+ All Categories
Home > Documents > Molecular Pathways of Frontotemporal Lobar Degeneration

Molecular Pathways of Frontotemporal Lobar Degeneration

Date post: 03-Oct-2016
Category:
Upload: christine
View: 217 times
Download: 0 times
Share this document with a friend
20
Molecular Pathways of Frontotemporal Lobar Degeneration Kristel Sleegers, 1,2,3 Marc Cruts, 1,2,3 and Christine Van Broeckhoven 1,2,3 1 Neurodegenerative Brain Diseases Group, Department of Molecular Genetics, VIB, 2 Laboratory of Neurogenetics, Institute Born-Bunge, and 3 University of Antwerp, Universiteitsplein 1, B-2610, Antwerpen, Belgium; email: [email protected], [email protected], [email protected] Annu. Rev. Neurosci. 2010. 33:71–88 First published online as a Review in Advance on March 22, 2010 The Annual Review of Neuroscience is online at neuro.annualreviews.org This article’s doi: 10.1146/annurev-neuro-060909-153144 Copyright c 2010 by Annual Reviews. All rights reserved 0147-006X/10/0721-0071$20.00 Key Words progranulin, TDP-43, neurodegeneration, etiological heterogeneity Abstract Frontotemporal lobar degeneration (FTLD) is a neurodegenerative condition that predominantly affects behavior, social awareness, and language. It is characterized by extensive heterogeneity at the clinical, pathological, and genetic levels. Recognition of these levels of hetero- geneity is important for proper disease management. The identification of progranulin and TDP-43 as key proteins in a significant proportion of FTLD patients has provided the impetus for a wealth of studies prob- ing their role in neurodegeneration. This review highlights the most recent developments and future directions in this field and puts them in perspective of the novel insights into the neurodegenerative process, which have been gained from related disorders, e.g., the role of FUS in amyotrophic lateral sclerosis. 71 Annu. Rev. Neurosci. 2010.33:71-88. Downloaded from www.annualreviews.org by University of Connecticut on 10/29/12. For personal use only.
Transcript
Page 1: Molecular Pathways of Frontotemporal Lobar Degeneration

NE33CH04-VanBroeckhoven ARI 14 May 2010 16:24

Molecular Pathwaysof FrontotemporalLobar DegenerationKristel Sleegers,1,2,3 Marc Cruts,1,2,3

and Christine Van Broeckhoven1,2,3

1Neurodegenerative Brain Diseases Group, Department of Molecular Genetics, VIB,2Laboratory of Neurogenetics, Institute Born-Bunge, and 3University of Antwerp,Universiteitsplein 1, B-2610, Antwerpen, Belgium; email: [email protected],[email protected], [email protected]

Annu. Rev. Neurosci. 2010. 33:71–88

First published online as a Review in Advance onMarch 22, 2010

The Annual Review of Neuroscience is online atneuro.annualreviews.org

This article’s doi:10.1146/annurev-neuro-060909-153144

Copyright c© 2010 by Annual Reviews.All rights reserved

0147-006X/10/0721-0071$20.00

Key Words

progranulin, TDP-43, neurodegeneration, etiological heterogeneity

Abstract

Frontotemporal lobar degeneration (FTLD) is a neurodegenerativecondition that predominantly affects behavior, social awareness, andlanguage. It is characterized by extensive heterogeneity at the clinical,pathological, and genetic levels. Recognition of these levels of hetero-geneity is important for proper disease management. The identificationof progranulin and TDP-43 as key proteins in a significant proportion ofFTLD patients has provided the impetus for a wealth of studies prob-ing their role in neurodegeneration. This review highlights the mostrecent developments and future directions in this field and puts themin perspective of the novel insights into the neurodegenerative process,which have been gained from related disorders, e.g., the role of FUS inamyotrophic lateral sclerosis.

71

Ann

u. R

ev. N

euro

sci.

2010

.33:

71-8

8. D

ownl

oade

d fr

om w

ww

.ann

ualr

evie

ws.

org

by U

nive

rsity

of

Con

nect

icut

on

10/2

9/12

. For

per

sona

l use

onl

y.

Page 2: Molecular Pathways of Frontotemporal Lobar Degeneration

NE33CH04-VanBroeckhoven ARI 14 May 2010 16:24

FTD: frontotemporaldementia

SD: semanticdementia

PNFA: progressivenonfluent aphasia

MND: motor neurondisease

FTLD-tau:frontotemporal lobardegeneration with taupathology

Contents

INTRODUCTION . . . . . . . . . . . . . . . . . . 72ETIOLOGICAL

HETEROGENEITY. . . . . . . . . . . . . . 72TDP-43 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 73PROGRANULIN . . . . . . . . . . . . . . . . . . . . 75CAN THE UNDERLYING

ETIOLOGY BE ESTABLISHEDIN VIVO?. . . . . . . . . . . . . . . . . . . . . . . . . 76

CAN GRN MUTATIONS OTHERTHAN NULL ALLELEMUTATIONS CAUSEDISEASE? . . . . . . . . . . . . . . . . . . . . . . . . 78

PGRN AND TDP-43 . . . . . . . . . . . . . . . . 79TDP-43 AND FUS SUGGEST A

ROLE FOR ABERRANT RNAPROCESSING INNEURODEGENERATION . . . . . . 80

CONCLUDING REMARKS . . . . . . . . . 81

INTRODUCTION

Frontotemporal dementia (FTD), semantic de-mentia (SD), and progressive nonfluent aphasia(PNFA) are gravely disabling and irreversibleclinical conditions that are characterized byprogressive neuronal loss in the frontal and/ortemporal cortices, collectively referred to asfrontotemporal lobar degeneration (FTLD).Different patterns of neurodegeneration can bedistinguished, with predominant prefrontal andanterotemporal atrophy in FTD, atrophy of themiddle and inferotemporal cortex in SD, andasymmetric atrophy of the left frontal and tem-poral cortices in PNFA (Neary et al. 2005).These topological differences are reflected inthe clinical presentation. FTD, often referredto as behavioral variant FTLD, presents clini-cally with changes in behavior and social con-duct, including loss of social awareness, poorimpulse control, and stereotypic, ritualized be-havior. Patients with SD develop a loss of se-mantic (long-established) knowledge of faces,emotions, objects, and language, resulting inimpaired word comprehension with otherwise

fluent and grammatically faultless speech. Incontrast, patients with PNFA progressively losefluency of speech with intact word comprehen-sion. In later stages of FTLD, the distinct clin-ical phenotypes usually start to show overlap,possibly reflecting the progressive involvementof other brain regions. Moreover, motor neu-ron disease (MND) and parkinsonism compli-cate the disease in up to 15% of patients, andoverlap with symptoms of Alzheimer disease(AD), corticobasal syndrome (CBS), and pro-gressive supranuclear palsy (PSP) is not un-common (Boeve 2007). Onset age of FTLDranges on average from 45 to 65 years (Nearyet al. 2005), often affecting people who are mid-career and still raising a family. Because of in-sidious onset, clinical heterogeneity, and thecurrent lack of rapid and conclusive diagnostictests, erroneous referrals tend to prolong thediagnostic trajectory, with an average diagnos-tic delay of three to four years (Hodges et al.2003). At present, no cure exists. In addition toclinical heterogeneity, FTLD is markedly het-erogeneous at the pathological as well as geneticlevel. As long as this etiological heterogeneityis not fully characterized and recognized, it willseriously impede the development of diagnosticmarkers and of drugs and/or treatments. Break-throughs in recent years, however, have signifi-cantly advanced our understanding of the com-plexity and heterogeneity of FTLD, opening upavenues for further research, several of whichare highlighted in this review.

ETIOLOGICALHETEROGENEITY

The intraneuronal accumulation of filamen-tous, hyperphosphorylated microtubule associ-ated protein tau is the most thoroughly inves-tigated molecular signature of FTLD (FTLD-tau), driven by A. Pick’s (1892) report of charac-teristic lesions in the brain of an FTD patient,hence the eponym Pick’s disease (Pick 1892),and the affirmative observation more than 100years later that mutations in the gene encod-ing microtubule associated protein tau (MAPT)cause FTLD-tau (Hutton et al. 1998). Tau is

72 Sleegers · Cruts · Van Broeckhoven

Ann

u. R

ev. N

euro

sci.

2010

.33:

71-8

8. D

ownl

oade

d fr

om w

ww

.ann

ualr

evie

ws.

org

by U

nive

rsity

of

Con

nect

icut

on

10/2

9/12

. For

per

sona

l use

onl

y.

Page 3: Molecular Pathways of Frontotemporal Lobar Degeneration

NE33CH04-VanBroeckhoven ARI 14 May 2010 16:24

TDP-43: 43 kDatransactivatingresponsive sequenceDNA-binding protein

FTLD-TDP:frontotemporal lobardegeneration withTDP-43 pathology

FTLD-UPS:frontotemporal lobardegeneration with tau-and TDP-43-negativeinclusions, which areimmunoreactive toproteins of theubiquitin-proteasomesystem

abundantly expressed in the central nervous sys-tem, where it interacts with microtubules to sta-bilize the microtubule network and regulatesaxonal transport, mediated by three or four mi-crotubule binding repeat regions (3R or 4R) intau. Most MAPT mutations identified to dateare missense, deletion, or silent mutations orintronic mutations affecting splice sites, result-ing in decreased ability to bind microtubules,increased tendency to form filaments, or alteredratio of tau 4R and 3R isoforms by alterna-tive splicing of exon 10, which encodes the sec-ond of the four microtubule binding repeat re-gions (see the FTD mutation database, http://www.molgen.ua.ac.be/FTDmutations; forreview, see Rademakers et al. 2004). In addi-tion to FTLD-tau, tauopathy is a pathologicalhallmark of other neurodegenerative diseasesas well, including AD, CBS, PSP, and argy-rophilic grain disease (AGD). Although severalquestions remain to be resolved about the waysin which aberrant tau leads to neuronal death(Wolfe 2009), it indisputably plays a role in neu-rodegeneration.

In most FTLD patients who come to au-topsy, however, tau pathology cannot be de-tected. In most of these FTLD brains, neu-ronal cytoplasmic and intranuclear inclusionsare present that are immunoreactive to ubiqui-tin (Bergmann et al. 1996, Johnson et al. 2005,Josephs et al. 2004). Adding to this ambigu-ity was the observation that several multiplextau-negative FTLD families showed conclu-sive genetic linkage to the chromosomal region17q21 harboring MAPT, but they carried nomutations in MAPT (e.g., Rademakers et al.2002, van der Zee et al. 2006). The fog be-gan to clear with the 2006 discovery of mu-tations in progranulin (GRN), located 1.7 Mbcentromeric of MAPT, explaining disease inthese tau-negative, ubiquitin-positive FTLDfamilies linked to 17q21 (Baker et al. 2006,Cruts et al. 2006). Since then, the cause ofFTLD has rapidly been resolved in many pa-tients; with 68 GRN mutations in 210 familiescurrently known worldwide (Gijselinck et al.2008a), GRN mutations are as important a causeof FTLD as MAPT mutations (http://www.

molgen.ua.ac.be/FTDmutations) (Table 1).The story quickened further with the near-simultaneous discovery of pathological 43 kDaTAR DNA-binding protein (TDP-43) asthe principal component of the ubiquitin-immunoreactive inclusions in tau-negativeFTLD-brains (FTLD-TDP) (Arai et al. 2006,Neumann et al. 2006). Taken together, tauand TDP-43 pathology still does not fully ex-plain the occurrence of FTLD. Some FTLDbrains show inclusions that are immunoreactiveonly to proteins of the ubiquitin-proteasomesystem (FTLD-UPS), and some have no in-clusions (FTLD-ni) (Mackenzie et al. 2009).And other genes besides MAPT and GRNcause FTLD (http://www.molgen.ua.ac.be/FTDmutations): Valosin-containing proteingene (VCP) mutations cause FTLD-TDP inthe frame of a broader syndrome includingPaget’s disease of the bone and inclusion bodymyopathy (IBMPFD; Watts et al. 2004), raremutations in the charged multivesicular bodyprotein 2B gene (CHMP2B) are found inFTLD-UPS (Holm et al. 2007, Skibinski et al.2005, van der Zee et al. 2008), and numer-ous families with both FTLD-TDP and MNDshow linkage to a region on chromosome 9,which suggests that at least one other gene forFTLD exists (Gijselinck et al. 2010, Le Beret al. 2009, Morita et al. 2006, Valdmanis et al.2007, Vance et al. 2006) (Table 1). Neverthe-less, with the discovery of GRN and TDP-43,the knowledge of FTLD is rapidly crystalliz-ing, which has already led to a highly sensi-tive and specific biomarker for FTLD-TDPcaused by GRN mutations (Ghidoni et al. 2008,Finch et al. 2009, Sleegers et al. 2009) as a firststep toward personalized health care for FTLDpatients.

TDP-43

TDP-43 is a DNA-, RNA-, and protein-binding nucleoprotein implicated in theregulation of numerous processes, includingtranscription, splicing, cell cycle regula-tion, apoptosis, microRNA biogenesis, mRNAtransport to and local translation at the synapse,

www.annualreviews.org • Frontotemporal Lobar Degeneration 73

Ann

u. R

ev. N

euro

sci.

2010

.33:

71-8

8. D

ownl

oade

d fr

om w

ww

.ann

ualr

evie

ws.

org

by U

nive

rsity

of

Con

nect

icut

on

10/2

9/12

. For

per

sona

l use

onl

y.

Page 4: Molecular Pathways of Frontotemporal Lobar Degeneration

NE33CH04-VanBroeckhoven ARI 14 May 2010 16:24

Table 1 Genetic heterogeneity of FTLDa

Locus GeneMutationfrequency Mutation type Phenotype Pathologyb References

3p11 CHMP2B <1% missense FTLD FTLD-UPS Skibinski et al. 20059p13 VCP <1% missense, nonsense IBMPFD FTLD-TDP Watts et al. 20049p ? ? ? FTLD+MND FTLD-TDP Morita et al. 2006c

17q21 MAPT 5–10% missense, deletion,silent, splice-site

FTLD FTLD-tau Hutton et al. 1998

17q21 GRN 5–10% deletion, nonsense,frameshift, met1,missense, splice-site

FTLD FTLD-TDP Baker et al. 2006,Cruts et al. 2006

aAbbreviations: CHMP2B, charged multivesicular body protein 2B; VCP, valosin-containing protein; MAPT, microtubule associated protein tau; GRN,progranulin; FTLD, frontotemporal lobar degeneration; IBMPFD, inclusion body myopathy associated with Paget’s disease of bone and frontotemporaldementia; MND, motor neuron disease; UPS, ubiquitine-proteasome system; TDP, TDP-43.bNomenclature according to Mackenzie et al. 2009.cFirst family published.

Proteinopathy:pathologycharacterized by theabnormal aggregationof proteins, e.g.,TDP-43 or tau; afrequentneuropathologicalobservation inneurodegenerativediseases

ALS: amyotrophiclateral sclerosis

and scaffolding for nuclear bodies (for review,see Buratti & Baralle 2008). Structurally, TDP-43 contains two RNA-recognition motifs anda carboxyl (C)-terminal glycine-rich regioninvolved in protein-binding (Wang et al.2004). Its exact role in neurodegeneration isstill a topic of intense investigation. Inclusion-bearing cells (neurons, but also glia) often showa change in subcellular distribution of TDP-43,with loss of nuclear TDP-43 and cytoplasmicsequestration (Arai et al. 2006, Neumann et al.2006). These inclusions contain both full-length TDP-43 and N-terminally truncatedfragments, which are hyperphosphorylatedand ubiquitinated. These posttranslationalmodifications are late events in the patholog-ical process (Dormann et al. 2009, Neumann2009), and N-terminal truncation is not re-quired for aggregation (Dormann et al. 2009).Like tauopathy, TDP-43 proteinopathy is notunique to FTLD but is also a pathologicalhallmark of other neurodegenerative disor-ders, e.g., amyotrophic lateral sclerosis (ALS)with or without dementia (Arai et al. 2006,Neumann et al. 2006) and Perry syndrome,an autosomal dominant form of parkinsonism(Wider et al. 2009). Furthermore, investigatorshave observed secondary TDP-43 pathologyin various other neurodegenerative disor-ders, including AD, hippocampal sclerosis,

α-synucleinopathies [Parkinson disease (PD),dementia with Lewy bodies (DLB)] andHuntington disease (for review, see Geser et al.2009). Inclusions in cortical brain regions arerich in C-terminal fragments, whereas inclu-sions in spinal cord contain more full-lengthTDP-43 (Igaz et al. 2008, Neumann et al.2009), suggesting different pathomechanismsin at least some TDP-43 proteinopathies, butphosphorylation at serine residues 409 and410 is a shared feature of all known TDP-43proteinopathies (Neumann et al. 2009). Never-theless, the inclusions contain TDP-43, leavingthe possibility that TDP-43 accumulation isnot a primary event in the pathogenesis, butrather a by-product of neurodegenerationsequestered by other aggregated components.Strongest evidence that aberrant TDP-43is sufficient to trigger neurodegeneration,however, comes from the detection of au-tosomal dominant mutations in the geneencoding TDP-43 (TARDBP) in familialALS (Kabashi et al. 2008, Sreedharan et al.2008, Van Deerlin et al. 2008). To date, mostmutations identified reside in the C-terminalglycine-rich region of the gene (http://www.molgen.ua.ac.be/FTDmutations), which isnecessary for binding of heterogeneous nuclearribonucleoproteins (hnRNP) in exon skippingand splicing activity (Buratti et al. 2005).

74 Sleegers · Cruts · Van Broeckhoven

Ann

u. R

ev. N

euro

sci.

2010

.33:

71-8

8. D

ownl

oade

d fr

om w

ww

.ann

ualr

evie

ws.

org

by U

nive

rsity

of

Con

nect

icut

on

10/2

9/12

. For

per

sona

l use

onl

y.

Page 5: Molecular Pathways of Frontotemporal Lobar Degeneration

NE33CH04-VanBroeckhoven ARI 14 May 2010 16:24

Loss-of-functionmutation: anymutation that givesrise to a gene productwith a complete orpartial loss of itsfunction

PGRN (or GRN):progranulin(protein/gene)

Several mutations are predicted to affectphosphorylation by introducing new phospho-rylation sites or increasing phosphorylationat adjacent residues, suggesting a role for ab-normal phosphorylation in neurodegeneration(Neumann et al. 2009), although phosphory-lation was found to be a relatively late eventin the conversion of soluble to insolubleTDP-43 (Dormann et al. 2009). The obser-vation of pathological TDP-43 inclusions inthe cytoplasm suggests that a loss of normalfunction in the nucleus could result in neu-rodegeneration, e.g., through a dysfunctionalshuttling of TDP-43 to and from the nucleusand the cytoplasm (Winton et al. 2008a).Drosophila melanogaster depleted of TDP-43shows atrophic presynaptic terminals at theneuromuscular junction, impaired locomotion,and reduced life span, which can be rescued byexpressing human TDP-43; this observationis indicative of a pathogenic loss-of-functionmechanism (Feiguin et al. 2009). The patternof nuclear clearing of TDP-43 is in line witha loss-of-function mechanism, but it is notobvious how the intranuclear inclusions ofTDP-43, which are frequently observed inGRN and VCP mutation carriers (Mackenzieet al. 2006, Sampathu et al. 2006), for example,fit into this picture. Conversely, a toxic gainof function is also conceivable. In a yeastmodel, several TARDBP mutations acceleratedaggregation of TDP-43 and were more toxicthan wild-type TDP-43 expressed at equallyhigh levels, suggesting a toxic gain of function(Johnson et al. 2009). Other investigators(Igaz et al. 2009, Zhang et al. 2009) have alsoobserved cytotoxicity of C-terminal fragments.Overexpression of TDP-43 in rat substantianigra through an adeno-associated virus vectorbrought about a neurodegeneration patternthat resembles human TDP-43 proteinopa-thy as well as behavioral motor dysfunction(Tatom et al. 2009), an observation thatwas further strengthened by TDP-43 trans-genic mouse lines (whether overexpressingmutant or wild-type human TDP-43) thatdisplayed a neurodegenerative phenotype(Wegorzewska et al. 2009, Wils et al.

2010), with homozygous and hemizygouswild-type TDP-43 transgenic mice showingdose-dependent neurodegeneration (Wils et al.2010). On the other hand, both overexpressionof mutant human TDP-43 and knockdownof tardbp in Danio rerio embryos caused asimilar motor phenotype, which suggestedthat both gain- and loss-of-function of TDP-43 can induce neurodegeneration (Kabashiet al. 2010). Further proof could come fromthe observation of genetic variants affectinggene dosage, but so far, no copy-numbervariants have been detected in ALS or FTLD(Gijselinck et al. 2009). Of note, althoughTARDBP missense mutations appeared to beunique for ALS, missense mutations have beenreported in patients with a clinical diagnosisof FTLD with or without MND (Benajibaet al. 2009, Winton et al. 2008b), andKovacs et al. (2009) recently described a novelmissense mutation in a patient with pathologi-cally confirmed FTLD-TDP with concomitantsupranuclear palsy and choreatic movements,but without signs of MND. Although theevidence supporting a pathogenic nature ofsome of these variants is not yet conclusive(e.g., lack of family data for segregation analysisor functional assay), these data suggest that ge-netic screening for TARDBP mutations shouldbe considered in the broader spectrum ofneurodegeneration.

PROGRANULIN

Progranulin (PGRN) is a ubiquitously ex-pressed secreted precursor protein that containstandem repeats of a unique (10- or) 12-cysteine(Cys) motif, which are proteolytically cleavedto form seven granulin (grn) peptides (grn A-G) (He & Bateman 2003). Both full-lengthPGRN and the grn peptides are implicated ina wide variety of biological functions, startingat embryonic development, including cell cy-cle regulation, wound repair, tumor growth,and inflammation, sometimes with opposite ef-fects (e.g., Daniel et al. 2003, He et al. 2003,He & Bateman 2003, Plowman et al. 1992).The balance between PGRN and grn peptides

www.annualreviews.org • Frontotemporal Lobar Degeneration 75

Ann

u. R

ev. N

euro

sci.

2010

.33:

71-8

8. D

ownl

oade

d fr

om w

ww

.ann

ualr

evie

ws.

org

by U

nive

rsity

of

Con

nect

icut

on

10/2

9/12

. For

per

sona

l use

onl

y.

Page 6: Molecular Pathways of Frontotemporal Lobar Degeneration

NE33CH04-VanBroeckhoven ARI 14 May 2010 16:24

Nonsense mediatedmRNA decay: acellular surveillancemechanism thatdegrades aberrantmRNAs containingpremature translationtermination codons

Null allele mutation:a mutation in an allelethat preventstranscription of thegene and/ortranslation into afunctional protein

Haploinsufficiency:condition in which lossof one functional alleleof a gene is sufficientto cause an abnormalphenotype

is regulated by secretory leukocyte proteaseinhibitor (SLPI), which binds to PGRN to pre-vent proteolysis by elastase (Zhu et al. 2002).PGRN is abundantly expressed in mitoticallyactive tissues or upon injury in tissues thatare less mitotically active (Daniel et al. 2000).Elevated PGRN is a frequent observation in tu-mors (He & Bateman 2003). PGRN is also ex-pressed in the central nervous system, alreadyat the embryonic stage, and significant expres-sion has been observed in the pyramidal andgranule cells of the hippocampus, the Purkinjecells, and cortical neurons (Daniel et al. 2000,Daniel et al. 2003), but its exact role in the post-mitotic neurons of the adult nervous system isless well-characterized.

All pathogenic GRN mutations identifiedin FTLD so far are dominant loss-of-functionmutations, including whole gene deletions(Gijselinck et al. 2008b) and nonsense andframeshift mutations leading to prematuretermination codons with subsequent non-sense mediated mRNA decay of the mutanttranscript, as well as mutations in the signalpeptide leading to protein mislocalization anddegradation and mutations in the initiationcodon preventing translation (i.e., GRN nullallele mutations) (for review, see Cruts & VanBroeckhoven 2008, Gijselinck et al. 2008a).These dominant null allele mutations in GRNinvariably result in PGRN haploinsufficiency(i.e., loss of 50% PGRN), implying that PGRNis critical for survival of neurons in the adultbrain. Several observations further support thishypothesis. Investigators have documentedsignificantly increased levels of PGRN indiseased tissue in ALS (Malaspina et al. 2001,Irwin, Lippa & Rosso 2009), Creutzfeldt-Jakobdisease (Baker & Manuelidis 2003), and AD(Baker et al. 2006; Pereson et al. 2009). Fur-thermore, full-length PGRN enhanced axonaloutgrowth, and grn E promoted neuronalsurvival of cultured neurons (Van Damme et al.2008). Last, GRN mutation carriers can show awide range of clinical presentations, includingAD, PD, and CBS (e.g., Benussi et al. 2009,Brouwers et al. 2007, Le Ber et al. 2008, Rade-makers et al. 2007), which could be interpreted

to reflect a loss of neuroprotection againstother looming neurodegenerative injuries.

GRN null allele mutations have not yet beenobserved in patients with ALS, despite the pres-ence of a similar TDP-43 proteinopathy, whichsuggests that motor neurons are less sensitiveto a 50% reduction in PGRN. Partial loss ofGRN is clearly not detrimental for all functionsof PGRN because patients carrying a GRNnull allele mutation have no other apparent ab-normalities, e.g., in development, growth, orwound repair. There could be many reasonswhy GRN is not always harmful, includingfunctional redundancy in these important bio-logical processes, regulation of expression of thefunctional allele of the gene, differences in sig-nal transduction (Zanocco-Marani et al. 1999),differences in receptors (which are currentlyunknown), and/or the presence of strong modi-fying factors. Homozygous Grn knockout miceshow only a mild male-type behavioral pheno-type (Kayasuga et al. 2007), underscoring thatPGRN is not indispensable for many of the bi-ological processes in which it is involved. Ofnote, even though some patients develop firstsymptoms of FTLD in their early forties, somemutation carriers live well beyond 70 yearswithout any cognitive complaints, even withinone family (e.g., Brouwers et al. 2007), whichstrongly indicates modifying factors. Furtherexploration of these aspects will likely be in-strumental in identifying targets for therapy.

CAN THE UNDERLYINGETIOLOGY BE ESTABLISHEDIN VIVO?

In postmortem tissue, distinct pathologicalprofiles of FTLD-TDP can be discernedthat appear to correlate well with the clinicaland genetic subtypes (Mackenzie et al. 2006,Sampathu et al. 2006). Small neurites, cytoplas-mic inclusions, and intranuclear inclusions arefrequent in GRN mutation carriers and in FTDor PNFA patients. Intranuclear inclusions areless frequent in families linked to chromosome9 and patients with FTD-MND or SD, andthe latter often have a predominance of long

76 Sleegers · Cruts · Van Broeckhoven

Ann

u. R

ev. N

euro

sci.

2010

.33:

71-8

8. D

ownl

oade

d fr

om w

ww

.ann

ualr

evie

ws.

org

by U

nive

rsity

of

Con

nect

icut

on

10/2

9/12

. For

per

sona

l use

onl

y.

Page 7: Molecular Pathways of Frontotemporal Lobar Degeneration

NE33CH04-VanBroeckhoven ARI 14 May 2010 16:24

neurites. Patients with IBMPFD with a VCPmutation typically have more intranuclear thancytoplasmic inclusions (Mackenzie et al. 2006,Sampathu et al. 2006). Conversely, the abilityto distinguish between the different etiologicalpathways during life is less straightforward,even though this knowledge is crucial tomake the correct treatment decisions whendrugs targeting the pathological proteins aredeveloped. VCP mutations cause the raredisorder IBMPFD (Watts et al. 2004), butthis correlation is not perfect; even in carriersof an identical VCP mutation, investigatorshave observed marked heterogeneity in clinicalexpression, such as VCP p.Arg159His, whichpresented with FTD only in one family, withFTD and/or Paget’s disease of the bone in an-other family, and with predominant inclusionbody myopathy or Paget’s disease in a thirdfamily (van der Zee et al. 2009). Nevertheless,presence of Paget’s disease or inclusion bodymyopathy in a patient with FTD strongly sug-gests an underlying VCP mutation. Apart fromFTLD-TDP caused by rare mutations in VCP,however, no set of clinical symptoms is specificfor one genetic or pathological subtype ofFTLD (Boeve & Hutton 2008). Roughly halfof patients with FTD will have FTLD-tau, andthe other half will have FTLD-TDP (Formanet al. 2006, Johnson et al. 2005). The presenceof MND is thought to be more frequent inFTLD-TDP, but not in those patients carryinga GRN mutation (e.g., Pickering-Brown et al.2008). Several symptoms are present at anunusually high frequency in GRN mutationcarriers, including early ideomotor apraxia,early memory impairment, and hallucinations(Rademakers et al. 2007, Le Ber et al. 2008),the latter two sometimes leading to an initialclinical diagnosis of AD or DLB. Furthermore,onset age tends to be earlier in MAPT mutationcarriers than in GRN mutation carriers (Cruts& Van Broeckhoven 2008). However, none ofthese features allows perfect discrimination,and they might also be dependent on thepresence of modifying factors in families. Inaddition, GRN mutation carriers may have anatypical clinical presentation, such as CBS or

AD [15% in French series (Le Ber et al. 2008)],and go unnoticed when no relatives are affected.At neuroimaging of GRN mutation carriers,asymmetrical atrophy seems to be a frequentfeature (e.g., Beck et al. 2008, Le Ber et al. 2008,van der Zee et al. 2007). For positron emissiontomography, development of radioisotopes andtracers specific for tau or TDP-43 will likelybe an important step forward to allow in vivoimaging of the proteinopathies comparableto imaging of amyloid deposition in ADusing Pittsburgh compound B (Klunk et al.2004). The inconsistent results of biomarkerstudies in FTLD focusing on levels of tau incerebrospinal fluid (CSF) (Green et al. 1999,Grossman et al. 2005) may have been causedby unrecognized etiological heterogeneity inthe study population, with an expected 50%of tau-negative patients (Bian & Grossman2007). The recent advances, however, havestimulated the exploration of disease-specificbiomarkers. Foulds et al. (2008) reportedTDP-43 protein plasma levels to be elevated inapproximately half of the patients with clinicalFTD, compatible with what is expected basedon neuropathological studies. This biomarkerassay will most likely facilitate drug trials tar-geting TDP-43 pathology by allowing a morecareful trial design. Plasma TDP-43 levelswere also elevated in 20% of AD patients and8% of cognitively healthy individuals (Fouldset al. 2008). Similarly, TDP-43 levels in CSFwere elevated in patients with FTLD and ALSbut showed considerable overlap with controlindividuals (Kasai et al. 2009, Steinacker et al.2008). In contrast, reduced PGRN levels inserum and plasma proved to be up to 100%sensitive and specific for underlying GRNmutations (Finch et al. 2009, Ghidoni et al.2008, Sleegers et al. 2009) already in unaffectedmutation carriers. Because this biomarkeraccurately reflects the underlying disease entityand can already predict disease in a prodromalstage, it holds great promise for the futureof FTLD disease management. Furthermore,it may have an application in drug develop-ment aiming to restore PGRN levels becauseelevated levels are associated with various

www.annualreviews.org • Frontotemporal Lobar Degeneration 77

Ann

u. R

ev. N

euro

sci.

2010

.33:

71-8

8. D

ownl

oade

d fr

om w

ww

.ann

ualr

evie

ws.

org

by U

nive

rsity

of

Con

nect

icut

on

10/2

9/12

. For

per

sona

l use

onl

y.

Page 8: Molecular Pathways of Frontotemporal Lobar Degeneration

NE33CH04-VanBroeckhoven ARI 14 May 2010 16:24

cancers. A panel of rapid, noninvasive testscovering all distinct etiological entities shouldultimately facilitate the diagnostic process.

CAN GRN MUTATIONS OTHERTHAN NULL ALLELEMUTATIONS CAUSE DISEASE?

Large-scale sequencing projects of GRNhave uncovered other genetic variants thatoccur only, or more frequently, in pa-tients, but which are not manifestly null al-lele mutations (http://www.molgen.ua.ac.be/FTDmutations). The pathogenic potential ofsome of these genetic variants is still hotly de-bated. Patient-specific missense mutations havebeen detected, for example, in the evolution-arily highly conserved grn domains (Brouwerset al. 2008, van der Zee et al. 2007). Muta-tions that introduce or replace a Cys-residueare especially predicted to be pathogenic be-cause Cys-residues are required to stabilizethe fold of four stacked beta-hairpins charac-teristic for the grn domains through disulfidebridges between these residues (Hrabal et al.1996, Tolkatchev et al. 2000). These muta-tions could result in misfolding of the protein,which may lose its normal activity (Tolkatchevet al. 2008) or be destined for degradationin the endoplasmic reticulum (ER), resultingin reduced amounts of functional protein. Insupport of this hypothesis, pathogenic Cys-residue mutations in several other proteins con-taining disulfide bond-rich epidermal-growth-factor (EGF)-like domains [fibrillin-1 (FBN1),uromodulin (UMOD)] (Robinson et al. 2002,Scolari et al. 2004) indeed induce retentionof the mutant transcript in the ER becauseof abnormal protein folding (Bernascone et al.2006, Whiteman & Handford 2003). Alterna-tively, because gain or loss of a Cys-residueresults in an odd number of Cys-residues,the unpaired Cys-residue in turn could havean increased propensity to form multimersthrough aberrant disulfide bond formation, aswas recently reported for pathogenic muta-tions in NOTCH3, which caused cerebral auto-somal dominant arteriopathy with subcortical

infarcts and leukoencephalopathy (CADASIL)(Opherk et al. 2009). Pathogenic mutations inNOTCH3 almost all lead to an odd number ofCys-residues in one of its EGF-like domains(Federico et al. 2005, Joutel et al. 1997). In ayeast-two hybrid assay, a grn dimeric repeatconsisting of grn B and A was shown to bindhuman immunodeficiency virus (HIV) Tat pro-teins, and mutation of grn Cys-residues weak-ened this binding ability (Trinh et al. 1999),further highlighting the pathogenic potentialof Cys-residue mutations.

Other GRN missense mutations haveaffected highly conserved Pro-residues occur-ring in a loop of the protein backbone of thegrn domain. Molecular modeling predictedamino acid substitutions at this position toaffect folding and stability of the grn domain(Brouwers et al. 2008, van der Zee et al. 2007).Arguing against a putative pathogenic natureof GRN missense mutations is the fact thatsome, albeit different, missense mutationsalso occur in healthy control individuals.FTLD-TDP has not been pathologicallyconfirmed, and segregation data are sparse.One mutation, p.Arg432Cys, was observed inthree (genetically distantly) related patientswith clinical FTLD (van der Zee et al. 2007);p.Cys521Tyr was observed in four affectedrelatives from a PNFA family but also inseveral unaffected siblings, although cognitivetests of carriers predicted a mild cognitiveimpairment (Cruchaga et al. 2009). However,in cultured cells, two predicted pathogenicmutations, p.Pro248Leu and p.Arg432Cys,showed less efficient transport through thesecretory pathway, which led to a significant(70% and 45%) reduction in PGRN secretion(Shankaran et al. 2008). This partial loss offunctional protein suggests that these raremutant alleles act as low penetrant risk factors,as opposed to the autosomal dominant natureof null allele mutations that create completehaploinsufficiency. This hypothesis is very wellcompatible with the presence of some of thesemutations in control individuals and with thelack of segregation data. Of note, the frequencyof these missense mutations in both patients

78 Sleegers · Cruts · Van Broeckhoven

Ann

u. R

ev. N

euro

sci.

2010

.33:

71-8

8. D

ownl

oade

d fr

om w

ww

.ann

ualr

evie

ws.

org

by U

nive

rsity

of

Con

nect

icut

on

10/2

9/12

. For

per

sona

l use

onl

y.

Page 9: Molecular Pathways of Frontotemporal Lobar Degeneration

NE33CH04-VanBroeckhoven ARI 14 May 2010 16:24

and healthy individuals is usually rare, withminor allele frequencies less than 1%. In ad-dition, missense mutations are also detected inother neurodegenerative disorders, such as AD(Brouwers et al. 2008), PD (Nuytemans et al.2008), and ALS (Schymick et al. 2007, Sleegerset al. 2008), which again suggests that theserare mutant alleles might increase susceptibilityfor neurodegeneration by failing neurotrophicproperties. In line with this speculation, invivo data on circulating protein levels inplasma or serum show a partial reduction ofPGRN in some predicted pathogenic missensemutation carriers [e.g., unrelated carriers ofp.Cys139Arg had partially reduced PGRNlevels in two independent studies (Finch et al.2009, Sleegers et al. 2009)], with levels inter-mediate between noncarriers and null mutationcarriers, regardless of their clinical phenotype.Because of their low frequency and possibly lowpenetrance, definite proof of the pathogenicityof some of these mutations relies on continuedfunctional experiments, which could, amongothers, address the question of whether loss ofone grn peptide is sufficient to affect diseaserisk, and if so, which grn peptides are relevantfor neuroprotection, whether acting alone orin multimers. Of note, the ability to adopt agrn fold is necessary but not sufficient for bio-logical activity of the grn peptides (Tolkatchevet al. 2008), which suggests that not everygrn domain will be equally sensitive to aminoacid substitutions. Common GRN geneticvariants have also been implicated in diseaserisk, affecting onset age and survival in patientswith ALS (Sleegers et al. 2008) among others,and a common 3′ UTR variant affected riskof FTLD by influencing a microRNA bindingsite (Rademakers et al. 2008); however, otherinvestigators have been unable to replicatethese findings (Rollinson et al. 2010). Becauseof their potential therapeutic implications,however, these findings warrant follow-up.

PGRN AND TDP-43

One of the puzzles that remain to be resolved iswhether and how loss of PGRN is linked with

the pathological accumulation of TDP-43.Mutant TARDBP is clearly sufficient to induceTDP-43 pathology in ALS patients, but muta-tions in TARDBP are at best a rare occurrencein FTLD (Benajiba et al. 2009, Gijselinck et al.2009, Kovacs et al. 2009, Winton et al. 2008b),and conversely, despite similarities at thepathological level between FTLD-TDP andALS, GRN null allele mutations have not beenfound in ALS (Schymick et al. 2007, Sleegerset al. 2008). VCP mutations, which are asso-ciated with predominant neuronal intranuclearTDP-43 inclusions (Mackenzie et al. 2006,Sampathu et al. 2006), altered localization ofTDP-43 between nucleus and cytosol, and aminority of TDP-43-positive inclusions wasalso VCP-immunoreactive (Gitcho et al. 2009);for GRN mutations, however, no such evidenceexists at present. Knockdown of neuronal GRNexpression using small interfering RNA incell culture models led to abnormal caspase3-mediated cleavage of TDP-43 (Zhang et al.2007), but this finding has been challengedby others (Shankaran et al. 2008). Brains of7–8-month-old PGRN knockout mice showedno evidence for accumulation of C-terminalfragments of TDP-43 or of phosphorylatedTDP-43 (Dormann et al. 2009). In an axotomymodel in mice, TDP-43 showed increasedexpression and relocalization to the cytosolfollowing acute neuronal injury, while aneuronal decrease in PGRN was observed(Moisse et al. 2009). The increase in TDP-43upon injury indicates a physiological role inneuronal repair. The concomitant decrease inneuronal PGRN is less well interpretable butcould reflect secretion of PGRN, given theneurotrophic properties of secreted PGRN(Moisse et al. 2009). However, Matzilevichet al. (2002) also reported a delayed responseof PGRN expression to acute neuronal injury,which suggests that it has a more promi-nent role in long-term neuronal survival(Ahmed et al. 2007). In surrounding microglia,expression of PGRN was upregulated inresponse to axotomy, which is compatible witha role in inflammation and repair (Ahmed et al.2007). Nevertheless, this occurrence does not

www.annualreviews.org • Frontotemporal Lobar Degeneration 79

Ann

u. R

ev. N

euro

sci.

2010

.33:

71-8

8. D

ownl

oade

d fr

om w

ww

.ann

ualr

evie

ws.

org

by U

nive

rsity

of

Con

nect

icut

on

10/2

9/12

. For

per

sona

l use

onl

y.

Page 10: Molecular Pathways of Frontotemporal Lobar Degeneration

NE33CH04-VanBroeckhoven ARI 14 May 2010 16:24

imply a direct functional link between TDP-43and PGRN. Given the possible role of grn pep-tides in HIV Tat protein binding (Trinh et al.1999), and the ability of TDP-43 to bind to theHIV TAR DNA element to which Tat proteinsbind to activate gene expression (Ou et al.1995), Ahmed et al. (2007) have speculated thatPGRN and TDP-43 may be able to interact di-rectly; this awaits further exploration, however.

TDP-43 AND FUS SUGGEST AROLE FOR ABERRANT RNAPROCESSING INNEURODEGENERATION

Much may be learned from searching forunifying themes in related neurodegenerativedisorders. For FTLD-TDP, ALS seems tobe particularly relevant. Like FTLD, ALS isa mechanistically heterogeneous disorder, butone of the major ALS pathologies is TDP-43proteinopathy (Arai et al. 2006, Neumann et al.2006). Substantial overlap also exists at the clin-ical level. FTD is complicated by MND in a sig-nificant proportion of patients, and up to 50%of ALS patients develop symptoms of fron-totemporal dysfunction (Lomen-Hoerth et al.2003). Moreover, multiplex families in whichFTLD-TDP and ALS cosegregate have beenlinked to a region on chromosome 9p, whichimplies that mutations in one gene could in-duce both clinical phenotypes (Le Ber et al.2009, Morita et al. 2006, Valdmanis et al. 2007,Vance et al. 2006; Gijselinck et al. 2010). Thesedata support the notion that FTLD-TDP andALS are closely related conditions in a con-tinuum of neurodegeneration and that scien-tific breakthroughs in one area may have a rip-ple effect extending to other disorders alongthe continuum. Dominant familial ALS linkedto chromosome 16 was recently shown to re-sult from pathogenic mutations in FUS (fusionin sarcoma) (Kwiatkowski et al. 2009, Vanceet al. 2009). FUS is an RNA/DNA process-ing protein like TDP-43, and its implicationin ALS bears remarkable resemblance to TDP-43 (Sleegers & Van Broeckhoven 2009). LikeTDP-43, FUS is associated with hnRNP, and

its functions include DNA repair, transcrip-tion, RNA splicing, transport of mRNA to andrapid local RNA translation at the synapse, andmicroRNA processing (for review, see Lagier-Tourenne & Cleveland 2009). Most mutationsidentified in FUS to date reside in the C-terminus (Kwiatkowski et al. 2009, Vance et al.2009), resembling the C-terminal clustering ofpathogenic mutations in TARDBP. In spinalcord and brain tissue of FUS mutation carri-ers, cytoplasmic inclusions containing mutantFUS protein could be observed, and in vitroexperiments confirmed relocation of mutantFUS to the cytoplasm (Kwiatkowski et al. 2009,Vance et al. 2009). FUS mutation carriers didnot show TDP-43 pathology, which impliesthe involvement of different pathways that leadto neurodegeneration. Mutations could lead toa loss of normal function or a gain of toxicfunction of FUS in the nucleus, resulting in(potentially general) impaired RNA process-ing. Instead of RNA metabolism per se, a spe-cific RNA target of FUS could be crucial forneuron viability; identifying such RNA targetsmay provide a link between FUS and TDP-43 but also among other ALS-associated RNAprocessing proteins such as ELP3 (Simpsonet al. 2008). Mislocalization of mutant FUS tothe cytoplasm could create a loss of its func-tion in the nucleus, but cytoplasmic aggre-gates of FUS could also be toxic to the cellor lead to sequestration of other proteins inthe cytoplasm. Last, mutant FUS or TDP-43could perturb normal mRNA transport to thedendrites and/or impair rapid RNA transla-tion at the postsynaptic site (e.g., in responseto stimulation by neurotrophic factors). Fu-ture research will likely shed more light on themechanisms by which aberrant FUS as well asTDP-43 affect neuron viability (Figure 1). Theidentification of aberrant TDP-43 and FUS inmotor neuron degeneration provides furthersupport for the hypothesis that impaired RNAprocessing plays a crucial role in neurodegener-ation (Gallo et al. 2005). Examples of impairedRNA processing seem especially numerous inMND (for review, see Simpson et al. 2008).Overexpression of TDP-43 in vitro enhances

80 Sleegers · Cruts · Van Broeckhoven

Ann

u. R

ev. N

euro

sci.

2010

.33:

71-8

8. D

ownl

oade

d fr

om w

ww

.ann

ualr

evie

ws.

org

by U

nive

rsity

of

Con

nect

icut

on

10/2

9/12

. For

per

sona

l use

onl

y.

Page 11: Molecular Pathways of Frontotemporal Lobar Degeneration

NE33CH04-VanBroeckhoven ARI 14 May 2010 16:24

the transcript inclusion of exon 7 of survival ofmotor neuron 2 (SMN2) (Bose et al. 2008), en-coding an RNA-binding protein implicated inthe neurodegenerative disorder spinal muscu-lar atrophy (Lefebvre et al. 1995). In FTLD,RNA processing is already indirectly implicatedthrough alternative splicing of MAPT exon 10.Thus it is also of interest that synthetic mul-timers of grn repeats have been implicated inRNA processing by modulating transcriptionelongation through an interaction with cyclinT1 (Hoque et al. 2003). Broader involvementof RNA processing in FTLD is likely becauseit is involved in numerous biological processesspecific for proper neuronal functioning, in-cluding synaptic plasticity (Steward & Schuman2001). This, and the overlap between ALS andFTLD-TDP, raises the question of whetherFUS could also be involved in FTLD (Sleegers& Van Broeckhoven 2009). Although the re-search is in its early days, Mackenzie (2009) re-cently reported that some FTLD brains thatwere previously classified as FTLD-UPS be-cause of the presence of tau- as well as TDP-43-negative inclusions harbor FUS-positive in-clusions; likewise, we have recently identified anFTLD patient carrying a FUS missense muta-tion (p.Met254Val) affecting an evolutionarilyconserved residue in the glycine-rich region ofFUS, which is predicted to affect protein func-tion (Van Langenhove et al. 2010).

CONCLUDING REMARKS

The identification of mutations in MAPT,GRN, VCP, and CHMP2B in FTLD patientsand the observation of pathological intraneu-ronal accumulation of tau, TDP-43, and FUSprotein in brains of FTLD patients demon-strate the diversity of molecular pathways in-volved in this condition. Rare variants inTARDBP and FUS in FTLD patients may fur-ther contribute to the genetic heterogeneity ofFTLD, and at least one other genetic causefor FTLD (on 9p) remains to be uncovered.These recent observations suggest that the fieldof FTLD research might be on the verge of anew wave of discoveries. Although the search

12

3

4

56

Presynaptic

RNA-containinggranules

RNA

DNA

Aggregatedproteins

Normalprotein

Mutantproteins

Postsynaptic

Figure 1Schematic representation of possible mechanisms by which mutant TARDBPor FUS might affect neuronal viability. (1) Mutant TDP-43 or FUS could leadto a loss of normal function or a gain of toxic function in the nucleus, resultingin impaired RNA processing; (2) a specific RNA target (possibly shared by FUSand TDP-43) could be crucial for neuron viability; (3) mislocalization ofmutant FUS or TDP-43 to the cytoplasm could create a loss of its function inthe nucleus; (4) cytoplasmic aggregates of FUS or TDP-43 (full-length orC-terminal fragments, ubiquitinated and phosphorylated) could be toxic to thecell; (5) mutant FUS or TDP-43 could perturb normal mRNA transport or (6)impair local RNA translation at the synapse in response to synaptic stimulation.

for mechanistic links between these pathwaysmay provide novel insights into the neurode-generative process, recognition of the etiolo-gial diversity will be crucial to advance patientcare. It allows studies focusing on more homo-geneous etiological subclasses, which can fa-cilitate development of diagnostic proceduresand drugs, but it may also increase our un-derstanding of disease mechanisms. The firstgenome-wide association study on FTLD, e.g.,including only patients with FTLD-TDP, re-cently uncovered common genetic variation

www.annualreviews.org • Frontotemporal Lobar Degeneration 81

Ann

u. R

ev. N

euro

sci.

2010

.33:

71-8

8. D

ownl

oade

d fr

om w

ww

.ann

ualr

evie

ws.

org

by U

nive

rsity

of

Con

nect

icut

on

10/2

9/12

. For

per

sona

l use

onl

y.

Page 12: Molecular Pathways of Frontotemporal Lobar Degeneration

NE33CH04-VanBroeckhoven ARI 14 May 2010 16:24

in the gene TMEM106B as a putative riskfactor for FTLD-TDP (Van Deerlin et al.2010). Further characterization of this trans-membrane protein of unknown function may

provide one more piece of the puzzle of FTLD.Hopefully, in the not-too-distant future thevarious etiological entities underlying FTLDwill be fully characterized.

DISCLOSURE STATEMENT

The authors are not aware of any affiliations, memberships, funding, or financial holdings thatmight be perceived as affecting the objectivity of this review.

ACKNOWLEDGMENTS

Research in the authors’ research group was funded in part by the Special Research Fund of theUniversity of Antwerp, the Fund for Scientific Research Flanders (FWO-V), the InteruniversityAttraction Poles program (IAP) P6/43 of the Belgian Science Policy Office, the Medical Founda-tion Queen Elisabeth, the Foundation for Alzheimer Research (SAO/FRMA), the Association forFrontotemporal Dementias (AFTD), and a Methusalem Excellence Grant of the Flemish Govern-ment, Flanders, Belgium, and a Zenith award from the Alzheimer’s Association USA to C.V.B., aMarie-Therese de Lava Fund of the King Baudouin Foundation Belgium to M.C., and a Santkinaward from the National Alzheimer League Belgium to M.C. and K.S. K.S. is a postdoctoralfellow of the FWO-V.

LITERATURE CITED

Ahmed Z, Mackenzie IR, Hutton ML, Dickson DW. 2007. Progranulin in frontotemporal lobar degenerationand neuroinflammation. J. Neuroinflamm. 4:7

Arai T, Hasegawa M, Akiyama H, Ikeda K, Nonaka T, et al. 2006. TDP-43 is a component of ubiquitin-positive tau-negative inclusions in frontotemporal lobar degeneration and amyotrophic lateral sclerosis.Biochem. Biophys. Res. Commun. 351(3):602–11

Baker CA, Manuelidis L. 2003. Unique inflammatory RNA profiles of microglia in Creutzfeldt-Jakob disease.Proc. Natl. Acad. Sci. USA 100:675–79

Baker M, Mackenzie IR, Pickering-Brown SM, Gass J, Rademakers R, et al. 2006. Mutations in progranulincause tau-negative frontotemporal dementia linked to chromosome 17. Nature 442(7105):916–19

Beck J, Rohrer JD, Campbell T, Isaacs A, Morrison KE, et al. 2008. A distinct clinical, neuropsychologicaland radiological phenotype is associated with progranulin gene mutations in a large UK series. Brain131(Pt. 3):706–20

Benajiba L, Le Ber I, Camuzat A, Lacoste M, Thomas-Anterion C, et al. 2009. TARDBP mutations inmotoneuron disease with frontotemporal lobar degeneration. Ann. Neurol. 65(4):470–73

Benussi L, Ghidoni R, Pegoiani E, Moretti DV, Zanetti O, Binetti G. 2009. Progranulin Leu271LeufsX10 isone of the most common FTLD and CBS associated mutations worldwide. Neurobiol. Dis. 33(3):379–85

Bergmann M, Kuchelmeister K, Schmid KW, Kretzschmar HA, Schroder R. 1996. Different variants of fron-totemporal dementia: a neuropathological and immunohistochemical study. Acta Neuropathol. 92(2):170–79

Bernascone I, Vavassori S, Di Pentima A, Santambrogio S, Lamorte G, et al. 2006. Defective intracellulartrafficking of uromodulin mutant isoforms. Traffic 7:1567–79

Bian H, Grossman M. 2007. Frontotemporal lobar degeneration: recent progress in antemortem diagnosis.Acta Neuropathol. 114(1):23–29

Boeve BF. 2007. Links between frontotemporal lobar degeneration, corticobasal degeneration, progressivesupranuclear palsy, and amyotrophic lateral sclerosis. Alzheimer Dis. Assoc. Disord. 21:S31–38

82 Sleegers · Cruts · Van Broeckhoven

Ann

u. R

ev. N

euro

sci.

2010

.33:

71-8

8. D

ownl

oade

d fr

om w

ww

.ann

ualr

evie

ws.

org

by U

nive

rsity

of

Con

nect

icut

on

10/2

9/12

. For

per

sona

l use

onl

y.

Page 13: Molecular Pathways of Frontotemporal Lobar Degeneration

NE33CH04-VanBroeckhoven ARI 14 May 2010 16:24

Boeve BF, Hutton M. 2008. Refining frontotemporal dementia with Parkinsonism linked to chromosome 17:introducing FTDP-17 (MAPT) and FTDP-17 (PGRN). Arch. Neurol. 65(4):460–64

Bose JK, Wang IF, Hung L, Tarn WY, Shen CK. 2008. TDP-43 overexpression enhances exon 7 inclusionduring the survival of motor neuron pre-mRNA splicing. J. Biol. Chem. 283(43):28852–59

Brouwers N, Nuytemans K, Van Der Zee J, Gijselinck I, Engelborghs S, et al. 2007. Alzheimer and parkinsondiagnoses in progranulin null mutation carriers in an extended founder family. Arch. Neurol. 64(10):1436–46

Brouwers N, Sleegers K, Engelborghs S, Maurer-Stroh S, Gijselinck I, et al. 2008. Genetic variability inprogranulin contributes to risk for clinically diagnosed Alzheimer disease. Neurology 71(9):656–64

Buratti E, Baralle FE. 2008. Multiple roles of TDP-43 in gene expression, splicing regulation, and humandisease. Front. Biosci. 13:867–78

Buratti E, Brindisi A, Giombi M, Tisminetzky S, Ayala YM, Baralle FE. 2005. TDP-43 binds heterogeneousnuclear ribonucleoprotein A/B through its C-terminal tail: an important region for the inhibition of cysticfibrosis transmembrane conductance regulator exon 9 splicing. J. Biol. Chem. 280(45):37572–84

Cruchaga C, Fernandez-Seara MA, Seijo-Martınez M, Samaranch L, Lorenzo E, et al. 2009. Cortical atro-phy and language network reorganization associated with a novel progranulin mutation. Cereb. Cortex19(8):1751–60

Cruts M, Gijselinck I, Van Der Zee J, Engelborghs S, Wils H, et al. 2006. Null mutations in progranulin causeubiquitin-positive frontotemporal dementia linked to chromosome 17q21. Nature 442(7105):920–24

Cruts M, Van Broeckhoven C. 2008. Loss of progranulin function in frontotemporal lobar degeneration.Trends Genet. 24(4):186–94

Daniel R, Daniels E, He Z, Bateman A. 2003. Progranulin (acrogranin/PC cell-derived growth factor/granulin-epithelin precursor) is expressed in the placenta, epidermis, microvasculature, and brain during murinedevelopment. Dev. Dyn. 227(4):593–99

Daniel R, He Z, Carmichael KP, Halper J, Bateman A. 2000. Cellular localization of gene expression forprogranulin. J. Histochem. Cytochem. 48(7):999–1009

Dormann D, Capell A, Carlson AM, Shankaran SS, Rodde R, et al. 2009. Proteolytic processing of TARDNA binding protein-43 by caspases produces C-terminal fragments with disease defining propertiesindependent of progranulin. J. Neurochem. 110(3):1082–94

Federico A, Bianchi S, Dotti MT. 2005. The spectrum of mutations for CADASIL diagnosis. Neurol. Sci.26(2):117–24

Feiguin F, Godena VK, Romano G, D’Ambrogio A, Klima R, Baralle FE. 2009. Depletion of TDP-43 affectsDrosophila motoneurons terminal synapsis and locomotive behavior. FEBS Lett. 583(10):1586–92

Finch N, Baker M, Crook R, Swanson K, Kuntz K, et al. 2009. Plasma progranulin levels predict pro-granulin mutation status in frontotemporal dementia patients and asymptomatic family members. Brain132(Pt. 3):583–91

Forman MS, Farmer J, Johnson JK, Clark CM, Arnold SE, et al. 2006. Frontotemporal dementia: clinico-pathological correlations. Ann. Neurol. 59(6):952–62

Foulds P, McAuley E, Gibbons L, Davidson Y, Pickering-Brown SM, et al. 2008. TDP-43 protein in plasmamay index TDP-43 brain pathology in Alzheimer’s disease and frontotemporal lobar degeneration. ActaNeuropathol. 116(2):141–46

Gallo JM, Jin P, Thornton CA, Lin H, Robertson J, et al. 2005. The role of RNA and RNA processing inneurodegeneration. J. Neurosci. 25(45):10372–75

Geser F, Martinez-Lage M, Kwong LK, Lee VM, Trojanowski JQ. 2009. Amyotrophic lateral sclerosis,frontotemporal dementia and beyond: the TDP-43 diseases. J. Neurol. 256(8):1205–14

Ghidoni R, Benussi L, Glionna M, Franzoni M, Binetti G. 2008. Low plasma progranulin levels predictprogranulin mutations in frontotemporal lobar degeneration. Neurology 71(16):1235–39

Gijselinck I, Engelborghs S, Maes G, Cuijt I, Peeters K, et al. 2010. A genome wide linkage study in a multiplexFTLD-ALS family identifies two loci at chromosomes 9 and 14. Arch. Neurol. In press

Gijselinck I, Sleegers K, Engelborghs S, Robberecht W, Martin JJ, et al. 2009. Neuronal inclusion proteinTDP-43 has no primary genetic role in FTD and ALS. Neurobiol. Aging 30(8):1329–31

Gijselinck I, Van Broeckhoven C, Cruts M. 2008a. Granulin mutations associated with frontotemporal lobardegeneration and related disorders: an update. Hum. Mut. 29(12):1373–86

www.annualreviews.org • Frontotemporal Lobar Degeneration 83

Ann

u. R

ev. N

euro

sci.

2010

.33:

71-8

8. D

ownl

oade

d fr

om w

ww

.ann

ualr

evie

ws.

org

by U

nive

rsity

of

Con

nect

icut

on

10/2

9/12

. For

per

sona

l use

onl

y.

Page 14: Molecular Pathways of Frontotemporal Lobar Degeneration

NE33CH04-VanBroeckhoven ARI 14 May 2010 16:24

Gijselinck I, Van Der Zee J, Engelborghs S, Goossens D, Peeters K, et al. 2008b. Progranulin locus deletionin frontotemporal dementia. Hum. Mutat. 29(1):53–58

Gitcho MA, Strider J, Carter D, Taylor-Reinwald L, Forman MS, et al. 2009. VCP mutations causing fron-totemporal lobar degeneration disrupt localization of TDP-43 and induce cell death. J. Biol. Chem.284(18):12384–98

Green AJ, Harvey RJ, Thompson EJ, Rossor MN. 1999. Increased tau in the cerebrospinal fluid of patientswith frontotemporal dementia and Alzheimer’s disease. Neurosci. Lett. 259(2):133–35

Grossman M, Farmer J, Leight S, Work M, Moore P, et al. 2005. Cerebrospinal fluid profile in frontotemporaldementia and Alzheimer’s disease. Ann. Neurol. 57(5):721–29

He Z, Bateman A. 2003. Progranulin (granulin-epithelin precursor, PC-cell-derived growth factor, acrogranin)mediates tissue repair and tumorigenesis. J. Mol. Med. 81(10):600–12

He Z, Ong CH, Halper J, Bateman A. 2003. Progranulin is a mediator of the wound response. Nat. Med.9(2):225–29

Hodges JR, Davies R, Xuereb J, Kril J, Halliday G. 2003. Survival in frontotemporal dementia. Neurology61:349–54

Holm IE, Englund E, Mackenzie IR, Johannsen P, Isaacs AM. 2007. A reassessment of the neuropathologyof frontotemporal dementia linked to chromosome 3. J. Neuropathol. Exp. Neurol. 66:884–91

Hoque M, Young TM, Lee CG, Serrero G, Mathews MB, Pe’ery T. 2003. The growth factor granulin interactswith cyclin T1 and modulates P-TEFb-dependent transcription. Mol. Cell. Biol. 23(5):1688–702

Hrabal R, Chen Z, James S, Bennett HP, Ni F. 1996. The hairpin stack fold, a novel protein architecture fora new family of protein growth factors. Nat. Struct. Biol. 3(9):747–52

Hutton M, Lendon CL, Rizzu P, Baker M, Froelich S, et al. 1998. Association of missense and 5′-splice-sitemutations in tau with the inherited dementia FTDP-17. Nature 393(6686):702–5

Igaz LM, Kwong LK, Chen-Plotkin A, Winton MJ, Unger TL, et al. 2009. Expression of TDP-43 C-terminal fragments in vitro recapitulates pathological features of TDP-43 proteinopathies. J. Biol. Chem.284(13):8516–24

Igaz LM, Kwong LK, Xu Y, Truax AC, Uryu K, et al. 2008. Enrichment of C-terminal fragments in TARDNA-binding protein-43 cytoplasmic inclusions in brain but not in spinal cord of frontotemporal lobardegeneration and amyotrophic lateral sclerosis. Am. J. Pathol. 173(1):182–94

Irwin D, Lippa CF, Rosso A. 2009. Progranulin (PGRN) expression in ALS: an immunohistochemical study.J. Neurol. Sci. 276(1–2):9–13

Johnson BS, Snead D, Lee JJ, McCaffery JM, Shorter J, Gitler AD. 2009. TDP-43 is intrinsicallyaggregation-prone and ALS-linked mutations accelerate aggregation and increase toxicity. J. Biol. Chem.284(30):20,329–39

Johnson JK, Diehl J, Mendez MF, Neuhaus J, Shapira JS, et al. 2005. Frontotemporal lobar degeneration:demographic characteristics of 353 patients. Arch. Neurol. 62(6):925–30

Josephs KA, Holton JL, Rossor MN, Godbolt AK, Ozawa T, et al. 2004. Frontotemporal lobar degenerationand ubiquitin immunohistochemistry. Neuropathol. Appl. Neurobiol. 30(4):369–73

Joutel A, Vahedi K, Corpechot C, Troesch A, Chabriat H, et al. 1997. Strong clustering and stereotypednature of Notch3 mutations in CADASIL patients. Lancet 350(9090):1511–15

Kabashi E, Lin L, Tradewell ML, Dion PA, Bercier V, et al. 2010. Gain and loss of function of ALS-relatedmutations of TARDBP (TDP-43) cause motor deficits in vivo. Hum. Mol. Genet. 19(4):671–83

Kabashi E, Valdmanis PN, Dion P, Spiegelman D, McConkey BJ, et al. 2008. TARDBP mutations in indi-viduals with sporadic and familial amyotrophic lateral sclerosis. Nat. Genet. 40(5):572–74

Kasai T, Tokuda T, Ishigami N, Sasayama H, Foulds P, et al. 2009. Increased TDP-43 protein in cerebrospinalfluid of patients with amyotrophic lateral sclerosis. Acta Neuropathol. 117(1):55–62

Kayasuga Y, Chiba S, Suzuki M, Kikusui T, Matsuwaki T, et al. 2007. Alteration of behavioural phenotype inmice by targeted disruption of the progranulin gene. Behav. Brain Res. 185(2):110–18

Klunk WE, Engler H, Nordberg A, Wang Y, Blomqvist G, et al. 2004. Imaging brain amyloid in Alzheimer’sdisease with Pittsburgh Compound-B. Ann. Neurol. 55(3):306–19

Kovacs GG, Murrell JR, Horvath S, Haraszti L, Majtenyi K, et al. 2009. TARDBP variation associated withfrontotemporal dementia, supranuclear gaze palsy, and chorea. Mov. Disord. 24(12):1843–47

84 Sleegers · Cruts · Van Broeckhoven

Ann

u. R

ev. N

euro

sci.

2010

.33:

71-8

8. D

ownl

oade

d fr

om w

ww

.ann

ualr

evie

ws.

org

by U

nive

rsity

of

Con

nect

icut

on

10/2

9/12

. For

per

sona

l use

onl

y.

Page 15: Molecular Pathways of Frontotemporal Lobar Degeneration

NE33CH04-VanBroeckhoven ARI 14 May 2010 16:24

Kwiatkowski TJ Jr, Bosco DA, Leclerc AL, Tamrazian E, Vanderburg CR, et al. 2009. Mutations in theFUS/TLS gene on chromosome 16 cause familial amyotrophic lateral sclerosis. Science 323(5918):1205–8

Lagier-Tourenne C, Cleveland DW. 2009. Rethinking ALS: the FUS about TDP-43. Cell 136(6):1001–4Le Ber I, Camuzat A, Berger E, Hannequin D, Laquerriere A, et al. 2009. Chromosome 9p-linked families

with frontotemporal dementia associated with motor neuron disease. Neurology 72(19):1669–76Le Ber I, Camuzat A, Hannequin D, Pasquier F, Guedj E, et al. 2008. Phenotype variability in progranulin

mutation carriers: a clinical, neuropsychological, imaging and genetic study. Brain 131(Pt. 3):732–46Lefebvre S, Burglen L, Reboullet S, Clermont O, Burlet P, et al. 1995. Identification and characterization of

a spinal muscular atrophy-determining gene. Cell 80(1):155–65Lomen-Hoerth C, Murphy J, Langmore S, Kramer JH, Olney RK, Miller B. 2003. Are amyotrophic lateral

sclerosis patients cognitively normal? Neurology 60(7):1094–97Mackenzie IR. 2009. TDP-43, FUS and new neuropathological insights. Presented at Int. Res. Workshop on

Frontotemporal Dementia in ALS, 3rd, London, Ontario, Can.Mackenzie IR, Baborie A, Pickering-Brown S, Du Plessis D, Jaros E, et al. 2006. Heterogeneity of ubiquitin

pathology in frontotemporal lobar degeneration: classification and relation to clinical phenotype. Acta.Neuropathol. 112(5):539–49

Mackenzie IR, Neumann M, Bigio EH, Cairns NJ, Alafuzoff I, et al. 2009. Nomenclature for neuropathologicsubtypes of frontotemporal lobar degeneration: consensus recommendations. Acta Neuropathol. 117(1):15–18

Malaspina A, Kaushik N, de Belleroche J. 2001. Differential expression of 14 genes in amyotrophic lateralsclerosis spinal cord detected using gridded cDNA arrays. J. Neurochem. 77(1):132–45

Matzilevich DA, Rall JM, Moore AN, Grill RJ, Dash PK. 2002. High-density microarray analysis of hip-pocampal gene expression following experimental brain injury. J. Neurosci. Res. 67(5):646–63

Moisse K, Volkening K, Leystra-Lantz C, Welch I, Hill T, Strong MJ. 2009. Divergent patterns of cytoso-lic TDP-43 and neuronal progranulin expression following axotomy: implications for TDP-43 in thephysiological response to neuronal injury. Brain. Res. 1249:202–11

Morita M, Al-Chalabi A, Andersen PM, Hosler B, Sapp P, et al. 2006. A locus on chromosome 9p conferssusceptibility to ALS and frontotemporal dementia. Neurology 66(6):839–44

Neary D, Snowden J, Mann D. 2005. Frontotemporal dementia. Lancet Neurol. 4(11):771–80Neumann M. 2009. Molecular neuropathology of TDP-43 proteinopathies. Int. J. Mol. Sci. 10(1):232–46Neumann M, Kwong LK, Lee EB, Kremmer E, Flatley A, et al. 2009. Phosphorylation of S409/410 of TDP-

43 is a consistent feature in all sporadic and familial forms of TDP-43 proteinopathies. Acta Neuropathol.117(2):137–49

Neumann M, Sampathu DM, Kwong LK, Truax AC, Micsenyi MC, et al. 2006. Ubiquitinated TDP-43 infrontotemporal lobar degeneration and amyotrophic lateral sclerosis. Science 314(5796):130–33

Nuytemans K, Pals P, Sleegers K, Engelborghs S, Corsmit E, et al. 2008. Progranulin variability has no majorrole in Parkinson disease genetic etiology. Neurology 71(15):1147–51

Opherk C, Duering M, Peters N, Karpinska A, Rosner S, et al. 2009. CADASIL mutations enhance sponta-neous multimerization of NOTCH3. Hum. Mol. Genet. 18(15):2761–67

Ou SH, Wu F, Harrich D, Garcıa-Martınez LF, Gaynor RB. 1995. Cloning and characterization of a novelcellular protein, TDP-43, that binds to human immunodeficiency virus type 1 TAR DNA sequencemotifs. J. Virol. 69(6):3584–96

Pereson S, Wils H, Kleinberger G, McGowan E, Vandewoestyne M, et al. 2009. Progranulin expression corre-lates with dense-core amyloid plaque burden in Alzheimer disease mouse models. J. Pathol. 219(2):173–81

Pick A. 1892. Uber die beziehungen der senilen hirnatrophie zur aphasie. Prag. Med. Wochenschr. 17:165–67Pickering-Brown SM, Rollinson S, Du Plessis D, Morrison KE, Varma A, et al. 2008. Frequency and clinical

characteristics of progranulin mutation carriers in the Manchester frontotemporal lobar degenerationcohort: comparison with patients with MAPT and no known mutations. Brain 131(Pt. 3):721–31

Plowman GD, Green JM, Neubauer MG, Buckley SD, McDonald VL, et al. 1992. The epithelin precursorencodes two proteins with opposing activities on epithelial cell growth. J. Biol. Chem. 267(18):13073–78

www.annualreviews.org • Frontotemporal Lobar Degeneration 85

Ann

u. R

ev. N

euro

sci.

2010

.33:

71-8

8. D

ownl

oade

d fr

om w

ww

.ann

ualr

evie

ws.

org

by U

nive

rsity

of

Con

nect

icut

on

10/2

9/12

. For

per

sona

l use

onl

y.

Page 16: Molecular Pathways of Frontotemporal Lobar Degeneration

NE33CH04-VanBroeckhoven ARI 14 May 2010 16:24

Rademakers R, Baker M, Gass J, Adamson J, Huey ED, et al. 2007. Phenotypic variability associated withprogranulin haploinsufficiency in patients with the common 1477C → T (Arg493X) mutation: an inter-national initiative. Lancet Neurol. 6(10):857–68

Rademakers R, Cruts M, Dermaut B, Sleegers K, Rosso SM, et al. 2002. Tau negative frontal lobe dementiaat 17q21: significant finemapping of the candidate region to a 4.8 cM interval. Mol. Psychiatry 7(10):1064–74

Rademakers R, Cruts M, Van Broeckhoven C. 2004. The role of tau (MAPT) in frontotemporal dementiaand related tauopathies. Hum. Mutat. 24(4):277–95

Rademakers R, Eriksen JL, Baker M, Robinson T, Ahmed Z, et al. 2008. Common variation in the miR-659binding-site of GRN is a major risk factor for TDP43-positive frontotemporal dementia. Hum. Mol.Genet. 17(23):3631–42

Robinson PN, Booms P, Katzke S, Ladewig M, Neumann L, et al. 2002. Mutations of FBN1 and genotype-phenotype correlations in Marfan syndrome and related fibrillinopathies. Hum. Mutat. 20:153–61

Rollinson S, Rohrer JD, Van Der Zee J, Sleegers K, Mead S, et al. 2010. No association of PGRN 3′UTRrs5848 in frontotemporal lobar degeneration. Neurobiol. Aging. In press

Sampathu DM, Neumann M, Kwong LK, Chou TT, Micsenyi M, et al. 2006. Pathological heterogeneity offrontotemporal lobar degeneration with ubiquitin-positive inclusions delineated by ubiquitin immuno-histochemistry and novel monoclonal antibodies. Am. J. Pathol. 169(4):1343–52

Schymick JC, Yang Y, Andersen PM, Vonsattel JP, Greenway M, et al. 2007. Progranulin mutations andamyotrophic lateral sclerosis or amyotrophic lateral sclerosis-frontotemporal dementia phenotypes.J. Neurol. Neurosurg. Psychiatry 78(7):754–56

Scolari F, Caridi G, Rampoldi L, Tardanico R, Izzi C, et al. 2004. Uromodulin storage diseases: clinical aspectsand mechanisms. Am. J. Kidney Dis. 44:987–99

Shankaran SS, Capell A, Hruscha AT, Fellerer K, Neumann M, et al. 2008. Missense mutations in theprogranulin gene linked to frontotemporal lobar degeneration with ubiquitin-immunoreactive inclusionsreduce progranulin production and secretion. J. Biol. Cell. 283(3):1744–53

Simpson CL, Lemmens R, Miskiewicz K, Broom WJ, Hansen VK, et al. 2008. Variants of the elongatorprotein 3 (ELP3) gene are associated with motor neuron degeneration. Hum. Mol. Genet. 18(3):472–81

Skibinski G, Parkinson NJ, Brown JM, Chakrabarti L, Lloyd SL, et al. 2005. Mutations in the endosomalESCRTIII-complex subunit CHMP2B in frontotemporal dementia. Nat. Genet. 37:806–8

Sleegers K, Brouwers N, Maurer-Stroh S, van Es MA, Van Damme P, et al. 2008. Progranulin geneticvariability contributes to amyotrophic lateral sclerosis. Neurology 71(4):253–59

Sleegers K, Brouwers N, Van Damme P, Engelborghs S, Gijselinck I, et al. 2009. Serum biomarker forprogranulin-associated frontotemporal lobar degeneration. Ann. Neurol. 65(5):603–9

Sleegers K, Van Broeckhoven C. 2009. Motor-neuron disease: rogue gene in the family. Nature 458(7237):415–17

Sreedharan J, Blair IP, Tripathi VB, Hu X, Vance C, et al. 2008. TDP-43 mutations in familial and sporadicamyotrophic lateral sclerosis. Science 319(5870):1668–72

Steinacker P, Hendrich C, Sperfeld AD, Jesse S, von Arnim CA, et al. 2008. TDP-43 in cerebrospinalfluid of patients with frontotemporal lobar degeneration and amyotrophic lateral sclerosis. Arch. Neurol.65(11):1481–87

Steward O, Schuman EM. 2001. Protein synthesis at synaptic sites on dendrites. Annu. Rev. Neurosci. 24:299–325

Tatom JB, Wang DB, Dayton RD, Skalli O, Hutton ML, et al. 2009. Mimicking aspects of frontotemporallobar degeneration and Lou Gehrig’s disease in rats via TDP-43 overexpression. Mol. Ther. 17(4):607–13

Tolkatchev D, Malik S, Vinogradova A, Wang P, Chen Z, et al. 2008. Structure dissection of human pro-granulin identifies well-folded granulin/epithelin modules with unique functional activities. Protein Sci.17(4):711–24

Tolkatchev D, Ng A, Vranken W, Ni F. 2000. Design and solution structure of a well-folded stack of twobeta-hairpins based on the amino-terminal fragment of human granulin A. Biochemistry 39(11):2878–86

86 Sleegers · Cruts · Van Broeckhoven

Ann

u. R

ev. N

euro

sci.

2010

.33:

71-8

8. D

ownl

oade

d fr

om w

ww

.ann

ualr

evie

ws.

org

by U

nive

rsity

of

Con

nect

icut

on

10/2

9/12

. For

per

sona

l use

onl

y.

Page 17: Molecular Pathways of Frontotemporal Lobar Degeneration

NE33CH04-VanBroeckhoven ARI 14 May 2010 16:24

Trinh DP, Brown KM, Jeang KT. 1999. Epithelin/granulin growth factors: extracellular cofactors for HIV-1and HIV-2 Tat proteins. Biochem. Biophys. Res. Commun. 256(2):299–306

Valdmanis PN, Dupre N, Bouchard JP, Camu W, Salachas F, et al. 2007. Three families with amyotrophiclateral sclerosis and frontotemporal dementia with evidence of linkage to chromosome 9p. Arch. Neurol.64(2):240–45

Vance C, Al-Chalabi A, Ruddy D, Smith BN, Hu X, et al. 2006. Familial amyotrophic lateral sclerosis withfrontotemporal dementia is linked to a locus on chromosome 9p13.2–21.3. Brain 129(Pt. 4):868–76

Vance C, Rogelj B, Hortobagyi T, De Vos KJ, Nishimura AL, et al. 2009. Mutations in FUS, an RNAprocessing protein, cause familial amyotrophic lateral sclerosis type 6. Science 323(5918):1208–11

Van Damme P, Van Hoecke A, Lambrechts D, Vanacker P, Bogaert E, et al. 2008. Progranulin functions as aneurotrophic factor to regulate neurite outgrowth and enhance neuronal survival. J. Cell. Biol. 181(1):37–41

Van Deerlin VM, Leverenz JB, Bekris LM, Bird TD, Yuan W, et al. 2008. TARDBP mutations in amyotrophiclateral sclerosis with TDP-43 neuropathology: a genetic and histopathological analysis. Lancet Neurol.7(5):409–16

Van Deerlin VM, Sleiman PMA, Martinez-Lage M, Chen-Plotkin A, Wang L-S, et al. 2010. Commonvariants at 7p21 are associated with frontotemporal lobar degeneration with TDP-43 inclusions. Nat.Genet. 42:234–39

Van Der Zee J, Le Ber I, Maurer-Stroh S, Engelborghs S, Gijselinck I, et al. 2007. Mutations other than nullmutations producing a pathogenic loss of progranulin in frontotemporal dementia. Hum. Mut. 2(4):416

Van Der Zee J, Pirici D, Van Langenhove T, Engelborghs S, Vandenberghe R, et al. 2009. Clinical hetero-geneity in 3 unrelated families linked to VCP p.Arg159His. Neurology 73(8):626–32

Van Der Zee J, Rademakers R, Engelborghs S, Gijselinck I, Bogaerts V, et al. 2006. A Belgian an-cestral haplotype harbours a highly prevalent mutation for 17q21-linked tau-negative FTLD. Brain129(Pt. 4):841–52

Van Der Zee J, Urwin H, Engelborghs S, Bruyland M, Vandenberghe R, et al. 2008. CHMP2B C-truncatingmutations in frontotemporal lobar degeneration are associated with an aberrant endosomal phenotype invitro. Hum. Mol. Genet. 17(2):313–22

Van Langenhove T, van der Zee J, Sleegers K, Engelborghs S, Vandenberghe R, et al. 2010. Genetic contri-bution of FUS to frontotemporal lobar degeneration. Neurology 74(5):366–71

Wang HY, Wang IF, Bose J, Shen CK. 2004. Structural diversity and functional implications of the eukaryoticTDP gene family. Genomics 83(1):130–39

Watts GD, Wymer J, Kovach MJ, Mehta SG, Mumm S, et al. 2004. Inclusion body myopathy associated withPaget disease of bone and frontotemporal dementia is caused by mutant valosin-containing protein. Nat.Genet. 36:377–81

Wegorzewska I, Bell S, Cairns NJ, Miller TM, Baloh RH. 2009. TDP-43 mutant transgenic mice developfeatures of ALS and frontotemporal lobar degeneration. Proc. Natl. Acad. Sci. USA 106(44):18809–14

Whiteman P, Handford PA. 2003. Defective secretion of recombinant fragments of fibrillin-1: implicationsof protein misfolding for the pathogenesis of Marfan syndrome and related disorders. Hum. Mol. Genet.12:727–37

Wider C, Dickson DW, Stoessl AJ, Tsuboi Y, Chapon F, et al. 2009. Pallidonigral TDP-43 pathology inPerry syndrome. Parkinsonism Relat. Disord. 15(4):281–86

Wils H, Kleinberger G, Janssens J, Pereson S, Joris G, et al. 2010. TDP-43 transgenic mice develop spasticparalysis and neuronal inclusions characteristic of ALS and frontotemporal lobar degeneration. Proc. Natl.Acad. Sci. USA 107:3858–63

Winton MJ, Igaz LM, Wong MM, Kwong LK, Trojanowski JQ, Lee VM. 2008a. Disturbance of nuclear andcytoplasmic TAR DNA-binding protein (TDP-43) induces disease-like redistribution, sequestration, andaggregate formation. J. Biol. Chem. 283(19):13302–9

Winton MJ, Van Deerlin VM, Kwong LK, Yuan W, Wood EM, et al. 2008b. A90V TDP-43 variant resultsin the aberrant localization of TDP-43 in vitro. FEBS Lett. 582(15):2252–56

Wolfe MS. 2009. Tau mutations in neurodegenerative diseases. J. Biol. Chem. 284(10):6021–25Zanocco-Marani T, Bateman A, Romano G, Valentinis B, He ZH, Baserga R. 1999. Biological activities and

signaling pathways of the granulin/epithelin precursor. Cancer Res. 59(20):5331–40

www.annualreviews.org • Frontotemporal Lobar Degeneration 87

Ann

u. R

ev. N

euro

sci.

2010

.33:

71-8

8. D

ownl

oade

d fr

om w

ww

.ann

ualr

evie

ws.

org

by U

nive

rsity

of

Con

nect

icut

on

10/2

9/12

. For

per

sona

l use

onl

y.

Page 18: Molecular Pathways of Frontotemporal Lobar Degeneration

NE33CH04-VanBroeckhoven ARI 14 May 2010 16:24

Zhang YJ, Xu YF, Cook C, Gendron TF, Roettges P, et al. 2009. Aberrant cleavage of TDP-43 enhancesaggregation and cellular toxicity. Proc. Natl. Acad. Sci. USA 106(18):7607–12

Zhang YJ, Xu YF, Dickey CA, Buratti E, Baralle F, et al. 2007. Progranulin mediates caspase-dependentcleavage of TAR DNA binding protein-43. J. Neurosci. 27(39):10530–34

Zhu J, Nathan C, Jin W, Sim D, Ashcroft GS, et al. 2002. Conversion of proepithelin to epithelins: roles ofSLPI and elastase in host defense and wound repair. Cell 111(6):867–78

88 Sleegers · Cruts · Van Broeckhoven

Ann

u. R

ev. N

euro

sci.

2010

.33:

71-8

8. D

ownl

oade

d fr

om w

ww

.ann

ualr

evie

ws.

org

by U

nive

rsity

of

Con

nect

icut

on

10/2

9/12

. For

per

sona

l use

onl

y.

Page 19: Molecular Pathways of Frontotemporal Lobar Degeneration

NE33-FM ARI 14 May 2010 19:47

Annual Review ofNeuroscience

Volume 33, 2010Contents

Attention, Intention, and Priority in the Parietal LobeJames W. Bisley and Michael E. Goldberg � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � 1

The Subplate and Early Cortical CircuitsPatrick O. Kanold and Heiko J. Luhmann � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � �23

Fly Motion VisionAlexander Borst, Juergen Haag, and Dierk F. Reiff � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � �49

Molecular Pathways of Frontotemporal Lobar DegenerationKristel Sleegers, Marc Cruts, and Christine Van Broeckhoven � � � � � � � � � � � � � � � � � � � � � � � � � � � � �71

Error Correction, Sensory Prediction, and Adaptationin Motor ControlReza Shadmehr, Maurice A. Smith, and John W. Krakauer � � � � � � � � � � � � � � � � � � � � � � � � � � � � � �89

How Does Neuroscience Affect Our Conception of Volition?Adina L. Roskies � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � 109

Watching Synaptogenesis in the Adult BrainWolfgang Kelsch, Shuyin Sim, and Carlos Lois � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � 131

Neurological ChannelopathiesDimitri M. Kullmann � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � 151

Emotion, Cognition, and Mental State Representation in Amygdalaand Prefrontal CortexC. Daniel Salzman and Stefano Fusi � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � 173

Category Learning in the BrainCarol A. Seger and Earl K. Miller � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � 203

Molecular and Cellular Mechanisms of Learning Disabilities:A Focus on NF1C. Shilyansky, Y.S. Lee, and A.J. Silva � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � 221

Wallerian Degeneration, WldS, and NmnatMichael P. Coleman and Marc R. Freeman � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � 245

v

Ann

u. R

ev. N

euro

sci.

2010

.33:

71-8

8. D

ownl

oade

d fr

om w

ww

.ann

ualr

evie

ws.

org

by U

nive

rsity

of

Con

nect

icut

on

10/2

9/12

. For

per

sona

l use

onl

y.

Page 20: Molecular Pathways of Frontotemporal Lobar Degeneration

NE33-FM ARI 14 May 2010 19:47

Neural Mechanisms for Interacting with a World Fullof Action ChoicesPaul Cisek and John F. Kalaska � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � 269

The Role of the Human Prefrontal Cortex in Social Cognitionand Moral JudgmentChad E. Forbes and Jordan Grafman � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � 299

Sodium Channels in Normal and Pathological PainSulayman D. Dib-Hajj, Theodore R. Cummins, Joel A. Black,and Stephen G. Waxman � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � 325

Mechanisms of Synapse and Dendrite Maintenance and TheirDisruption in Psychiatric and Neurodegenerative DisordersYu-Chih Lin and Anthony J. Koleske � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � 349

Connecting Vascular and Nervous System Development: Angiogenesisand the Blood-Brain BarrierStephen J. Tam and Ryan J. Watts � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � 379

Motor Neuron Diversity in Development and DiseaseKevin C. Kanning, Artem Kaplan, and Christopher E. Henderson � � � � � � � � � � � � � � � � � � � � � � 409

The Genomic, Biochemical, and Cellular Responses of the Retina inInherited Photoreceptor Degenerations and Prospects for theTreatment of These DisordersAlexa N. Bramall, Alan F. Wright, Samuel G. Jacobson, and Roderick R. McInnes � � � 441

Genetics and Cell Biology of Building Specific Synaptic ConnectivityKang Shen and Peter Scheiffele � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � 473

Indexes

Cumulative Index of Contributing Authors, Volumes 24–33 � � � � � � � � � � � � � � � � � � � � � � � � � � � 509

Cumulative Index of Chapter Titles, Volumes 24–33 � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � 513

Errata

An online log of corrections to Annual Review of Neuroscience articles may be found athttp://neuro.annualreviews.org/

vi Contents

Ann

u. R

ev. N

euro

sci.

2010

.33:

71-8

8. D

ownl

oade

d fr

om w

ww

.ann

ualr

evie

ws.

org

by U

nive

rsity

of

Con

nect

icut

on

10/2

9/12

. For

per

sona

l use

onl

y.


Recommended