Você está na página 1de 15

Neurobiology of Aging 34 (2013) 248 –262

www.elsevier.com/locate/neuaging

Rho guanine nucleotide exchange factor is an NFL mRNA destabilizing


factor that forms cytoplasmic inclusions in amyotrophic lateral sclerosis
Cristian A. Droppelmanna, Brian A. Kellera, Danae Campos-Meloa, Kathryn Volkeninga,b,
Michael J. Stronga,b,*
a
Molecular Brain Research Group, Robarts Research Institute, University of Western Ontario, London, Ontario, Canada
b
Department of Clinical Neurological Sciences, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada

Received 6 December 2011; received in revised form 26 May 2012; accepted 24 June 2012

Abstract

Amyotrophic lateral sclerosis (ALS) is an adult-onset progressive disorder of unknown etiology characterized by the selective degen-
eration of motor neurons. Recent evidence supports the hypothesis that alterations in RNA metabolism in motor neurons can explain the
development of protein inclusions, including neurofilamentous aggregates, observed in this pathology. In mice, p190RhoGEF, a guanine
nucleotide exchange factor, is involved in neurofilament protein aggregation in an RNA-triggered transgenic model of motor neuron disease.
Here, we observed that rho guanine nucleotide exchange factor (RGNEF), the human homologue of p190RhoGEF, binds low molecular
weight neurofilament mRNA and affects its stability via 3= untranslated region destabilization. We observed that the overexpression of
RGNEF in a stable cell line significantly decreased the level of low molecular weight neurofilament protein. Furthermore, we observed
RGNEF cytoplasmic inclusions in ALS spinal motor neurons that colocalized with ubiquitin, p62/sequestosome-1, and TAR (trans-active
regulatory) DNA-binding protein 43 (TDP-43). Our results provide further evidence that RNA metabolism pathways are integral to ALS
pathology. This is also the first described link between ALS and an RNA binding protein with aggregate formation that is also a central cell
signaling pathway molecule.
© 2013 Elsevier Inc. All rights reserved.

Keywords: Amyotrophic lateral sclerosis; Rho guanine nucleotide exchange factor; RGNEF; p190RhoGEF; NFL; RNA stability; RNA binding protein;
Protein aggregates

1. Introduction Hays, 2004), TAR DNA binding protein of 43 kDa (TDP-


43) (Arai et al., 2006; Neumann et al., 2006), fused in
Amyotrophic lateral sclerosis (ALS) is an adult-onset
sarcoma/translocated in liposarcoma (FUS/TLS) (Kwiat-
progressive disorder characterized by the selective degen-
kowski et al., 2009; Vance et al., 2009), copper/zinc super-
eration of motor neurons resulting in paralysis and death 3
oxide dismutase 1 (SOD1) (Shaw et al., 2008; Stieber et al.,
to 5 years after onset in most patients (Strong et al., 2005).
There are no treatments that will arrest disease progression 2000), ubiquitin (Arai et al., 2006; Neumann et al., 2006),
and the cause remains elusive. Protein aggregate formation and 14-3-3 (Kawamoto et al., 2004). Additionally, motor
in motor neurons is a neuropathological hallmark of this neurons also show a selective decrease in the levels of
disease. These aggregates contain proteins such as neuro- polyadenylated mRNA, low molecular weight neurofila-
filament (NF) (Kondo et al., 1986), peripherin (He and ment (NFL) mRNA, ␣-internexin mRNA, and peripherin
mRNA (Bergeron et al., 1994; Wong et al., 2000). These
data, together with the participation of TDP-43 and FUS/
* Corresponding author at: Room C7-120, University Hospital, LHSC,
TLS as RNA binding proteins (Buratti et al., 2001; Crozat et
339 Windermere Road, London, Ontario N6A 5A5, Canada. Tel.: ⫹1 519
663 3874; fax: ⫹1 519 663 3609. al., 1993), supports the hypothesis that alterations in RNA
E-mail address: Michael.Strong@schulich.uwo.ca (M.J. Strong). metabolism in motor neurons can lead to the development

0197-4580/$ – see front matter © 2013 Elsevier Inc. All rights reserved.
http://dx.doi.org/10.1016/j.neurobiolaging.2012.06.021
C.A. Droppelmann et al. / Neurobiology of Aging 34 (2013) 248 –262 249

of protein aggregates in ALS (Strong, 2010). This hypoth- 2.2. Anti-RGNEF antibody production
esis has been supported with a recent report showing a
pathological association of an expanded hexanucleotide re- A polyclonal goat antibody against RGNEF that recog-
peat in the noncoding region of C9ORF72 with ALS (Deje- nizes a sequence on the RNA binding domain of the protein
sus-Hernandez et al., 2011; Renton et al., 2011). was produced by Twenty-First Century Biochemicals (Mar-
The NF proteins are a highly conserved family of neu- lborough, MA, USA). The specificity of the antibody was
ronal intermediate filament proteins. There are 3 members analyzed by competition assay with a blocking peptide
of the NF family, a 68-kDa form (NFL), a 160-kDa form identical to the antigenic peptide used to produce the anti-
(middle molecular weight NF), and a 200-kDa form (high body (Supplementary Fig. 1).
molecular weight NF). NF subunits assemble as heteropo- 2.3. RGNEF and p190RhoGEF plasmids
lymers in which the initial polymerization of NFL subunits
is required for normal NF architecture formation (for review The full RGNEF coding sequence (GenBank BC157846)
see Strong, 1999). In transgenic mouse models overexpress- was cloned by polymerase chain reaction (PCR) using Pfu
ing peripherin or high molecular weight NF in which there DNA polymerase (Fermentas Canada, Inc., Burlington,
is targeted disruption of the NFL gene, selective death of Canada). The commercial RGNEF clone MHS1768-
motor neurons is observed, along with motor dysfunction 99865695 (Open Biosystems Lafayette, Colorado, USA)
and the formation of intermediate filament aggregates, in- was used as a template. The RGNEF coding sequence was
cluding intraneuronal NF aggregates (Beaulieu et al., 1999, ligated to pcDNA3.1-myc-His (Invitrogen, Life Technolo-
2000). These results highlight the importance of NF stoi- gies Inc., Burlington, Canada) between the KpnI/XhoI sites
chiometry in motor neurons and the importance of NFL (pcDNA-RGNEF-myc; Supplementary Fig. 2A). A deletion
mRNA stability as a possible cause of the protein aggre- in the 3= end of the RGNEF sequence (from nucleotide 3769
gates observed in ALS (Szaro and Strong, 2010; Thyagara- to stop codon) including the entire region that encodes the
jan et al., 2007). RNA binding domain was generated. The product was also
In mice, it has been observed that p190RhoGEF, a gua- incorporated into pcDNA3.1-myc-His between the KpnI/
nine nucleotide exchange factor (GEF) (van Horck et al., XhoI sites to create pcDNA-RGNEF-⌬CT-myc (Supple-
2001) interacts with a destabilizing region of NFL mRNA mentary Fig. 2B). In addition, a fragment of an alternatively
providing stability to the transcript (Cañete-Soler et al., spliced form of RGNEF with a shorter RNA binding do-
2001). This protein is involved in NF protein aggregation main lacking exons 29 and 33 (BD-⌬exon29/33) was cloned
observed in an RNA-triggered transgenic model of motor from HEK293T cDNA using primers that recognize coding
neuron disease (Lin et al., 2005; Nie et al., 2002). Previ- exons 25 to 34 of RGNEF. To generate glutathione S-trans-
ously, we showed that the RNA binding domain of the ferase (GST) fusion proteins, the sequences corresponding
human homologue of p190RhoGEF, called rho guanine to the RNA binding domain of RGNEF (RGNEF-BD) and
nucleotide exchange factor (RGNEF), can interact with BD-⌬exon29/33 were cloned into pGEX-5X-3 (GE Health-
NFL mRNA (Volkening et al., 2010). Here, we show that care Life Sciences, Baie d’Urfe, Quebec, Canada) between
full length RGNEF is an RNA binding protein that acts as EcoRI/XhoI sites (Supplementary Fig. 2C and D). The plas-
an NFL mRNA stability factor via 3= untranslated region mid pcDNA-p190RhoGEF was kindly provided by Dr.
(UTR) destabilization and reduces NFL protein levels when Wouter H. Moolenaar (Amsterdam, Netherlands).
overexpressed in HEK293T cells. Moreover, we show that
RGNEF also presents a pathogenic phenotype including 2.4. RGNEF stable cell line
aggregate formation in motor neurons of ALS patients. A stable cell line of HEK293T cells expressing full
length RGNEF-myc (HEK293T-RGNEF(⫹)) was created
using the Jump-in Fast Gateway Targeted Integration Sys-
2. Methods tem (Invitrogen) according to the manufacturer’s instruc-
tions. This technology uses PhiC31 integrase-mediated recom-
2.1. Cases bination to stably integrate DNA sequences of choice at
Postmortem frozen and formalin-fixed, paraffin-embed- specific genomic locations called pseudo-attP sites in mamma-
ded tissues were collected as part of the ALS protocol at lian cells (Thyagarajan et al., 2001,Thyagarajan et al. 2008).
London Health Sciences Centre (London, Ontario, Canada). The cells, with the gene of interest successfully incorporated
Six control cases (4 male, 2 female, age range 61–74 years), into their genome, were selected using hygromycin B.
13 sporadic ALS (sALS; 8 male, 5 female, age range 45– 80
2.5. Western blots and immunocytochemistry
years) cases, and 3 familial ALS (fALS; 2 male, 1 female,
age range 56 –71 years) cases without known mutations in HEK293T cells were transfected with pcDNA-RGNEF-
the coding sequence of SOD1, FUS/TLS, and TDP-43 and myc, pcDNA-RGNEF-⌬CT-myc, or pcDNA3.1-myc-His
without C9ORF72 expanded repeats were used in these (control) using Lipofectamine 2000 (Invitrogen) according
studies. to the manufacturer’s protocols. After 24 hours, the cells
250 C.A. Droppelmann et al. / Neurobiology of Aging 34 (2013) 248 –262

were lysed in NP-40 buffer and SDS-PAGE was performed 2.8. RNA isolation, relative quantitative PCR
using 100 ␮g of protein extracts and then transferred to a and RT-PCR
nitrocellulose membrane. Mouse anti-myc (1:2500; Cedar-
Total RNA from transfected HEK293T cells was isolated
lane Laboratories, Burlington, NC, USA), rabbit anti-NFL
using Trizol reagent (Invitrogen). Reverse transcription was
(1:1000; Invitrogen), mouse anti-actin (1:400; Santa Cruz
performed using the Superscript II reverse transcriptase
Biotechnologies, Santa Cruz, CA, USA), and rabbit anti-
system (Invitrogen). Relative quantitative RT-PCR for lu-
GAPDH (1:2500; Abcam, Cambridge, UK) antibodies were
ciferase mRNA and NFL mRNA stability experiments were
used for Western blotting. For immunocytochemistry,
performed by coamplifying firefly luciferase and renilla
HEK293T-RGNEF(⫹) cells were fixed in 4% paraformal- luciferase or RGNEF and 18S (as internal standard) during
dehyde for 15 minutes and washed with phosphate-buffered the linear range of both reactions. Every experiment was
saline (PBS). Cells were incubated with mouse anti-myc performed in triplicate. Both luciferases are expressed from
(1:1000; Cedarlane Laboratories) for 1 hour at room tem- the same vector (pmirGLO vector; Promega, Madison, WI,
perature and then with anti-mouse Alexa Fluor 543 (1:1000; USA) and renilla luciferase works as the control for trans-
Invitrogen). Finally, the cells were incubated with Hoechst fection efficiency. The experiment and data analysis for the
to visualize nuclei. luciferase mRNA stability experiment was performed in the
same way as the luciferase reporter assays. For the detection
2.6. GST fused protein purification of NFL mRNA in HEK293T cells and RNA- immunopre-
cipitation experiments, standard RT-PCR protocols were
GST-RGNEF-BD, GST-BD-⌬exon29/33, and GST pro-
followed without quantification. PCR primers for firefly
teins were purified from BL21DE3 E. coli (Invitrogen)
luciferase were forward 5=-CAA GAC TAT AAG ATT
using the GSTrap 4B purification system (GE Healthcare
CAA TCT GCC CTG CTG-3= and reverse 5=-GAT GTT
Life Sciences) according to the manufacturer’s instructions.
GGG GTG TTG CAG CAG GAT-3= (Amplicon: 561 base
The purified protein was confirmed by SDS-PAGE stained
pair [bp]). PCR primers for renilla luciferase were forward
with Coomassie blue and verified by molecular size by
5=-GAG CAA CGC AAA CGC ATG ATC ACT G-3= and
comparison with molecular weight standards. reverse 5=-TTC AGC AGC TCG AAC CAA GCG GT-3=
(Amplicon: 302 bp). PCR primers for NFL were forward
2.7. RNA-immunoprecipitation-PCR (RNA-IP-PCR) 5=-CAA GAC CCT GGA AAT CGA AG-3= and reverse
HEK293T cells were transfected with pcDNA-RGNEF- 5=-TCT TGG ACA TGG CTG GTG TA-3= (Amplicon: 405
myc, pcDNA-RGNEF-⌬CT-myc, or pcDNA3.1-myc-His bp) and PCR. primers for 18S were forward 5= AGT TGG
(control) vector using Lipofectamine 2000 (Invitrogen). Af- TGG AGC GAT TTG TC3= and reverse 5= TTC CTC GTT
ter 24 hours, whole cell homogenates were prepared using CAT GGG GAA TA3= (Amplicon: 297 bp.).
NP-40 buffer with proteinase inhibitors (Complete Protease
2.9. GEF activity
Inhibitor; Roche, Mississauga, Ontario, Canada) and RNase
inhibitors (RNAaseOUT; Invitrogen). Cell lysate (1 mg) The GEF activity in HEK293T cells transfected with
was mixed with 10 ␮L Pansorbin (Calbiochem-Millepore, pcDNA-RGNEF-myc and HEK293T transfected with
Billerica, MA, USA) and 1 ␮g of antibody (mouse anti-myc pcDNA3.1-myc-His (control) cells was determined using the
or IgG as control) and incubated for 1 hour at room tem- “Rho Activation Assay Biochem Kit” (Cytoskeleton Inc., Den-
perature. After centrifugation at 10,000g, pellets were ver, CO, USA) according to the manufacturer’s instructions.
washed 3 times in 1 mL cold RNase-free NP-40 buffer (PBS
containing 1% NP-40 and proteinase inhibitors and RNase 2.10. Luciferase reporter assay
inhibitors as above), and then 3 times in cold PBS (with The reporter plasmid was constructed by inserting the 3=
proteinase inhibitors and RNase inhibitors as above). The UTR of human or mouse NFL mRNA between the NheI/
RNA was resuspended in 40 ␮L of nuclease-free water and SalI sites downstream of the firefly luciferase coding se-
then reverse transcription (RT)-PCR was performed to de- quence in the pmirGLO vector after PCR amplification
tect immunoprecipitated NFL mRNA. Alternatively, RNA- using Pfu DNA polymerase. Five constructs were made:
IP-PCR was performed by mixing 2 ␮g of purified GST- pmirGLO-mouse NFL 3= UTR (GenBank NM_010910;
RGNEF-BD, GST-BD-⌬exon29/33, or GST protein and 2 bases 1–286); pmirGLO-NFL 3= UTR-Short (UTR-S; en-
␮g of proteinase K-treated total RNA isolated from control coding a segment of the human NFL 3= UTR analogous to
and ALS patients using anti-GST antibody (Amersham-GE the murine NFL 3= UTR; bases 1–286 of human 3= UTR);
Healthcare Life Sciences). For all experiments, a negative pmirGLO-NFL 3= UTR-Medium (UTR-M; GenBank;
RT reaction (without reverse transcriptase) and negative BC039237; bases 1–1380); pmirGLO-NFL 3= UTR-Long
PCR (without cDNA template) were used as negative con- (UTR-L; GenBank; NM_006158; bases 1–1838); and pmir-
trols. HEK293T cDNA from total RNA was used as tem- GLO-NFL 3= UTR M⌬S (3= UTR-M with bases 1–286
plate for positive controls. deleted, resulting in a sequence of 1094 bp) (Supplementary
C.A. Droppelmann et al. / Neurobiology of Aging 34 (2013) 248 –262 251

Fig. 3). HEK293T or Neuro2A cells were plated 24 hours (1:100), mouse anti-ubiquitin (1:5000; Chemicon, Temecula,
prior to transfection in 96-well plates at 9 ⫻ 103 cells per CA, USA), mouse anti-TDP-43 (1:500; Abnova, Walnut, CA,
well. Transfections were performed with Lipofectamine USA), rabbit anti-p62/Sequestosome-1 (1:1000; Enzo Life Sci-
2000 according to the manufacturer’s instructions. Cells ences, Brockville, Ontario, Canada), and rabbit anti-14-3-3
were cotransfected with 2.83 fmol of pcDNA-RGNEF- (1:250; Santa Cruz Biotechnology) were used as primary an-
myc, pcDNA-p190RhoGEF, pcDNA-RGNEF-⌬CT-myc, or tibodies. Anti-RGNEF was detected using secondary anti-
pcDNA-TDP-43 (Strong et al., 2007) and 3.47 fmol of body conjugated to Alexa Fluor 488 (1:500; Invitrogen).
pmirGLO with the corresponding NFL 3= UTR. Luciferase Anti-TDP-43, anti-p62/Sequestosome-1, anti-ubiquitin, and
activity was measured 24 hours after transfection using the anti-14-3-3 were detected using secondary antibody conju-
Dual-Glo Luciferase Assay System (Promega) in a Turner gated to Alexa Fluor 546 (1:500; Invitrogen). Slides were
Biosystems Luminometer Modulus (Promega). The pmir- viewed with a Nikon 510 Confocal microscope and local-
GLO vector expresses both luciferases with different pro- ization determined by capture and examination of single
moters eliminating the necessity of transfecting 2 vectors plane images in sequential mode for colocalization.
for normalizing luciferase activity. Thus firefly luciferase
2.13. Colocalization images and schematic diagram
activity was normalized to renilla luciferase activity to ad-
of proteins
just for variations in transfection efficiency among experi-
ments. Data were also normalized to the RGNEF, An intensity correlation analysis (Li et al., 2004) using
p190RhoGEF, or TDP-43 effect on pmirGLO without NFL ImageJ software was performed for the colocalization im-
3= UTR (empty vector) to obtain the specific effect over ages. The colocalized pixels are shown as product of the
NFL 3= UTR. To present destabilization as negative values differences from the mean (PDM) images. PDM ⫽ (red
and stabilization as positive values, the data were presented intensity⫺mean red intensity) ⫻ (green intensity⫺mean
as 1-([NFL*]/[Vector*]), where: [NFL*] ⫽ (normalized green intensity). In the colocalization images blue color
luciferase activity of pmirGLO coupled NFL 3= UTR indicates a low level of colocalization while yellow and
alone)/(normalized luciferase activity pmirGLO-NFL cou- white indicate a high degree of colocalization. The sche-
pled NFL 3= UTR in presence of protein) and [Vector*] ⫽ matic representation of proteins was performed using the
(normalized luciferase activity pmirGLO alone)/(normal- Dog 2.0 software (Ren et al., 2009).
ized luciferase activity pmirGLO in presence of protein).
Each experiment was performed in triplicate. 2.14. Statistics
For the luciferase reporter assays experiments and NFL
2.11. siRNA experiments
protein quantification, analysis of variance with Newman–
An small inhibitory RNA (siRNA) against RGNEF Keuls post hoc test was performed. The results are presented
(SASI_HS01_00264015; Sigma) was transfected in HEK293T- as mean ⫾ standard error of the mean and p values corre-
RGNEF(⫹) cells using Lipofectamine 2000 according to spond to comparison with the control. Student t tests were
the manufacturer’s instructions. To maintain the siRNA performed for GEF activity analysis, the effect of RGNEF
expression for 7 days, 3 consecutive transfections were done over firefly luciferase mRNA and the NFL mRNA levels in
before lysing the cells and performing the Western blot. In HEK293T-RGNEF(⫹). Data are presented as mean ⫾ stan-
parallel, as control, HEK293T cells and HEK293T-RG- dard error of the mean and the p values correspond to the
NEF(⫹) cells were transfected using a control siRNA comparison with the control. A value of p ⬍ 0.05 was
(Misssion siRNA Universal Negative Control No. 1; Sigma, considered statistically significant for all experiments.
Oakville, Ontario, Canada). Each experiment was per-
formed in triplicate. 3. Results
2.12. Immunohistochemistry 3.1. Human RGNEF shows a high similarity with
murine p190RhoGEF
Archival paraffin-embedded sections of lumbar spinal
cord from control and ALS cases were serially sectioned at An amino acid (aa) sequence comparison analysis was
6-␮m thickness. For 3,4 diaminobenzide (DAB)-immuno- performed to determine the similarity between the murine
histochemistry, tissue sections were deparaffinized using p190RhoGEF (GenBank U73199) and its human homolog,
standard protocols and then immunolabeled using a goat RGNEF (GenBank BC157846). These proteins share the
anti-RGNEF (1:500; optimum pH: 7.4) antibody. The anti- same conserved domains (Fig. 1A). After Basic Local
gen–antibody complex was visualized using the Vectastain Alignment Search Tool analysis (BLAST utility; blast.
Elite ABC kit (Vector, Burlington, Ontario, Canada) and an ncbi.nlm.nih.gov), we observed an 80% identity between
Olympus BX45 microscope with Image-Pro Plus software, p190RhoGEF and RGNEF at the protein level. The GEF
version 7 (Leeds, Minneapolis, MN, USA). For confocal domain (mouse: aa 847–1039; human: aa 850 –1042 accord-
immunofluorescence microscopy the sections were simi- ing the conserved domains utility; www.ncbi.nlm.nih.gov/
larly processed but double-labeled. Goat anti-RGNEF Structure/cdd/wrpsb.cgi) showed 92% identity (Fig. 1B).
252 C.A. Droppelmann et al. / Neurobiology of Aging 34 (2013) 248 –262

Fig. 1. Mouse p190RhoGEF and human rho guanine nucleotide exchange factor (RGNEF) show a high level of identity in their sequences. (A) Schematic
of the conserved domains shared between p190RhoGEF and RGNEF. Domains include a cystein rich zinc finger domain (yellow), RhoGEF domain (blue),
Pleckstrin homology (PH) domain (green), and RNA binding domain (red). These proteins share the same conserved domains. (B) Alignment of the GEF
domain (between brackets) of RGNEF (human; H) and p190RhoGEF (mouse; M). Conserved amino acids are shown in blue. (C) Alignment of the RNA
binding domain (between brackets) of RGNEF (human; H) and p190RhoGEF (mouse; M). Conserved amino acids are shown in red.

The RNA binding domain (mouse: aa 1254 –1610; Cañete- mRNA (Fig. 2A, lane 3). The same was observed for control
Soler et al., 2001; human: aa 1257–1614; Volkening et al., cells (transfected with the empty vector; Fig. 2A, lane 1)
2010) showed 76.2% identity (Fig. 1C). These results show and cells expressing RGNEF-myc or RGNEF-⌬CT-myc
a great similarity in the functional domains of p190RhoGEF with control IgG IP (Fig. 2A, lanes 4 and 5). To evaluate if
and RGNEF; thus, it is reasonable to predict similar func- this interaction was direct or mediated by other proteins, we
tional properties of both proteins. performed RNA-IP using purified GST-RGNEF-BD (Sup-
plementary Fig. 2C and Supplementary Fig 3D) and purified
3.2. RGNEF binds to NFL mRNA
proteinase K-treated total RNA from spinal cord of control,
We previously showed that the RNA binding domain of sALS and fALS patients. Purified GST-BD-⌬exon29/33,
RGNEF (RGNEF-BD) interacts with NFL mRNA (Volk- which expresses a GST fusion protein with a shorter RNA
ening et al., 2010). Here, we analyzed the interaction of the binding domain but with an equivalent mass than GST-
full length RGNEF protein with human NFL transcript. Full RGNEF-BD (Supplementary Fig. 2B and Supplementary
length myc-tagged RGNEF was transiently expressed in Fig 3D), and purified GST alone, were used to confirm
HEK293T cells (Supplementary Figs. 2A andSupplemen- specificity. We observed interaction only with GST-
tary Fig 4A) and RNA-IP-PCR was performed to detect RGNEF-BD in control, sALS, and fALS patients (Fig. 2B).
interaction with the NFL mRNA that is endogenously ex- These results clearly show that full length RGNEF interacts
pressed in these HEK293T cells (Supplementary Fig. 4B). with NFL mRNA and suggest that the basic interaction of
An RGNEF version without the RNA binding domain this mRNA with the RNA binding domain of RGNEF does
(RGNEF-⌬CT-myc) (Supplementary Figs. 2B and Supple- not appear to require any intermediate proteins.
mentary Fig 4C) was used as a control for interaction 3.3. RGNEF has GEF activity and acts as an NFL mRNA
specificity. Our results showed specific interaction of full destabilizing factor
length RGNEF (containing the RNA binding domain) with
NFL mRNA in transfected cells using an anti-myc antibody To analyze the cellular roles of RGNEF, we examined
IP (Fig. 2A, lane 2), while cells transiently expressing whether RGNEF exhibits GEF activity as well as whether it
RGNEF-⌬CT-myc do not show any interaction with NFL can act as a human NFL mRNA stability factor. As RGNEF
C.A. Droppelmann et al. / Neurobiology of Aging 34 (2013) 248 –262 253

Fig. 2. Rho guanine nucleotide exchange factor (RGNEF) binds to neurofilament (NFL) mRNA. (A) RNA-immunoprecipitation-polymerase chain reaction
(RNA-IP-PCR) experiment analyzing the interaction of full length RGNEF (RGNEF-myc) and RGNEF-⌬CT-myc (RGNEF lacking RNA binding domain),
with endogenous NFL mRNA in HEK293T cells. Lysates of cells transfected with plasmids pcDNA-RGNEF-myc, pcDNA-RGNEF-⌬CT-myc and
pcDNA3.1-myc-His (control) were immunoprecipitated using anti-myc antibody and IgG (control). NFL mRNA was detected in the immunoprecipitated
pellets by RT-PCR. Interaction was only observed in cells expressing RGNEF-myc (lane 2). There was no interaction detected with RGNEF-⌬CT-myc (lane
3), empty vector (lane 1) or in cells expressing RGNEF-myc and BD-⌬exon-myc and immunoprecipitated with IgG (lanes 4, 5). RT-PCR reaction controls
were no RT negative control, no cDNA negative control, and positive control using HEK293T cDNA. (B) RNA-IP-PCR experiment analyzing the interaction
of purified GST-RGNEF-BD, GST-BD-⌬exon29/33 and GST with NFL mRNA using purified total RNA obtained from control, sporadic amyotrophic lateral
sclerosis (sALS) and familial amyotrophic lateral sclerosis (fALS) spinal cords. Anti-GST antibody was used for the immunoprecipitation and NFL mRNA
was detected in the pellet by RT-PCR. Interaction was observed with the complete RNA binding domain of RGNEF (GST-RGNEF-BD) but not with GST
alone or GST-BD-⌬exon29/33. RT-PCR reaction controls were no RT negative control, no cDNA negative control, and positive control using HEK293T
cDNA.

has a conserved GEF domain, we predicted that RGNEF ciferases’ mRNAs, in HEK293T cells cotransfected with
would have the capacity to activate RhoA protein as GEF pcDNA-RGNEF-myc and pmirGLO fused to NFL 3= UTR
activity in the cell is required for activation of RhoA (Nal- constructs (3= UTR-S: ⫺0.0834 ⫾ 0.0194; 3= UTR-M:
bant et al., 2009). We used a RhoA activation assay that ⫺0.1027 ⫾ 0.0174; 3= UTR-L: ⫺0.1333 ⫾ 0.0429: 3=
determines the amount of activated RhoA (GTP-RhoA) in a UTR-M⌬S: ⫺0.1827 ⫾ 0.0383; all p ⬍ 0.05; negative
cellular lysate based on the capacity of GTP-RhoA to bind values indicate downregulation; Fig. 4A). To determine if
to Rhotekin binding domain (RBD) (Ren et al., 1999). We the presence of the RNA binding domain is critical in the
found that HEK293T cells transiently expressing RGNEF- observed effect over the NFL 3= UTR, a cotransfection of
myc showed increased GTP-RhoA interaction with RBD, pcDNA-RGNEF-⌬CT-myc and pmirGLO fused to NFL 3=
and consequently, more GEF activity, than control cells UTR (Short, Medium, or Long) was performed. Using this
transfected with the empty vector (3.4-fold; p ⬍ 0.05; Fig. mutant RGNEF, the downregulating effect previously ob-
3). These data show that RGNEF has the ability to function served with the full-length RGNEF was completely abol-
as a GEF protein as would be predicted. ished, and an upregulation of luciferase activity could even
Next, we evaluated the effect of RGNEF on the stability be observed (Supplementary Fig. 5). For a control of the
of human NFL via the 3= UTR by using a firefly luciferase model, we examined the interaction of TDP-43 with human
reporter gene coupled to the 3= UTR of NFL mRNA. The NFL-3= UTR as we previously described (Strong et al.,
pmirGLO system was used so that the 2 luciferases (firefly 2007). HEK293T cells were cotransfected with pmirGLO-
and renilla) were expressed from 1 construct avoiding ex- NFL 3= UTR-S and pcDNA-TDP-43 and an increase in the
pression alterations with transfection efficiency variability. luciferase activity was detected (0.0656 ⫾ 0.0095, p ⬍
Four constructs were screened, pmirGLO-NFL 3= UTR-S 0.05; positive value indicates upregulation; Fig. 4A), sug-
(286 bp), pmirGLO-NFL 3= UTR-M (1380 bp), pmirGLO- gesting stabilization of the NFL 3= UTR-S, which is con-
NFL 3= UTR-L (1838 bp) and pmirGLO-NFL 3= UTR- sistent with our previous reports (Strong et al., 2007; Volk-
M⌬Short (3= UTR-M⌬S; 1094 bp). The purpose of 3= UTR- ening et al., 2009).
M⌬S construct was to eliminate putative determinants of Because of the apparent contradiction with the data re-
stabilization that could be present in the 3= UTR-S (Cañete- garding mouse p190RhoGEF, which was found to stabilize
Soler et al., 1998). There was specific downregulation of NFL mRNA (Cañete-Soler et al., 2001), we decided to
luciferase activity, suggesting destabilization of the lu- evaluate the effect of p190RhoGEF over the luciferase re-
254 C.A. Droppelmann et al. / Neurobiology of Aging 34 (2013) 248 –262

Fig. 3. Rho guanine nucleotide exchange factor (RGNEF) shows guanine nucleotide exchange factor (GEF) activity in HEK293T cells. (A) The GEF activity
in HEK293T transiently expressing RGNEF-myc was evaluated using the capacity of activated RhoA (RhoA-GTP) to bind to Rhotekin binding domain
(RBD). A pull-down experiment using RBD-agarose beads in lysates of cells transfected with pcDNA-RGNEF-myc (RGNEF) and pcDNA3.1-myc-His
(control) was performed. The upper panel of (A) shows the presence of RGNEF in cells transfected with pcDNA-RGNEF-myc to confirm expression. In the
middle panel (pull down experiment), cells overexpressing RGNEF show a higher amount of active RhoA than the empty vector control. The lower panel
shows the input control for the experiment (1/6 of the protein amount used in the pull-down experiment). (B) Quantification of 3 GEF activity experiments
calculated as the RhoA in the pull-down against the input amount of RhoA. The results are presented as mean ⫾ standard error of the mean (* ⫽ p ⬍ 0.05).

porter gene coupled to mouse NFL 3= UTR in our experi- aimed to determine whether RGNEF acts analogously to
mental model. We observed an upregulation of the lucifer- p190RhoGEF and therefore has a heterologous effect on the
ase activity, suggesting stabilization of the mRNA, in mouse NFL 3= UTR. We observed that RGNEF can down-
HEK293T cells cotransfected with pcDNA-p190RhoGEF regulate the luciferase reporter gene coupled to mouse NFL
and pmirGLO-mouse NFL 3= UTR (0.0922 ⫾ 0.0524, p ⬍ 3= UTR (⫺0.1059 ⫾ 0.0563; p ⬍ 0.01; Fig. 4B). As a
0.05; Fig. 4A), totally in agreement with the published data. control, we evaluated the effect of p190RhoGEF on the
Interestingly, when we performed the same experiment us- mouse 3= UTR. As expected, we observed upregulation of
ing pmirGLO-NFL 3= UTR-S (91% of identity with mouse luciferase activity (0.1057 ⫾ 0.0556; p ⬍ 0.01; Fig. 4B).
NFL 3= UTR) the result was practically identical (0.0971 ⫾ These results support the above conclusion about the intrin-
0.0532; p ⬍ 0.05; Fig. 4A). These results suggest that the sic properties of these proteins, demonstrated that the de-
destabilizing and stabilizing capacity of RGNEF and stabilizing effect of RGNEF can be observed in neuronal
p190RhoGEF respectively are intrinsic properties of these cell lines, and that RGNEF can act in a heterologous way.
proteins. The decrease in luciferase activity in the reporter gene
Because we conducted the experiments in a nonneural assay and the interaction of RGNEF with NFL mRNA both
cell line, we used mouse Neuro2A cells to evaluate whether suggest, but do not prove, a direct effect of RGNEF on NFL
the RGNEF effect over the luciferase coupled to NFL 3= mRNA stability. To clarify this, we measured the effect of
UTR could be observed in a neuronal cell line. Further, we RGNEF on the levels of firefly luciferase coupled NFL 3=
C.A. Droppelmann et al. / Neurobiology of Aging 34 (2013) 248 –262 255

Fig. 4. Rho guanine nucleotide exchange factor (RGNEF) acts as a destabilizing factor of neurofilament (NFL) mRNA. (A) Reporter gene assay showing
decreased normalized luciferase activity (see Methods) in HEK293T cells cotransfected with pcDNA-RGNEF-myc and pmirGLO linked to 1 of 4 different
lengths of human 3= untranslated region (UTR) NFL mRNA (S ⫽ 3= UTR Short; M ⫽ 3= UTR Medium; L ⫽ 3= UTR Long; M⌬S ⫽ 3= UTR M⌬S). Also,
we observed an increase in the normalized luciferase activity in cells cotransfected with pcDNA- TAR DNA binding protein of 43 kDa (TDP-43) and
pmirGLO NFL 3= UTR-S or with pcDNA-p190RhoGEF and pmirGLO-mouse NFL 3= UTR or pmirGLO NFL 3= UTR-S. p values (* p ⬍ 0.05; ** p ⬍ 0.01;
*** p ⬍ 0.001) refer to comparison with control experiment (luciferase activity of the vector without NFL 3= UTR in presence of RGNEF-myc or
p190RhoGEF or TDP-43). (B) Reporter gene assay showing decreased normalized luciferase activity in Neuro2A cell cotransfected with pcDNA-RGNEF-
myc and pmirGLO-mouse NFL 3= UTR and increased luciferase activity in cells cotransfected with pcDNA-p190RhoGEF and pmirGLO-mouse NFL 3=
UTR. p values (** p ⬍ 0.01) refer to comparison to control experiment (luciferase activity of the vector without NFL 3= UTR in presence of RGNEF-myc
or p190RhoGEF). (C) The level of normalized luciferase mRNA (see Methods) in HEK293T cells cotransfected with pmirGLO-NFL 3= UTR-M⌬S and
pcDNA-RGNEF-myc. We observed a significant reduction of luciferase-3= UTR-M⌬S mRNA in the presence of RGNEF-myc. p value (** p ⬍ 0.01) refer
to comparison with control experiment (luciferase mRNA levels of the vector without NFL 3= UTR in presence of RGNEF-myc).
256 C.A. Droppelmann et al. / Neurobiology of Aging 34 (2013) 248 –262

UTR-M⌬S mRNA. We used the 3= UTR-M⌬S construct active aggregates were observed: skein-like (Fig. 6B and D)
because it showed the highest reduction in luciferase activ- and punctate (Fig. 6C and E). There was no apparent rela-
ity (Fig. 4A), allowing easier detection of variation in the tionship between the type of aggregate observed and ALS
firefly luciferase mRNA levels. We observed that in the variant. When quantification was performed comparing
presence of RGNEF, there was a significant reduction in RGNEF and TDP-43 aggregates there was no statistically
the mRNA levels of luciferase-coupled NFL 3= UTR-M⌬S significant difference between the number of RGNEF and
(⫺0.3465 ⫾ 0.1116, p ⬍ 0.01; Fig. 4C). TDP-43 protein inclusions in ALS patients (Table 1). These
These results clearly show that RGNEF acts as an NFL results strongly suggest a pathogenic phenotype for RGNEF
mRNA destabilizing factor through a seemingly direct ef- in ALS motor neurons as has been postulated for TDP-43.
fect on the NFL 3= UTR. Taken together, RGNEF functions
as a GEF protein and it can also act as an mRNA binding 3.6. RGNEF colocalizes with ubiquitin, p62/
protein, imparting instability to human NFL mRNA. Sequestosome-1, and TDP-43 in motor neurons from
ALS patients
3.4. NFL protein levels are decreased in a stable cell line
overexpressing full length RGNEF To determine if RGNEF could coaggregate with other
proteins known to be present in pathogenic aggregates, we
To evaluate the NFL mRNA destabilizing activity evaluated the colocalization of RGNEF with ubiquitin,
of RGNEF on NFL protein levels, we created the TDP-43 and p62/Sequestosome-1 (a marker of neurodegen-
HEK293T-RGNEF(⫹) stable cell line that overexpresses eration that participates in the ubiquitin pathway) (Kuusisto
full length RGNEF-myc (Fig. 5A). We obtained a clone et al., 2001, 2008; Pikkarainen et al., 2008, 2010; Seppänen
with a highly significant decrease in NFL protein levels et al., 2009). We observed a high degree of colocalization
(18.28-fold; p ⬍ 0.001) (Fig. 5B and C). This effect over between RGNEF and ubiquitin in ALS patients (Fig. 7A).
NFL protein levels was seen in other clones generated by The same was observed for RGNEF and p62/Sequesto-
the same manner (Supplementary Fig. 6). To determine some-1 (Fig. 7B). In control motor neurons, RGNEF did not
whether the decrease of NFL protein levels was due to an colocalize with ubiquitin and p62/Sequestosome-1 (Fig. 7A
effect of RGNEF on the NFL mRNA levels, we analyzed and B). Some RGNEF inclusions colocalized with TDP-43
the NFL mRNA levels in HEK293T-RGNEF(⫹) cells. We in ALS, while others did not (Fig. 7C). Interestingly, a weak
observed a significant decrease of the NFL mRNA levels in but consistent colocalization of RGNEF and TDP-43 was also
this cell line (Fig. 5D). observed in the nucleus of healthy motor neurons (Fig. 7C).
To evaluate if the decrease of NFL proteins levels was Because 14-3-3 is known to interact with p190RhoGEF (Zhai
caused specifically by RGNEF, we performed a rescue exper- et al., 2001) and NFL mRNA (Ge et al., 2007), we aimed to
iment using a specific siRNA against RGNEF. After treating determine whether it colocalizes with RGNEF. We observed a
HEK293T-RGNEF(⫹) cells with the siRNA against RGNEF, very weak colocalization of 14-3-3 with RGNEF aggregates in
we observed a significant increase of NFL proteins levels the ALS cases that were analyzed (Supplementary Fig. 7).
together with a decrease of RGNEF protein levels due to the However we did observe colocalization between RGNEF and
siRNA (Fig. 5E and F). The percentage of NFL protein recov- 14-3-3 in Nissl body-like structures, where RGNEF is present
ered after the siRNA treatment is in agreement with the pre- in some motor neurons of control patients (Supplementary
viously reported recovery rate of NFL protein levels observed Fig. 7).
after an inducible shutdown of its expression (Millecamps et When coupled with data suggesting that the presence of
al., 2007). aggregates is pathological, particularly those that are
These results clearly indicate that the NFL mRNA de- TDP-43 and/or FUS/TLS immunoreactive (Arai et al.,
stabilizing activity of RGNEF can impact NFL protein lev- 2006; Deng et al., 2010; Kwiatkowski et al., 2009; Neu-
els, suggesting that RGNEF may have an important impact mann et al., 2006; Vance et al., 2009), these results strongly
on NF homeostasis in motor neurons. suggest that RGNEF aggregates observed in ALS are also
pathological. Further, this suggests that a common pathway
3.5. RGNEF forms cytoplasmic inclusions in motor of aggregate formation may exist in ALS-affected motor
neurons from ALS patients neurons.
To analyze the intracellular localization of RGNEF, we
examined the presence of this protein in motor neurons of
4. Discussion
ALS and control spinal cords by immunohistochemistry.
We observed homogeneous cytoplasmic RGNEF immuno- The major finding in this study is that RGNEF, the
reactivity in spinal motor neurons of control tissue (Fig. human homolog of the mouse protein p190RhoGEF, is a
6A). In contrast, intensely immunoreactive intracytoplasmic bifunctional protein that appears to be involved in the pa-
accumulations of RGNEF were clearly observed in every thology of ALS. This manifests as aggregates and skeins
case of ALS examined, regardless of ALS variant (sALS or that colocalize with other proteins that have been described
fALS). Two morphological variants of RGNEF-immunore- as pathological in ALS (TDP-43, ubiquitin, p62/Sequesto-
C.A. Droppelmann et al. / Neurobiology of Aging 34 (2013) 248 –262 257

Fig. 5. Neurofilament (NFL) protein levels are decreased in a stable cell line expressing full length RGNEF. (A) Rho guanine nucleotide exchange factor
(RGNEF)-myc was detected using an anti-myc antibody in the stable cell line HEK293-RGNEF(⫹) and observed through confocal microscopy. RGNEF (red)
was localized almost exclusively in the cytoplasm, however, a very low amount of RGNEF was observed in the nucleus. Hoechst (blue) was used as a nuclear
marker (scale bar ⫽ 5 ␮m). (B) The amount of NFL protein in HEK293T-RGNEF(⫹) clone A was analyzed by Western blot and compared with the level
of NFL in HEK293T cells (control). A clear reduction in NFL protein levels was observed in the stable cell line compared with control cells. (C)
Quantification of 3 Western blot experiments comparing the NFL protein levels between control and HEK293T-RGNEF(⫹) clone A cells. The results are
presented as mean ⫾ standard error of the mean. p value compares HEK293T-RGNEF(⫹) cells with control, standardized against GAPDH (***p ⬍ 0.001).
(D) The amount of NFL mRNA in HEK293T-RGNEF(⫹) clone A was analyzed by relative quantitative polymerase chain reaction (PCR) and compared with
the level of NFL in HEK293T cells (control). A clear reduction in NFL mRNA levels was observed in the stable cell line compared with control cells as
observed in the quantification of 3 experiments (***p ⬍ 0.001). (E) Western blot experiment showing the effect of an siRNA against RGNEF over the NFL
protein levels. HEK293T and HEK293T-RGNEF(⫹) clone A cells were treated with a control siRNA and also HEK293T-RGNEF(⫹) clone A cells were
treated with an siRNA against RGNEF. The amount of NFL is clearly increased in the presence of the siRNA against RGNEF, while the levels of RGNEF
are significantly decreased. (F) Quantification of 3 Western blot experiments comparing the NFL protein levels between HEK293T and HEK293T-
RGNEF(⫹) clone A cells treated with control siRNA and HEK293T-RGNEF(⫹) clone A treated with siRNA against RGNEF (***p ⬍ 0.001).

some-1). We have shown that RGNEF is an RNA binding We have demonstrated that full length RGNEF interacts
protein that acts as an RNA destabilizing factor for human with NFL mRNA and that this interaction does not appear to
NFL mRNA via its 3= UTR, and can regulate NFL protein require a protein mediator. This builds upon our initial
levels in cells. We have also determined that RGNEF can observation that the RNA binding domain of RGNEF could
actively function as a GEF protein. interact with NFL mRNA in whole cell lysates from spinal
258 C.A. Droppelmann et al. / Neurobiology of Aging 34 (2013) 248 –262

Fig. 6. Rho guanine nucleotide exchange factor (RGNEF) forms cytoplasmic inclusions in motor neurons of amyotrophic lateral sclerosis (ALS) patients.
Immunohistochemistry of RGNEF in motor neurons of control (A), sporadic ALS (sALS) (B, C) and familial ALS (fALS) (D, E) spinal cords. RGNEF forms
extensive skein-like aggregates (B, D) and punctate aggregates (D, E) in ALS motor neurons (Scale bar ⫽ 10 ␮m).

cord (containing all protein and RNA) in ALS but not levels. This suggests that this cellular model could share
control cases (Volkening et al., 2010). Taken together, these several characteristics with ALS tissues at the level of NFL
data strongly suggest that the presence of an RNA binding mRNA regulation involving RGNEF. Analyzing the putative
protein complex-based control mechanism in control spinal proteins that could participate in the regulation of this interac-
cord, or its absence in ALS spinal cord, prevents RGNEF tion with NFL mRNA in this cellular model will be crucial to
interaction with NFL mRNA. Interestingly, using HEK293T understand the role of this protein in ALS pathology.
as cellular model, interaction between RGNEF and NFL The effect of RGNEF on NFL mRNA stability was
mRNA was observed, including regulation of NFL mRNA analyzed using the NFL mRNA 3= UTR. This was previ-
levels via the 3= UTR, as well as regulation of NFL protein ously reported as a widely described target for RNA binding
proteins that regulates the stability of NFL in mice and
humans (Cañete-Soler et al., 1998, 2001; Ge et al., 2005,
Table 1
2007; Lin et al., 2004; Strong et al., 2007; Volkening et al.,
Quantification of RGNEF and TDP-43 neuronal cytoplasmic inclusions
in motor neurons from control, sALS and fALS patients 2009). We used 3 different NFL mRNA 3= UTRs: 3=
UTR-S, analogous to the mouse NFL 3= UTR, and 3=
Neuronal cytoplasmic inclusions (%)
UTR-M and 3= UTR-L reported in sequence databases. We
Control sALS fALS
observed a destabilizing effect on all of these NFL 3= UTRs
RGNEF 0 8.83 ⫾ 3.43 8.15 ⫾ 1.61 with an effect directly proportional to the length of the 3=
TDP-43 0 16.23 ⫾ 9.16 9.52 ⫾ 3.99
UTR. This result might indicate the presence of more than
Neuronal cytoplasmic inclusions from spinal cord motor neurons of con- 1 binding motif for RGNEF on NFL 3= UTR-M and -L. The
trol, sALS, and fALS patients (n ⫽ 3) were quantified. For the quantifi-
cation, only cells with a visible nucleus were considered. No statistically
destabilizing effect of RGNEF over NFL 3= UTR was com-
significant difference between the number of RGNEF and TDP-43 neuro- pletely abolished when an RGNEF version was used that
nal cytoplasmic inclusions in sALS and fALS was observed. Note that the did not contain the RNA binding domain (RGNEF-⌬CT-
error in the TDP-43 quantification is higher than for RGNEF due to the fact myc). This version of RGNEF does not interact with NFL
that not all ALS cases quantified in this study display TDP-43-positive mRNA, but a stabilizing effect can be observed. This could
aggregates.
Key: ALS, amyotrophic lateral sclerosis; fALS, familial ALS; RGNEF, rho
be explained by the presence of positive regulating factors
guanine nucleotide exchange factor; sALS, sporadic ALS; TDP-43, TAR that compete with RGNEF now gaining access to NFL 3=
DNA binding protein of 43 kDa; UTR in absence of the RNA binding domain of RGNEF.
C.A. Droppelmann et al. / Neurobiology of Aging 34 (2013) 248 –262 259

Fig. 7. Rho guanine nucleotide exchange factor (RGNEF) colocalizes with ubiquitin, p62/Sequestosome-1, and partially with TAR DNA binding protein of
43 kDa (TDP-43). (A) Immunofluorescence using antibodies against RGNEF (green) and ubiquitin (red) in amyotrophic lateral sclerosis (ALS) and control
motor neurons was performed. The merge and colocalization analysis (product of the differences from the mean [PDM] image) show that RGNEF aggregates
show significant colocalization with ubiquitin (yellow). In control motor neurons, RGNEF does not colocalize with ubiquitin. (B) Immunofluorescence using
antibodies against RGNEF (green) and p62/Sequestosome-1 (red) in ALS and control motor neurons was performed. The merge and colocalization analysis
(PDM image) show that RGNEF aggregates show significant colocalization with p62/Sequestosome-1 in ALS, but not in controls. (C) Immunofluorescence
using antibodies against RGNEF (green) and TDP-43 (red) in ALS and control motor neurons was performed. The merge and colocalization analysis (PDM
image) show that RGNEF aggregates show partial colocalization with TDP-43. In control motor neurons RGNEF shows a weak colocalization with TDP-43
in the nucleus (scale bar ⫽ 10 ␮m). In the PDM images, blue color indicates low level of colocalization while yellow and white indicates high colocalization.
260 C.A. Droppelmann et al. / Neurobiology of Aging 34 (2013) 248 –262

Interestingly, removal of the first 286 bases of the 3= A remarkable finding in this study is the observation of
UTR-M (3= UTR-M⌬S) resulted in greater downregulation of extensive RGNEF protein aggregates in all cases of ALS
luciferase signal. Together with the stabilizing effect of studied. The immunoreactivity was consistent with that ob-
TDP-43 on 3= UTR-S, this result supports the hypothesis that served for proteins such as FUS/TLS (Kwiatkowski et al.,
there is a presence of alternate stability determinants within 2009; Vance et al., 2009) and TDP-43 (Arai et al., 2006;
this region, which is consistent with our previous reports for Neumann et al., 2006), which strongly suggests that
14-3-3 (Ge et al., 2007) and TDP-43 (Volkening et al., 2009). RGNEF is a new element in the pathophysiology of ALS.
Considering these results, 2 very important future directions The colocalization of RGNEF with ubiquitin and p62/Se-
will be to determine the regions of NFL 3= UTR that interact questosome-1 aggregates strongly supports the pathological
with RGNEF, and to determine the levels of in vivo NFL nature of these aggregates. Notably, RGNEF showed some
expression of each mRNA bearing different 3= UTR variants. colocalization with TDP-43 in ALS affected motor neurons.
It was interesting to observe that the destabilizing effect This is interesting because RGNEF and TDP-43 both inter-
of RGNEF on human NFL 3= UTR was opposite to the act with NFL mRNA (Strong et al., 2007; Volkening et al.,
effect described for murine p190RhoGEF (Cañete-Soler et 2009). TDP-43 can interact with multiple RNA targets (Co-
al., 2001). We first hypothesized that the observed differ- lombrita et al., 2012; Sephton et al., 2011; Tollervey et al.,
ence between our work and that previously described for 2011; Xiao et al., 2011) and multiple proteins (Freibaum et
p190RhoGEF was technical in nature—a protection assay al., 2010). It is probable that RGNEF can display this same
versus our luciferase assay using live cells. Due to this, we attribute. Therefore, it is possible that when different inter-
decided to analyze the effect of p190RhoGEF over the acting partners (RNAs and/or proteins) are involved,
mouse NFL 3= UTR using our system. Notably, we con- RGNEF and TDP-43 form spatially separated aggregates
firmed the results of Cañete-Soler et al. (2001) and we also and when similar interacting partners are involved, both
observed that p190RhoGEF can act to stabilize human NFL 3= proteins aggregate in the same compartment. While the
UTR. Further, we observed that RGNEF can act to destabilize mechanism by which RGNEF is induced to form cytosolic
mouse NFL 3= UTR in mouse neuronal cells. Consequently, aggregates remains to be determined, we can hypothesize a
these results strongly suggest that p190RhoGEF is intrinsically function for RGNEF in RNA transport because a similar
a stabilizing protein and RGNEF is a destabilizing protein. The mechanism has been proposed for TDP-43 (Strong, 2010;
destabilizing effect of RGNEF was confirmed using a stable Strong et al., 2007). Additionally, RGNEF and TDP-43
cell line expressing RGNEF-myc. In this cell line, the NFL show weak colocalization in the nucleus of healthy control
protein levels were greatly decreased, an effect produced by motor neurons, suggesting a physiological nuclear role for a
the downregulation of NFL mRNA levels and specifically small pool of RGNEF that may be related with its RNA
dependent on the presence of RGNEF because when RG- binding protein function.
NEF levels were diminished by an siRNA, the NFL levels Our results put RGNEF into the small group of RNA
increased. binding proteins that form aggregates in ALS, along with
Considering that ALS is not a natural pathology in ro- TDP-43 (Arai et al., 2006; Neumann et al., 2006), FUS/TLS
dents, the differences between p190RhoGEF and RGNEF (Deng et al., 2010; Kwiatkowski et al., 2009; Vance et al.,
emphasize the importance of understanding the molecular 2009), and mutant SOD1 (Bruijn et al., 1997, 1998; Rosen
mechanism of NFL mRNA regulation in humans. The mo- et al., 1993; Stieber et al., 2000). In this form, RGNEF is
tor neuron death in the reported mouse model involving another important component of the cellular phenotype ob-
p190RhoGEF was explained by its resulting from the disas- served in ALS, and may be a prominent contributor to the
sembly of a putative stabilizing complex where p190RhoGEF disease pathogenesis given its effect on NFL mRNA. It
is participating when NFL mRNA or protein are overexpressed is noteworthy that the murine homologue to RGNEF,
(Lin et al., 2005). In humans, however, we hypothesize that the p190RhoGEF, can act as an antiapoptotic factor in neuronal
cause of the pathology is a result of the aggregation of RGNEF cells (Wu et al., 2003), suggesting that the sequestration of
that could sequester NFL mRNA, induce neurofilamentous RGNEF into aggregates in ALS may contribute to neuronal
aggregates, and subsequently, motor neuron death. It is note- death. The presence of aggregates of RGNEF, an RNA binding
protein that can bind and destabilize NFL transcripts, could
worthy that RGNEF has been shown to interact with NFL
also explain the lower levels of NFL mRNA detected in ALS
mRNA only in ALS cases (Volkening et al., 2010). We do
motor neurons (Wong et al., 2000), as well as provide a
not yet know whether NFL protein is involved in ALS
possible mechanism for NF aggregate formation together with
pathogenesis involving RGNEF; however this will be an
a link between ALS and Rho signaling pathways.
area of focus for future studies. Therefore, it is possible that
the mouse model provided accurate clues relating to the
involvement of p190RhoGEF in motor neuron cell death,
Disclosure statement
but we speculate that the mechanism involved is different in
humans due to the opposing roles of RGNEF and The authors declare that they have no competing inter-
p190RhoGEF in NFL mRNA stabilization. ests.
C.A. Droppelmann et al. / Neurobiology of Aging 34 (2013) 248 –262 261

Acknowledgements Seeley, W.W., Josephs, K.A., Coppola, G., Geschwind, D.H., Wszolek,
Z.K., Feldman, H., Knopman, D.S., Petersen, R.C., Miller, B.L.,
The authors thank Dr. Rosa Rademakers for the Dickson, D.W., Boylan, K.B., Graff-Radford, N.R., Rademakers, R.,
C9ORF72 expanded repeats genotyping of ALS patients 2011. Expanded GGGGCC hexanucleotide repeat in noncoding region
of C9ORF72 causes chromosome 9p-linked FTD and ALS. Neuron 72,
and Dr. Wouter H. Moolenaar for kindly providing pcDNA-
245–256.
p190RhoGEF plasmid. This work was supported by the Deng, H.X., Zhai, H., Bigio, E.H., Yan, J., Fecto, F., Ajroud, K., Mishra,
Canadian Institute of Health Research (CIHR), The McFeat M., Ajroud-Driss, S., Heller, S., Sufit, R., Siddique, N., Mugnaini, E.,
Family Fund, and the ALS Society of Canada. Siddique, T., 2010. FUS-immunoreactive inclusions are a common
feature in sporadic and non-SOD1 familial amyotrophic lateral sclero-
Appendix A. Supplementary data sis. Ann. Neurol. 67, 739 –748.
Freibaum, B.D., Chitta, R.K., High, A.A., Taylor, J.P., 2010. Global
Supplementary data associated with this article can be analysis of TDP-43 interacting proteins reveals strong association with
found, in the online version, http://dx.doi.org/10.1016/j. RNA splicing and translation machinery. J. Proteome Res. 9, 1104 –
neurobiolaging.2012.06.021. 1120.
Ge, W.W., Volkening, K., Leystra-Lantz, C., Jaffe, H., Strong, M.J., 2007.
14-3-3 protein binds to the low molecular weight neurofilament (NFL)
References mRNA 3’ UTR. Mol. Cell. Neurosci. 34, 80 – 87.
Ge, W.W., Wen, W., Strong, W., Leystra-Lantz, C., Strong, M.J., 2005.
Arai, T., Hasegawa, M., Akiyama, H., Ikeda, K., Nonaka, T., Mori, H., Mutant copper-zinc superoxide dismutase binds to and destabilizes
Mann, D., Tsuchiya, K., Yoshida, M., Hashizume, Y., Oda, T., 2006. human low molecular weight neurofilament mRNA. J. Biol. Chem.
TDP-43 is a component of ubiquitin-positive tau-negative inclusions in 280, 118 –124.
frontotemporal lobar degeneration and amyotrophic lateral sclerosis. He, C.Z., Hays, A.P., 2004. Expression of peripherin in ubiquinated inclu-
Biochem. Biophys. Res. Commun. 351, 602– 611. sions of amyotrophic lateral sclerosis. J. Neurol. Sci. 217, 47–54.
Beaulieu, J.M., Jacomy, H., Julien, J.P., 2000. Formation of intermediate Kawamoto, Y., Akiguchi, I., Nakamura, S., Budka, H., 2004. 14-3-3
filament protein aggregates with disparate effects in two transgenic proteins in Lewy body-like hyaline inclusions in patients with sporadic
mouse models lacking the neurofilament light subunit. J. Neurosci. 20, amyotrophic lateral sclerosis. Acta Neuropathol. 108, 531–537.
5321–5328. Kondo, A., Iwaki, T., Tateishi, J., Kirimoto, K., Morimoto, T., Oomura, I.,
Beaulieu, J.M., Nguyen, M.D., Julien, J.P., 1999. Late onset of motor 1986. Accumulation of neurofilaments in a sporadic case of amyo-
neurons in mice overexpressing wild-type peripherin. J. Cell Biol. 147, trophic lateral sclerosis. Jpn. J. Psychiatry Neurol. 40, 677– 684.
531–544. Kuusisto, E., Kauppinen, T., Alafuzoff, I., 2008. Use of p62/SQSTM1
Bergeron, C., Beric-Maskarel, K., Muntasser, S., Weyer, L., Somerville, antibodies for neuropathological diagnosis. Neuropathol. Appl. Neuro-
M.J., Percy, M.E., 1994. Neurofilament light and polyadenylated biol. 34, 169 –180.
mRNA levels are decreased in amyotrophic lateral sclerosis motor Kuusisto, E., Salminen, A., Alafuzoff, I., 2001. Ubiquitin-binding protein
neurons. J. Neuropathol. Exp. Neurol. 53, 221–230. p62 is present in neuronal and glial inclusions in human tauopathies
Bruijn, L.I., Becher, M.W., Lee, M.K., Anderson, K.L., Jenkins, N.A., and synucleinopathies. Neuroreport 12, 2085–2090.
Copeland, N.G., Sisodia, S.S., Rothstein, J.D., Borchelt, D.R., Price, Kwiatkowski, T.J., Jr, Bosco, D.A., Leclerc, A.L., Tamrazian, E., Vander-
D.L., Cleveland, D.W., 1997. ALS-linked SOD1 mutant G85R medi- burg, C.R., Russ, C., Davis, A., Gilchrist, J., Kasarskis, E.J., Munsat,
ates damage to astrocytes and promotes rapidly progressive disease T., Valdmanis, P., Rouleau, G.A., Hosler, B.A., Cortelli, P., de Jong,
with SOD1-containing inclusions. Neuron 18, 327–338. P.J., Yoshinaga, Y., Haines, J.L., Pericak-Vance, M.A., Yan, J.,
Bruijn, L.I., Houseweart, M.K., Kato, S., Anderson, K.L., Anderson, S.D., Ticozzi, N., Siddique, T., McKenna-Yasek, D., Sapp, P.C., Horvitz,
Ohama, E., Reaume, A.G., Scott, R.W., Cleveland, D.W., 1998. Ag- H.R., Landers, J.E., Brown, R.H., Jr., 2009. Mutations in the FUS/TLS
gregation and motor neuron toxicity of an ALS-linked SOD1 mutant gene on chromosome 16 cause familial amyotrophic lateral sclerosis.
independent from wild-type SOD1. Science 281, 1851–1854. Science 323, 1205–1208.
Buratti, E., Dörk, T., Zuccato, E., Pagani, F., Romano, M., Baralle, F.E., Li, Q., Lau, A., Morris, T.J., Guo, L., Fordyce, C.B., Stanley, E.F., 2004.
2001. Nuclear factor TDP-43 and SR proteins promote in vitro and in A syntaxin 1, Galpha(o), and N-type calcium channel complex at a
vivo CFTR exon 9 skipping. EMBO J. 20, 1774 –1784. presynaptic nerve terminal: analysis by quantitative immunocolocal-
Cañete-Soler, R., Schwartz, M.L., Hua, Y., Schlaepfer, W.W., 1998. Sta- ization. J. Neurosci. 24, 4070 – 4081.
bility determinants are localized to the 3’-untranslated region and Lin, H., Zhai, J., Cañete-Soler, R., Schlaepfer, W.W., 2004. 3’ untranslated
3’-coding region of the neurofilament light subunit mRNA using a region in a light neurofilament (NF-L) mRNA triggers aggregation of
tetracycline-inducible promoter. J. Biol. Chem. 273, 12650 –12654. NF-L and mutant superoxide dismutase 1 proteins in neuronal cells.
Cañete-Soler, R., Wu, J., Zhai, J., Shamim, M., Schlaepfer, W.W., 2001. J. Neurosci. 24, 2716 –2726.
p190RhoGEF Binds to a destabilizing element in the 3’ untranslated Lin, H., Zhai, J., Schlaepfer, W.W., 2005. RNA-binding protein is involved
region of light neurofilament subunit mRNA and alters the stability of in aggregation of light neurofilament protein and is implicated in the
the transcript. J. Biol. Chem. 276, 32046 –32050. pathogenesis of motor neuron degeneration. Hum. Mol. Genet. 14,
Colombrita, C., Onesto, E., Megiorni, F., Pizzuti, A., Baralle, F.E., Buratti, 3643–3659.
E., Silani, V., Ratti, A., 2012. TDP-43 and FUS RNA-binding proteins Millecamps, S., Gowing, G., Corti, O., Mallet, J., Julien, J.P., 2007.
bind distinct sets of cytoplasmic messenger RNAs and differently Conditional NF-L transgene expression in mice for in vivo analysis of
regulate their post-transcriptional fate in motoneuron-like cells. J. Biol. turnover and transport rate of neurofilaments. J. Neurosci. 27, 4947–
Chem. 287, 15635–15647. 4956.
Crozat, A., Aman, P., Mandahl, N., Ron, D., 1993. Fusion of CHOP to a Nalbant, P., Chang, Y.C., Birkenfeld, J., Chang, Z.F., Bokoch, G.M., 2009.
novel RNA-binding protein in human myxoid liposarcoma. Nature 363, Guanine nucleotide exchange factor-H1 regulates cell migration via
640 – 644. localized activation of RhoA at the leading edge. Mol. Biol. Cell 20,
Dejesus-Hernandez, M., Mackenzie, I.R., Boeve, B.F., Boxer, A.L., Baker, 4070 – 4082.
M., Rutherford, N.J., Nicholson, A.M., Finch, N.A., Flynn, H., Adam- Neumann, M., Sampathu, D.M., Kwong, L.K., Truax, A.C., Micsenyi,
son, J., Kouri, N., Wojtas, A., Sengdy, P., Hsiung, G.Y., Karydas, A., M.C., Chou, T.T., Bruce, J., Schuck, T., Grossman, M., Clark, C.M.,
262 C.A. Droppelmann et al. / Neurobiology of Aging 34 (2013) 248 –262

McCluskey, L.F., Miller, B.L., Masliah, E., Mackenzie, I.R., Feldman, Strong, M.J., 2010. The evidence for altered RNA metabolism in amyo-
H., Feiden, W., Kretzschmar, H.A., Trojanowski, J.Q., Lee, V.M., trophic lateral sclerosis (ALS). J. Neurol. Sci. 288, 1–12.
2006. Ubiquitinated TDP-43 in frontotemporal lobar degeneration and Strong, M.J., Kesavapany, S., Pant, H.C., 2005. The pathobiology of
amyotrophic lateral sclerosis. Science 314, 130 –133. amyotrophic lateral sclerosis: a proteinopathy? J. Neuropathol. Exp.
Nie, Z., Wu, J., Zhai, J., Lin, H., Ge, W., Schlaepfer, W.W., Cañete-Soler, Neurol. 64, 649 – 664.
R., 2002. Untranslated element in neurofilament mRNA has neuro- Strong, M.J., Volkening, K., Hammond, R., Yang, W., Strong, W., Ley-
pathic effect on motor neurons of transgenic mice. J. Neurosci. 22, stra-Lantz, C., Shoesmith, C., 2007. TDP43 is a human low molecular
7662–7670. weight neurofilament (hNFL) mRNA-binding protein. Mol. Cell. Neu-
Pikkarainen, M., Hartikainen, P., Alafuzoff, I., 2008. Neuropathologic rosci. 35, 320 –327.
features of frontotemporal lobar degeneration with ubiquitin-positive Szaro, B.G., Strong, M.J., 2010. Post-transcriptional control of neurofila-
inclusions visualized with ubiquitin-binding protein p62 immunohisto- ments: New roles in development, regeneration and neurodegenerative
chemistry. J. Neuropathol. Exp. Neurol. 67, 280 –298. disease. Trends Neurosci. 33, 27–37.
Pikkarainen, M., Hartikainen, P., Alafuzoff, I., 2010. Ubiquitinated p62- Thyagarajan, A., Strong, M.J., Szaro, B.G., 2007. Post-transcriptional
positive, TDP-43-negative inclusions in cerebellum in frontotemporal
control of neurofilaments in development and disease. Exp. Cell Res.
lobar degeneration with TAR DNA binding protein 43. Neuropathol-
313, 2088 –2097.
ogy 30, 197–199.
Thyagarajan, B., Liu, Y., Shin, S., Lakshmipathy, U., Scheyhing, K., Xue,
Ren, J., Wen, L., Gao, X., Jin, C., Xue, Y., Yao, X., 2009. DOG 1.0:
H., Ellerström, C., Strehl, R., Hyllner, J., Rao, M.S., Chesnut, J.D.,
illustrator of protein domain structures. Cell Res. 19, 271–273.
2008. Creation of engineered human embryonic stem cell lines using
Ren, X.D., Kiosses, W.B., Schwartz, M.A., 1999. Regulation of the small
phiC31 integrase. Stem Cells 26, 119 –126.
GTP-binding protein Rho by cell adhesion and the cytoskeleton.
EMBO J. 18, 578 –585. Thyagarajan, B., Olivares, E.C., Hollis, R.P., Ginsburg, D.S., Calos, M.P.,
Renton, A.E., Majounie, E., Waite, A., Simón-Sánchez, J., Rollinson, S., 2001. Site-specific genomic integration in mammalian cells mediated
Gibbs, J.R., Schymick, J.C., Laaksovirta, H., van Swieten, J.C., Myl- by phage phiC31 integrase. Mol. Cell. Biol. 21, 3926 –3934.
lykangas, L., Kalimo, H., Paetau, A., Abramzon, Y., Remes, A.M., Tollervey, J.R., Curk, T., Rogelj, B., Briese, M., Cereda, M., Kayikci, M.,
Kaganovich, A., Scholz, S.W., Duckworth, J., Ding, J., Harmer, D.W., König, J., Hortobágyi, T., Nishimura, A.L., Zupunski, V., Patani, R.,
Hernandez, D.G., Johnson, J.O., Mok, K., Ryten, M., Trabzuni, D., Chandran, S., Rot, G., Zupan, B., Shaw, C.E., Ule, J., 2011. Charac-
Guerreiro, R.J., Orrell, R.W., Neal, J., Murray, A., Pearson, J., Jansen, terizing the RNA targets and position-dependent splicing regulation by
I.E., Sondervan, D., Seelaar, H., Blake, D., Young, K., Halliwell, N., TDP-43. Nat. Neurosci. 14, 452– 458.
Callister, J.B., Toulson, G., Richardson, A., Gerhard, A., Snowden, J., van Horck, F.P., Ahmadian, M.R., Haeusler, L.C., Moolenaar, W.H.,
Mann, D., Neary, D., Nalls, M.A., Peuralinna, T., Jansson, L., Isoviita, Kranenburg, O., 2001. Characterization of p190RhoGEF, a RhoA-
V.M., Kaivorinne, A.L., Holtta-Vuori, M., Ikonen, E., Sulkava, R., specific guanine nucleotide exchange factor that interacts with micro-
Benatar, M., Wuu, J., Chio, A., Restagno, G., Borghero, G., Sabatelli, tubules. J. Biol. Chem. 276, 4948 – 4956.
M., Heckerman, D., Rogaeva, E., Zinman, L., Rothstein, J.D., Sendt- Vance, C., Rogelj, B., Hortobágyi, T., De Vos, K.J., Nishimura, A.L.,
ner, M., Drepper, C., Eichler, E.E., Alkan, C., Abdullaev, Z., Pack, Sreedharan, J., Hu, X., Smith, B., Ruddy, D., Wright, P., Ganesal-
S.D., Dutra, A., Pak, E., Hardy, J., Singleton, A., Williams, N.M., ingam, J., Williams, K.L., Tripathi, V., Al-Saraj, S., Al-Chalabi, A.,
Heutink, P., Pickering-Brown, S., Morris, H.R., Tienari, P.J., Traynor, Leigh, P.N., Blair, I.P., Nicholson, G., de Belleroche, J., Gallo, J.M.,
B.J., Traynor, B.J., 2011. A hexanucleotide repeat expansion in Miller, C.C., Shaw, C.E., 2009. Mutations in FUS, an RNA processing
C9ORF72 is the cause of chromosome 9p21-linked ALS-FTD. Neuron protein, cause familial amyotrophic lateral sclerosis type 6. Science
72, 257–268. 323, 1208 –1211.
Rosen, D.R., Siddique, T., Patterson, D., Figlewicz, D.A., Sapp, P., Hen- Volkening, K., Leystra-Lantz, C., Strong, M.J., 2010. Human low molec-
tati, A., Donaldson, D., Goto, J., O’Regan, J.P., Deng, H.X., etAl, etA.l, ular weight neurofilament (NFL) mRNA interacts with a predicted
1993. Mutations in Cu/Zn superoxide dismutase gene are associated p190RhoGEF homologue (RGNEF) in humans. Amyotroph. Lateral
with familial amyotrophic lateral sclerosis. Nature 362, 59 – 62. Scler. 11, 97–103.
Sephton, C.F., Cenik, C., Kucukural, A., Dammer, E.B., Cenik, B., Han, Volkening, K., Leystra-Lantz, C., Yang, W., Jaffee, H., Strong, M.J., 2009.
Y., Dewey, C.M., Roth, F.P., Herz, J., Peng, J., Moore, M.J., Yu, G.,
Tar DNA binding protein of 43 kDa (TDP-43), 14-3-3 proteins and
2011. Identification of neuronal RNA targets of TDP-43-containing
copper/zinc superoxide dismutase (SOD1) interact to modulate NFL
ribonucleoprotein complexes. J. Biol. Chem. 286, 1204 –1215.
mRNA stability. Implications for altered RNA processing in amyo-
Seppänen, A., Pikkarainen, M., Hartikainen, P., Hofmann, S.C., Majamaa,
trophic lateral sclerosis (ALS). Brain Res. 1305, 168 –182.
K., Alafuzoff, I., 2009. Expression of collagen XVII and ubiquitin-
Wong, N.K., He, B.P., Strong, M.J., 2000. Characterization of neuronal
binding protein p62 in motor neuron disease. Brain Res. 1247, 171–
intermediate filament protein expression in cervical spinal motor neu-
177.
Shaw, B.F., Lelie, H.L., Durazo, A., Nersissian, A.M., Xu, G., Chan, P.K., rons in sporadic amyotrophic lateral sclerosis (ALS). J. Neuropathol.
Gralla, E.B., Tiwari, A., Hayward, L.J., Borchelt, D.R., Valentine, J.S., Exp. Neurol. 59, 972–982.
Whitelegge, J.P., 2008. Detergent-insoluble aggregates associated with Wu, J., Zhai, J., Lin, H., Nie, Z., Ge, W.W., García-Bermejo, L., Muschel,
amyotrophic lateral sclerosis in transgenic mice contain primarily full- R.J., Schlaepfer, W.W., Cañete-Soler, R., 2003. Cytoplasmic retention
length, unmodified superoxide dismutase-1. J. Biol. Chem. 283, 8340 – sites in p190RhoGEF confer anti-apoptotic activity to an EGFP-tagged
8350. protein. Brain Res. Mol. Brain Res. 117, 27–38.
Stieber, A., Gonatas, J.O., Gonatas, N.K., 2000. Aggregates of mutant Xiao, S., Sanelli, T., Dib, S., Sheps, D., Findlater, J., Bilbao, J., Keith, J.,
protein appear progressively in dendrites, in periaxonal processes of Zinman, L., Rogaeva, E., Robertson, J., 2011. RNA targets of TDP-43
oligodendrocytes, and in neuronal and astrocytic perikarya of mice identified by UV-CLIP are deregulated in ALS. Mol. Cell. Neurosci.
expressing the SOD1(G93A) mutation of familial amyotrophic lateral 47, 167–180.
sclerosis. J. Neurol. Sci. 177, 114 –123. Zhai, J., Lin, H., Shamim, M., Schlaepfer, W.W., Cañete-Soler, R., 2001.
Strong, M.J., 1999. Neurofilament metabolism in sporadic amyotrophic Identification of a novel interaction of 14 –3-3 with p190RhoGEF.
lateral sclerosis. J. Neurol. Sci. 169, 170 –177. J. Biol. Chem. 276, 41318 – 41324.

Você também pode gostar