the rna-binding protein hud: a regulator of neuronal differentiation, maintenance and plasticity

12
The RNA-binding protein HuD: a regulator of neuronal differentiation, maintenance and plasticity Julie Desche ˆ nes-Furry, 1 Nora Perrone-Bizzozero, 2 and Bernard J. Jasmin 1,3 * Summary mRNA stability is increasingly recognized as being essential for controlling the expression of a wide variety of transcripts during neuronal development and synaptic plasticity. In this context, the role of AU-rich elements (ARE) contained within the 3 0 untranslated region (UTR) of transcripts has now emerged as key because of their high incidence in a large number of cellular mRNAs. This important regulatory element is known to significantly modulate the longevity of mRNAs by interacting with available stabilizing or destabilizing RNA-binding pro- teins (RBP). Thus, in parallel with the emergence of ARE, RBP are also gaining recognition for their pivotal role in regulating expression of a variety of mRNAs. In the nervous system, the member of the Hu family of ARE- binding proteins known as HuD, has recently been implicated in multiple aspects of neuronal function including the commitment and differentiation of neuronal precursors as well as synaptic remodeling in mature neurons. Through its ability to interact with ARE and stabilize multiple transcripts, HuD has now emerged as an important regulator of mRNA expression in neurons. The present review is designed to provide a comprehen- sive and updated view of HuD as an RBP in the nervous system. Additionally, we highlight the role of HuD in multiple aspects of a neuron’s life from early differentia- tion to changes in mature neurons during learning paradigms and in response to injury and regeneration. Finally, we describe the current state of knowledge concerning the molecular and cellular events regulating the expression and activity of HuD in neurons. BioEssays 28:822–833, 2006. ß 2006 Wiley Periodicals, Inc. Introduction In recent years, the role of the 3 0 untranslated region (3 0 UTR) has become increasingly recognized in modulating mRNA stability, localization and translation in several cell biological aspects. More specifically, the 3 0 UTR can control mRNA expression at the earliest stages of oogenesis, embryogen- esis and tissue development (see for review Refs 1,2). Post- transcriptional events have been implicated in the regulation of short-lived mRNAs encoding specific cytokines, growth- factors and immediate-early response genes (see for review Ref. 3). Importantly, post-transcriptional mechanisms are not limited to short-lived transcripts since they are also important for the control of various mRNAs encoding proteins involved in metabolic activities including for example, the regulation of parathyroid hormone mRNA stability by calcium and phos- phate. (4) In parallel to the discovery of additional mRNAs being regulated via post-transcriptional mechanisms, there is also an increase in the number of diseases of the cardiovascular, immune and nervous systems that are associated with mutations in the 3 0 UTR of specific mRNAs or with misregula- tion of RNA-binding proteins (RBP) that interact with 3 0 UTRs (see for review Refs 3,5–8). Within the nervous system, the role of RBP in all aspects of neuronal function is not only becoming evident but is also 1 Department of Cellular and Molecular Medicine and Centre for Neuromuscular Disease, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada. 2 Department of Neurosciences, University of New Mexico, School of Medicine, Albuquerque, New Mexico. 3 Molecular Medicine Program, Ottawa Health Research Institute, Ottawa, Ontario, Canada. Funding agencies. Work in the Jasmin laboratory is supported by the Canadian Institutes of Health Research, the Muscular Dystrophy Association of America and the Association Franc ¸aise contre les Myopathies. Work in Perrone-Bizzozero’s laboratory is supported by the National Institutes of Health; Grant number: NS30255. *Correspondence to: Bernard J. Jasmin, Department of Cellular and Molecular Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON, K1H 8M5. E-mail: [email protected] DOI 10.1002/bies.20449 Published online in Wiley InterScience (www.interscience.wiley.com). 822 BioEssays 28.8 BioEssays 28:822–833, ß 2006 Wiley Periodicals, Inc. Abbreviations: 3 0 UTR, 3 0 untranslated region; AChE, acetylcholines- terase; ARE, AU-rich element; CARM1, co-activator associated methyltransferase 1; CPEB, cytoplasmic polyadenylation element binding; DRG, dorsal root ganglion; ELAV, embryonic lethal abnor- mal-vision; FMRP, Fragile-X mental retardation protein; hnRNPA1 or K, heteronuclear ribonucleoprotein A1 or K; IMP1, insulin-like growth factor mRNA binding protein 1; KSRP, K homology-type splicing regulatory protein; MARCKS, myristoylated alanine-rich C kinase substrate; PABP, poly(A)-binding protein; RBP, RNA-binding protein; RNP, ribonucleoprotein; RRM, RNA-recognition motif; SCG, superior cervical ganglion; SNP, single nucleotide polymorphism; ZBP, zipcode- binding protein. Review articles

Upload: julie-deschenes-furry

Post on 06-Jun-2016

214 views

Category:

Documents


2 download

TRANSCRIPT

The RNA-binding protein HuD:a regulator of neuronal differentiation,maintenance and plasticityJulie Deschenes-Furry,1 Nora Perrone-Bizzozero,2 and Bernard J. Jasmin1,3*

SummarymRNA stability is increasingly recognized as beingessential for controlling the expression of a wide varietyof transcripts during neuronal development and synapticplasticity. In this context, the role of AU-rich elements(ARE)containedwithin the30 untranslated region (UTR)oftranscripts has nowemerged as key because of their highincidence in a large number of cellular mRNAs. Thisimportant regulatory element is known to significantlymodulate the longevity of mRNAs by interacting withavailable stabilizing or destabilizing RNA-binding pro-teins (RBP). Thus, in parallel with the emergence of ARE,RBP are also gaining recognition for their pivotal role in

regulating expression of a variety of mRNAs. In thenervous system, the member of the Hu family of ARE-binding proteins known as HuD, has recently beenimplicated in multiple aspects of neuronal functionincluding the commitment anddifferentiation of neuronalprecursors as well as synaptic remodeling in matureneurons. Through its ability to interact with ARE andstabilize multiple transcripts, HuD has now emerged asan important regulator of mRNA expression in neurons.The present review is designed to provide a comprehen-sive and updated view of HuD as an RBP in the nervoussystem. Additionally, we highlight the role of HuD inmultiple aspects of a neuron’s life from early differentia-tion to changes in mature neurons during learningparadigms and in response to injury and regeneration.Finally, we describe the current state of knowledgeconcerning the molecular and cellular events regulatingthe expression and activity of HuD in neurons.BioEssays28:822–833, 2006. � 2006 Wiley Periodicals, Inc.

Introduction

In recent years, the role of the 30untranslated region (30UTR)

has become increasingly recognized in modulating mRNA

stability, localization and translation in several cell biological

aspects. More specifically, the 30UTR can control mRNA

expression at the earliest stages of oogenesis, embryogen-

esis and tissue development (see for review Refs 1,2). Post-

transcriptional events have been implicated in the regulation of

short-lived mRNAs encoding specific cytokines, growth-

factors and immediate-early response genes (see for review

Ref. 3). Importantly, post-transcriptional mechanisms are not

limited to short-lived transcripts since they are also important

for the control of variousmRNAs encoding proteins involved in

metabolic activities including for example, the regulation of

parathyroid hormone mRNA stability by calcium and phos-

phate.(4) In parallel to the discovery of additionalmRNAs being

regulated via post-transcriptional mechanisms, there is also

an increase in the number of diseases of the cardiovascular,

immune and nervous systems that are associated with

mutations in the 30UTR of specific mRNAs or with misregula-

tion of RNA-binding proteins (RBP) that interact with 30UTRs

(see for review Refs 3,5–8).

Within the nervous system, the role of RBP in all aspects of

neuronal function is not only becoming evident but is also

1Department of Cellular and Molecular Medicine and Centre for

Neuromuscular Disease, Faculty of Medicine, University of Ottawa,

Ottawa, Ontario, Canada.2Department of Neurosciences, University of New Mexico, School of

Medicine, Albuquerque, New Mexico.3Molecular Medicine Program, Ottawa Health Research Institute,

Ottawa, Ontario, Canada.

Funding agencies. Work in the Jasmin laboratory is supported by the

Canadian Institutes of Health Research, the Muscular Dystrophy

Association of America and the Association Francaise contre les

Myopathies. Work in Perrone-Bizzozero’s laboratory is supported by

the National Institutes of Health; Grant number: NS30255.

*Correspondence to: Bernard J. Jasmin, Department of Cellular and

Molecular Medicine, University of Ottawa, 451 Smyth Road, Ottawa,

ON, K1H 8M5. E-mail: [email protected]

DOI 10.1002/bies.20449

Published online in Wiley InterScience (www.interscience.wiley.com).

822 BioEssays 28.8 BioEssays 28:822–833, � 2006 Wiley Periodicals, Inc.

Abbreviations: 30UTR, 30 untranslated region; AChE, acetylcholines-

terase; ARE, AU-rich element; CARM1, co-activator associated

methyltransferase 1; CPEB, cytoplasmic polyadenylation element

binding; DRG, dorsal root ganglion; ELAV, embryonic lethal abnor-

mal-vision; FMRP, Fragile-X mental retardation protein; hnRNP A1 or

K, heteronuclear ribonucleoprotein A1 or K; IMP1, insulin-like growth

factor mRNA binding protein 1; KSRP, K homology-type splicing

regulatory protein; MARCKS, myristoylated alanine-rich C kinase

substrate; PABP, poly(A)-binding protein; RBP, RNA-binding protein;

RNP, ribonucleoprotein; RRM, RNA-recognition motif; SCG, superior

cervical ganglion; SNP, single nucleotide polymorphism; ZBP, zipcode-

binding protein.

Review articles

gaining importance. This is likely related to the fact that

amongst cell types, neurons present an architectural chal-

lenge because of their relatively small cell body size and

extensive network of projections and connections. Aswe learn

more about the basic mechanisms underlying neurogenesis,

neurite elongation, synapse formation and plasticity, we find

that RNA-binding proteins are not only involved in all these

events but that they are in fact essential and, hence, act as

key components, to ensure their appropriate unfolding and

completion. Consequently, it is not surprising to find direct

associations between neurodegenerative diseases and con-

ditions such as spinal muscular atrophy, amyotrophic lateral

sclerosis and Fragile X syndrome, and perturbation in RNA

regulation and/or RBP activity (see for review Refs 3,5,6,8,9).

There are a number of well-characterized RBP that have

been shown to assume specific roles in normal neuronal

development and function (see Table 1).(10–13) Among

these RBP, the cytoplasmic polyadenylation element binding

(CPEB) protein, zipcode-binding protein (ZBP), Fragile-Xmental

retardation protein (FMRP) and Staufen are important for

mRNA transport along dendrites or axons.(14–19) In addition to

transporting specific target transcripts, CPEB, ZBPandFMRP

all have translational regulatory roles such that translation is

inhibited during transport and activated at the final destination

by specific signals.(20–24) Besides regulating transport and

translation, RBP are known to control mRNA degradation.

To date, the majority of these, including specifically AUF1

(seeTable 1), havebeen shown to stimulate or facilitatemRNA

decay.(25,26) In comparison, theHu family of RBP, ofwhichHuD

is the focus of this review, are among the few known proteins

that stabilize transcripts in the cytoplasm.

Over the last 15 years, there has therefore been grow-

ing interest in the function and regulation of the Hu family of

RBP, which is directly involved in development of paraneo-

plastic encephalomyelitis and sensory neuronopathy syn-

dromes.(31,33,36) These syndromes result from the expression

of neuronal proteins by tumor cells in the body, typically small-

cell lung cancer tumors. Ectopic expression of these ordinarily

immune-privileged proteins results in production of onconeur-

al antibodies that attack the nervous system and lead to the

development of autoimmune neurologic diseases. The anti-

body produced in the above disorder was termed anti-Hu

based on the name of the patient in which the antibody was

discovered.(31,37) The Hu antibody was used to identify HuD

and other Hu proteins (see further).(33)

Amongst the different family members, HuD is recognized

as oneof the earliestmarkers of neuronal cells aswell as being

an essential regulator of neuronal differentiation and survi-

val.(8,27,38–43) Of particular interest and of potential clinical

relevance is the recent association between polymorphisms

in the HuD gene and age-at-onset of Parkinson’s disease.(44)

A total of nine single nucleotide polymorphisms (SNP) within

the human HuD locus region were genotyped in this study,

of which two, one in intron 2 and one in exon 8, showed

Table 1. Neuronal RNA-binding proteins

RBP Domains Function and recognized element

Hu (HuB, HuC and HuD) . 3 RRM . mRNA stabilization

. Cell-cycle regulation and neurite extension

. ARE (AUUUA, see text)(8,27–31)

AUF1 (p37, p40, p42, p45) . 2 RRM . mRNA destabilization

. ARE(25,26,32)

Cytoplasmic polyadenylation element binding (CPEB) protein . 2 RRM . Polyadenylation and translation activation (derepression)

. mRNA dendritic transport

. Synaptic plasticity

. CPE (UUUUUAU)(18,20,23)

Musashi (Msi1 and 2) . 2 RRM . Translation repression

. Neuronal development(10,12)

Zipcode-binding protein (ZBP1 and 2) . 1 RRM . mRNA transport

. 4 KH . Translation repression

. Growth-cone dynamics

. Zipcode(14,17,19,21,24)

Human K homology-type splicing regulatory protein (KSRP) or rat . 4 KH . mRNA dendritic transport

MARTA-1 . mRNA degradation(15,16,33)

NOVA-1 and NOVA-2 . 3 KH . Splicing regulation

. Neuronal survival

. UCA(U/C) stem-loop repeats(8,11,31)

Fragile X mental retardation protein (FMRP) . 2 KH . Translation repression and mRNA transport

. 1 RGG . Synaptic structure and plasticity

. G quartet structure(8,34,35)

Staufen (Stau1 and 2) . 5 dsRBD . Dendritic mRNA targeting

. Double-stranded RNA(13)

Review articles

BioEssays 28.8 823

moderately to strongly significant effects on age-at-onset of

Parkinson’s disease. Here, we therefore present a compre-

hensive and updated view of HuD and its role in multiple

aspects of a neuron’s life including the initial phenotypic

commitment and differentiation, as well as synaptic remodel-

ing during learning paradigms and in response to injury and

regeneration. Additionally, we describe the current state of

knowledge concerning the molecular and cellular events

regulating the expression, activity and function of HuD in

neurons.

The HuD Gene, its transcripts and

encoded proteins

As mentioned, HuD was the first of the Hu proteins to be

identified as theanti-Huantigen in patientswith paraneoplastic

encephalomyelitis and sensory neuronopathy.(31,33,36) The Hu

protein family also includes HuB (Hel-N1), HuC (Hel-N3) and

HuR (HuA).(33) Whereas HuR is ubiquitously expressed

and HuB is found in neurons and gonads, expression of HuC

and HuD is restricted to neurons. Due to the presence of three

highly conserved RNA-recognition motifs (RRM) (see below

and for review 45,46), HuD shares a high degree of homology

with the Drosophila proteins ELAV (responsible for the

Embryonic Lethal Abnormal Vision phenotype; see Fig. 1)

that is required for the normal development and maintenance

of the nervous system.(33,47)

HuD is encoded by a relatively large gene (�146 Kb)

located on chromosome1 in humans (1p34) and chromosome

4 in mice (49.5 cM). As shown in Fig. 2, some of the exons are

separatedby large intronic regions. The localizationof theHuD

gene in the mouse genome was based on syntenic regions

between human and mouse, suggesting that the flanking

genes are conserved between species.(48) The gene consists

of three putative non-coding exon 1 variants, termed 1a, 1b

and1c, andseveral commoncodingexons (2, 3, 4, 5and8; see

Fig. 2).(30,48) The first and second RRM are encoded by exons

2 and 3, and exons 4 and 5, respectively, while the third RRM is

encodedbyexon 8. TheHuDgene is also subject to alternative

splicing of exons 6 and 7 which affects the length of the

hinge region linking the second and third RRM. This results in

the three different transcripts and molecular forms, termed

HuDpro, HuD and HuDmex(30) of which HuDpro and HuD are the

major variants.(47–50) The resulting proteins have molecular

masses ranging between37and43 kDaandare characterized

by the three RRMs and unique nuclear export signals located

within the hinge region.(40)With theexceptionof theN-terminal

domain and the hinge region, the protein sequences between

the different Hu proteins are very similar (70 to 85% identity;

see Fig. 1).(47)

HuD’s RNA-recognition motifs (RRM)

and target transcripts

As shown in Figs 1 and 2, all Hu proteins contain three RRM.

RRMs are found in various proteins involved in RNA proces-

sing and turnover, including hnRNPA1 involved in splicing and

transport of RNA, poly(A)-binding protein (PABP) involved in

transcript stability and translation, and the Drosophila ELAV

protein involved in RNA splicing (see for review 45,46). The

Figure 1. The murine Hu family of ARE-

binding proteins shares a high degree of

homology with the Drosophila protein ELAV.

HuD, HuB, HuC, HuR and ELAV protein

sequences are aligned demonstrating the high

degree of homology especially within the RRM

(highlighted in yellow). RNPmotifs that bind the

RNA are indicated by underlined blue letters.

The exons encoding each protein region are

indicated above the sequence alignment and

the splicing between the exons is indicated by

the slanted bar. The underlined and red residue

(R236) corresponds to the methylated arginine

in HuD and HuR.

Review articles

824 BioEssays 28.8

RRM is a conserved structure of�80–90 residues containing

two consensus ribonucleoprotein (RNP) motifs separated by

25–35 amino acids that interact directly with the RNA. The

RNP motifs, octameric RNP-1 and hexameric RNP-2, each

contain three conserved aromatic residues that are implicated

in RNA interactions in a number of different RBP including

proteins that bind pre-mRNA, mRNA, pre-rRNA as well as

small nuclear and heteronuclear RNAs. The consensus

structure of RRM consists of b1-a1-b2-b3-a2-b4 and the

location of the RNPmotifs in the first and third b-strands of theRRM is highly conserved.(45,51) Although the structure of RRM

iswell preserved, only a few residues foundmostly in the RNP

motifs are highly conserved. Consequently, it is the variable

regions between the RNP motifs and RRM that tend to lend

sequence specificity to the RBP. Similarly to other RRM-

containing RBP, the first and second HuD RRM consist of the

consensus RRM structure and form a cleft between them

where the RNA is bound specifically between the b-sheets.(52)

Crystal structureexperiments revealed that the residueswithin

the first and second HuD RRM that interact with the RNA, are

conserved between HuD and ELAV proteins.(52)

Hu proteins recognize and bind specifically to well-

described AU-rich elements (ARE) found within the 30UTR of

approximately 1 in 20 human genes(53,54) and directly

implicated in RNA turnover.(30,53,55,56) The AREs are sepa-

rated into three classes based on their sequence and

structure. Class I AREs consist of one to three dispersed

AUUUAmotifs separated byU-rich regionswhile class II AREs

consist of multiple clusters of AUUUA motifs. Class III AREs,

although less-well defined, consist mainly of U-rich sequences

and do not contain the AUUUA pentamer.(57) The different

classes of AREs appear to instruct distinct modes of mRNA

decay. Class I and III ARE-directed mRNA degradation

involves simultaneous deadenylation of mRNAs resulting in

a pool of transcripts of homogeneous lengths, termed

distributive deadenylation, which is followed by degradation

of the mRNA body. In contrast, Class II ARE-directed mRNA

degradation consists of complete deadenylation of one

transcript resulting in a pool of mRNAs of heterogeneous

lengths, termed processive deadenylation.(58) Recently, HuD-

RNA crystal structure and in vitro binding assays have

determined that the consensus sequence recognized by

RRM1 and RRM2 is X-U/C-U-X-X-U/C?-U-U/C, where the

‘‘?’’ denotes questionable binding of theC in this location.(52,59)

Consequently, HuD has been demonstrated to bind and

stabilize transcripts with CU- and U-rich sequences as well

as class II and III AU-rich elements. The structure of the RRM

allows for the binding of any number of domains and RNA

conformations.(52) Similarly to HuR, HuD, while able to bind all

classes of ARE, retains some sequence specificity and

preference such that it will not bind a transcript based solely

on the presence of an ARE.(60)

Unlikemany other ARE-binding proteins that principally act

to destabilize transcripts, such as AUF1 family members

(Table 1), Hu proteins act to stabilize ARE-containing

transcripts thereby significantly prolonging their half-

life.(39,42,61,62) In vitro binding studies performed with HuD

have demonstrated that the RNA is bound primarily by the first

RRM while the second RRM, which also binds the RNA,

functions mostly to stabilize the RNA–protein complex.(52,63)

Figure 2. HuD gene, transcript and protein organization. A: HuD exon and intron organization is depicted. Green boxes correspond to

three putative alternative exon1 variants, blue boxes correspond to coding region exons and red boxes correspond to alternatively spliced

coding region exons. Note the large intronic regions separating exons 1 through 4. B: Alternative splicing of exons 6 and 7 results in the

production of three alternative transcripts (HuDpro, HuD and HuDmex) which have untranslated regions (UTR and exon 1 shown in green) of

varying and unknown lengths. The promoter region driving gene transcription and the predominant exon 1 variant alongwith the resulting 50

untranslated region, remain unknown.C:Protein organization of the differentHuD transcripts. Thevarying length of the hinge region linking

the second and third RRM results from alternative splicing of exons 6 and 7.

Review articles

BioEssays 28.8 825

The third RRM, in addition to participating in maintaining

RNA–protein complex stability, binds to poly(A) tails.(64–67) In

the latter case, the length of the poly(A) tail correlates with the

overall binding efficiency of Hu proteins.(64) In contrast, the

third RRM of mouse HuB and HuC could not bind to poly(A)

ribohomopolymers and generally showed very little RNA-

binding activity.(68) There are, however, some inconsistencies

between studies as another study performed with mouse HuC

demonstrated that the third RRM could specifically bind to

poly(A)-sepharose beads.(66) The different results obtained

with each Hu family member may simply reflect different

technical approaches. In that context, the studies performed

with HuD used complete in vitro transcribed GAP-43 tran-

scripts with differing lengths of poly(A) tails as opposed to

poly(A) nucleotide chains, and could consequently be con-

sidered a more accurate representation of the binding activity

of the third RRM.(64) Alternatively, it is possible that the third

RRM is also involved in protein–protein interactions rather

than direct RNA-binding, as suggested by Kasashima et al.

(2002), who showed that HuD, HuB and HuC could form

dimers via the third RRM.

As the variety of cis-acting elements recognized by the

RRM have increased, so have the number of transcripts that

interact with HuD. Accordingly, HuD has been shown to bind

and stabilize several developmentally regulated transcripts

including c-fos, c-myc, N-myc, p21waf1, neuroserpin and

MARCKS.(69–73) Interestingly, and in agreement with its role

in neuronal differentiation, many of the transcripts bound by

HuD play a key role in the formation of neuronal processes

such as GAP-43 and tau.(39,62,74) In this context, our studies

have recently shown that HuD also binds and stabilizes

acetylcholinesterase (AChE) transcripts during neuronal

differentiation.(42) Since in addition to its well-recognized

function in neurotransmission, AChE can also stimulate

neurite outgrowth (Refs 42,75,76 and references therein), it

appears therefore that HuD binds and regulates the stability of

a subset of transcripts with similar functional roles that is key

for neuronal development and plasticity.

Although the principal function of HuD is to stabilize its

target transcripts and increase their half-life, several studies

performed with HuD or other members of the Hu family

suggest that HuD is also involved in other aspects of mRNA

regulation (see Figure 3). The variable linker domain sepa-

rating the second and third RRMs has been suggested

to contain a novel nuclear export signal that allows HuD

to shuttle target transcripts out of the nucleus into the

cytoplasm.(29,40,77) In addition, the first and second RRM and

part of the linker domain have been shown to interact with the

primary mRNA export receptor TAP/NXF1 in cultured neuro-

nal cells.(78) In fact, previous studies have shown that neurite

elongation is severely impeded when shuttling of HuD is

blocked, suggesting that HuD plays an important role in

localizing its target transcripts to the cytoplasm.(40) Once in the

cytoplasm, HuD and its cargo are targeted to axons, in the

case of tau mRNA, and growth cones, in the case of GAP-43

mRNA.(79,80)

Specific subcellular localization of the HuD-mRNA gran-

ules appears to involve the KIF3A microtubule-associated

motor protein.(79) Along these lines, HuDwas shown to bind in

an RNA-dependent manner to insulin-like growth factor

mRNA-binding protein 1 (IMP1).(40,81) IMP1 is the mouse

ortholog of chicken ZBP1 which is known to have a role in

mRNA transport and translation regulation (see Table 1).(82)

Furthermore, HuD has also been shown to colocalize with

Figure 3. HuD function and regulation within

neurons. A: HuD gene transcription within

neurons may be controlled by neuron-specific

transcription factors. HuD transcript may also

be bound by itself. B: HuD binds its target

transcript within the cytoplasm and possibly

within the nucleus prior to nuclear export. Post-

translational modification of HuD, such as

methylation, inhibits HuD–RNA interactions.

C: HuD competes for target transcripts with

otherRBP that are predominantly destabilizing.

The abundance of the different RBP deter-

mines the eventual fate of transcripts. D: HuDstabilizes target transcripts and protects them

from decay. E: HuD binds to its target tran-

scripts in the axon hillock, along the axonand in

the growth cone. HuD transport may involve

molecular motors and interactions with other

RNA-transport proteins (see Table 1).

Review articles

826 BioEssays 28.8

ribosomes in dendrites of pyramidal neurons of the CA1 and

CA3 regions of the hippocampus.(83) Finally, HuD-bound

mRNAs are also targeted to ribosomes and polysomes,

similarly to HuB and neurofilament-M mRNA.(80,81,84) These

studies suggest therefore that HuD could also be involved,

either directly or indirectly through binding with other RBP

with translational functions, in stimulating translation of its

target transcripts. In this context, a recent study has shown

that HuB can interact with hnRNP K and both can bind to

p21 mRNA 30UTR.(85) This later work also demonstrated

that hnRNP K alone decreased the translation of this

transcript, whereas HuB could block this effect and indirectly

increase translation. Given the similarities in Hu protein

function and target transcripts, it is possible that HuD has

similar functions.

Functional role of HuD in neurons

The neuronal Hu proteins are known to be some of the earliest

markers of the neuronal phenotype since they appear to

suppress neuroblast proliferation and promote neuronal

differentiation.(47,68,86–88) In situ hybridization experiments

performed in developing and adult mouse and rat cerebral and

cerebellar cortex revealed a distinct expression pattern for

mRNAs encodingHuB, HuC andHuD.(47,89) During embryonic

development, HuD expression can be localized to cells exiting

the cell cycle in the ventricular zone, and to those migrating in

the intermediate zones and undergoing terminal differentia-

tion.(41,47,89) In the adult brain, HuD-positive neurons corre-

spond primarily to projection neurons found in the neocortex,

hippocampus, entorhinal cortex and cerebellum. Similarly,

HuD is very strongly expressed in the ventral motoneurons of

the spinal cord, the sensory neurons of the dorsal root ganglia

(DRG) and sympathetic neurons in their ganglia.(41,89) At the

subcellular level, HuD is detected in the cell body, axon and

growth cones.(47,79,80) Generally, HuB and HuD have a similar

pattern of expression which is somewhat different from HuC.

Although expression of HuB and HuD overlap, they appear to

have similar(68,85) and opposing(90,91) functions during neuro-

nal development depending on the context. For example, they

can both stimulate neuronal differentiation and neurite out-

growth but have opposing roles in progenitor cell self-renewal,

such that HuB positively and HuD negatively affects this

capacity.

Studies in which the expression level of HuD has been

manipulated byeither overexpression or downregulation, have

highlighted the significance of HuD in stabilizing various

mRNA in neurons. Specifically, HuD binds a subset of

transcripts that encode proteins involved in neuronal develop-

ment. Several recent studies have shown that overexpression

of HuD increases the rate and length of neurite outgrowth in

several different types of neuronal cells, including in both

primary cultures and neuronal cell lines. By contrast,

decreased HuD expression results in an inhibition of neurite

extension.(38,39,41,62) Together, these results indicate that HuD

is an important regulator of neurite elongation and morpholo-

gical differentiation.(8,92) Expression of HuD in non-neuronal

cells does not stimulate the development of processes,

suggesting that pluripotent cells must already be engaged

along a neuronal cell fate in order for HuD to be effective in

promoting morphological differentiation.(38,41) This effect is

specific to neuronal Hu proteins, as HuB and HuC can also

stimulatemorphological differentiation, while HuR is unable to

stimulate neurite extension in neural crest cells.(41,68,84)

Recently, HuD transgenic and HuD knockout mice have

beengeneratedand characterized.(91,93) HuD transgenicmice

specifically expressed higher total amounts of HuD in the

forebrain, particularly in all subregions of the hippocampus

including in dentate granule cells that do not normally express

HuD.(47,83,89) Interestingly, in both HuD-overexpressing and

-deficient mice, there are no apparent morphological or

structural defects in the adult brain. However, extension of

some cranial nerves is transiently impaired during embry-

ogenesis in HuD-deficient mice. In comparison, loss-of-

function alleles of Drosophila elav are embryonic-lethal and

hypomorphic mutations result in morphological and structural

defects of the eye.(94,95) Given that there are three neuronal Hu

proteins with similar functions and one ELAV, it is possible that

expression of the other members of the Hu family are

compensating for the loss of HuD in knockout animals.

Although there are no apparent morphological defects, HuD-

deficient mice display motor/sensory defects such as an

abnormal clasping reflex of the hindlimbs when suspended by

the tail.(91) The abnormal clasping reflex suggests a defect in

sensory and motor functions which is also confirmed by poor

performance in rotarod experiments.(91) Both of these mouse

models of aberrant HuD expression have initially been used to

confirm some of the in vitro and cell culture data and to further

elucidate the importance of HuD to neuronal development and

function.

During early developmental stages, HuD appears to be

involved at multiple levels of cell lineage commitment,

differentiation and survival. Studies using HuD knockout mice

showed that, during embryogenesis, the absence of HuD is

related to an increase in the progenitor cells’ ability to renew,

and to a decrease in their ability to exit the cell cycle and

morphologically differentiate into neurons.(91) Cells that are

not leaving the cell cycle are also more likely to undergo

apoptosis since levels of apoptosis are elevated in HuD-

deficient mice. In adult mice, the number of subventricular

zone progenitor cells is greater in the HuD knockout mice.

These studies suggest therefore, that HuD is important for

promoting the exit from the cell cycle, for negative regulation of

proliferation and stimulation of the differentiation process.(91)

In addition, an increased level of HuD in cultures of neural crest

cells precipitates neurotrophin dependence.(41,96) Similarly,

HuD overexpression in mouse embryonic stem cells can

Review articles

BioEssays 28.8 827

double the number of cells with long neurites and expressing

GAP-43 only when the cells are induced to differentiate with

retinoic acid.(38) The results obtained with HuD knockout mice

and from cultured neuronal cell lines, together with the

preferential expression of HuD in projection neurons, strongly

suggest that HuD through its stabilizing effect on several

transcripts, is essential for the proper development and

maintenance of neuronal extensions.

The significance of HuD in the regulated expression of

mRNAs and proteins involved in the growth and formation of

neuronal projections, suggests that it is also important to other

aspects of axonal and dendritic functions in the adult nervous

system.A fewdistinct andcomplementary studieshaveclearly

demonstrated that spatial learning tasks can enhance HuD

expression in hippocampal neurons resulting in a concomitant

increase in GAP-43 transcript levels.(97,98) Another experi-

mental paradigm, single trial contextual fear conditioning, also

demonstrated an association between HuD expression and

learning.(83) In the latter study, HuD expression was increased

in different regions of the hippocampus namely, in the hilar

region of the dentate gyrus, as well as in the CA3 and CA1

regions. Along similar lines, increased expression of HuD in

the hippocampus of transgenic mice overexpressing HuD,

stimulates GAP-43 mRNA expression and stabilization(93) as

well as AChE mRNA expression.(99) More specifically, over-

expression of HuD in dentate granule cells, where HuD is not

normally expressed, results in a marked increase in GAP-43

mRNA levels via post-transcriptional mRNA stabilization of

newly synthesized, but normally degraded, transcripts.(93)

Consequently, atypical expression of HuD in neurons may

have important regulatory effects on the expression of its

target transcripts and, thus, a significant downstreameffect on

neuronal function.

Given its significant role in morphological differentiation,

HuD has also been suggested to have a role in axonal

regeneration. Recent findings have shown that following nerve

crush of DRG sensory neurons and facial motoneurons, HuD

protein and transcript levels increasewithin 7 days of the injury

and remain elevated for up to 21 days.(100,101) This increase in

HuD expression is accompanied by a dramatic increase in the

mRNA levels of GAP-43, a known target of HuD. In

regenerating neurons of the DRG, HuD protein colocalized

with GAP-43 transcripts as well as with ribosomal RNA.(100) A

recent study has also demonstrated the co-localization of HuD

and GAP-43 mRNA in growth cones.(80) In this context, we

have found that exogenousexpressionof humanHuD in the rat

superior cervical ganglion (SCG) results in themaintenance of

AChE and GAP-43 mRNA levels following axotomy (unpub-

lished data). Together, these findings demonstrate that,

in addition to its role during neuronal development, HuD

expression and function correlate with plasticity of the nervous

system both in response to injury and during learning. Further

studies performed with the HuD-transgenic and -deficient

mice are necessary to strengthen these correlations into direct

or indirect involvement of HuD.

Molecular events regulating expression

of HuD and its function

Despite the well-established involvement of HuD in the control

of a subset of neuronal transcripts involved in neuronal

differentiation and plasticity, there appears to be a lack of

information concerning thenatureof themolecular andcellular

mechanisms that regulate expression of HuD in neurons. This

is important since a number of recent studies point to mRNA

stability as a consequence of functional antagonism between

stabilizing and destabilizing proteins (see for reviewRef. 102).

This is best exemplified by studies performed with the

ubiquitous Hu family member HuR, whose tissue expression

overlaps with AUF1, a family of four isoforms that are

predominantly destabilizing (Table 1).(32,103) In addition, many

transcripts that havebeen identifiedas targets forHuRarealso

AUF1 targets, such as p21, c-fos, TNF-alpha and cyclin

D1.(102,104) The antagonistic effects of these proteins have

been demonstrated using siRNA to individually decrease the

expression of HuR or AUF1, resulting in increased association

of the target transcript with the opposite protein and

subsequent decreased or increased stability, respectively.(104)

Using a similar approach, competition for transcript-binding

siteswas also demonstratedwithin the AUF1 family.(105) In this

context, it is important to note that AUF1 has also been

reported to have mRNA-stabilizing activity.(106,107) Thus,

it appears that competition for target transcripts and the

eventual outcome for their longevity are largely determined by

the relative abundance of the different RBP.(102,104) Accord-

ingly, it becomes important to gain a better understanding of

the events presiding over HuD expression in neurons.

Transcriptional and post-transcriptionalregulatory mechanisms.The HuD gene is divided into eight coding exons beginning

with exon 2 (see Figs 1 and 2).(48) A comparison of the

available cDNA sequences in GenBank (NM_010488,

BC052451 and BC052451) shows the presence of three

alternate non-coding exon1 splice variants termed exon1a, 1b

and 1c (see Fig. 2).(48) Presently, it is unclear whether a single

promoter is responsible for transcription of this large gene

with splicing of exon1 variants occurring subsequently or

whether there are multiple promoter choices driving specific

expression of exon 1 variants. Although the HuD 50 regulatory

region of the mammalian gene has yet to be isolated and

characterized, studies performed with the Xenopus laevis

HuD homologue, elrD, have shown that there are two possible

promoter regions upstream of exon 2.(108) The most 50

promoter region is upstream of a non-coding exon 1 and the

second promoter region is located within the first intron. Both

Review articles

828 BioEssays 28.8

promoter regions have been shown to drive transcription in a

neuronal-dependent fashion and to contain neuron-specific

transcription factor binding sites (E-box and neuron-restrictive

silencer factor site). Similarly, characterization of the human

HuB 50 regulatory region demonstrates the presence of cis-

acting elements that can direct tissue-specific expression in

neurons.(109) Analysis of the zebrafish HuC 50 regulatory

region resulted in the identification of multiple E-box se-

quences, a putative MyT1-binding site and two GC boxes that

could also drive neuron-specific expression of HuC.(110,111)

The Drosophila ELAV 50 flanking region including enhancer

domains within an intronic region, also confers neural-specific

expression patterns to this gene.(112) Accordingly, the specific

and early neuronal expression of HuD alongwith the presence

of alternate 50 regulatory regions strongly suggest that

expression of HuD is regulated by transcriptional mechanisms

involving selective cis-acting elements and trans-acting

factors that are specific to neurons (seeRef. 87). In agreement

with this view, a recent study demonstrated that the transcrip-

tional rate of HuD is significantly altered in neurons subjected

to changes in the levels of thyroid hormone.(113)

In addition to transcriptional regulatory events, the relative

abundance of HuD may also be regulated by post-transcrip-

tional mechanisms (see Figure 3). Northern blot analysis has

revealed the presence of two HuD transcripts of �3.7 and

4.4 kb in neurons.(114) These are thought to originate from

alternative polyadenylation or splicing to a putative down-

stream non-coding exon (exon 9).(48) Alignment of available

cDNA sequences indeed reveals the presence of alternate

30UTRs. Amongst the different Hu proteins, the 30UTRs

are unique to each transcript.(47) In contrast, specific se-

quences coding for stem-loop structures and putative ARE

within the HuD 30UTR are highly conserved between different

species.(28) In addition, the 30UTR of the Drosophila homo-

logues (ELAV and SxL) are important for the appropriate

regulation of these transcripts.(115,116) More specifically, HuB,

ELAVand SxL have been shown to autoregulate their expres-

sion through interactions with their own 30UTR.(116–118)

Recent studies have shown that ELAV expression is depen-

dent on the presence of a long UTR at its 30 end.(115) An

ELAV-binding site was in fact found within the elav mRNA

located within an alternative non-coding 30exon.(118) The

authors of this study proposed that ELAV protein binds to

elav mRNA 30UTR when protein levels reach a certain

threshold and, in this case, block further protein production.

Such a similar negative autoregulatory loop also can be

assigned to SxL protein, which can downregulate its own

expression by binding to specific regions within the

sxl 30UTR.(116) Together, these observations are coherent

with the notion that, in addition to transcriptional events

(see paragraph above), post-transcriptional mechanisms

operating through the 30UTR are also important for control-

ling HuD expression.

Post-translational regulatory mechanismsAlthough the abundance of RBP is important to ultimately

control the fate of specific transcripts, other factors such as the

RBPaffinity and function are also implicated. In a recent study,

wehaveshown thatHuDdidnot actively bind tooneof its target

transcripts until neuronal differentiation was stimulated, even

though significant levels of HuD protein were present in

undifferentiated PC12 cells and these levels did not change

upon differentiation (see Refs 42,49,62). This suggests that

additional factors affect the ability of HuD to stabilize target

transcripts. In this context, several recent studies have begun

to elucidate the pathways regulating post-translational mod-

ification of the Hu family of RNA-binding proteins including

phosphorylation andmethylation, and to determine the impact

of these modifications on their activity.(54,62,119) Notably, NGF-

induced differentiation and stress-related stimuli can indirectly

regulate HuD and HuR function, respectively, by a PKC-

mediated signaling cascade.(62,120) Recently, a series of

indirect experiments led to the suggestion that threonine

phosphorylation of the neuronal Hu proteins (HuB, HuC and

HuD) by PKCa isozyme results in the re-distribution of the

proteins and in an increased stabilization of GAP-43 mRNA,

a well-known HuD target.(54)

In addition to phosphorylation, a recent study has shown

that arginine residues in the HuR hinge region are methylated

by the methyltransferase CARM1 (coactivator-associated

arginine methyltransferase 1) in response to lipopolysacchar-

ide.(119) Arginine methylation is another common modification

of some RBP that results in modification in the pattern of

protein–protein interactions. Along those lines, it has also

recently been demonstrated in PC12 cells that HuD is

methylated on the corresponding arginine (Arg236, see Fig. 1)

by the same methyltransferase, and that methylation results

in decreased RNA-binding activity of HuD.(121) In these

studies, methylation-resistant mutant HuD increased expres-

sion of some target transcripts such as p21cip1/waf1, and

resulted in increased neuronal differentiation of PC12 cells.

Although these cell culture studies suggest that phosphoryla-

tion and methylation can regulate HuD function directly or

indirectly, it should be noted that these studies have been

performed in vitro. Whether post-translational modifications

are also occurring in vivo still remains to be determined.

Nonetheless, these studies indicate that post-translational

modifications are also likely to be important in regulating the

binding and functional activity of HuD on target transcripts.

Finally, the fate of target transcripts may also depend on

other factors that might modulate the binding activity and

specificity of HuD. These include the structure and sequence

of the target transcripts and the interaction with additional

RBPs. The transcript itself can, in many cases, modify the

function of RBP (see for review Refs 52,122). An example of

this was given above with the poly(A) tail since the presence

and length of the poly(A) tail can alter the binding of theRRM to

Review articles

BioEssays 28.8 829

the ARE.(64,67) The presence of HuD in RNP granules and the

suggested multimerization of HuD implies that interactions

with other proteins or RBP can modulate the activity of HuD

and its specificity in a manner analogous to the interactions of

transcription factors on promoter elements.(47,79,85,123) This

may explain in part the identification of numerous RBPs

interacting with a single transcript such asGAP-43, AChE and

the early-response genes c-myc and c-fos (see for review

102).(42,61,62,124)

Conclusion and perspective

To date, there is compelling evidence indicating that, through

its ability to bind and stabilize a specific subset of transcripts,

HuD plays an essential role in the development and

maintenance of neuronal phenotype. The downstream effects

of this specific activity include the stimulation of morphological

differentiation of newly committed and developing neurons.

Additionally, HuDexpression andbindingactivity have recently

been shown to increase during nerve regeneration as well as

with learning andmemory. The ability of HuD to directly control

these key molecular and cellular events indicates that HuD

may act as an important regulator of neuronal differentiation

and function. Therefore, a thorough understanding of the

mechanisms affecting the expression, activity and function of

HuD in neurons appears warranted in order to increase our

basic knowledge of the events regulating neuronal differentia-

tion, maintenance and plasticity. Ultimately, this knowledge

could prove useful for the development of novel therapeutic

strategies aimed at manipulating, pharmacologically or

through gene therapy approaches, the levels of RBP that

may be beneficial for the treatment of several neurological

diseases and conditions.

Acknowledgments

We thank members of the Jasmin laboratory for fruitful

discussions.

References1. Bashirullah A, Cooperstock RL, Lipshitz HD. 1998. RNA localization in

development. Annu Rev Biochem 67:335–394.

2. King ML, Zhou Y, Bubunenko M. 1999. Polarizing genetic information

in the egg: RNA localization in the frog oocyte. Bioessays 21:546–

557.

3. Hollams EM, Giles KM, Thomson AM, Leedman PJ. 2002. MRNA

stability and the control of gene expression: implications for human

disease. Neurochem Res 27:957–980.

4. Naveh-Many T, Bell O, Silver J, Kilav R. 2002. Cis and trans acting

factors in the regulation of parathyroid hormone (PTH) mRNA stability

by calcium and phosphate. FEBS Lett 529:60–64.

5. Conne B, Stutz A, Vassalli JD. 2000. The 30 untranslated region of

messenger RNA: a molecular ‘hotspot’ for pathology? Nat Med 6:637–

641.

6. Mendell JT, Dietz HC. 2001. When the message goes awry: disease-

producing mutations that influence mRNA content and performance.

Cell 107:411–414.

7. Misquitta CM, Iyer VR, Werstiuk ES, Grover AK. 2001. The role of 30-

untranslated region (30-UTR) mediated mRNA stability in cardiovascular

pathophysiology. Mol Cell Biochem 224:53–67.

8. Perrone-Bizzozero N, Bolognani F. 2002. Role of HuD and other RNA-

binding proteins in neural development and plasticity. J Neurosci Res

68:121–126.

9. Gallo JM, Jin P, Thornton CA, Lin H, Robertson J, et al. 2005. The role of

RNA and RNA processing in neurodegeneration. J Neurosci 25:10372–

10375.

10. Imai T, Tokunaga A, Yoshida T, Hashimoto M, Mikoshiba K, et al. 2001.

The neural RNA-binding protein Musashi1 translationally regulates

mammalian numb gene expression by interacting with its mRNA. Mol

Cell Biol 21:3888–3900.

11. Jensen KB, Dredge BK, Stefani G, Zhong R, Buckanovich RJ, et al.

2000. Nova-1 regulates neuron-specific alternative splicing and is

essential for neuronal viability. Neuron 25:359–371.

12. Okano H, Imai T, Okabe M. 2002. Musashi: a translational regulator of

cell fate. J Cell Sci 115:1355–1359.

13. Roegiers F, Jan YN. 2000. Staufen: a common component of mRNA

transport in oocytes and neurons? Trends Cell Biol 10:220–224.

14. Bassell GJ, Oleynikov Y, Singer RH. 1999. The travels of mRNAs

through all cells large and small. FASEB J 13:447–454.

15. Rehbein M, Wege K, Buck F, Schweizer M, Richter D, et al. 2002.

Molecular characterization of MARTA1, a protein interacting with the

dendritic targeting element of MAP2 mRNAs. J Neurochem 82:1039–

1046.

16. Gherzi R, Lee KY, Briata P, Wegmuller D, Moroni C, et al. 2004. A KH

domain RNA binding protein, KSRP, promotes ARE-directed mRNA

turnover by recruiting the degradation machinery. Mol Cell 14:571–583.

17. Farina KL, Singer RH. 2002. The nuclear connection in RNA transport

and localization. Trends Cell Biol 12:466–472.

18. Huang YS, Carson JH, Barbarese E, Richter JD. 2003. Facilitation of

dendritic mRNA transport by CPEB. Genes Dev 17:638–653.

19. Zhang HL, Eom T, Oleynikov Y, Shenoy SM, Liebelt DA, et al. 2001.

Neurotrophin-induced transport of a beta-actin mRNP complex in-

creases beta-actin levels and stimulates growth cone motility. Neuron

31:261–275.

20. Mendez R, Richter JD. 2001. Translational control by CPEB: a means to

the end. Nat Rev Mol Cell Biol 2:521–529.

21. Ross AF, Oleynikov Y, Kislauskis EH, Taneja KL, Singer RH. 1997.

Characterization of a beta-actin mRNA zipcode-binding protein. Mol

Cell Biol 17:2158–2165.

22. Si K, Giustetto M, Etkin A, Hsu R, Janisiewicz AM, et al. 2003. A

neuronal isoform of CPEB regulates local protein synthesis and

stabilizes synapse-specific long-term facilitation in aplysia. Cell 115:

893–904.

23. Wu L, Wells D, Tay J, Mendis D, Abbott MA, et al. 1998. CPEB-mediated

cytoplasmic polyadenylation and the regulation of experience-dependent

translation of alpha-CaMKII mRNA at synapses. Neuron 21:1129–1139.

24. Huttelmaier S, Zenklusen D, Lederer M, Dictenberg J, Lorenz M, et al.

2005. Spatial regulation of beta-actin translation by Src-dependent

phosphorylation of ZBP1. Nature 438:512–515.

25. Guhaniyogi J, Brewer G. 2001. Regulation of mRNA stability in

mammalian cells. Gene 265:11–23.

26. Wilson GM, Brewer G. 1999. The search for trans-acting factors

controlling messenger RNA decay. Prog Nucleic Acid Res Mol Biol

62:257–291.

27. Antic D, Keene JD. 1997. Embryonic lethal abnormal visual RNA-

binding proteins involved in growth, differentiation, and posttranscrip-

tional gene expression. Am J Hum Genet 61:273–278.

28. Good PJ. 1995. A conserved family of elav-like genes in vertebrates.

Proc Natl Acad Sci USA 92:4557–4561.

29. Keene JD. 1999. Why is Hu where? Shuttling of early-response-gene

messenger RNA subsets. Proc Natl Acad Sci USA 96:5–7.

30. Liu J, Dalmau J, Szabo A, Rosenfeld M, Huber J, et al. 1995.

Paraneoplastic encephalomyelitis antigens bind to the AU-rich elements

of mRNA. Neurology 45:544–550.

31. Musunuru K, Darnell RB. 2001. Paraneoplastic neurologic disease

antigens: RNA-binding proteins and signaling proteins in neuronal

degeneration. Annu Rev Neurosci 24:239–262.

32. Gouble A, Morello D. 2000. Synchronous and regulated expression of

two AU-binding proteins, AUF1 and HuR, throughout murine develop-

ment. Oncogene 19:5377–5384.

Review articles

830 BioEssays 28.8

33. Szabo A, Dalmau J, Manley G, Rosenfeld M, Wong E, et al. 1991. HuD,

a paraneoplastic encephalomyelitis antigen, contains RNA-binding

domains and is homologous to Elav and Sex-lethal. Cell 67:325–333.

34. Antar LN, Bassell GJ. 2003. Sunrise at the synapse: the FMRP mRNP

shaping the synaptic interface. Neuron 37:555–558.

35. Bardoni B, Mandel JL. 2002. Advances in understanding of fragile X

pathogenesis and FMRP function, and in identification of X linked

mental retardation genes. Curr Opin Genet Dev 12:284–293.

36. Dalmau J, Furneaux HM, Gralla RJ, Kris MG, Posner JB. 1990.

Detection of the anti-Hu antibody in the serum of patients with small

cell lung cancer—a quantitative western blot analysis. Ann Neurol 27:

544–552.

37. Graus F, Elkon KB, Cordon-Cardo C, Posner JB. 1986. Sensory

neuronopathy and small cell lung cancer. Antineuronal antibody that

also reacts with the tumor. Am J Med 80:45–52.

38. Anderson KD, Sengupta J, Morin M, Neve RL, Valenzuela CF, et al.

2001. Overexpression of HuD accelerates neurite outgrowth and

increases GAP-43 mRNA expression in cortical neurons and retinoic

acid-induced embryonic stem cells in vitro. Exp Neurol 168:250–258.

39. Aranda-Abreu GE, Behar L, Chung S, Furneaux H, Ginzburg I. 1999.

Embryonic lethal abnormal vision-like RNA-binding proteins regulate

neurite outgrowth and tau expression in PC12 cells. J Neurosci 19:

6907–6917.

40. Kasashima K, Terashima K, Yamamoto K, Sakashita E, Sakamoto H.

1999. Cytoplasmic localization is required for the mammalian ELAV-like

protein HuD to induce neuronal differentiation. Genes Cells 4:667–683.

41. Wakamatsu Y, Weston JA. 1997. Sequential expression and role of Hu

RNA-binding proteins during neurogenesis. Development 124:3449–3460.

42. Deschenes-Furry J, Belanger G, Perrone-Bizzozero N, Jasmin BJ. 2003.

Post-transcriptional regulation of Acetylcholinesterase mRNAs in nerve

growth factor-treated PC12 cells by the RNA-binding protein HuD. J Biol

Chem 278:5710–5717.

43. Figueroa A, Cuadrado A, Fan J, Atasoy U, Muscat GE, et al. 2003. Role

of HuR in skeletal myogenesis through coordinate regulation of muscle

differentiation genes. Mol Cell Biol 23:4991–5004.

44. Noureddine MA, Qin XJ, Oliveira SA, Skelly TJ, van der WJ, et al. 2005.

Association between the neuron-specific RNA-binding protein ELAVL4

and Parkinson disease. Hum Genet 117:27–33.

45. Burd CG, Dreyfuss G. 1994. Conserved structures and diversity of

functions of RNA-binding proteins. Science 265:615–621.

46. Kenan DJ, Query CC, Keene JD. 1991. RNA recognition: towards

identifying determinants of specificity. Trends Biochem Sci 16:214–220.

47. Okano HJ, Darnell RB. 1997. A hierarchy of Hu RNA binding proteins in

developing and adult neurons. J Neurosci 17:3024–3037.

48. Inman MV, Levy S, Mock BA, Owens GC. 1998. Gene organization and

chromosome location of the neural-specific RNA binding protein Elavl4.

Gene 208:139–145.

49. Steller U, Kohls S, Muller B, Soller R, Muller R, et al. 1996. The RNA

binding protein HuD: rat cDNA and analysis of the alternative spliced

mRNA in neuronal differentiating cell lines P19 and PC12. Brain Res Mol

Brain Res 35:285–296.

50. Tora M, Barbera VM, Real FX. 2000. Detection of HuD transcripts by

means of reverse transcriptase and polymerase chain reaction:

implications for the detection of minimal residual disease in patients

with small cell lung cancer. Cancer Lett 161:157–164.

51. Birney E, Kumar S, Krainer AR. 1993. Analysis of the RNA-recognition

motif and RS and RGG domains: conservation in metazoan pre-mRNA

splicing factors. Nucleic Acids Res 21:5803–5816.

52. Wang X, Tanaka Hall TM. 2001. Structural basis for recognition of AU-

rich element RNA by the HuD protein. Nat Struct Biol 8:141–145.

53. Bakheet T, Williams BR, Khabar KS. 2003. ARED 2.0: an update of AU-

rich element mRNA database. Nucleic Acids Res 31:421–423.

54. Pascale A, Amadio M, Scapagnini G, Lanni C, Racchi M, et al. 2005.

Neuronal ELAV proteins enhance mRNA stability by a PKCalpha-

dependent pathway. Proc Natl Acad Sci USA 102:12065–12070.

55. Chung S, Jiang L, Cheng S, Furneaux H. 1996. Purification and

properties of HuD, a neuronal RNA-binding protein. J Biol Chem

271:11518–11524.

56. Bakheet T, Williams BR, Khabar KS. 2006. ARED 3.0: the large and

diverse AU-rich transcriptome. Nucleic Acids Res 34:D111–D114.

57. Chen CY, Shyu AB. 1995. AU-rich elements: characterization and

importance in mRNA degradation. Trends Biochem Sci 20:465–470.

58. Xu N, Chen CY, Shyu AB. 1997. Modulation of the fate of cytoplasmic

mRNA by AU-rich elements: key sequence features controlling mRNA

deadenylation and decay. Mol Cell Biol 17:4611–4621.

59. Park-Lee S, Kim S, Laird-Offringa IA. 2003. Characterization of the

interaction between neuronal RNA-binding protein HuD and AU-rich

RNA. J Biol Chem 278:39801–39808.

60. Toba G, Qui J, Koushika SP, White K. 2002. Ectopic expression of

Drosophila ELAV and human HuD in Drosophila wing disc cells reveals

functional distinctions and similarities. J Cell Sci 115:2413–2421.

61. Deschenes-Furry J, Belanger G, Mwanjewe J, Lunde JA, Parks RJ, et al.

2005. The RNA-binding protein HuR binds to acetylcholinesterase

transcripts and regulates their expression in differentiating skeletal

muscle cells. J Biol Chem 280:25361–25368.

62. Mobarak CD, Anderson KD, Morin M, Beckel-Mitchener A, Rogers SL,

et al. 2000. The RNA-binding protein HuD is required for GAP-43 mRNA

stability, GAP-43 gene expression, and PKC-dependent neurite out-

growth in PC12 cells. Mol Biol Cell 11:3191–3203.

63. Park S, Myszka DG, Yu M, Littler SJ, Laird-Offringa IA. 2000. HuD RNA

Recognition Motifs Play Distinct Roles in the Formation of a Stable

Complex with AU-Rich RNA. Mol Cell Biol 20:4765–4772.

64. Beckel-Mitchener AC, Miera A, Keller R, Perrone-Bizzozero NI. 2002.

Poly(A) Tail Length-dependent Stabilization of GAP-43 mRNA by the

RNA-binding Protein HuD. J Biol Chem 277:27996–28002.

65. Anderson KD, Morin MA, Beckel-Mitchener A, Mobarak CD, Neve RL,

et al. 2000. Overexpression of HuD, but not of its truncated form HuD

IþII, promotes GAP-43 gene expression and neurite outgrowth in PC12

cells in the absence of nerve growth factor. J Neurochem 75:1103–

1114.

66. Abe R, Sakashita E, Yamamoto K, Sakamoto H. 1996. Two different RNA

binding activities for the AU-rich element and the poly(A) sequence of

the mouse neuronal protein mHuC. Nucleic Acids Res 24:4895–4901.

67. Ma WJ, Chung S, Furneaux H. 1997. The Elav-like proteins bind to AU-

rich elements and to the poly(A) tail of mRNA. Nucleic Acids Res

25:3564–3569.

68. Akamatsu W, Okano HJ, Osumi N, Inoue T, Nakamura S, et al. 1999.

Mammalian ELAV-like neuronal RNA-binding proteins HuB and HuC

promote neuronal development in both the central and the peripheral

nervous systems. Proc Natl Acad Sci USA 96:9885–9890.

69. Chagnovich D, Cohn SL. 1996. Binding of a 40-kDa protein to the N-

myc 30-untranslated region correlates with enhanced N-myc expression

in human neuroblastoma. J Biol Chem 271:33580–33586.

70. Cuadrado A, Navarro-Yubero C, Furneaux H, Kinter J, Sonderegger P,

et al. 2002. HuD binds to three AU-rich sequences in the 30-UTR of

neuroserpin mRNA and promotes the accumulation of neuroserpin

mRNA and protein. Nucleic Acids Res 30:2202–2211.

71. Joseph B, Orlian M, Furneaux H. 1998. p21(waf1) mRNA contains a

conserved element in its 30-untranslated region that is bound by the

Elav-like mRNA-stabilizing proteins. J Biol Chem 273:20511–20516.

72. Manohar CF, Short ML, Nguyen A, Nguyen NN, Chagnovich D, et al.

2002. HuD, a neuronal-specific RNA-binding protein, increases the in

vivo stability of MYCN RNA. J Biol Chem 277:1967–1973.

73. Wein G, Rossler M, Klug R, Herget T. 2003. The 30-UTR of the mRNA

coding for the major protein kinase C substrate MARCKS contains a

novel CU-rich element interacting with the mRNA stabilizing factors HuD

and HuR. Eur J Biochem 270:350–365.

74. Chung S, Eckrich M, Perrone-Bizzozero N, Kohn DT, Furneaux H. 1997.

The Elav-like proteins bind to a conserved regulatory element in the 30-

untranslated region of GAP-43 mRNA. J Biol Chem 272:6593–6598.

75. Layer PG, Willbold E. 1995. Novel functions of cholinesterases in

development, physiology and disease. Prog Histochem Cytochem 29:

1–94.

76. Soreq H, Seidman S. 2001. Acetylcholinesterase—new roles for an old

actor. Nat Rev Neurosci 2:294–302.

77. Fan XC, Steitz JA. 1998. HNS, a nuclear-cytoplasmic shuttling

sequence in HuR. Proc Natl Acad Sci USA 95:15293–15298.

78. Saito K, Fujiwara T, Katahira J, Inoue K, Sakamoto H. 2004. TAP/NXF1,

the primary mRNA export receptor, specifically interacts with a neuronal

RNA-binding protein HuD. Biochem Biophys Res Commun 321:291–297.

Review articles

BioEssays 28.8 831

79. Aronov S, Aranda G, Behar L, Ginzburg I. 2002. Visualization of

translated tau protein in the axons of neuronal P19 cells and

characterization of tau RNP granules. J Cell Sci 115:3817–3827.

80. Smith CL, Afroz R, Bassell GJ, Furneaux HM, Perrone-Bizzozero NI,

et al. 2004. GAP-43 mRNA in growth cones is associated with HuD and

ribosomes. J Neurobiol 61:222–235.

81. Atlas R, Behar L, Elliott E, Ginzburg I. 2004. The insulin-like growth

factor mRNA binding-protein IMP-1 and the Ras-regulatory protein

G3BP associate with tau mRNA and HuD protein in differentiated P19

neuronal cells. J Neurochem 89:613–626.

82. Nielsen J, Christiansen J, Lykke-Andersen J, Johnsen AH, Wewer UM,

et al. 1999. A family of insulin-like growth factor II mRNA-binding

proteins represses translation in late development. Mol Cell Biol 19:

1262–1270.

83. Bolognani F, Merhege MA, Twiss J, Perrone-Bizzozero NI. 2004.

Dendritic localization of the RNA-binding protein HuD in hippocampal

neurons: association with polysomes and upregulation during con-

textual learning. Neurosci Lett 371:152–157.

84. Antic D, Lu N, Keene JD. 1999. ELAV tumor antigen, Hel-N1,

increases translation of neurofilament M mRNA and induces

formation of neurites in human teratocarcinoma cells. Genes Dev

13:449–461.

85. Yano M, Okano HJ, Okano H. 2005. Involvement of Hu and

heterogeneous nuclear ribonucleoprotein K in neuronal differentiation

through p21 mRNA post-transcriptional regulation. J Biol Chem 280:

12690–12699.

86. Barami K, Iversen K, Furneaux H, Goldman SA. 1995. Hu protein

as an early marker of neuronal phenotypic differentiation by sub-

ependymal zone cells of the adult songbird forebrain. J Neurobiol 28:

82–101.

87. Marusich MF, Furneaux HM, Henion PD, Weston JA. 1994. Hu neuronal

proteins are expressed in proliferating neurogenic cells. J Neurobiol 25:

143–155.

88. Weickert CS, Webster MJ, Colvin SM, Herman MM, Hyde TM, et al.

2000. Localization of epidermal growth factor receptors and putative

neuroblasts in human subependymal zone. J Comp Neurol 423:359–372.

89. Clayton GH, Perez GM, Smith RL, Owens GC. 1998. Expression of

mRNA for the elav-like neural-specific RNA binding protein, HuD, during

nervous system development. Brain Res Dev Brain Res 109:271–280.

90. Okano H, Akamatsu W, Yano M, Mitsuhashi T, Fujihara H, et al. 2005.

RNA-Binding Protein Hu Family Regulates Neural Stem Cell Proliferation

and Differentiation. Abstract viewer/Itinerary Planner. Washington, DC:

Society for Neuroscience Program No. 598.13.

91. Akamatsu W, Fujihara H, Mitsuhashi T, Yano M, Shibata S, et al. 2005.

The RNA-binding protein HuD regulates neuronal cell identity and

maturation. Proc Natl Acad Sci USA 102:4625–4630.

92. Malter JA. 2001. Regulation of mRNA Stability in the Nervous System

and Beyond. J Neurosci Res 66:311–316.

93. Bolognani F, Tanner DC, Merhege M, Deschenes-Furry J, Jasmin B,

et al. 2006. In vivo post-transcriptional regulation of GAP-43 mRNA by

overexpression of the RNA-binding protein HuD. J Neurochem 96:790–

801.

94. Campos AR, Grossman D, White K. 1985. Mutant alleles at the locus

elav in Drosophila melanogaster lead to nervous system defects. A

developmental-genetic analysis. J Neurogenet 2:197–218.

95. Jimenez F, Campos-Ortega JA. 1987. Genes in subdivision 1B of the

Drosophila melanogaster X-chromosome and their influence on neural

development. J Neurogenet 4:179–200.

96. Dalmau J, Furneaux HM, Rosenblum MK, Graus F, Posner JB. 1991.

Detection of the anti-Hu antibody in specific regions of the nervous

system and tumor from patients with paraneoplastic encephalomyelitis/

sensory neuronopathy. Neurology 41:1757–1764.

97. Pascale A, Gusev PA, Amadio M, Dottorini T, Govoni S, et al. 2004.

Increase of the RNA-binding protein HuD and posttranscriptional up-

regulation of the GAP-43 gene during spatial memory. Proc Natl Acad

Sci USA 101:1217–1222.

98. Quattrone A, Pascale A, Nogues X, Zhao W, Gusev P, et al. 2001.

Posttranscriptional regulation of gene expression in learning by the

neuronal ELAV-like mRNA-stabilizing proteins. Proc Natl Acad Sci USA

98:11668–11673.

99. Deschenes-Furry JL, Mousavi K, Smith LE, Parks RJ, Perrone-Bizzonero

N, et al. 2003. Post-transcriptional regulation of acetylcholinesterase

mRNAs in akotomized neurons. Abstract viewer/Itinerary Planner.

Washington, DC: Society for Neuroscience Program No. 471.15.

100. Anderson KD, Merhege MA, Morin M, Bolognani F, Perrone-Bizzozero

NI. 2003. Increased expression and localization of the RNA-binding

protein HuD and GAP-43 mRNA to cytoplasmic granules in DRG

neurons during nerve regeneration. Exp Neurol 183:100–108.

101. Anderson KD, Steward O. 2003. In vivo Expression of the Neuronal-

specific RNA-binding Protein HuD in Reference to a Potential Target

mRNA (GAP-43) in the Mouse Facial Nucleus During Regeneration.

Abstract viewer/Itinerary Planner. Washington, DC: Society for Neu-

roscience Program No. 78.17.

102. Barreau C, Paillard L, Osborne HB. 2005. AU-rich elements and

associated factors: are there unifying principles? Nucleic Acids Res

33:7138–7150.

103. Lu JY, Schneider RJ. 2004. Tissue distribution of AU-rich mRNA-binding

proteins involved in regulation of mRNA decay. J Biol Chem 279:

12974–12979.

104. Lal A, Mazan-Mamczarz K, Kawai T, Yang X, Martindale JL, et al. 2004.

Concurrent versus individual binding of HuR and AUF1 to common

labile target mRNAs. EMBO J 23:3092–3102.

105. Raineri I, Wegmueller D, Gross B, Certa U, Moroni C. 2004. Roles of

AUF1 isoforms, HuR and BRF1 in ARE-dependent mRNA turnover

studied by RNA interference. Nucleic Acids Res 32:1279–1288.

106. Kiledjian M, DeMaria CT, Brewer G, Novick K. 1997. Identification of

AUF1 (heterogeneous nuclear ribonucleoprotein D) as a component of

the alpha-globin mRNA stability complex. Mol Cell Biol 17:4870–4876.

107. Sela-Brown A, Silver J, Brewer G, Naveh-Many T. 2000. Identification of

AUF1 as a parathyroid hormone mRNA 30-untranslated region-binding

protein that determines parathyroid hormone mRNA stability. J Biol

Chem 275:7424–7429.

108. Nassar F, Wegnez M. 2001. Characterization of two promoters of the

Xenopus laevis elrD gene. Biochem Biophys Res Commun 283:392–

398.

109. King PH. 1996. Cloning the 50 flanking region of neuron-specific Hel-N1:

evidence for positive regulatory elements governing cell-specific

transcription. Brain Res 723:141–147.

110. Park HC, Kim CH, Bae YK, Yeo SY, Kim SH, et al. 2000. Analysis of

upstream elements in the HuC promoter leads to the establishment of

transgenic zebrafish with fluorescent neurons. Dev Biol 227:279–293.

111. Zhao C, He X, Tian C, Meng A. 2006. Two GC-rich boxes in huC

promoter play distinct roles in controlling its neuronal specific

expression in zebrafish embryos. Biochem Biophys Res Commun

342:214–220.

112. Yao KM, White K. 1994. Neural specificity of elav expression: defining a

Drosophila promoter for directing expression to the nervous system.

J Neurochem 63:41–51.

113. Cuadrado A, Navarro-Yubero C, Furneaux H, Munoz A. 2003. Neuronal

HuD gene encoding a mRNA stability regulator is transcriptionally

repressed by thyroid hormone. J Neurochem 86:763–773.

114. Abe R, Uyeno Y, Yamamoto K, Sakamoto H. 1994. Tissue-specific

expression of the gene encoding a mouse RNA binding protein

homologous to human HuD antigen. DNA Res 1:175–180.

115. Samson ML. 1998. Evidence for 30 untranslated region-dependent

autoregulation of the Drosophila gene encoding the neuronal nuclear

RNA-binding protein ELAV. Genetics 150:723–733.

116. Yanowitz JL, Deshpande G, Calhoun G, Schedl PD. 1999. An N-terminal

truncation uncouples the sex-transforming and dosage compensation

functions of sex-lethal. Mol Cell Biol 19:3018–3028.

117. Abe R, Yamamoto K, Sakamoto H. 1996. Target specificity of neuronal

RNA-binding protein, Mel-N1: direct binding to the 30 untranslated

region of its own mRNA. Nucleic Acids Res 24:2011–2016.

118. Borgeson CD, Samson ML. 2005. Shared RNA-binding sites for

interacting members of the Drosophila ELAV family of neuronal proteins.

Nucleic Acids Res 33:6372–6383.

119. Li H, Park S, Kilburn B, Jelinek MA, Henschen-Edman A, et al. 2002.

Lipopolysaccharide-induced methylation of HuR, an mRNA-stabilizing

protein, by CARM1. Coactivator-associated arginine methyltransferase.

J Biol Chem 277:44623–44630.

Review articles

832 BioEssays 28.8

120. Wang W, Fan J, Yang X, Furer-Galban S, Lopez dS I, et al. 2002. AMP-

activated kinase regulates cytoplasmic HuR. Mol Cell Biol 22:3425–3436.

121. Fujiwara T, Mori Y, Chu DL, Koyama Y, Miyata S, et al. 2006. CARM1

Regulates Proliferation of PC12 Cells by Methylating HuD. Mol Cell Biol

26:2273–2285.

122. Williamson JR. 2000. Induced fit in RNA-protein recognition. Nat Struct

Biol 7:834–837.

123. Kasashima K, Sakashita E, Saito K, Sakamoto H. 2002. Complex

formation of the neuron-specific ELAV-like Hu RNA-binding proteins.

Nucleic Acids Res 30:4519–4526.

124. Irwin N, Chao S, Goritchenko L, Horiuchi A, Greengard P, et al.

2002. Nerve growth factor controls GAP-43 mRNA stability via the

phosphoprotein ARPP-19. Proc Natl Acad Sci USA 99:12427–

12431.

Review articles

BioEssays 28.8 833