TMP 7 DD1

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 12

[Cell Cycle 6:23, 2970-2981, 1 December 2007]; ©2007 Landes Bioscience

Report

N-Terminal Proteolysis of Full-Length Mutant Huntingtin in an


Inducible PC12 Cell Model of Huntington’s Disease
Tamara Ratovitski1,† ABSTRACT
Masayuki Nakamura1,†,‡ Proteolytic cleavage of mutant huntingtin may play a key role in the pathogenesis
of Huntington’s disease; however the steps in huntingtin proteolysis are not fully under-
James D’Ambola1

.
stood. Huntingtin was shown to be cleaved by caspases and calpains within a region

E
Ekaterine Chighladze1 between 460–600 amino acids from the N-terminus. Two smaller N-terminal fragments

UT
produced by unknown protease have been previously described as cp-A and cp-B. To
Yideng Liang1 further investigate the huntingtin proteolytic pathway, we used an inducible PC12 cell

RIB
Wenfei Wang1 model expressing full-length huntingtin with either normal or expanded polyglutamine.
This cell model recapitulates several steps of huntingtin proteolysis: proteolysis mediated
Rona Graham2 by caspases within the region previously mapped for caspase cleavage, and cleavage

IST
Michael R. Hayden2 generating two novel N-terminal fragments (cp-1 approximately 90–105 residues long
and cp-2 extending beyond 115–129 epitope of huntingtin). Interestingly, the deletion
David R. Borchelt3 of amino acids 105–114 (mapped previously as a cleavage site for cp-A) failed to

D
Ricky R. Hirschhorn1,4 affect the production of cp-1 or cp-2. Therefore, we conclude that these new fragments

OT
are distinct from previously described cp-A and cp-B. We demonstrate that cp-1 and
Christopher A. Ross1,5,* cp-2 fragments are produced and accumulate within nuclear and cytoplasmic inclusions
1Division of Neurobiology; Department of Psychiatry; 5Departments of Neurology
prior to huntingtin-induced cell toxicity, and these fragments can be formed by caspase-
ON
and Neuroscience; Johns Hopkins University School of Medicine; Baltimore,
independent proteolytic cleavage of huntingtin in PC12 cells. In addition, inhibition of
Maryland USA calpains leads to decreased subsequent degradation of cp-1 and cp-2 fragments, and
accelerated formation of inclusions. Further delineation of huntingtin cleavage events may
.D
2Department of Medical Genetics; Centre for Molecular Medicine and Therapeutics;

Child and Family Research Institute; University of British Columbia; Vancouver,


lead to novel therapeutic targets for HD.
British Columbia, Canada
CE

3Department of Neuroscience; Santa Fe Health Alzheimer’s Disease Center;


IEN

McKnight Brain Institute; University of Florida; Gainesville, Florida USA


4Department of Biology; Hood College; Frederick, Maryland USA
ABBREVIATIONS
HD, Huntington’s disease; Htt, huntingtin; polyQ, polyglutamine; ER, endoplasmic
SC

†These authors equally contributed to this work.


reticulum; aa, amino acid; Dox, doxycycline; NG108-15, neuroblastoma-glioma; PC12,
‡Current Address: Department of Psychiatry; Kagoshima University Graduate pheochromocytoma; tTA, transactivator; TRE, tet-responsive element; DMEM, Dulbecco’s
BIO

School of Medical and Dental Sciences; Sakuragaoka, Kagoshima, Japan modified Eagle’s medium; FBS, fetal bovine serum; NGF, nerve growth factor; HEK,
*Correspondence to: Christopher A. Ross and Tamara Ratovitski; CMSC 8-121; 600 human embryonic kidney; NER, nuclear extraction reagent; DAPI, 4',6-Diamidino-2-phe-
North Wolfe Street; Baltimore, Maryland 21287 USA; Fax: 410.614.0013; Email: nylindol; GAPDH, glyceraldehyde-3-phosphate dehydrogenase; PARP, poly ADP-ribose
caross@jhu.edu/ tratovi1@jhmi.edu polymerase; FA, formic acid; UPS, ubiquitin-proteasome system; GFP, green fluorescent
ES

Original manuscript submitted: 07/31/07 protein; CMV, cytomegalovirus; NS, non-specific


Revised manuscript submitted: 08/31/07
ND

Manuscript accepted: 09/02/07


INTRODUCTION
Previously published online as a E-publication:
LA

http://www.landesbioscience.com/journals/cc/article/4992 Huntington’s disease (HD) is a progressive neurodegenerative disorder caused by CAG


repeat expansion (coding for polyglutamine) within the coding region of the huntingtin
KEY WORDS gene product (Htt). There is a threshold of 36 repeats for HD, and an inverse correla-
07

tion between the length of the polyglutamine (polyQ) expansion and the age of onset.1,2
Huntington’s Disease mechanism, proteolysis,
20

Clinically HD is characterized by movement disorder and cognitive and behavioral


protein degradation, caspases, calpains
changes, progressing to death within 15–20 years from onset. The symptoms of HD are
©

likely to be caused by both neuronal death and dysfunction.


ACKNOWLEDGEMENTS
Htt is an approximately 350 kD ubiquitously expressed protein that has been localized
See page XXX. to several subcellular compartments, including nuclear and cytoplasmic fractions. It has
been found in association with organelles, including mitochondria, ER/Golgi, endosomes,
presynaptic and clathrin-coated vesicles and plasma membrane.3-8 Cellular toxicity in HD
may involve interference of expanded Htt with gene transcription, vesicle transport, mito-
chondrial function, Ca2+ homeostasis, and protein degradation machinery via proteasome
or autophagy.9-15 The nature of cytotoxic species of expanded Htt is unknown, but there is
emerging evidence suggesting that they might be generated via Htt proteolysis. The main

2970 Cell Cycle 2007; Vol. 6 Issue 23


N-terminal Proteolysis of Huntingtin in PC12 Cell Model

which can be detected with antibodies


to Htt peptides 1–17 and 81–90 but not
115-129.20
Htt can be cleaved between residues
513 and 586 by caspases 2, 3, 6 and 7
(Fig. 1), and this cleavage has been linked
to cell toxicity.33-38 The importance of
caspase 6 mediated cleavage of Htt at
position 586 have been confirmed in
vivo: YAC 128 mice expressing mutant
Htt resistant to cleavage at this site
do not demonstrate a phenotype in
any of the endpoints previously estab-

E .
lished in the YAC128 model, including

UT
motor, cognitive and neuropathological
deficits.39 Cleavage of Htt by calpains

RIB
(e.g., at residues 469 and 536, Fig. 1)
has also been reported to contribute
to toxicity.27,40-43 At least two smaller

IST
N-terminal fragments (cp-A and cp-B)
were previously described in reference 18:

D
Cp-A was defined as a fragment produced
by cleavage in the region between resi-

OT
dues 82 and 129, which accumulates
within nuclear inclusions in mouse-rat
ON neuroblastoma-glioma (NG108-15)
stable cells expressing expanded forms
Figure 1. Generation of inducible PC12 cell model. (A) The “tet-off” system was used in which the expres- of Htt. Deletion of amino acids 105–
.D
sion of Htt can be induced by removing doxycycline. This facilitates binding of tTA to the tet-responsive 114, or alteration within this region
element (TRE) controlling Htt expression. The full-length human Htt cDNA with either 23Q or 148Q was
prevented production of the cp-A frag-
CE

cloned into pTet-splice vector (pTet-splice-FL-Htt-23/148Q). *Stable lines generated with these vectors
expressed Htt with 21Q or 126Q respectively, as confirmed by genotyping. The N-terminal c-myc tag ment. Cp-B fragment is longer (up to
was included for Htt detection. The tTA expression was driven by the cytomegalovirus (CMV) promoter. 214 amino acids) and was found in
IEN

(B) Schematic of Htt proteolysis. Within the sequence indicated are putative cp-A/cp-B (18) and cp-1/cp-2 cytoplasmic inclusions. Cp-A fragment
cleavage sites (hatched pattern); identified caspase and calpain sites (solid boxes). Below are shown was suggested to be produced by an
epitopes recognized by Htt antibodies.
SC

unknown aspartic protease. Certain lyso-


somal proteases (cathepsin D, B and L)
BIO

pathological feature of all CAG repeat disorders is the formation were found to contribute to Htt degradation and accumulation of
of nuclear and cytoplasmic inclusions in neurons of affected brain stable N-terminal fragments in clonal striatal cells, but it is not clear
regions. Inclusions in brains of HD patients can be stained with anti- whether they mediate Htt clearance, or facilitate generation of toxic
ES

bodies to ubiquitin, and with antibodies directed at the N-terminal, fragments.44,45


but not C-terminal, epitopes of Htt.16-21 This suggests that inclu- Although data from mouse and cell studies support the role of
ND

sions contain truncated N-terminal fragments of Htt. Although Htt proteolysis in HD pathogenesis, what steps generate the toxic
the inclusion bodies themselves might be neuroprotective,10,22 the fragments, the lengths of the fragments, and whether fragments
LA

N-terminal fragments of Htt in cells may be pathogenic. are produced prior to or subsequent to cell toxicity are poorly
Cellular models of HD suggest that short N-terminal fragments understood. There is considerable mounting evidence for the role
accumulate in the nucleus, and are more toxic than longer fragments of specific sized fragments in the pathogenesis of disease. Mouse
07

or full-length Htt.18,21-27 Mouse model studies also support a role HD models described above suggest that the 552 amino acid (aa),
of Htt proteolysis in HD pathogenesis. Transgenic mice expressing 513 aa and the 117 aa (“shortstop”) mutant Htt fragments are not
20

N-terminal fragments of Htt generally have more severe behavioral toxic. In contrast the 586 aa, the exon 1 (R6/2 mice) and the N171
©

and pathologic phenotypes than mice expressing full-length Htt.28-30 (N171-82Q) mutant Htt fragments are toxic. The 586 aa fragment
One exception—the “shortstop” mice, expressing a short N-terminal is produced in vivo from both endogenous and mutant htt. However,
Htt fragment only 117 amino acids long31—do not demonstrate a it is not clear whether cleavage in the caspase and calpain region
phenotype compared to wild type mice. Inclusions are present, but is always necessary and precedes the formation of cp-A and cp-B
these do not correlate with neuronal loss. A recently described induc- - like fragments, or whether these proteolytic pathways may occur
ible mouse model, expressing full-length expanded Htt, accumulate a in parallel. Elucidating the steps in Htt proteolysis may facilitate the
60 kD Htt N-terminal cleavage product in the nucleus,32 and display identification of novel targets for development of HD therapeutics.
relatively severe phenotype. Mice expressing the first 171 amino We have generated a stable inducible cell model of HD, in which
acids of Htt (Htt-N171-82Q) also accumulate nuclear inclusions differentiated rat pheochromocytoma (PC12) cells are induced to

www.landesbioscience.com Cell Cycle 2971


N-terminal Proteolysis of Huntingtin in PC12 Cell Model

express full-length Htt with either normal or expanded polygluta- (Calbiochem) and calpain1/2-specific, MDL128170, 10 µM
mine repeats. This cell model reveals several steps of Htt proteolysis: (Calbiochem), or calpeptin, 50 µM or 75 µM (Calbiochem). Fresh
proteolysis mediated by caspases within the region previously mapped inhibitors were added every three days. Proteasome inhibitors lacta-
for caspase cleavage, and cleavage generating two novel N-terminal cystin (Calbiochem); MG132 (Calbiochem) or U102 (Biomol) were
fragments cp-1 and cp-2. These short N-terminal fragments accumu- added to growth media at indicated concentrations for the last 24 h
late within nuclear and cytoplasmic inclusions, and their formation of cell incubation. For transient transfection experiments human
precedes Htt-induced cell toxicity. We provide evidence that cp-1 embryonic kidney (HEK) 293FT cells (Invitrogen) were grown
and cp-2 fragments generated in differentiated PC12 cells can be in DMEM with 10% FBS, 100 µg/ml Geneticin, 100 units/ml
formed by caspase-independent proteolytic cleavage, consistent with penicillin and 100 units/ml streptomycin, and transfected with Htt
the parallel cleavage model in this cell system. Epitope mapping of constructs using Lipofectamine 2000 (Invitrogen) according to the
these fragments with antibodies to Htt suggest that they are similar manufacturer’s protocol. Proteasome reporter cell line GFPu-1 was
in length to previously described cp-A and cp-B fragments. However from American Type Culture Collection (ATCC).
the deletion of amino acids 105–114 (implicated in generation of Subcellular fractionation and dissociation of aggregates. PC12

E .
cp-A) failed to affect the production of cp-1 and cp-2 fragments in cells were first fractionated to enrich for cytoplasmic and nuclear

UT
this PC12 cell model, suggesting distinct proteolytic events. proteins using 0.5% Nonidet P-40 (protocol 1), or Nuclear and
Cytoplasmic Extraction reagent (NE-PER) from Pierce (protocol 2).

RIB
MATERIALS AND METHODS Nuclear pellets and soluble cytoplasmic fractions C were obtained
after centrifugation at 500 g (protocol 1), or at 15,000 g (protocol 2).
Plasmids and mutagenesis. Full-length constructs for inducible Nuclear pellets were solubilized using SDS buffer (protocol 1) or

IST
expression of normal (pTet-splice-FL-Htt-23Q) or expanded (pTet- Nuclear Extraction Reagent (NER, Pierce, protocol 2), and super-
splice-FL-Htt-148Q) Htt were described previously in reference 32. natants containing soluble nuclear fractions N and pellets were

D
The deletions of residues 105–114 and substitutions of alanines at collected after centrifugation at 15,000 g. Cytoplasmic fraction C did
positions 109–113 were introduced into pTet-N171-148Q construct not produce any pellet upon solubilization in SDS buffer. The pellets

OT
(described in Tanaka et al., 2006) by site-directed mutagenesis using from nuclear fractions were further dissociated using formic acid to
QuikChange II XL kit (Stratagene) according to manufacturer’s produce formic acid-soluble nuclear fractions F.18,23
protocol. The resulting constructs were digested with XhoI and DNA
fragments encoding N-terminal Htt fragments (N171) were ligated
ONCell toxicity measurements. Stable PC12 cell lines carrying either
FL-Htt-21Q or FL-Htt-126Q were seeded on collagen I coated
into pTet-splice-FL-23Q after digestion with XhoI. Htt expression 24-well plates (BD Biosciences) at a concentration 105 cells/ml
.D
constructs used for transient transfection, including wild-type and in differentiation media either with or without doxycycline. Cell
caspase-resistant (quint mutation) forms of normal and expanded viability was measured by trypan blue exclusion assay. Results repre-
CE

Htt N-terminal fragment (N1212-15Q and N1212-138Q), were sent the average of four wells. Staining with annexin V-EGFP and
previously described in references 34 and 40. propidium iodide of PC12 cells was performed using the annexin
IEN

Cell culture and transfection. To establish inducible stable V-EGFP apoptosis detection kit (BioVision Research Products)
PC12 cell lines expressing full-length normal or polyQ expanded according to the manufacturer’s protocol, on day 8 after induction of
SC

Htt, we used Tet-off PC12 cells (Clontech), stably expressing the Htt expression and differentiation. Five fields (200–500 cells each)
tetracycline transactivator (tTA). Cells were co-transfected with Htt were counted blindly for each condition.
BIO

constructs and pTK-Hyg vector (Clontech) in the ratio 20:1 using Immunofluorescence. PC12 cells, induced to express FL-Htt-126Q
Lipofectamine 2000 (Invitrogen) according to the manufacturer’s for 8 days, were fixed with 4% paraformaldehyde for 15 min, perme-
procedure, and were plated on collagen I treated culture dishes alized with 0.5% Triton X-100 (Sigma) for 10 min, blocked in
ES

(Biocoat, BD Biosciences). The stable clones were selected in 10% normal goat serum (Sigma) for 30 min, and incubated with
Dulbecco’s modified Eagle’s medium (DMEM) supplemented with monoclonal anti c-myc antibody, polyclonal antibody to Htt epit-
ND

5% Tet System approved fetal bovine serum (FBS, Clontech), 10% opes 497–513, or Htt epitopes 55–66 (1 h at room temperature),
horse serum (Invitrogen), 100 µg/ml Geneticin (Invitrogen), 200 µg/ followed by goat anti-mouse IgG FITC-conjugated (Vector), or goat
LA

ml hygromycin (Roche), 200 ng/ml doxycycline (Dox, Invitrogen), anti-rabbit Cy3-conjugated (Vector) secondary antibodies (1 h at
100 units/ml penicillin and 100 units/ml streptomycin. After three room temperature). Nuclei were stained with 4', 6-Diamidino-2-ph
to four weeks of selection at 37˚C in a humidified 5% CO2 incu- enylindol (DAPI, Vector).
07

bator, the antibiotic resistant clones were isolated and screened for Western blot analysis and antibodies. Polyclonal antibodies to
Htt expression by Western blot analysis using anti-c-myc antibody Htt-htt 55–65 and htt 81–90 were described previously in reference
20

9E10 (Roche). Stable cell lines were maintained in the same medium. 20. Briefly, they were produced in rabbit against a series of synthetic
©

The polyQ repeat length in selected lines, confirmed by genotyping, peptides, the number of each peptide corresponds to the residue
was 21Q for normal Htt and 126Q for expanded Htt (lines S6 number of wild-type human htt with 23Q (Genbank Accession
and S6-5). For neuronal differentiation PC12 cells were grown in #NM-002111).20 Antibody to Htt epitope 497–513 was also gener-
DMEM with 1% horse serum, supplemented with nerve growth ated in rabbit against the synthetic peptide. Htt 115–129 antibody
factor (NGF, 50 ng/ml, Roche). To study the effects of inhibitors (against residues 115–129 of Htt) is from Chemicon International
in PC12 cells, the following inhibitors were added to differentiation (cat. # MAB5490), antibody to DLND neoepitope of Htt-htt552
media: caspase pan-specific, Z-VAD, 25 µM (Calbiochem), caspase and antibody to DSVD neoepitope of Htt-htt513 are a kind gift
2-specific, Z-VDVAD, 10 µM (Calbiochem), caspase 3-specific, from Sophie Roy (Merck Canada Co. Ltd., Canada). For Western
AcDMQD, 25 µM (Calbiochem), caspase 6-specific Z-VEID, 10 µM blotting analysis, PC12 or HEK293 cells were lysed in M-PER buffer

2972 Cell Cycle 2007; Vol. 6 Issue 23


N-terminal Proteolysis of Huntingtin in PC12 Cell Model

(Pierce) with protease inhibitors (Protease Inhibitor Cocktail III,


Calbiochem), unless otherwise indicated, and protein concentrations
were estimated using BCA method (Biorad). Lysates (60 µg for PC12
cells, and 30 µg for HEK293 cells) were fractionated on NuPAGE
4–12% Bis-Tris polyacrylamide gels (Invitrogen), transferred to
nitrocellulose membranes, and probed with antibodies against Htt,
c-myc (9E10, Roche), and actin (Sigma) or GAPDH (Santa Cruz
Biotechnology) for loading control. β-tubulin (Sigma) and poly
ADP-ribose polymerase (PARP, Biomol) antibodies were used as
fractionation control. Immunoblots were developed with peroxi-
dase-conjugated secondary antibodies (Amersham), and enhanced
chemiluminescence (ECL-Plus detection reagent, Amersham).

E .
RESULTS

UT
PC12 cells expressing FL-Htt-126Q produce N-terminal frag-
ments. To study proteolysis of Htt, we have developed several stable

RIB
inducible PC12 cell lines expressing full-length Htt with either
normal (21Q) or expanded (126Q) polyglutamines. We used the

IST
Tet-off system in which the expression of Htt can be induced by
removing doxycycline (Fig. 1A). PC12 cells were differentiated using
NGF for up to 14 days, and Htt expression was evaluated with either

D
anti-c-myc antibodies, or antibodies recognizing the epitope between

OT
residues 55 and 66 of Htt (Fig. 1B). Differentiated PC12 cells (line
S6) expressed full-length Htt with 126Q, which was detectable after
four days upon induction (Fig. 2A). Two partially resolved fragments
migrating at ~60 kDa (cp-1 and cp-2) were detected at the same time
ON
with antibody to Htt. The same fragments were also detected with
.D
antibody to c-myc (data not shown), indicating that they represent
N-terminal Htt cleavage products. These fragments showed gel
CE

mobility similar to N-terminal fragments identified in transgenic


mice expressing full-length Htt with expanded polyglutamine,32
IEN

and are consistent with the size predicted for cp-A and cp-B frag-
ments detected in NG108 cells and clonal striatal cells expressing
Htt.18,44 However, unlike cp-A and cp-B, cp-1 and cp-2 frag-
SC

ments were readily detected without proteasome inhibition. PC12 Figure 2. PC12 cells expressing FL-Htt-126Q produce N-terminal fragments.
cells expressing full-length Htt with normal polyglutamine repeat PC12 cells (line S6) were differentiated using NGF and induced to express
BIO

(FL-Htt-21Q) produced very low levels of comparable fragments FL-Htt-126Q by removal of doxycycline for indicated time periods. Total cell
(data not shown). extracts (60 µg per lane) were analyzed by Western blotting with antibody to
Since caspase mediated cleavage of Htt has been described previ- residues 55-66 of Htt (A), neoepitope antibody to caspase 2/3/7 cleavage
ES

ously in references 33–38, we used neoepitope antibodies to caspase sites in Htt (DLND) htt552 (B), neoepitope antibody to caspase 3 site in Htt
(DSVD) htt513 (C), or antibody to GAPDH for loading control. FL-Htt-126Q is
2/3/7 site in Htt (DLND) htt552 to further characterize Htt proteo- indicated by arrowhead. Short N-terminal fragments cp-1/cp-2 (arrows) run
ND

lytic fragments produced by PC12 cells expressing expanded Htt. 552 as partially resolved doublet. Larger caspase generated fragments (arrows)
aa fragments were visible starting from day 4 upon induction, and are detected with htt552 and htt513 antibodies. Non-specific bands are
LA

their mobility was consistent with the region previously mapped for marked with NS.
caspase 2 cleavage (Fig. 2B). This antibody also had some reactivity
with uncleaved Htt. Some caspase 3 generated fragments were also
07

detected using neoepitope antibody to caspase 3 site of Htt (DSVD)


20

htt513 (Fig. 2C), however the signal was low. Thus, we showed that fragments were stabilized, and could be detected without proteasome
our stable PC12 cell model reveals several steps of Htt proteolysis inhibition. To investigate whether our fragments are identical to cp-A
©

including caspase-mediated proteolysis within the caspase/calpain and cp-B, we have generated PC12 cell lines expressing FL-Htt-126Q
cleavage domain, and N-terminal proteolysis producing shorter frag- with “cp-A cleavage site” deletions. Using at least three independent
ments (Fig. 1B). cell lines, we demonstrate that deletion of residues 105–114 does not
N-Terminal cp-1/cp-2 fragments may be distinct from cp-A/ affect the formation of 60 kD N-terminal fragments in stable PC12
cp-B. The domain containing the cleavage site for cp-A was previ- lines expressing FL-Htt-126Q (Fig. 3A and data not shown). The
ously mapped to the region between residues 105 and 114 in substitution of alanines at positions 109–113 (CENI to AAAA) also
NG108-1518 and striatal cells,44 expressing expanded Htt. However, did not alter the production of short N-terminal fragments in three
cp-A cleavage product was detected previously only in the presence of independent PC12 lines (data not shown). Therefore, we conclude
proteasome inhibitors, whereas in our PC12 cell model cp-1 and cp-2 that cp-1 and cp-2 fragments are distinct from the previously

www.landesbioscience.com Cell Cycle 2973


N-terminal Proteolysis of Huntingtin in PC12 Cell Model

described cp-A and cp-B.


To map cp-1 and cp-2 fragments
we used antibodies recognizing
different Htt epitopes, and detected
the fragments in human embryonic
kidney (HEK) 293 cells transiently
expressing truncated Htt (N1212-15Q
and N1212-138Q), or FL-Htt-148Q
(Fig. 3B, right panel). We found that
HEK293 cells transfected with either
N1212-138Q, or FL-Htt-148Q
produce fragments which co-migrate
with cp-1 and cp-2 fragments from

E .
PC12 detected with either antibody to

UT
Htt, or anti-c-myc antibody (Fig. 3B,
left panel). The fragments produced

RIB
from N1212-138Q in HEK293 cells
(Fig. 3B, lanes 6 and 9) migrated
slightly faster than those produced

IST
from FL-Htt-148Q (lanes 7 and 10),
which is consistent with the different

D
number of glutamines. Notably, two
N-terminal fragments produced from

OT
N1212-15Q construct (lanes 5 and
8) can be easily separated on a gel,
ON whereas fragments with expanded
polyglutamine (138Q) appear as a
single band unresolved in these condi-
.D
tions. In HEK293 cells both cp-1
and cp-2 were detected with anti-
CE

bodies recognizing N-terminal c-myc


tag (data not shown) and epitopes
IEN

between residues 55 and 66 of Htt


(Fig. 3B, lanes 5–7), and between
SC

residues 81 and 90 of Htt (Fig. 3B,


lanes 8–10). However, only cp-2 frag-
BIO

ment was detected with the antibody


against an epitope between residues
Figure 3. N-terminal cp-1/ cp-2 fragments may be distinct from cp-A/cp-B. (A) “Cp-A cleavage site” deletion 115 and 129 of Htt (Fig. 3B, lanes
ES

(18) does not affect the formation of cp-1/cp-2 fragments in PC12 cells. Western blot of total cell extracts 12 and 13). Since cp-1 and cp-2 frag-
(60 µg per lane) from stable PC12 cells induced to express wild type FL-Htt-126Q (line S6-5, left panel), or ments with expanded polyglutamine
ND

mutant FL-Htt-153Q with amino acids 105-114 removed (right panel) for the indicated number of days. Htt is
detected with anti-c-myc antibody, FL-Htt is indicated with arrowheads, cp-1/cp-2 fragments are indicated by
can not be resolved on this gel, we
arrows. Left panel was assembled from 2 blots. One representative deletion mutant PC12 cell line out of three only observed a substantial decrease in
LA

is shown. (B) Epitope mapping of cp-1/cp-2 fragments. Left panel- Western blot of total cell extracts (60 µg signal from detection of cp-2 fragment
per lane) from stable PC12 cells induced to express FL-Htt-126Q for eight days (lanes 2 and 4), or un-induced (lane 13), consistent with the lack
(lanes 1 and 3). Full-length Htt (arrowheads) and cp-1/cp-2 fragments (arrows) are detected with antibody to of recognition of cp-1 by this anti-
07

residues 55-66 of Htt (lane 2), or anti-c-myc antibody (lane 4). Right panel- Western blot of total cell extracts
(30 µg per lane) from HEK293 cells transfected with either FL-Htt-148Q (lanes 7 and 10), truncated Htt con-
body. Thus epitope mapping indicate
that cleavage producing cp-1 fragment
20

structs (N1212-15Q or N1212-138Q, lanes 5, 8, 12 and lanes 6, 9, 13 respectively), or non-transfected


cells (Cont). Full-length and truncated Htt (arrowheads) and cp-1/cp-2 fragments (arrows) are detected with occurs between the epitopes 81–90,
©

antibody to residues 55-66 of Htt (lanes 5–7), with antibody to residues 81–90 of Htt (lanes 8–10), or with and 115–129 of Htt. Based on muta-
antibody to residues 115–129 of Htt (lanes 11–13). HEK293 cells transfected with expanded Htt constructs genesis studies in PC12 cells, described
produce fragments which co-migrated with cp-1/cp-2 fragments detected in PC12. Notably, two N-terminal
fragments produced from N1212-15Q construct (lane 5) can be easily separated on a gel, whereas fragments
above, the cleavage site producing cp-1
with expanded polyglutamine (138Q) appear as a single band unresolved on this gel. Lanes 5–7 are from the is not likely to lie between residues 105
same blot, which is also shown on Figure 6C. Non-specific bands are marked with NS. and 114. Combined, these results map
the position of cleavage generating
cp-1 to be between the epitope 81–90
and residue 105 of Htt. The exact
position of the site of cleavage gener-

2974 Cell Cycle 2007; Vol. 6 Issue 23


N-terminal Proteolysis of Huntingtin in PC12 Cell Model

E .
UT
RIB
D IST
OT
ON
.D
CE
IEN

Figure 4. N-terminal fragments of Htt are found in nuclear inclusions and accumulate in the aggregate fraction in PC12 cells. (A) Immunofluorescent
SC

detection of N-terminal fragments in PC12 cells (line S6) induced to express FL-Htt-126Q for 8 days. C-myc epitope is shown in green; epitopes between
residues 497–513 or residues 55–66 of Htt are shown in red; the nuclear staining (DAPI) is shown in blue. The right panel represents the merged image.
BIO

(B–D) Disaggregation of nuclear inclusions in PC12 cells. Two procedures were used in parallel to fractionate and dissociate aggregated Htt fragments
(B, see experimental procedures). Enrichment for cytoplasmic and nuclear proteins in fractions C and N respectively was confirmed by the presence of
β-tubulin and PARP in corresponding fractions obtained using both protocols (C). (D) Western blot analysis of three fractions obtained from PC12 cells (line S6)
differentiated and induced to express FL-Htt-126Q for 8 days. Htt was detected with antibodies to c-myc, or to residues 55–66 of Htt. FL- Htt-126Q is
ES

indicated with arrowheads, cp-1/cp-2 fragments - with arrows. Fractionation of material obtained with other two independent cell lines produced similar
results (data not shown).
ND
LA

ating cp-2 fragment (which extends beyond 115–129 epitope of Htt of nuclear inclusions, whereas cytoplasmic aggregates may contain
according to epitope mapping) remains to be identified. diverse mixture of Htt derivatives, including longer fragments.
07

Short N-terminal fragments build up nuclear inclusions and To confirm that short N-terminal fragments are incorporated into
accumulate in aggregate fraction. To determine the subcellular nuclear aggregates, we used (previously described in refs. 18 and 23)
20

localization of short N-terminal fragments in PC12 cells expressing biochemical solubilization of aggregates with formic acid (FA, Fig.
expanded Htt, we performed immunofluorescence staining with 4B–D). Using antibodies to c-myc or to Htt (epitope 55–66 aa), we
©

antibodies recognizing different Htt epitopes (Fig. 4A). Our results detected FL-Htt-126Q protein mostly in SDS soluble cytoplasmic
indicate that most nuclear inclusions can be labeled with antibodies fraction C, and to a lesser extent in SDS soluble nuclear fraction
recognizing N-terminal epitopes (c-myc and an epitope between N (Fig. 4D). FA-soluble nuclear fraction F was mostly composed
amino acids 55–66), but not with antibodies against further of N-terminal fragments co-migrating with cp-1 and cp-2 frag-
C-terminal epitope (amino acids 497–513). In contrast, most of the ments detected in the soluble cytoplasmic fraction. Fraction F also
C-terminal epitopes were detected in cytoplasmic aggregates, or were contained small amounts of longer fragments probably generated
diffusely spread around the cytoplasm. This staining pattern is consis- by caspases, but did not contain detectable levels of FL-Htt-126Q.
tent with the idea that N-terminal fragments are main components The presence of some FL-Htt-126Q in soluble nuclear fraction N

www.landesbioscience.com Cell Cycle 2975


N-terminal Proteolysis of Huntingtin in PC12 Cell Model

cells that have lost membrane integrity. About 12% of the cells
were positive for either annexin V alone, or for both dyes on day 8
after induction of expression of expanded Htt, compared to ~2% in
control (Fig. 5B).
N-terminal fragments can be formed independently of caspase
cleavage. Differentiated PC12 cells accumulate two kinds of expanded
Htt cleavage products: approximately 100 kD caspase generated frag-
ments, and shorter N-terminal fragments potentially produced by
some unknown protease. The results of the time-course experiment
(Fig. 2) are consistent with both sequential and parallel models of Htt
cleavage. To further investigate whether the initial cleavage of Htt by
caspases /calpains is necessary for generation of cp-1 and cp-2 frag-
ments, differentiated PC12 cells, induced to express FL-Htt-126Q,

E .
were treated with caspase or calpain inhibitors (Fig. 6A). Specific

UT
inhibitors to caspase 2, 3, and 6, as well as pan-specific caspase inhib-
itor and specific calpain 1/2 inhibitor (MDL 28170) were added to

RIB
PC12 cells on day 2 after induction of Htt expression. On day 8
upon induction the cells were harvested and Htt fragments were visu-
alized with either anti-c-myc antibody, or antibodies to Htt (data not

IST
shown). Our data indicate that inhibition of caspases 2, 3 or 6 fails
to affect the levels of N-terminal fragments, however treatment of

D
PC12 cells with calpain 1/2 inhibitor results in a significant increase
in levels of cp-1 and cp-2 fragments (Fig. 6A, lane 5). To verify that

OT
caspase inhibitors efficiently blocked caspase-mediated cleavage of
Htt, neoepitope antibodies to caspase 2/3/7 cleavage site in Htt

Figure 5. Increased cell death in PC12 cells, expressing FL Htt 126Q.


ON
(DLND) htt552 were used (Fig. 6B, lanes 6–10). The production
of 552 aa caspase generated fragments was inhibited by pan-specific
(A) Cell toxicity measured by trypan blue exclusion assay. Cells expressing
caspase inhibitor (Fig. 6B, lane 6), or caspase 2 specific inhibitor
.D
FL-Htt-126Q (line S6) show significant toxicity on day 12 after induction of
Htt expression (n = 4, *p < 0.05, FL-Htt-126Q versus FL-Htt-21Q). (B) Cell (Fig. 6, lane 7), which confirms that in this system it is caspase 2 that
toxicity and apoptosis measured by staining with annexin V-EGFP and prop- is cleaving Htt at residue 552, not caspase 3. The levels of N-terminal
CE

idium iodide. On day 8 after induction of FL-Htt-126Q expression, live PC12 fragments remained unchanged (Fig. 6B, lanes 1 and 2).
cells (line S6-5) were double stained with annexinV-EGFP and propidium Independent evidence in favor of this hypothesis came from
IEN

iodide. About 12% of the cells are positive for either annexin V alone, or
for both dyes compared to ~2% in control. Five fields (200–500 cells each)
transient transfection experiments with HEK293 cells using Htt
were counted for each condition (n = 5, **p < 0.005, dox- versus dox+). N1212-Quint constructs with deleted cleavage sites for caspases 2,
SC

3 and 6.34 As described in the previous section, cp-1 and cp-2 frag-
ments are generated from Htt N1212 constructs with either 15Q or
BIO

138Q in HEK293 cells (Fig. 3B and Fig. 6C, upper panel). Caspase
might be a result of slight contamination of the nuclear fraction 2 fragments of Htt, visualized with neoepitope antibody htt552,
with cytoplasmic proteins, as revealed by detection of low levels of were only produced from the wild type Htt constructs, but not from
ES

β-tubulin in the nuclear fraction (Fig. 4C). Since most of the cyto- constructs with deleted caspase sites (compare lanes 1 and 2, with
plasmic inclusions had perinuclear localization (Fig. 3 and Fig. 8B), lanes 3 and 4, Fig. 6C, lower panel). However, generation of cp-1 and
ND

it is possible that these aggregates were co-purified with the nuclear cp-2 fragments was not affected by deletions of caspase cleavage sites
pellet (see experimental procedures), and thus FA-soluble fraction F in Htt (Fig. 6C, upper panel, lanes 3 and 4). These results, combined
LA

may contain proteins from both nuclear and cytoplasmic inclusions. with the data obtained with caspase inhibitors in PC12 cells are
In contrast, PC12 cells expressing Htt with normal repeat length highly suggestive of the N-terminal cleavage of Htt occurring via a
did not accumulate any Htt species in FA-soluble fraction (data not caspase-independent pathway in these cell systems.
07

shown). Inhibition of calpain increases accumulation of N-terminal frag-


FL-Htt-126Q induces moderate toxicity in PC12 cells. The effect ments and accelerates aggregation. Treatment of PC12 cells with
20

of expanded Htt expression on PC12 cell viability was measured by calpain inhibitor MDL28170 leads to an increase in accumulation of
©

trypan blue exclusion assay. Cells expressing FL-Htt-126Q (line cp-1 and cp-2 fragments (Fig. 6A). Since some of calpain inhibitors
S6) begin to show cell toxicity on day 12 after induction of expres- may also inhibit the proteasome activity, we further investigated the
sion and differentiation (Fig. 5A). Cells expressing Htt with normal effects of both classes of inhibitors on the production of cp-1 and cp-2
repeat length (21Q) did not show toxicity greater than baseline. fragments. When PC12 cells, induced to express FL-Htt-126Q, were
Because of only a moderate degree of cell toxicity observed on day treated with two different concentrations of calpain inhibitor MDL
12, an apoptotic marker, annexin V, was used to evaluate earlier 28170, or another calpain inhibitor, calpeptin, levels of accumulated
changes in cells expressing expanded Htt (Line S6-5, Fig. 5B). On cp-1 and cp-2 fragments increased in a dose-dependent manner (Fig.
day 8 after induction of Htt expression, live cells were double stained 7A, lanes 2–4 and 11–13, respectively). Proteasome inhibitors lacta-
with annexinV-EGFP and propidium iodine, the latter penetrates cystin and MG132 also elevated levels of cp-1 and cp-2 fragments

2976 Cell Cycle 2007; Vol. 6 Issue 23


N-terminal Proteolysis of Huntingtin in PC12 Cell Model

Figure 6. N-terminal fragments are formed


independently of caspase cleavage in PC12
and HEK293 cells. (A) Western blot analysis
of total cell extracts (60 µg per lane) from
PC12 cells induced to express FL-Htt-126Q
(line S6-5) for eight days in the presence of
the following inhibitors: caspase pan-specific,
Z-VAD, 25 µM (lane 1), caspase 2-specific,
Z-VDVAD, 10 µM (lane 2), caspase 3-specific,
AcDMQD, 25 µM (lane 3), caspase 6-specific
Z-VEID, 10 µM (lane 4), and calpain1/2-spe-
cific, MDL128170, 10 µM (lane 5). Control
extract was loaded in lane 6. Htt proteins are
detected with anti-c-myc antibody. Blot was re-
probed with the antibody to GAPDH for load-

.
ing control. (B) Total cell extracts (60 µg per

E
lane) from PC12 cells treated as described

UT
in (A) (lanes 1–4), and control extract (lane
5), analyzed by Western blot with anti-c-myc

RIB
antibody. Blot was re-probed with neoepitope
antibodies to caspase 2/3/7 cleavage site
in Htt (DLND) htt552 (lanes 6–10), and with

IST
antibody to actin for loading control. (C)
Western blot analysis of total cell extracts (30
µg per lane) from HEK293 cells transfected

D
with either wild type FL-Htt construct (lane
5), truncated Htt-N1210 constructs (lane 1

OT
and 2) or mutant Htt-N1212-Quint constructs
with deleted cleavage sites for caspases 2,
3, and 6 -D513A, D530A, D552A, D586A,
ON and D589A (lanes 3 and 4). Htt proteins are
detected with antibody to Htt (upper panel),
or htt552 antibody (lower panel). FL-Htt and
.D
Htt-N1212 are indicated with arrowheads,
cp-1/cp-2 fragments- with arrows. Non-spe-
cific bands are marked with NS. Parts of blot
CE

(C) are also shown on Figure 3B.


IEN
SC
BIO
ES

(Fig. 7A, lanes 5 and 6); however U102, an inhibitor of caspase-like to non-treated cells. However, calpain inhibitor MDL28170
ND

(PGPH) activity of the proteasome, failed to do so (Fig. 7A, lane 7). (25 µM) does not increase GFPu fluorescence, and thus it does not
It should be noted that accumulation of larger (~100 kD) Htt frag- efficiently inhibit proteasome activity (Fig. 7B). Therefore the effect
LA

ments (possibly generated by caspase or calpain cleavage), was also of MDL28170 on the accumulation of Htt fragments is likely due to
increased with both calpain and proteasome inhibition. a specific inhibition of calpains.
Since both calpain inhibitors and proteasome inhibitors affect Increased accumulation of cp-1 and cp-2 fragments in PC12
07

cp-1 and cp-2 fragments in a similar way, it was essential to rule out cells treated with calpain inhibitors may reflect increased production
the possibility that these effects of calpain inhibitors may be due to of these fragments, or reduced degradation. To evaluate these two
20

a non-specific inhibition of the ubiquitin-proteasome system (UPS). possibilities, PC12 cells expressing FL-Htt-126Q were treated with
©

This was achieved by using the green fluorescent protein (GFP) protein synthesis inhibitor cycloheximide for 17 hours. As expected,
proteasome reporter cell line GFPu-1.46 GFPu-1 HEK293 cells the basal levels of Htt were reduced by cycloheximide. Notably, a
express a reporter consisting of a short degron, CL1,47 fused to the proportional increase in cp-1/cp-2 fragments accumulation with
C-terminus of GFP. GFPu is unstable compared to GFP, however in calpain inhibition was still observed in the absence of protein
conditions of inhibition or impairment of UPS, GFPu is stabilized, synthesis in cells treated with cyclohexamide (data not shown).
and therefore GFPu fluorescence can be used as a measure of UPS Thus, the accumulation of cp-1 and cp-2 fragments with calpain
activity.46 We found that, in fact, treatment of GFPu-1 cells with inhibition does not seem to be caused by increased Htt synthesis,
proteasome inhibitors (lactacystin and MG132) leads to impairment and therefore it may result from altered clearance of cp-1 and cp-2
of UPS, measured by a robust increase in fluorescence, compared fragments. To test the effect of calpain inhibitors on cp-1 and cp-2

www.landesbioscience.com Cell Cycle 2977


N-terminal Proteolysis of Huntingtin in PC12 Cell Model

Since our data suggest that cp-1 and


cp-2 fragments may be substrates for
calpains, we next tested whether incuba-
tion of PC12 lysates with calpain1 and
2 will facilitate cleavage of expanded Htt
fragments in vitro (Fig. 8C). Clearly, both
full-length Htt and its cp-1 and cp-2
fragments are efficiently digested upon
addition of calpain 1 or calpain 2 at higher
concentration, evidenced by a decrease
in FL-Htt and fragments levels, and by
appearance of lower molecular weight
bands representing Htt cleavage products.

E .
This increase of degradation was blocked

UT
by calpain inhibitors. Notably, the longer
fragments (~100 kD) potentially gener-

RIB
ated by caspase/calpain cleavage were also
affected by calpains in vitro.
Overall, inhibition of calpains leads to

IST
abnormal degradation of cp-1 and cp-2
fragments, and, as a result, to a dramatic

D
increase in accumulated fragments, and
accelerated formation of inclusions. Thus,

OT
calpain mediated cleavage may be a first
step in the clearance of cp-1/cp-2, followed
ON by their complete breakdown by protea-
some machinery.
.D
DISCUSSION
CE

Numerous reports indicate the important


role of Htt proteolysis in the pathogenesis
IEN

of HD.15-31 Studies of Htt cleavage have


Figure 7. Calpain and proteasome inhibitors increase accumulation of Htt N-terminal fragments in PC12 been focused on two domains within the
cells. (A) Western blot analysis of total cell extracts (60 µg per lane) from PC12 cells induced to express
SC

Htt sequence that are highly susceptible to


FL-Htt-126Q (line S6-5) for eight days, and treated with the following inhibitors for 24 h: calpain inhibi-
proteolysis: cleavage by caspases or calpains
tor MDL128170, 10 µM (lane 3) or 25 µM (lanes 4 and 8); calpeptin, 50 µM (lane 13) or 75 µM
BIO

(lane 12); lactacystin, 10 µM (lane 5); MG132, 0.5 µM (lane 6); U102, 10 µM (lane 7), non-treated within a region between about 460 and
control cells (lanes 2, 9, 11), or non-induced cells (lanes 1 and 10). Lanes 1–5 are from the same blot. 600 amino acids from the N-terminus,
Htt proteins are detected with anti-c-myc antibody. Blots were re-probed with antibody to actin for load- and cleavage by unknown proteases near
ing control. FL-Htt is indicated by arrowheads, caspase fragments—by arrows, cp-1/cp-2 fragments—by the N-terminus (Fig. 1B). The application
ES

double arrows. (B) GFPu fluorescence of GFPu-1 HEK293 cells treated with lactacystin (10 µM), MG132
of previously described stable cell models
(0.5 µM), or MDL28170 (25 µM).
ND

of HD to the study of Htt proteolysis has


been limited, because most of these cell
degradation, we determined fragments levels during 21 h in the lines express truncated forms of Htt, which do not contain both
LA

presence of cycloheximide (Fig. 8A). When the protein synthesis is Htt domains prone to proteolysis.24-26,48 In two reports focused on
blocked, a decline in the steady state levels of a protein reflects the the characterization of the N-terminal cleavage step, the mapping
07

rate of its degradation. Our time course experiments demonstrate of the cleavage sites were accomplished using either truncated Htt
that the rate of cp-1 and cp-2 clearance is markedly diminished in constructs, or transient overexpression of full-length Htt in condi-
20

the presence of calpain inhibitors, and is decreased even more in the tions involving proteasome inhibition.18,44
presence of lactacysin (Fig. 8A). To further investigate the Htt proteolytic pathway and the
©

As described above, cp-1 and cp-2 fragments mostly accumulate possible relationships among different cleavage events, an inducible
in the aggregate fraction (Fig. 4). To determine whether reduced PC12 cell model expressing full-length expanded Htt was developed.
degradation of the fragments caused by calpain inhibition will result In our model both caspase-derived fragments and short N-terminal
in more aggregation, we performed immunofluorescent staining of fragments cp-1 and cp-2 accumulate prior to formation of inclu-
PC12 cells, treated with calpain inhibitor (Fig. 8B). We found that sions and Htt induced toxicity. Unlike previous reports, the novel
these cells form much more aggregates than untreated cells, most of N-terminal fragments cp-1 and cp-2 described here are detectable
the inclusions being localized to nuclear and perinuclear compartments. without proteasome inhibition, which may be more relevant to

2978 Cell Cycle 2007; Vol. 6 Issue 23


N-terminal Proteolysis of Huntingtin in PC12 Cell Model

cell physiology. The generation of


these cp-1 and cp-2 fragments is not
affected by the deletion of residues
105–114. Therefore, our cp-1 and
cp-2 fragments appear to be distinct
from the cp-A and cp-B fragments
described by Trottier’s group.18 They
appear very similar to the short
N-terminal fragments observed in
our inducible mouse model of HD
(described previously, ref. 32).
Caspase- mediated cleavage of
Htt has been described in vitro,

E .
and fragments derived from caspase

UT
cleavage accumulate in brains of HD
patients.33,36 In the YAC 128 trans-

RIB
genic mouse model of HD, alteration
of the Htt caspase 6 cleavage site at
residue 586 completely ameliorate

IST
pathology, indicating the impor-
tance of caspase 6 mediated cleavage

D
of Htt.39 However it is not clear
whether initial cleavage of Htt by

OT
caspases is a necessary first step
in a sequential proteolytic pathway
ON generating short N-terminal frag-
ments, or whether these fragments
can be produced independently from
.D
caspase cleavage. Our new data indi-
cate that in PC12 cells Htt can be
CE

cleaved by caspase 2 at residue 552,


as well as caspase 3 at residue 513.
IEN

This cleavage event does not seem


to affect the production of shorter
SC

N-terminal fragments of Htt cp-1


and cp-2, consistent with the parallel
BIO

cleavage model in these system. This


suggests that in PC12 cells caspase 2
derived Htt fragments (552 aa long)
ES

and shorter cp-1 and cp-2 frag-


Figure 8. Inhibition of calpain increases stability of cp1/2 fragments and accelerates aggregation in PC12 ments may have diverse functional
ND

cells expressing FL-Htt-126Q. (A) Cp-1/cp-2 levels determined by Western blotting with anti-c-myc antibody of roles, and may be generated, active,
proteins from PC12 cells (line S6-5) treated with calpain inhibitor MDL28170 or lactacystin in the presence
of cycloheximide, or cells treated with cycloheximide alone (control) for indicated time periods. Cells were
or cleared in different sub-cellular
LA

harvested on day 8 post induction. One representative blot out of three is shown. Equal loading was con- compartments. Experiments with
firmed by staining of the blots after transfer with Ponceau-S-red (not shown). The graph shows quantification of HEK293 cells, which overexpress
cp-1/cp-2 protein levels from 3 experiments described above (n = 3). Results are means ± standard deviation Htt N1212-Quint constructs with
07

and expressed as % of initial level. (B) Immunofluorescent detection of Htt aggregates in PC12 cells induced to deleted cleavage sites for caspases 2,
express FL-Htt-126Q (line S6-5) for eight days in the absence or presence of calpain inhibitor (MDL28170, 10
3 and 6, also failed to demonstrate a
20

µM). Htt expression is detected with anti-c-myc antibody (green), nuclei are stained with DAPI (blue). Aggregates
are indicated with arrows. One representative field is shown for each condition. The graph shows quantification correlation between cleavage of Htt
©

of aggregation. Five fields combined containing 200–400 cells were counted on each of three different plates by caspases and the formation of
(n = 3, ***p < 0.005 with calpain inhibitor versus control). (C) Cleavage of FL-Htt-126Q and cp-1/cp-2 frag- cp-1 and cp-2 fragments. However,
ments by calpain-1 and -2 in vitro. PC12 cell lysates (100 µg) from cells expressing FL-Htt-126Q (line S6-5) for it should be noted that blocking
eight days were incubated with the indicated amounts of calpain 1 and 2 for 30 min at 30˚C in the absence or
presence of calpain inhibitor (MDL28170, 25 µM). Htt proteins are detected with anti-c-myc antibody. Note the
caspase cleavage sites might facilitate
decrease in FL-Htt (arrowhead) and cp-1/cp-2 fragments (arrows), and the appearance of low molecular weight cleavage at other sites (generating
bands with higher concentration of calpains. shorter N-terminal fragments),
which are normally not favorable
within the context of normal Htt in
vivo. Notably, we found that caspase

www.landesbioscience.com Cell Cycle 2979


N-terminal Proteolysis of Huntingtin in PC12 Cell Model

6 is expressed at very low levels by PC12 cells (our unpublished Acknowledgements


observations), therefore the possible connection between caspase 6 This work was supported by HDSA Coalition for the Cure, High
mediated cleavage and the N-terminal proteolysis of Htt remains to Q Foundation, NINDS 16375, NIH NS038144-08, and Hood
be evaluated in more physiologically relevant cell systems and mouse College Board of Associates Grant to RRH. We thank Lisa Ellerby
models. (the Buck Institute for Age Research, Novato, CA) for constructs
Two calpain cleavage sites have been identified at amino acids 469 and discussions, and greatly appreciate a gift of htt552 and htt513
and 536 of Htt.40 Alteration of these sites reduced Htt toxicity and antibodies from Sophie Roy (Merck Canada Co. Ltd., Canada)
aggregation in cells, suggesting that calpains may generate toxic Htt References
fragments.40 Our data indicate that calpain may be also involved in 1. A novel gene containing a trinucleotide repeat that is expanded and unstable on Huntington’s
disease chromosomes. The Huntington’s Disease Collaborative Research Group. Cell 1993;
other steps of Htt proteolysis and clearance. We suggest that short 72:971-83.
N-terminal Htt fragments cp-1 and cp-2 are degraded via calpain 2. Ross CA, Margolis RL, Rosenblatt A, Ranen NG, Becher MW, Aylward E. Huntington
dependent pathway in PC12 cells, and that inhibition of calpains disease and the related disorder, dentatorubral-pallidoluysian atrophy (DRPLA). Medicine
(Baltimore) 1997; 76:305-38.
results in increased accumulation of the cp-1 and cp-2 fragments,

.
3. Sharp AH, Loev SJ, Schilling G, Li SH, Li XJ, Bao J, Wagster MV, Kotzuk JA, Steiner JP,

E
and increased aggregation. Because two specific calpain inhibitors Lo A, et al. Widespread expression of Huntington’s disease gene (IT15) protein product.

UT
caused similar changes in the levels of cp-1 and cp-2, and these Neuron 1995; 14:1065-74.
inhibitors did not affect the UPS, their effect on cp-1 and cp-2 frag- 4. Velier J, Kim M, Schwarz C, Kim TW, Sapp E, Chase K, Aronin N, DiFiglia M. Wild-type
and mutant huntingtins function in vesicle trafficking in the secretory and endocytic path-

RIB
ments accumulation is likely due to a specific inhibition of calpains, ways. Exp Neurol 1998; 152:34-40.
rather than possible non-specific effects on other proteases or protea- 5. DiFiglia M, Sapp E, Chase K, Schwarz C, Meloni A, Young C, Martin E, Vonsattel JP,
some. This raises the possibility that cp-1/cp-2 fragments may be Carraway R, Reeves SA, et al. Huntingtin is a cytoplasmic protein associated with vesicles

IST
in human and rat brain neurons. Neuron 1995; 14:1075-81.
processed by calpains before subsequent clearance by the proteasome. 6. Yanai A, Huang K, Kang R, Singaraja RR, Arstikaitis P, Gan L, Orban PC, Mullard A,
A similar multi-step degradation pathway has been described for a Cowan CM, Raymond LA, Drisdel RC, Green WN, Ravikumar B, Rubinsztein DC,
number of transcription factors, including p53.49,50 Toxic mutant

D
El-Husseini A, Hayden MR. Palmitoylation of huntingtin by HIP14 is essential for its traf-
ficking and function. Nat Neurosci 2006; 9:824-31.
Htt fragments resistant to degradation by proteasome were found to 7. Suopanki J, Gotz C, Lutsch G, Schiller J, Harjes P, Herrmann A, Wanker EE. Interaction

OT
be accumulated in HD neurons.51-53 Thus our findings that calpains of huntingtin fragments with brain membranes-clues to early dysfunction in Huntington’s
may mediate mutant Htt clearance are consistent with the idea that disease. J Neurochem 2006; 96:870-84.

activation of calpains in HD neurons27 may represent a cellular


protective mechanism that facilitates the clearance of toxic mutant
ON
8. Kegel KB, Sapp E, Yoder J, Cuiffo B, Sobin L, Kim YJ, Qin ZH, Hayden MR, Aronin
N, Scott DL, Isenberg G, Goldmann WH, DiFiglia M. Huntingtin associates with acidic
phospholipids at the plasma membrane. J Biol Chem 2005; 280:36464-73.
Htt fragments. 9. Ross CA, Poirier MA. Protein aggregation and neurodegenerative disease. Nat Med 2004;
.D
10(Suppl):S10-7.
The proteases that generate cp-1 and cp-2 fragments of Htt in
10. Ross CA, Poirier MA. Opinion: What is the role of protein aggregation in neurodegenera-
PC12 cells remain to be identified. The involvement of an aspartyl tion? Nat Rev Mol Cell Biol 2005; 6:891-8.
CE

protease in the generation of cp-A fragment have been suggested 11. Berman SB, Greenamyre JT. Update on Huntington’s disease. Curr Neurol Neurosci Rep
previously in ref. 18. However, in our PC12 cells, expressing 2006; 6:281-6.
IEN

12. Borrell-Pages M, Zala D, Humbert S, Saudou F. Huntington’s disease: From huntingtin


FL-Htt-126Q, levels of cp-1/cp-2 fragments were not affected by function and dysfunction to therapeutic strategies. Cell Mol Life Sci 2006; 63:2642-60.
aspartyl protease inhibitor pepstatin A at 5 or 100 µM (data not 13. Sadri-Vakili G, Cha JH. Mechanisms of disease: Histone modifications in Huntington’s
shown), indicating that cp-1/cp-2 are formed via distinct proteolytic disease. Nat Clin Pract Neurol 2006; 2:330-8.
SC

14. Li S, Li XJ. Multiple pathways contribute to the pathogenesis of Huntington disease. Mol
pathway. Neurodegener 2006; 1:19.
In our current model, expression of mutant full-length Htt
BIO

15. Tarlac V, Storey E. Role of proteolysis in polyglutamine disorders. J Neurosci Res 2003;
induces relatively modest toxicity in PC12 cells, compared with 74:406-16.
greater cell death in our previously described inducible PC12 cell 16. Becher MW, Kotzuk JA, Sharp AH, Davies SW, Bates GP, Price DL, Ross CA. Intranuclear
neuronal inclusions in Huntington’s disease and dentatorubral and pallidoluysian atro-
model expressing a short N-terminal (N63) Htt fragment.25 This
ES

phy: Correlation between the density of inclusions and IT15 CAG triplet repeat length.
is consistent with the observations that N-terminal Htt fragments Neurobiol Dis 1998; 4:387-97.
are usually more toxic than full-length Htt. In the PC12 cell model 17. DiFiglia M, Sapp E, Chase KO, Davies SW, Bates GP, Vonsattel JP, Aronin N. Aggregation
ND

of huntingtin in neuronal intranuclear inclusions and dystrophic neurites in brain. Science


described here, cp-1 and cp-2 fragments are generated prior to 1997; 277:1990-3.
toxicity, suggesting the possibility of causal relationship. Calpain 18. Lunkes A, Lindenberg KS, Ben-Haiem L, Weber C, Devys D, Landwehrmeyer GB, Mandel
LA

inhibitors had a profound effect on cell toxicity even in uninduced JL, Trottier Y. Proteases acting on mutant huntingtin generate cleaved products that differ-
entially build up cytoplasmic and nuclear inclusions. Mol Cell 2002; 10:259-69.
PC12 cells, so their effects on toxicity in relation to levels of cp-1 19. Mende-Mueller LM, Toneff T, Hwang SR, Chesselet MF, Hook VY. Tissue-specific proteol-
and cp-2 were not interpretable. Future studies will be focused on the
07

ysis of Huntingtin (htt) in human brain: Evidence of enhanced levels of N- and C-terminal
mapping of the exact sites of the cleavage events producing cp-1 and htt fragments in Huntington’s disease striatum. J Neurosci 2001; 21:1830-7.
20

20. Schilling G, Klevytska A, Tebbenkamp AT, Juenemann K, Cooper J, Gonzales V, Slunt H,


cp-2 fragments, and testing the effects of alterations of these sites on
Poirer M, Ross CA, Borchelt DR. Characterization of huntingtin pathologic fragments in
mutant Htt induced toxicity. This will make it possible to evaluate human Huntington disease, transgenic mice, and cell models. J Neuropathol Exp Neurol
©

whether proteolytic cleavage of Htt producing short N-terminal 2007; 66:313-20.


fragments (cp-1 and cp-2) is a critical step in a pathogenic pathway 21. Sun B, Fan W, Balciunas A, Cooper JK, Bitan G, Steavenson S, Denis PE, Young Y, Adler
B, Daugherty L, Manoukian R, Elliott G, Shen W, Talvenheimo J, Teplow DB, Haniu
leading to HD. M, Haldankar R, Wypych J, Ross CA, Citron M, Richards WG. Polyglutamine repeat
In conclusion, Htt cleavage is emerging as a complex pathogenic length-dependent proteolysis of huntingtin. Neurobiol Dis 2002; 11:111-22.
process, generating several toxic fragments potentially relevant to HD 22. Kim M, Lee HS, LaForet G, McIntyre C, Martin EJ, Chang P, Kim TW, Williams M,
Reddy PH, Tagle D, Boyce FM, Won L, Heller A, Aronin N, DiFiglia M. Mutant hun-
pathogenesis. The novel inducible PC12 cell model described here tingtin expression in clonal striatal cells: Dissociation of inclusion formation and neuronal
will be used for further elucidation of these cleavage events, which survival by caspase inhibition. J Neurosci 1999; 19:964-73.
may provide potential therapeutic targets for HD within the pathway 23. Lunkes A, Mandel JL. A cellular model that recapitulates major pathogenic steps of
Huntington’s disease. Hum Mol Genet 1998; 7:1355-61.
of Htt proteolysis.

2980 Cell Cycle 2007; Vol. 6 Issue 23


N-terminal Proteolysis of Huntingtin in PC12 Cell Model

24. Cooper JK, Schilling G, Peters MF, Herring WJ, Sharp AH, Kaminsky Z, Masone J, Khan 46. Bence NF, Sampat RM, Kopito RR. Impairment of the ubiquitin-proteasome system by
FA, Delanoy M, Borchelt DR, Dawson VL, Dawson TM, Ross CA. Truncated N-terminal protein aggregation. Science 2001; 292:1552-5.
fragments of huntingtin with expanded glutamine repeats form nuclear and cytoplasmic 47. Gilon T, Chomsky O, Kulka RG. Degradation signals for ubiquitin system proteolysis in
aggregates in cell culture. Hum Mol Genet 1998; 7:783-90. Saccharomyces cerevisiae. EMBO J 1998; 17:2759-66.
25. Igarashi S, Morita H, Bennett KM, Tanaka Y, Engelender S, Peters MF, Cooper JK, Wood 48. Saudou F, Finkbeiner S, Devys D, Greenberg ME. Huntingtin acts in the nucleus to induce
JD, Sawa A, Ross CA. Inducible PC12 cell model of Huntington’s disease shows toxicity apoptosis but death does not correlate with the formation of intranuclear inclusions. Cell
and decreased histone acetylation. Neuroreport 2003; 14:565-8. 1998; 95:55-66.
26. Zhou H, Cao F, Wang Z, Yu ZX, Nguyen HP, Evans J, Li SH, Li XJ. Huntingtin forms 49. Kubbutat MH, Vousden KH. Proteolytic cleavage of human p53 by calpain: A potential
toxic NH2-terminal fragment complexes that are promoted by the age-dependent decrease regulator of protein stability. Mol Cell Biol 1997; 17:460-8.
in proteasome activity. J Cell Biol 2003; 163:109-18. 50. Wei W, Yang H, Cao P, Menconi M, Chamberlain C, Petkova V, Hasselgren PO.
27. Gafni J, Ellerby LM. Calpain activation in Huntington’s disease. J Neurosci 2002; Degradation of C/EBPbeta in cultured myotubes is calpain-dependent. J Cell Physiol 2006;
22:4842-9. 208:386-98.
28. Mangiarini L, Sathasivam K, Seller M, Cozens B, Harper A, Hetherington C, Lawton M, 51. Waelter S, Boeddrich A, Lurz R, Scherzinger E, Lueder G, Lehrach H, Wanker EE.
Trottier Y, Lehrach H, Davies SW, Bates GP. Exon 1 of the HD gene with an expanded CAG Accumulation of mutant huntingtin fragments in aggresome-like inclusion bodies as a result
repeat is sufficient to cause a progressive neurological phenotype in transgenic mice. Cell of insufficient protein degradation. Mol Biol Cell 2001; 12:1393-407.
1996; 87:493-506. 52. Venkatraman P, Wetzel R, Tanaka M, Nukina N, Goldberg AL. Eukaryotic proteasomes
29. Schilling G, Becher MW, Sharp AH, Jinnah HA, Duan K, Kotzuk JA, Slunt HH, Ratovitski cannot digest polyglutamine sequences and release them during degradation of polygluta-

.
T, Cooper JK, Jenkins NA, Copeland NG, Price DL, Ross CA, Borchelt DR. Intranuclear mine-containing proteins. Mol Cell 2004; 14:95-104.

E
inclusions and neuritic aggregates in transgenic mice expressing a mutant N-terminal frag- 53. Holmberg CI, Staniszewski KE, Mensah KN, Matouschek A, Morimoto RI. Inefficient

UT
ment of huntingtin. Hum Mol Genet 1999; 8:397-407. degradation of truncated polyglutamine proteins by the proteasome. EMBO J 2004;
30. Rubinsztein DC. Lessons from animal models of Huntington’s disease. Trends Genet 2002; 23:4307-18.
18:202-9.

RIB
31. Slow EJ, Graham RK, Osmand AP, Devon RS, Lu G, Deng Y, Pearson J, Vaid K, Bissada
N, Wetzel R, Leavitt BR, Hayden MR. Absence of behavioral abnormalities and neurode-
generation in vivo despite widespread neuronal huntingtin inclusions. Proc Natl Acad Sci

IST
USA 2005; 102:11402-7.
32. Tanaka Y, Igarashi S, Nakamura M, Gafni J, Torcassi C, Schilling G, Crippen D, Wood JD,
Sawa A, Jenkins NA, Copeland NG, Borchelt DR, Ross CA, Ellerby LM. Progressive phe-
notype and nuclear accumulation of an amino-terminal cleavage fragment in a transgenic

D
mouse model with inducible expression of full-length mutant huntingtin. Neurobiol Dis
2006; 21:381-91.

OT
33. Wellington CL, Ellerby LM, Hackam AS, Margolis RL, Trifiro MA, Singaraja R,
McCutcheon K, Salvesen GS, Propp SS, Bromm M, Rowland KJ, Zhang T, Rasper D, Roy
S, Thornberry N, Pinsky L, Kakizuka A, Ross CA, Nicholson DW, Bredesen DE, Hayden
MR. Caspase cleavage of gene products associated with triplet expansion disorders generates
truncated fragments containing the polyglutamine tract. J Biol Chem 1998; 273:9158-67.
34. Wellington CL, Singaraja R, Ellerby L, Savill J, Roy S, Leavitt B, Cattaneo E, Hackam A,
ON
Sharp A, Thornberry N, Nicholson DW, Bredesen DE, Hayden MR. Inhibiting caspase
.D
cleavage of huntingtin reduces toxicity and aggregate formation in neuronal and nonneuro-
nal cells. J Biol Chem 2000; 275:19831-8.
CE

35. Kim YJ, Yi Y, Sapp E, Wang Y, Cuiffo B, Kegel KB, Qin ZH, Aronin N, DiFiglia M.
Caspase 3-cleaved N-terminal fragments of wild-type and mutant huntingtin are pres-
ent in normal and Huntington’s disease brains, associate with membranes, and undergo
IEN

calpain-dependent proteolysis. Proc Natl Acad Sci USA 2001; 98:12784-9.


36. Wellington CL, Ellerby LM, Gutekunst CA, Rogers D, Warby S, Graham RK, Loubser O,
van Raamsdonk J, Singaraja R, Yang YZ, Gafni J, Bredesen D, Hersch SM, Leavitt BR, Roy
S, Nicholson DW, Hayden MR. Caspase cleavage of mutant huntingtin precedes neurode-
SC

generation in Huntington’s disease. J Neurosci 2002; 22:7862-72.


37. Hermel E, Gafni J, Propp SS, Leavitt BR, Wellington CL, Young JE, Hackam AS,
BIO

Logvinova AV, Peel AL, Chen SF, Hook V, Singaraja R, Krajewski S, Goldsmith PC, Ellerby
HM, Hayden MR, Bredesen DE, Ellerby LM. Specific caspase interactions and amplifica-
tion are involved in selective neuronal vulnerability in Huntington’s disease. Cell Death
Differ 2004; 11:424-38.
ES

38. Sawa A, Nagata E, Sutcliffe S, Dulloor P, Cascio MB, Ozeki Y, Roy S, Ross CA, Snyder
SH. Huntingtin is cleaved by caspases in the cytoplasm and translocated to the nucleus
via perinuclear sites in Huntington’s disease patient lymphoblasts. Neurobiol Dis 2005;
ND

20:267-74.
39. Graham RK, Deng Y, Slow EJ, Haigh B, Bissada N, Lu G, Pearson J, Shehadeh J, Bertram
L, Murphy Z, Warby SC, Doty CN, Roy S, Wellington CL, Leavitt BR, Raymond LA,
LA

Nicholson DW, Hayden MR. Cleavage at the caspase-6 site is required for neuronal dys-
function and degeneration due to mutant huntingtin. Cell 2006; 125:1179-91.
40. Gafni J, Hermel E, Young JE, Wellington CL, Hayden MR, Ellerby LM. Inhibition of
07

calpain cleavage of huntingtin reduces toxicity: Accumulation of calpain/caspase fragments


in the nucleus. J Biol Chem 2004; 279:20211-20.
41. Kim M, Roh JK, Yoon BW, Kang L, Kim YJ, Aronin N, DiFiglia M. Huntingtin is degraded
20

to small fragments by calpain after ischemic injury. Exp Neurol 2003; 183:109-15.
42. Bizat N, Hermel JM, Boyer F, Jacquard C, Creminon C, Ouary S, Escartin C, Hantraye P,
©

Kajewski S, Brouillet E. Calpain is a major cell death effector in selective striatal degenera-
tion induced in vivo by 3-nitropropionate: Implications for Huntington’s disease. J Neurosci
2003; 23:5020-30.
43. Goffredo D, Rigamonti D, Tartari M, De Micheli A, Verderio C, Matteoli M, Zuccato C,
Cattaneo E. Calcium-dependent cleavage of endogenous wild-type huntingtin in primary
cortical neurons. J Biol Chem 2002; 277:39594-8.
44. Kim YJ, Sapp E, Cuiffo BG, Sobin L, Yoder J, Kegel KB, Qin ZH, Detloff P, Aronin N,
DiFiglia M. Lysosomal proteases are involved in generation of N-terminal huntingtin frag-
ments. Neurobiol Dis 2006; 22:346-56.
45. Qin ZH, Wang Y, Kegel KB, Kazantsev A, Apostol BL, Thompson LM, Yoder J, Aronin N,
DiFiglia M. Autophagy regulates the processing of amino terminal huntingtin fragments.
Hum Mol Genet 2003; 12:3231-44.

www.landesbioscience.com Cell Cycle 2981

You might also like