNP C is 98 Fukuda, T., Naiki, T., Saito, M. and Irie, K. (2009) hnRNP K interacts with RNA binding
required for postimplantation mouse development but is dispensable for cell viability. motif protein 42 and functions in the maintenance of cellular ATP level during stress
Mol. Cell. Biol. 20, 4094–4105 conditions. Genes Cells 14, 113–128


c The Authors Journal compilation 
c 2010 Biochemical Society
390 S. P. Han, Y. H. Tang and R. Smith

99 Habelhah, H., Shah, K., Huang, L., Ostareck-Lederer, A., Burlingame, A. L., Shokat, 122 Melton, A. A., Jackson, J., Wang, J. and Lynch, K. W. (2007) Combinatorial control of
K. M., Hentze, M. W. and Ronai, Z. (2001) ERK phosphorylation drives cytoplasmic signal-induced exon repression by hnRNP l and PSF. Mol. Cell. Biol. 27, 6972–6984
accumulation of hnRNP-K and inhibition of mRNA translation. Nat. Cell Biol. 3, 123 Motta-Mena, L. B., Heyd, F. and Lynch, K. W. Context-dependent regulatory mechanism
325–330 of the splicing factor hnRNP L. Mol. Cell 37, 223–234
100 Mukhopadhyay, N. K., Kim, J., Cinar, B., Ramachandran, A., Hager, M. H., Di Vizio, D., 124 Hung, L. H., Heiner, M., Hui, J. Y., Schreiner, S., Benes, V. and Bindereif, A. (2008)
Adam, R. M., Rubin, M. A., Raychaudhuri, P., De Benedetti, A. and Freeman, M. R. Diverse roles of hnRNP L in mammalian mRNA processing: a combined microarray and
(2009) Heterogeneous nuclear ribonucleoprotein K is a novel regulator of androgen RNAi analysis. RNA 14, 284–296
receptor translation. Cancer Res. 69, 2210–2218 125 Soderberg, M., Raffalli-Mathieu, F. and Lang, M. A. (2002) Inflammation modulates the
101 Bomsztyk, K., Denisenko, O. and Ostrowski, J. (2004) hnRNP K: one protein multiple interaction of heterogeneous nuclear ribonucleoprotein (hnRNP) I/polypyrimidine tract
processes. BioEssays 26, 629–638 binding protein and hnRNP L with the 3 untranslated region of the murine inducible
102 Moumen, A., Masterson, P., O’Connor, M. J. and Jackson, S. P. (2005) hnRNP K: an nitric-oxide synthase mRNA. Mol. Pharmacol. 62, 423–431
HDM2 target and transcriptional coactivator of p53 in response to DNA damage. Cell 126 Majumder, M., Yaman, I., Gaccioli, F., Zeenko, V. V., Wang, C. P., Caprara, M. G.,
123, 1065–1078 Venema, R. C., Komar, A. A., Snider, M. D. and Hatzoglou, M. (2009) The hnRNA-
103 Dzwonek, A., Mikula, M. and Ostrowski, J. (2006) The diverse involvement of binding proteins hnRNP L and PTB are required for efficient translation of the Cat-1
heterogeneous nuclear ribonucleoprotein K in mitochondrial response to insulin. FEBS arginine/lysine transporter mRNA during amino acid starvation. Mol. Cell. Biol. 29,
Lett. 580, 1839–1845 2899–2912
104 Mikula, M., Dzwonek, A., Karczmarski, J., Rubel, T., Dadlez, M., Wyrwicz, L. S., 127 Markovtsov, V., Nikolic, J. M., Goldman, J. A., Turck, C. W., Chou, M. Y. and Black, D. L.
Bomsztyk, K. and Ostrowski, J. (2006) Landscape of the hnRNP K protein–protein (2000) Cooperative assembly of an hnRNP complex induced by a tissue-specific
interactome. Proteomics 6, 2395–2406 homolog of polypyrimidine tract binding protein. Mol. Cell. Biol. 20, 7463–7479
105 Honore, B., Rasmussen, H. H., Vorum, H., Dejgaard, K., Liu, X. D., Gromov, P., Madsen, 128 Soulard, M., Dellavalle, V., Siomi, M. C., Pinolroma, S., Codogno, P., Bauvy, C., Bellini,
P., Gesser, B., Tommerup, N. and Celis, J. E. (1995) Heterogeneous nuclear M., Lacroix, J. C., Monod, G., Dreyfuss, G. and Larsen, C. J. (1993) hnRNP G: sequence
ribonucleoprotein-H, ribonucleoprotein-H , and ribonucleoprotein-F are members of a and characterization of a glycosylated RNA-binding protein. Nucleic Acids Res. 21,
ubiquitously expressed subfamily of related but distinct proteins encoded by genes 4210–4217
mapping to different chromosomes. J. Biol. Chem. 270, 28780–28789 129 Hofmann, Y. and Wirth, B. (2002) hnRNP-G promotes exon 7 inclusion of survival motor
106 Honore, B. (2000) The hnRNP 2H9 gene, which is involved in the splicing reaction, is a neuron (SMN) via direct interaction with Htra2-beta 1. Human Mol. Gen. 11, 2037–2049
multiply spliced gene. Biochim. Biophys. Acta 1492, 108–119 130 Nasim, M. T., Chernova, T. K., Chowdhury, H. M., Yue, B. G. and Eperon, I. C. (2003)
107 Honore, B., Baandrup, U. and Vorum, H. (2004) Heterogeneous nuclear HnRNP G and Tra2 beta: opposite effects on splicing matched by antagonism in RNA
ribonucleoproteins F and H/H show differential expression in normal and selected binding. Human Mol. Gen. 12, 1337–1348
131 Shin, K. H., Kim, R. H., Kang, M. K. and Park, N. H. (2008) hnRNP G elicits
cancer tissues. Exp. Cell Res. 294, 199–209
tumor-suppressive activity in part by upregulating the expression of Txnip. Biochem.
108 Alkan, S. A., Martincic, K. and Milcarek, C. (2006) The hnRNPs F and H2 bind to similar
Biophys. Res. Comm. 372, 880–885
sequences to influence gene expression. Biochem. J. 393, 361–371
132 Shin, K. H., Kang, M. K. and Park, N. H. (2008) Heterogeneous nuclear
109 Caputi, M. and Zahler, A. M. (2001) Determination of the RNA binding specificity of the
ribonucleoprotein G, nitric oxide, and oral carcinogenesis. Nitric Oxide 19, 125–132
heterogeneous nuclear ribonucleoprotein (hnRNP) H/H /F/2H9 family. J. Biol. Chem.
133 Elliott, D. J., Venables, J. P., Newton, C. S., Lawson, D., Boyle, S., Eperon, I. C. and
276, 43850–43859
Cooke, H. J. (2000) An evolutionarily conserved germ cell-specific hnRNP is encoded
110 Veraldi, K. L., Arhin, G. K., Martincic, K., Chung-Ganster, L. H., Wilusz, J. and Milcarek,
by a retrotransposed gene. Human Mol. Gen. 9, 2117–2124
C. (2001) hnRNP F influences binding of a 64-kilodalton subunit of cleavage stimulation
134 Lingenfelter, P. A., Delbridge, M. L., Thomas, S., Hoekstra, H. E., Mitchell, M. J., Graves,
factor to mRNA precursors in mouse B cells. Mol. Cell. Biol. 21, 1228–1238
J. A. M. and Disteche, C. M. (2001) Expression and conservation of processed copies of
111 Chou, M. Y., Rooke, N., Turck, C. W. and Black, D. L. (1999) hnRNP H is a component of
the RBMX gene. Mammal. Genome 12, 538–545
a splicing enhancer complex that activates a c-src alternative exon in neuronal cells.
135 Carpenter, B., MacKay, C., Alnabulsi, A., MacKay, M., Telfer, C., Melvin, W. T. and
Mol. Cell. Biol. 19, 69–77
Murray, G. I. (2006) The roles of heterogeneous nuclear ribonucleoproteins in tumour
112 Patton, J. G., Mayer, S. A., Tempst, P. and Nadalginard, B. (1991) Characterization and
development and progression. Biochim. Biophys. Acta 1765, 85–100
molecular-cloning of polypyrimidine tract-binding protein: a component of a complex
136 Shin, K. H., Kang, M. K., Kim, R. H., Christensen, R. and Park, N. H. (2006)
necessary for pre-messenger RNA splicing. Genes Dev. 5, 1237–1251
Heterogeneous nuclear ribonucleoprotein G shows tumor suppressive effect against oral
113 Oh, Y. L., Hahm, B., Kim, Y. K., Lee, H. K., Lee, J. W., Song, O. K., Tsukiyama-Kohara, K., squamous cell carcinoma cells. Clin. Cancer Res. 12, 3222–3228
Kohara, M., Nomoto, A. and Jang, S. K. (1998) Determination of functional domains in 137 Matter, N., Marx, M., Weg-Remers, S., Ponta, H., Herrlich, P. and Konig, H. (2000)
polypyrimidine-tract-binding protein. Biochem. J. 331, 169–175 Heterogeneous ribonucleoprotein A1 is part of an exon-specific splice-silencing
114 Vitali, F., Henning, A., Oberstrass, F. C., Hargous, Y., Auweter, S. D., Erat, M. and Allain, complex controlled by oncogenic signaling pathways. J. Biol. Chem. 275, 35353–35360
F. H. T. (2006) Structure of the two most C-terminal RNA recognition motifs of PTB using 138 Min, H. S., Chan, R. C. and Black, D. L. (1995) The generally expressed hnRNP-F is
segmental isotope labeling. EMBO J. 25, 150–162 involved in a neural-specific pre-messenger-RNA splicing event. Genes Dev. 9,
115 Sharma, S., Kohlstaedt, L. A., Damianov, A., Rio, D. C. and Black, D. L. (2008) 2659–2671
Polypyrimidine tract binding protein controls the transition from exon definition to an 139 Michelotti, E. F., Tomonaga, T., Krutzsch, H. and Levens, D. (1995) Cellular nucleic-acid
intron defined spliceosome. Nat. Struct. Mol. Biol. 15, 183–191 binding-protein regulates the CT element of the human c-myc protooncogene. J. Biol.
116 Matlin, A. J., Southby, J., Gooding, C. and Smith, C. W. J. (2007) Repression of Chem. 270, 9494–9499
alpha-actinin SM exon splicing by assisted binding of PTB to the polypyrimidine tract. 140 Perrotti, D., Bonatti, S., Trotta, R., Martinez, R., Skorski, T., Salomoni, P., Grassilli, E.,
RNA 13, 1214–1223 Iozzo, R. V., Cooper, D. R. and Calabretta, B. (1998) TLS/FUS, a pro-oncogene involved
117 Lamichhane, R., Daubner, G. M., Thomas-Crusells, J., Auweter, S. D., Manatschal, C., in multiple chromosomal translocations, is a novel regulator of BCR/ABL-mediated
Austin, K. S., Valniuk, O., Allain, F. H. and Rueda, D. (2010) RNA looping by PTB: leukemogenesis. EMBO J. 17, 4442–4455
evidence using FRET and NMR spectroscopy for a role in splicing repression. Proc. Natl. 141 Yang, L., Embree, L. J. and Hickstein, D. D. (2000) TLS–ERG leukemia fusion protein
Acad.Sci. U.S.A. 107, 4105–4110 inhibits RNA splicing mediated by serine-arginine proteins. Mol. Cell. Biol. 20,
118 Xue, Y., Zhou, Y., Wu, T., Zhu, T., Ji, X., Kwon, Y., Zhang, C., Yeo, G., Black, D. L., Sun, 3345–3354
H. et al. (2009) Genome-wide analysis of PTB–RNA interactions reveals a strategy used 142 Panagopoulos, I., Storlazzi, C. T., Fletcher, C. D. M., Fletcher, J. A., Nascimento, A.,
by the general splicing repressor to modulate exon inclusion or skipping. Mol. Cell 36, Domanski, H. A., Wejde, J., Brosjo, O., Rydholm, A., Isaksson, M. et al. (2004) The
996–1006 chimeric FUS/CREB3L2 gene is specific for low-grade fibromyxoid sarcoma. Genes
119 Knoch, K. P., Bergert, H., Borgonovo, B., Saeger, H. D., Altkruger, A., Verkade, P. and Chromosomes Cancer 40, 218–228
Solimena, M. (2004) Polypyrimidine tract-binding protein promotes insulin secretory 143 Goransson, M., Andersson, M. K., Forni, C., Stahlberg, A., Andersson, C., Olofsson, A.,
granule biogenesis. Nat. Cell Biol. 6, 207–214 Mantovani, R. and Aman, P. (2009) The myxoid liposarcoma FUS-DDIT3 fusion
120 Bushell, M., Stoneley, M., Kong, Y. W., Hamilton, T. L., Spriggs, K. A., Dobbyn, H. C., oncoprotein deregulates NF-κB target genes by interaction with NFKBIZ. Oncogene 28,
Qin, X. L., Sarnow, P. and Willis, A. E. (2006) Polypyrimidine tract binding protein 270–278
regulates IRES-mediated gene expression during apoptosis. Mol. Cell 23, 401–412 144 Vance, C., Rogelj, B., Hortobagyi, T., De Vos, K. J., Nishimura, A. L., Sreedharan, J., Hu,
121 Hui, J. Y., Reither, G. and Bindereif, A. (2003) Novel functional role of CA repeats and X., Smith, B., Ruddy, D., Wright, P. et al. (2009) Mutations in FUS, an RNA processing
hnRNP L in RNA stability. RNA 9, 931–936 protein, cause familial amyotrophic lateral sclerosis type 6. Science 323, 1208–1211


c The Authors Journal compilation 
c 2010 Biochemical Society
hnRNP diversity 391

145 Kashima, T., Rao, N., David, C. J. and Manley, J. L. (2007) hnRNP A1 functions with 170 Pautz, A., Linker, K., Altenhofer, S., Heil, S., Schmidt, N., Art, J., Knauer, S., Stauber, R.,
specificity in repression of SMN2 exon 7 splicing. Human Mol. Genet. 16, Sadri, N., Pont, A. et al. (2009) Similar regulation of human inducible nitric-oxide
3149–3159 synthase expression by different isoforms of the RNA-binding protein AUF1. J. Biol.
146 Chen, H. H., Chang, J. G., Lu, R. M., Peng, T. Y. and Tarn, W. Y. (2008) The RNA binding Chem. 284, 2755–2766
protein hnRNP Q modulates the utilization of exon 7 in the survival motor neuron 2 171 Lin, J. Y., Shih, S. R., Pan, M. J., Li, C., Lue, C. F., Stollar, V. and Li, M. L. (2009) hnRNP
(SMN2) gene. Mol. Cell. Biol. 28, 6929–6938 A1 interacts with the 5 untranslated regions of enterovirus 71 and sindbis virus RNA
147 Mizukami, K., Ishikawa, M., Iwakiri, M., Ikonomovic, M. D., Dekosky, S. T., Kamma, H. and is required for viral replication. J. Virol. 83, 6106–6114
and Asada, T. (2005) Immunohistochemical study of the hnRNP A2 and B1 in the 172 Hamilton, B. J., Nichols, R. C., Tsukamoto, H., Boado, R. J., Pardridge, W. M. and Rigby,
hippocampal formations of brains with Alzheimer’s disease. Neurosci. Letters. 386, W. F. C. (1999) hnRNP A2 and hnRNP L bind the 3 UTR of glucose transporter 1 mRNA
111–115 and exist as a complex in vivo . Biochem. Biophys. Res. Commun. 261, 646–651
148 Singh, R. and Valcarcel, J. (2005) Building specificity with nonspecific RNA-binding 173 Paek, K. Y., Kim, C. S., Park, S. M., Kim, J. H. and Jang, S. K. (2008) RNA-binding
proteins. Nat. Struct. Mol. Biol. 12, 645–653 protein hnRNP D modulates internal ribosome entry site-dependent translation of
149 Samson, M. L. (2008) Rapid functional diversification in the structurally conserved hepatitis C virus RNA. J. Virol. 82, 12082–12093
ELAV family of neuronal RNA binding proteins. BMC Genomics 9, 392 174 Lau, P. P., Zhu, H. J., Nakamuta, M. and Chan, L. (1996) Cloning of an apobec-1
interacting protein that also binds apoB mRNA. Circulation 94, 3683–3683
150 Good, P. J. (1995) A conserved family of ELAV-like genes in vertebrates. Proc. Natl.
175 Levesque, K., Halvorsen, M., Abrahamyan, L., Chatel-Chaix, L., Poupon, V., Gordon, H.,
Acad. Sci. U.S.A. 92, 4557–4561
DesGroseillers, L., Gatignol, A. and Mouland, A. J. (2006) Trafficking of HIV-1 RNA is
151 Borgeson, C. D. and Samson, M. L. (2005) Shared RNA-binding sites for interacting
mediated by heterogeneous nuclear ribonucleoprotein A2 expression and impacts on
members of the Drosophila ELAV family of neuronal proteins. Nucleic Acids Res. 33,
viral assembly. Traffic 7, 1177–1193
6372–6383
176 Mahajan, M. C., Narlikar, G. J., Boyapaty, G., Kingston, R. E. and Weissman, S. M.
152 Han, S. P., Friend, L. R., Carson, J. H., Korza, G., Barbarese, E., Maggipinto, M., Hatfield,
(2005) Heterogeneous nuclear ribonucleoprotein C1/C2, MeCP1, and SWI/SNF form a
J. T., Rothnagel, J. A. and Smith, R. (2010) Differential subcellular distributions and
chromatin remodeling complex at the beta-globin locus control region. Proc. Natl. Acad.
trafficking functions of hnRNP A2/B1 spliceoforms. Traffic 11, 886–898 Sci. U.S.A. 102, 15012–15017
153 Backe, P. H., Messias, A. C., Ravelli, R. B. G., Sattler, M. and Cusack, S. (2005) X-ray 177 Chen, H., Hewison, M. and Adams, J. S. (2006) Functional characterization of
crystallographic and NMR studies of the third KH domain of hnRNP K in complex with heterogeneous nuclear ribonuclear protein C1/C2 in vitamin D resistance: a novel
single-stranded nucleic acids. Structure 13, 1055–1067 response element-binding protein. J. Biol. Chem. 281, 39114–39120
154 Du, Z. H., Fenn, S., Tjhen, R. and James, T. L. (2008) Structure of a construct of a human 178 Choi, H. S., Hwang, C. K., Song, K. Y., Law, P. Y., Wei, L. N. and Loh, H. H. (2009)
poly(C)-binding protein containing the first and second KH domains reveals insights Poly(C)-binding proteins as transcriptional regulators of gene expression. Biochem.
into its regulatory mechanisms. J. Biol. Chem. 283, 28757–28766 Biophys. Res. Commun. 380, 431–436
155 Whitson, S. R., LeStourgeon, W. M. and Krezel, A. M. (2005) Solution structure of the 179 Revil, T., Pelletier, J., Toutant, J., Cloutier, A. and Chabot, B. (2009) Heterogeneous
symmetric coiled coil tetramer formed by the oligomerization domain of hnRNP C: nuclear ribonucleoprotein K represses the production of pro-apoptotic Bcl-x(S) splice
implications for biological function. J. Mol. Biol. 350, 319–337 isoform. J. Biol. Chem. 284, 21458–21467
156 Steinert, P. M., Mack, J. W., Korge, B. P., Gan, S. Q., Haynes, S. R. and Steven, A. C. 180 Liu, Y. L., Bourgeois, C. F., Pang, S. C., Kudla, M., Dreumont, N., Kister, L., Sun, Y. H.,
(1991) Glycine loops in proteins: their occurrence in certain intermediate filament Stevenin, J. and Elliott, D. J. (2009) The germ cell nuclear proteins hnRNP G-T and
chains, loricrins and single-stranded RNA-binding proteins. Int. J. Biol. Macromol. 13, RBMY activate a testis-specific exon. PLoS Genet. 5
130–139 181 Kuninger, D. T., Izumi, T., Papaconstantinou, J. and Mitra, S. (2002) Human
157 Tiedge, H. (2006) K-turn motifs in spatial RNA coding. RNA Biol. 3, 133–139 AP-endonuclease 1 and hnRNP-L interact with a nCaRE-like repressor element in the
158 Zhang, Q. S., Manche, L., Xu, R. M. and Krainer, A. R. (2006) hnRNP A1 associates with AP-endonuclease 1 promoter. Nucleic Acids Res. 30, 823–829
telomere ends and stimulates telomerase activity. RNA 12, 1116–1128 182 Cosme, R. S. C., Yamamura, Y. and Tang, Q. Y. (2009) Roles of polypyrimidine tract
159 Huang, P. R., Tsai, S. T., Hsieh, K. H. and Wang, T. C. V. (2008) Heterogeneous nuclear binding proteins in major immediate-early gene expression and viral replication of
ribonucleoprotein A3 binds single-stranded telomeric DNA and inhibits telomerase human cytomegalovirus. J. Virol. 83, 2839–2850
extension in vitro . Biochim. Biophys. Acta 1783, 193–202 183 Rossbach, O., Hung, L. H., Schreiner, S., Grishina, I., Heiner, M., Hui, J. Y. and Bindereif,
160 David, C. J., Chen, M., Assanah, M., Canoll, P. and Manley, J. L. (2010) HnRNP proteins A. (2009) Auto- and cross-regulation of the hnRNP L proteins by alternative splicing.
controlled by c-Myc deregulate pyruvate kinase mRNA splicing in cancer. Nature 463, Mol. Cell. Biol. 29, 1442–1451
364–368 184 Guang, S. H., Felthauser, A. M. and Mertz, J. E. (2005) Binding of hnRNP L to the
161 Moran-Jones, K., Grindlay, J., Jones, M., Smith, R. and Norman, J. C. (2009) hnRNP A2 pre-mRNA processing enhancer of the herpes simplex virus thymidine kinase gene
regulates alternative mRNA splicing of TP53INP2 to control invasive cell migration. enhances both polyadenylation and nucleocytoplasmic export of intronless mRNAs.
Cancer Res. 69, 9219–9227 Mol. Cell. Biol. 25, 6303–6313
162 Gao, C. J., Guo, H. T., Wei, J. P., Mi, Z. Y., Wai, P. and Kuo, P. C. (2004) S-nitrosylation 185 Hwang, B., Lim, J. H., Hahm, B., Jang, S. K. and Lee, S. W. (2009) hnRNP L is required
for the translation mediated by HCV IRES. Biochem. Biophys. Res. Commun. 378,
of heterogeneous nuclear ribonucleoprotein A/B regulates osteopontin transcription in
584–588
endotoxin-stimulated murine macrophages. J. Biol. Chem. 279, 11236–11243
186 Hicks, G. G., Singh, N., Nashabi, A., Mai, S., Bozek, G., Klewes, L., Arapovic, D., White,
163 Tolnay, M., Vereshchagina, L. A. and Tsokos, G. C. (1999) Heterogeneous nuclear
E. K., Koury, M. J., Oltz, E. M. et al. (2000) Fus deficiency in mice results in defective
ribonucleoprotein DOE is a sequence-specific DNA-binding protein. Biochem. J. 338,
B-lymphocyte development and activation, high levels of chromosomal instability and
417–425
perinatal death. Nat. Genet. 24, 175–179
164 Guha, M., Pan, H., Fang, J. K. and Avadhani, N. G. (2009) Heterogeneous nuclear
187 Mourelatos, Z., Abel, L., Yong, J. S., Kataoka, N. and Dreyfuss, G. (2001) SMN
ribonucleoprotein A2 is a common transcriptional coactivator in the nuclear transcription
interacts with a novel family of hnRNP and spliceosomal proteins. EMBO J. 20,
response to mitochondrial respiratory stress. Mol. Biol. Cell. 20, 4107–4119 5443–5452
165 Najera, I., Krieg, M. and Karn, J. (1999) Synergistic stimulation of HIV-1 Rev-dependent 188 Liu, H. M., Aizaki, H., Choi, K. S., Machida, K., Ou, J. J. H. and Lai, M. M. C. (2009)
export of unspliced mRNA to the cytoplasm by hnRNP A1. J. Mol. Biol. 285, SYNCRIP (synaptotagmin-binding, cytoplasmic RNA-interacting protein) is a host factor
1951–1964 involved in hepatitis C virus RNA replication. Virology 386, 249–256
166 Zhao, T. T., Graber, T. E., Jordan, L. E., Cloutier, M., Lewis, S. M., Goulet, I., Cote, J. and 189 Grosset, C., Chen, C. Y. A., Xu, N. H., Sonenberg, N., Jacquemin-Sablon, H. and Shyu,
Holcik, M. (2009) hnRNP A1 regulates UV-induced NF-kappa B signalling through A. B. (2000) A mechanism for translationally coupled mRNA turnover: interaction
destabilization of cIAP1 mRNA. Cell Death Differ. 16, 244–252 between the poly(A) tail and a c-fos RNA coding determinant via a protein complex. Cell
167 Fahling, M., Mrowka, R., Steege, A., Martinka, P., Persson, P. B. and Thiele, B. J. (2006) 103, 29–40
Heterogeneous nuclear ribonucleoprotein-A2/B1 modulate collagen prolyl 190 Rossoll, W., Kroning, A. K., Ohndorf, U. M., Steegborn, C., Jablonka, S. and Sendtner,
4-hydroxylase, α (I) mRNA stability. J. Biol. Chem. 281, 9279–9286 M. (2002) Specific interaction of Smn, the spinal muscular atrophy determining gene
168 Palanisamy, V., Park, N. J., Wang, J. and Wong, D. T. (2008) AUF1 and HuR proteins product, with hnRNP-R and gry-rbp/hnRNP-Q: a role for Smn in RNA processing in
stabilize interleukin-8 mRNA in human saliva. J. Dent. Res. 87, 772–776 motor axons? Human Mol. Genet. 11, 93–105
169 Liao, B. S., Hu, Y. and Brewer, G. (2007) Competitive binding of AUF1 and TIAR to MYC 191 Fackelmayer, F. O. and Richter, A. (1994) Purification of 2 isoforms of hnRNP-U and
mRNA controls its translation. Nat. Struct. Mol. Biol. 14, 511–518 characterization of their nucleic-acid binding-activity. Biochemistry 33, 10416–10422


c The Authors Journal compilation 
c 2010 Biochemical Society
392 S. P. Han, Y. H. Tang and R. Smith

192 Kim, M. K. and Nikodem, V. M. (1999) hnRNP U inhibits carboxy-terminal domain 199 Heinrich, B., Zhang, Z. Y., Raitskin, O., Hiller, M., Benderska, N., Hartmann, A. M.,
phosphorylation by TFIIH and represses RNA polymerase II elongation. Mol. Cell. Biol. Bracco, L., Elliott, D., Ben-Ari, S., Soreq, H. et al. (2009) Heterogeneous nuclear
19, 6833–6844 ribonucleoprotein G regulates splice site selection by binding to CC(A/C)-rich regions
193 Kukalev, A., Nord, Y., Palmberg, C., Bergman, T. and Percipalle, P. (2005) Actin and in pre-mRNA. J. Biol. Chem. 284, 14303–14315
hnRNP U cooperate for productive transcription by RNA polymerase II. Nature Struct. 200 Singh, R., Valcarcel, J. and Green, M. R. (1995) Distinct binding specificities and
Mol. Biol. 12, 238–244 functions of higher eukaryotic polypyrimidine tract-binding proteins. Science 268,
194 Ishikawa, F., Matunis, M. J., Dreyfuss, G. and Cech, T. R. (1993) Nuclear proteins that 1173–1176
bind the premessenger RNA 3 splice-site sequence R(UUAG/G) and the human 201 Perez, I., McAfee, J. G. and Patton, J. G. (1997) Multiple RRMs contribute to
telomeric DNA-sequence D(TTAGGG)N. Mol. Cell. Biol. 13, 4301–4310 RNA-binding specificity and affinity for polypyrimidine tract binding protein.
195 Xu, N. H., Chen, C. Y. A. and Shyu, A. B. (2001) Versatile role for hnRNP D isoforms in Biochemistry 36, 11881–11890
the differential regulation of cytoplasmic mRNA turnover. Mol. Cell. Biol. 21, 6960–6971 202 Chan, R. C. C. and Black, D. L. (1997) The polypyrimidine tract binding protein binds
196 Eversole, A. and Maizels, N. (2000) In vitro properties of the conserved mammalian upstream of neural cell-specific c-src exon N1 to repress the splicing of the intron
protein hnRNP D suggest a role in telomere maintenance. Mol. Cell. Biol. 20, downstream. Mol. Cell. Biol. 17, 4667–4676
5425–5432 203 Amir-Ahmady, B., Boutz, P. L., Markovtsov, V., Phillips, M. L. and Black, D. L. (2005)
197 Reimann, I., Huth, A., Thiele, H. and Thiele, B. J. (2002) Suppression of Exon repression by polypyrimidine tract binding protein. RNA 11, 699–716
15-lipoxygenase synthesis by hnRNP E1 is dependent on repetitive nature of LOX mRNA 204 Michelotti, E. F., Michelotti, G. A., Aronsohn, A. I. and Levens, D. (1996)
3 -UTR control element DICE. J. Mol. Biol. 315, 965–974 Heterogeneous nuclear ribonucleoprotein K is a transcription factor. Mol. Cell. Biol. 16,
198 Holcik, M. and Liebhaber, S. A. (1997) Four highly stable eukaryotic mRNAs assemble 2350–2360
3 untranslated region RNA-protein complexes sharing cis and trans components. Proc. 205 Manley, J. L. and Krainer, A. R. (2010) A rational nomenclature for serine/arginine-rich
Natl. Acad. Sci. U.S.A. 94, 2410–2414 protein splicing factors (SR proteins). Genes Dev. 24, 1073–1074

Received 22 March 2010/29 June 2010; accepted 29 June 2010


Published on the Internet 27 August 2010, doi:10.1042/BJ20100396


c The Authors Journal compilation 
c 2010 Biochemical Society

You might also like