You are on page 1of 8

THE JOURNAL OF BIOLOGICAL CHEMISTRY Vol. 280, No. 19, Issue of May 13, pp.

19419 –19425, 2005


© 2005 by The American Society for Biochemistry and Molecular Biology, Inc. Printed in U.S.A.

Degradation of Trafficking-defective Long QT Syndrome Type II


Mutant Channels by the Ubiquitin-Proteasome Pathway*
Received for publication, March 2, 2005
Published, JBC Papers in Press, March 10, 2005, DOI 10.1074/jbc.M502327200

Qiuming Gong‡, David R. Keeney‡, Maurizio Molinari§, and Zhengfeng Zhou‡¶


From the ‡Division of Molecular Medicine, Department of Medicine, Oregon Health and Science University, Portland,
Oregon 97239 and §Institute for Research in Biomedicine, CH-6500 Bellinzona, Switzerland

Mutations in the human ether-a-go-go-related gene dominant negative suppression of wild type hERG channel
(hERG) cause chromosome 7-linked long QT syndrome function (4).

Downloaded from http://www.jbc.org/ at FUDAN UNIVERSITY SCHOOL OF PHARMACY on May 24, 2018
type II (LQT2). We have shown previously that LQT2 We have previously shown that the LQT2 mutation Y611H is
mutations lead to endoplasmic reticulum (ER) retention trafficking-defective (5). This mutant expresses only the imma-
and rapid degradation of mutant hERG proteins. In this ture form of hERG channel protein, which fails to reach the
study we examined the role of the ubiquitin-proteasome plasma membrane. The Y611H mutant protein is retained in
pathway in the degradation of the LQT2 mutation the endoplasmic reticulum (ER) and rapidly degraded. It is well
Y611H. We showed that proteasome inhibitors N-acetyl- recognized that newly synthesized proteins in the ER are under
L-leucyl-L-leucyl-L-norleucinal and lactacystin but not
stringent surveillance by a quality control system (6 – 8). The
lysosome inhibitor leupeptin inhibited the degradation ER quality control system ensures that only properly folded
of Y611H mutant channels. In addition, ER mannosidase
and assembled proteins are exported from the ER to the Golgi.
I inhibitor kifunensine and down-regulation of EDEM
Misfolded and unassembled proteins are retained in the ER
(ER degradation-enhancing ␣-mannosidase-like pro-
and eventually degraded by a process termed ER-associated
tein) also suppressed the degradation of Y611H mutant
channels. Proteasome inhibition but not mannosidase degradation (ERAD). According to current models, ERAD sub-
inhibition led to the accumulation of full-length hERG strates undergo retrotranslocation or dislocation from the ER
protein in the cytosol. The hERG protein accumulated to the cytosol, in which they are degraded by the proteasome
in the cytosol was deglycosylated. Proteasome inhibi- (8). In most cases, the degradation by the proteasome is pre-
tion also resulted in the accumulation of polyubiquiti- ceded by polyubiquitination. In addition, mannose trimming by
nated hERG channels. These results suggest that the ER mannosidase I and deglycosylation by cytosolic peptide:N-
degradation of LQT2 mutant channels is mediated by glycanase (PNGase) are important processes in proteasomal
the cytosolic proteasome in a process that involves man- degradation of glycoproteins (8). Recently EDEM (ER degrada-
nose trimming, polyubiquitination, and deglycosylation tion-enhancing ␣-mannosidase-like protein) has been shown to
of mutant channels. play a crucial role in the degradation of misfolded glycoproteins
(9, 10).
The importance of ERAD is underscored by the fact that
Long QT syndrome is a cardiac disorder characterized by proteasomal degradation of misfolded proteins has been shown
prolongation of QT intervals on the electrocardiogram and a in an increasing number of human diseases. The role of ERAD
high risk of sudden death due to ventricular arrhythmias. The in the degradation of misfolded proteins has been studied ex-
chromosome 7-linked form of the inherited long QT syndrome tensively in cystic fibrosis and ␣1-antitrypsin deficiency (11,
(long QT syndrome type II (LQT2))1 is caused by mutations of 12). Defective trafficking of mutant channels to the plasma
human ether-a-go-go-related gene (hERG) (1). hERG encodes membrane has been shown as the most common mechanism of
the pore-forming subunit of the rapidly activating-delayed rec- hERG channel dysfunction in LQT2. More than 15 LQT2 mu-
tifier K⫹ channel (IKr) in the heart (2, 3). Studies of LQT2 tations have been reported to cause defective trafficking of
mutant channels expressed heterologously in oocytes or mam- mutant channels (13, 14). These LQT2 mutations lead to mis-
malian cells have shown that LQT2 mutations cause hERG folding, ER retention, and the degradation of mutant hERG
channel dysfunction by multiple mechanisms including defec- channels (5, 13, 14). Although degradation of mutant hERG
tive protein trafficking, abnormal gating or permeation, and channels by the proteasome has been suggested (15, 16), little
is known about how mutant hERG channels are targeted to the
* This work was supported by Grant HL68854 (to Z. Z.) from the cytosolic proteasome for degradation. In this study, we ana-
National Institutes of Health and an award to Oregon Health and lyzed the role of the ubiquitin-proteasome pathway in the deg-
Science University under the Howard Hughes Medical Institute Bio- radation of the LQT2 mutant Y611H. Our results suggest that
medical Research Support Program for Medical Research. The costs of
publication of this article were defrayed in part by the payment of page the mutant hERG protein is targeted to the proteasome for
charges. This article must therefore be hereby marked “advertisement” degradation by dislocation from the ER membrane to the cy-
in accordance with 18 U.S.C. Section 1734 solely to indicate this fact. tosol. The proteasome-dependent degradation involves man-
¶ To whom correspondence should be addressed: Div. of Molecular
nose trimming, ubiquitination, and deglycosylation of
Medicine, NRC3, Oregon Health and Science University, 3181 S.W.
Sam Jackson Park Rd., Portland, OR 97239. Tel.: 503-494-2713; Fax: mutant channels.
503-494-7368; E-mail: zhouzh@ohsu.edu. EXPERIMENTAL PROCEDURES
1
The abbreviations used are: LQT2, long QT syndrome type II;
hERG, human ether-a-go-go-related gene; ER, endoplasmic reticulum; Reagents and Cell Transfection—The proteasome inhibitors lactacys-
ERAD, ER-associated degradation; ALLN, N-acetyl-L-leucyl-L-leucyl-L- tin and N-acetyl-L-leucyl-L-leucyl-L-norleucinal (ALLN) were purchased
norleucinal; PNGase, peptide:N-glycanase; GFP, green fluorescence from Kamiya Biomedical Co. (Seattle, WA) and Calbiochem, respec-
protein; siRNA, small interfering RNA; siGFP, GFP-directed siRNA; tively. The ER mannosidase I inhibitor kifunensine was purchased from
siEDEM, EDEM-directed siRNA. Toronto Research Chemicals, Inc. (North York, Ontario, Canada). Anti-

This paper is available on line at http://www.jbc.org 19419


19420 Proteasomal Degradation of Mutant hERG Channels
hERG antibody was raised against the hERG-thioredoxon fusion pro-
tein as described previously (5). Anti-ubiquitin monoclonal antibody
was purchased from Santa Cruz Biotechnology (Santa Cruz, CA). Anti-
calnexin antibody was purchased from Stressgen (Victoria, British Co-
lumbia, Canada). Expre35S35S protein labeling mix was purchased from
PerkinElmer Life Sciences. HEK293 cells stably expressing wild type
hERG or the Y611H mutant were cultured as described previously (5).
For down-regulation of EDEM, HEK293 cells were cotransfected with 6
FIG. 1. Effects of ALLN, lactacystin, and leupeptin on protein
␮g of pcDNA3-Y611H and 10 ␮g of pcDNA3-siEDEM or pcDNA3-siGFP
levels of wild type hERG and the Y611H mutant. HEK293 cells
utilizing Lipofectamine 2000 reagent (Invitrogen) (9).
stably transfected with wild type hERG or Y611H were treated with (⫹)
Western Blot Analysis—Western blot procedures were described pre- or without (⫺) 50 ␮M ALLN, 20 ␮M lactacystin (LACT), or 100 ␮M
viously (5). Briefly, cells were lysed in lysis buffer containing 50 mM leupeptin (LEUP) for 24 h. The cell lysates were subjected to SDS-
Tris-HCl, pH 7.4, 150 mM NaCl, 1 mM EDTA, 1% Triton X-100, and PAGE and immunoblotted with anti-hERG antibody.
proteinase inhibitors. After centrifugation at 14,000 ⫻ g for 10 min at
4 °C, cell lysates were subjected to 7.5% SDS-PAGE and electrophoreti-
cally transferred onto nitrocellulose membranes. hERG proteins were transcription kit (Ambion, Austin, TX) and biotin-16-UTP (Roche Ap-
detected with anti-hERG antibody (1:10000 dilution) and visualized plied Science). Thirty ␮g of total RNA or yeast tRNA were analyzed
with a horseradish peroxidase-conjugated second antibody and ECL with EDEM riboprobe, and 10 ␮g of total RNA or yeast tRNA were

Downloaded from http://www.jbc.org/ at FUDAN UNIVERSITY SCHOOL OF PHARMACY on May 24, 2018
detection kit. Deglycosylation of hERG proteins by PNGase was per- analyzed by actin riboprobe using RPAII and BrightStart BioDetect
formed as described previously (17). kits (Ambion). Yeast tRNA was used as a control for the complete
Metabolic Labeling and Immunoprecipitation—Cells in 35-mm digestion of the probes by RNase.
plates were pulse-labeled for 1 h in methionine- and cysteine-free Dul-
becco’s modified Eagle’s medium containing 110 ␮Ci/ml [35S]methi- RESULTS
onine/cysteine and chased in Dulbecco’s modified Eagle’s medium with The involvement of the proteasome in the degradation of
2 mM unlabeled methionine and cysteine for time intervals up to 24 h.
hERG channel was initially assessed by examining the effect of
For drug treatment, 50 ␮M ALLN, 20 ␮M lactacystin, 100 ␮M leupeptin,
or 100 ␮M kifunensine was added 1 h before pulse labeling and was proteasome inhibitors ALLN and lactacystin on the steady
continuously included in the culture medium during the pulse and the state protein levels of wild type hERG and the LQT2 mutant
chase periods. At the end of chase period, cells were lysed in 600 ␮l of Y611H. Fig. 1 shows Western blot analysis of hERG channel in
immunoprecipitation buffer containing 50 mM Tris-HCl, pH 7.4, 150 mM HEK293 cells stably transfected with wild type hERG or
NaCl, 1 mM EDTA, 1% Triton X-100, 1% sodium deoxycholate, 0.1% Y611H. Wild type hERG expressed two protein bands, an upper
SDS, 1 mg/ml bovine serum albumin, and protease inhibitors (100 ␮M
band of 155 kDa and a lower band of 135 kDa, whereas the
phenylmethylsulfonyl fluoride, 1 ␮g/ml pepstatin A, 1 ␮g/ml leupeptin,
4 ␮l/ml aprotinin). After centrifugation at 14,000 ⫻ g for 10 min at 4 °C, Y611H mutant expressed only the 135-kDa lower band. We
cell lysates were precleared by incubation with protein A-agarose beads have previously shown that the upper band is the fully glyco-
(Pierce). hERG antiserum (1:100 dilution) was then added, and the sylated, mature form of the channel protein located in the
mixture was incubated at 4 °C overnight. The antigen-antibody com- plasma membrane, and the lower band is a core-glycosylated,
plexes were isolated with protein A-agarose beads, subjected to 7.5% immature form of the channel protein located in the ER (5).
SDS-PAGE, and visualized with autoradiography. For quantitative
Exposure to 50 ␮M ALLN or 20 ␮M lactacystin for 24 h signif-
analysis, the dried gels were exposed to a K-screen and quantified by
phosphorimaging analysis using Quantity One soft ware (Bio-Rad, Mo- icantly increased protein levels of the immature form without
lecular Imager FX). an apparent increase in the mature form of wild type hERG or
Detection of Ubiquitinated hERG Proteins—Cells were lysed in 600 Y611H. The lysosome inhibitor leupeptin had no effect on pro-
␮l of immunoprecipitation buffer. Cell lysates containing an equal tein levels of wild type hERG or Y611H. These results strongly
amount of protein were immunoprecipitated with anti-hERG antibody suggest that the proteasome is involved in the degradation of
at 1:100 dilution as described above. The immunoprecipitates were
the immature form of hERG channels.
subjected to 5.5% SDS-PAGE and electrophoretically transferred onto
nitrocellulose membranes. The membranes were probed with mono- To further examine the role of the proteasome in the degra-
clonal anti-ubiquitin antibody at 1:500 dilution. The membranes were dation of hERG channels, we performed pulse-chase analysis.
also probed with anti-hERG antibody after stripping of the bound In these experiments, cells expressing wild type hERG or
anti-ubiquitin antibodies. Y611H were metabolically labeled with [35S]methionine and
Subcellular Fractionation—Cells were homogenized in 600 ␮l of ho- [35S]cysteine for 1 h and chased with unlabeled methionine and
mogenization buffer containing 10 mM Tris-HCl. pH 7.4, 1 mM EDTA,
cysteine for up to 24 h under control conditions or in the
and proteinase inhibitors with a Dounce homogenizer. Cells metaboli-
cally labeled by [35S]methionine/cysteine were homogenized in 600 ␮l of presence of 50 ␮M ALLN (Fig. 2A). Under control conditions,
homogenization buffer by freezing and thawing the cell suspension and wild type hERG was initially synthesized as the 135-kDa im-
passing it 10 times through a 25-gauge needle as described previously mature form, which was gradually converted to the 155-kDa
(18). Homogenates were centrifuged at 1000 ⫻ g for 10 min to remove mature form. The kinetics of hERG channel maturation and
unbroken cells and nuclei. The postnuclear supernatant was then cen- degradation were analyzed by phosphorimaging quantification.
trifuged at 10,000 ⫻ g for 30 min. The supernatant from the 10,000 ⫻
As shown in Fig. 2B, the conversion of wild type hERG from the
g centrifugation was further centrifuged at 100,000 ⫻ g for 60 min.
Membrane pellets from 10,000 ⫻ g and 100,000 ⫻ g spins were washed immature to the mature form became obvious at 2 h of chase
with homogenization buffer and solubilized in a buffer containing 50 and reached a maximum at 8 h. About 60% of the immature
mM Tris-HCl, pH 7.4, 1 mM EDTA, 150 mM NaCl, 1% Triton X-100, 1% form was converted to the mature form. In the presence of
sodium deoxycholate, 0.1% SDS, 1 mg/ml bovine serum albumin, and ALLN, there was a significant increase in the amount of la-
protease inhibitors. The supernatant from the 100,000 ⫻ g centrifuga- beled immature form of wild type hERG. The increase in the
tion (cytosolic fraction) was adjusted to a final concentration of 150 mM
immature form, however, did not result in an increase in the
NaCl, 1% Triton X-100, and 1 mg/ml bovine serum albumin. Solubilized
membranes and cytosolic fractions were analyzed by immunoprecipita- mature form during the chase. In fact, ALLN significantly
tion and immunoblot as described above. Equal fractions from each reduced the efficiency of hERG channel maturation, as only
pellet and supernatant were analyzed. about 20% of the immature form was converted to the mature
RNase Protection Assay—Twenty-four h after transfection with form in the presence of ALLN.
siRNAs, the cells were harvested and total RNA isolated using the In pulse-chase experiments, the Y611H mutant was initially
RNeasy method (Qiagen, Valencia, CA). RNase protection probes were
synthesized as the 135-kDa immature form and was not con-
made by subcloning a 452-nucleotide cDNA fragment corresponding to
1386 –1838 bp of EDEM and a 290-nucleotide cDNA fragment corre- verted to the mature form during the chase (Fig. 2C). Rather,
sponding to 466 –756 bp of actin into pCRII vector (Invitrogen). The the mutant protein underwent progressive degradation with
antisense RNA probes were transcribed using a MAXIscript in vitro complete disappearance of the 135-kDa band by 24 h. Kinetic
Proteasomal Degradation of Mutant hERG Channels 19421

FIG. 3. Effects of lactacystin and leupeptin on the degradation

Downloaded from http://www.jbc.org/ at FUDAN UNIVERSITY SCHOOL OF PHARMACY on May 24, 2018
of the Y611H mutant channel. Cells expressing Y611H mutant were
pulse-labeled for 1 h and chased for 8 h in the absence or presence of 20
␮M lactacystin (LACT) (A), or 100 ␮M leupeptin (LEUP) (B). hERG
channels were immunoprecipitated with anti-hERG antibody and sub-
jected to electrophoresis on SDS-polyacrylamide gel for analysis. Quan-
tified data are plotted as percentage of the value at time point 0, and the
data shown are means from two independent experiments.

of leupeptin, respectively. Thus, these pulse-chase results are


consistent with our Western blot data and suggest that the
cytosolic proteasome is involved in the degradation of the
Y611H mutant channel.
Degradation of ERAD substrate proteins by the cytosolic
proteasome implies that they must be dislocated from the ER to
the cytosol. It has been reported that some membrane proteins
can be dislocated from the ER to the cytosol in an intact form
when proteasome activity is inhibited (18 –20). To test whether
hERG channel proteins undergo similar dislocation, we per-
formed subcellular fractionation experiments (Fig. 4A). After
removal of unbroken cells and nuclei by centrifugation at
1000 ⫻ g, postnuclear supernatant was separated into three
fractions: 10,000 ⫻ g pellet (crude membranes), 100,000 ⫻ g
pellet (residual microsome membranes), and 100,000 ⫻ g su-
pernatant (cytosolic fraction) (18). Western blot analysis
showed that in the absence of ALLN, both wild type hERG and
FIG. 2. Pulse-chase analysis of wild type hERG and Y611H
Y611H mutant proteins were recovered only from the 10,000 ⫻
mutant channels. HEK293 cells stably transfected with wild type
hERG (A) or Y611H (C) were pulse-labeled for 1 h and chased for the g pellet membrane fraction. In the presence of ALLN, they were
indicated times in the absence or presence of 50 ␮M ALLN. The cells also present in the 100,000 ⫻ g supernatant cytosolic fraction.
were lysed and hERG was analyzed by immunoprecipitation with anti- As a control, the ER membrane protein calnexin was shown to
hERG antibody and SDS-PAGE. The amount of mature and immature be present only in the 10,000 ⫻ g pellet membrane fraction.
forms of hERG was quantified by phosphorimaging analysis and plotted
as percentage of the value at time point 0 (B and D). The data shown are Thus, these results suggest that an intact form of hERG chan-
means from two (wild type, B) or four (Y611H, D) independent nel can be dislocated from the ER to the cytosol. Because ERAD
experiments. substrate proteins dislocated to the cytosol have been reported
to undergo deglycosylation prior to degradation by the protea-
analysis shown in Fig. 2D revealed that the immature form of some (18, 21–23), we examined whether the hERG protein
Y611H was degraded with an estimated half-life of 4.5 h. ALLN recovered from the cytosolic fraction represents the deglycosy-
inhibited the degradation of the Y611H mutant channel, lated form. As shown in Fig. 4B, the Y611H mutant protein
thereby prolonging its half-life to 12 h (Fig. 2, C and D). How- recovered from the cytosolic fraction had a molecular mass of
ever, inhibition of the degradation did not cause conversion of 132 kDa and was resistant to PNGase treatment. In contrast,
the immature to the mature form. We also performed pulse- the mutant protein recovered from membrane fraction had a
chase experiments to study the effects of the specific protea- molecular mass of 135 kDa and was sensitive to PNGase, with
some inhibitor lactacystin and the lysosome inhibitor leupeptin its molecular mass being reduced to 132 kDa. These results
on the degradation of the Y611H mutant channel. In these suggest that Y611H mutant protein recovered from the cytoso-
experiments, cells expressing Y611H were labeled for 1 h and lic fraction represents the deglycosylated form.
chased for 8 h under control conditions or in the presence of 10 To demonstrate that the presence of hERG channels in the
␮M lactacystin or 100 ␮M leupeptin. As shown in Fig. 3, lacta- cytosol is due to dislocation of those retained in the ER, rather
cystin inhibited the degradation of the Y611H mutant. After than the failure of incorporation of newly synthesized hERG
8 h of chase, 26% of the 35S-labeled hERG protein remained channels into the ER, we performed pulse-chase experiments
under control conditions, whereas in the presence of lactacystin followed by cell fractionation. Cells expressing the Y611H mu-
68% of the 35S-labeled hERG protein remained. In contrast, tant were labeled for 1 h and chased for up to 24 h in the
leupeptin had no effect on the degradation of the Y611H mu- presence of 50 ␮M ALLN. hERG proteins were immunoprecipi-
tant. After 8 h of chase, 17 and 21% of the 35S-labeled hERG tated from the 10,000 ⫻ g pellet membrane fraction and the
protein remained under control conditions and in the presence 100,000 ⫻ g supernatant cytosolic fraction. As shown in Fig. 5,
19422 Proteasomal Degradation of Mutant hERG Channels

FIG. 6. Ubiquitination of wild type hERG and Y611H mutant


channels. A, untransfected HEK293 cells and wild type hERG- or
Y611H-transfected cells were treated with (⫹) or without (⫺) 50 ␮M
ALLN for 24 h. The cell lysates were analyzed by immunoprecipitation

Downloaded from http://www.jbc.org/ at FUDAN UNIVERSITY SCHOOL OF PHARMACY on May 24, 2018
(IP) with anti-hERG antibody followed by serial immunoblotting (IB)
with anti-ubiquitin (upper panel) and anti-hERG (lower panel) antibod-
ies. B, ALLN-treated cells were homogenized and fractionated. Samples
from membrane (M) and cytosolic (C) fractions were analyzed by im-
munoprecipitation with anti-hERG antibody followed by serial immu-
noblotting with anti-ubiquitin (upper panel) and anti-hERG (lower
panel) antibodies.

that newly synthesized hERG channels are incorporated into


the ER membrane and then dislocated from the ER into the
cytosol for proteasomal degradation. The results also show that
FIG. 4. A, accumulation of hERG channels in the cytosol. HEK293 despite the presence of hERG protein in the cytosolic fraction,
cells stably transfected with wild type hERG (top and bottom panels) or the majority of hERG protein was still present in the mem-
Y611H (middle panel) were treated with (⫹) or without (⫺) 50 ␮M
ALLN for 24 h. The cells were homogenized and subjected to subcellular
brane fraction during the chase, indicating that the rate of
fractionation as described under “Experimental Procedures.” Samples dislocation from the membrane to the cytosol is reduced in the
from different fractions were separated on SDS-polyacrylamide gel and presence of proteasome inhibitors.
immunoblotted with anti-hERG antibody (top and middle panels) or Many ERAD substrate proteins are conjugated with poly-
anti-calnexin antibody (bottom panel). B, effect of PNGase on hERG
ubiquitin chains that serve as a signal for proteasomal degra-
channels. Cells expressing the Y611H mutant were treated with 50 ␮M
ALLN for 24 h. The cells were then homogenized and subjected to dation. To demonstrate that the hERG channel is modified by
subcellular fractionation. Samples from 10,000 ⫻ g pellet (membrane polyubiquitin chains, cell lysates were immunoprecipitated
fraction) and 100,000 ⫻ g supernatant (cytosolic fraction) were treated with anti-hERG antibody and then immunoblotted with anti-
with (⫹) or without (⫺) PNGase and analyzed by immunoblotting with ubiquitin antibody. In these experiments, untransfected HEK
anti-hERG antibody.
cells and cells expressing wild type hERG or Y611H were
treated with or without ALLN for 24 h. As shown in Fig. 6A,
treatment of cells with ALLN resulted in the accumulation of
high molecular weight ubiquitinated hERG channels in wild
type hERG and in Y611H-transfected cells but not in untrans-
fected cells. Ubiquitinated hERG channels were barely detect-
able in untreated cells, suggesting that ubiquitinated hERG
channel protein is normally degraded by the proteasome in the
absence of proteasome inhibitor. To determine the subcellular
localization of the ubiquitinated hERG channels, cell fraction-
ation experiments were carried out in which hERG channels
were immunoprecipitated from membrane and cytosolic frac-
tions of ALLN-treated cells and then immunoblotted with anti-
ubiquitin antibody. As shown in Fig. 6B, ubiquitinated hERG
channels were present in both membrane and cytosolic frac-
tions. This result suggests that the ubiquitination of hERG
FIG. 5. Pulse-chase analysis of the Y611H mutant channel in
the membrane and cytosolic fractions. Cells expressing the Y611H channels starts before they are released into the cytosol.
mutant were pulse-labeled for 1 h and chased for the indicated times in The hERG channel protein is modified by N-linked glycosyl-
the presence of 50 ␮M ALLN. The cells were homogenized and fraction- ation (17, 24). Several studies have shown that the trimming of
ated by centrifugation. Samples from 10,000 ⫻ g pellet (membrane mannose by ER mannosidase I is required for proteasomal
fraction) and 100,000 ⫻ g supernatant (cytosolic fraction) were ana-
lyzed by immunoprecipitation with anti-hERG antibody and SDS- degradation of glycoproteins (25–29). We examined the role of
PAGE. Quantified data are plotted as percentage of the value of mem- mannose trimming in the degradation of the Y611H mutant
brane fraction at time point 0, and the data shown are means from two channel by using the ER mannosidase I inhibitor kifunensine.
independent experiments. As shown in Fig. 7, cells expressing the Y611H mutant were
metabolically labeled with [35S]methionine and [35S]cysteine
nearly all 35S-labeled hERG protein was present in the mem- for 1 h and chased with unlabeled methionine and cysteine for
brane fraction at the end of the pulse labeling. The amount of up to 24 h under control conditions or in the presence of 100 ␮M
hERG protein in the membrane fraction gradually decreased kifunensine (Fig. 7A). Similar to proteasome inhibitors, kifu-
during the chase, whereas the amount of hERG protein in the nensine inhibited the degradation of the Y611H mutant chan-
cytosolic fraction gradually increased, reaching a maximum at nel. In cell fractionation experiments, it was found that inhi-
8 h of chase. These pulse-chase experiments strongly suggest bition of degradation by kifunensine did not result in the
Proteasomal Degradation of Mutant hERG Channels 19423

FIG. 7. Effects of kifunensine on the degradation of the Y611H


mutant. Cells expressing the Y611H mutant were pulse-labeled for 1 h
and chased for the indicated times in the absence or presence of 100 ␮M

Downloaded from http://www.jbc.org/ at FUDAN UNIVERSITY SCHOOL OF PHARMACY on May 24, 2018
kifunensine. A, the cell lysates were subjected to immunoprecipitation
with anti-hERG antibody and SDS-PAGE. B, samples from membrane
and cytosolic fractions were analyzed by immunoprecipitation with
anti-hERG antibody and SDS-PAGE.

accumulation of hERG channel protein in the cytosol (Fig. 7B).


These results suggest that mannose trimming is an important
process in the degradation of the Y611H mutant channel.
Recently, it has been shown that EDEM plays an important
role in proteasomal degradation of misfolded glycoproteins (9,
10). EDEM is an ER type II membrane protein homologous to
␣-ER mannosidase but lacking mannosidase activity. To test
whether EDEM is involved in the degradation of mutant hERG FIG. 8. Effect of down-regulation of EDEM on the degradation
channels, we used RNA-mediated interference to reduce the of the Y611H mutant. The Y611H mutant was cotransfected with
intracellular concentration of EDEM. In these experiments, we siEDEM or siGFP into HEK293 cells. Twenty-four h after transfection,
the intracellular concentration of EDEM mRNA was determined by
cotransfected the Y611H mutant with an EDEM-directed RNase protection assay (A), and the cotransfected cells were pulse-
siRNA (siEDEM) or a control GFP-directed siRNA (siGFP) (9) labeled for 1 h and chased for the indicated times (B). The cell lysates
into HEK293 cells and performed pulse-chase analysis 24 h were subjected to immunoprecipitation with anti-hERG antibody and
after transfection. A RNase protection assay revealed a de- SDS-PAGE. The amount of hERG protein was quantified by phosphor-
imaging analysis and plotted as percentage of the value at time point 0
crease in EDEM mRNA in siEDEM-transfected cells (57 ⫾ 5%) (C). The data shown are means from three independent experiments.
compared with siGFP control (n ⫽ 3) (Fig. 8A). Pulse-chase
experiments showed that down-regulation of EDEM inhibited
the degradation of the Y611H mutant channel (Fig. 8, B and C). wild type hERG channels. In the case of the delta opioid recep-
Down-regulation of EDEM led to a prolongation of the half-life tor, proteasome inhibition leads to an increase in the cell sur-
of the Y611H mutant channel from 3.2 to 9.5 h, similar to that face expression of functional receptors (18). In hERG, however,
found in ALLN-treated samples. inhibition of the proteasome does not result in an increase in
the mature form. Similarly, proteasome inhibition increases
DISCUSSION the immature but not the mature form of wild type cystic
The present experiments demonstrate that degradation of fibrosis transmembrane conductance (11).
LQT2 mutant hERG channels is mediated by the ubiquitin- Degradation of ER luminal and membrane proteins by the
proteasome pathway. We show that the misfolded hERG cytosolic proteasome requires dislocation of substrates from the
protein is targeted to the proteasome for degradation by dislo- ER to the cytosol. This retrograde transport is believed to occur
cation from the ER membrane to the cytosol. The proteasome- through the Sec61p ER translocation complex (32, 33). How-
dependent degradation involves mannose trimming, ubiquiti- ever, the mechanism of dislocation and the link between dislo-
nation, and deglycosylation of mutant channels. cation and proteolysis by the proteasome are controversial. It
Our pulse-chase experiments revealed important informa- has been reported that dislocation and degradation by the
tion about the kinetics of the biogenesis of the wild type hERG proteasome are tightly coupled events in several ERAD sub-
channel. About 60% of the core-glycosylated immature form of strates, suggesting that active proteasome is required for the
wild type hERG is processed to the complex-glycosylated ma- extraction of ERAD substrates from the ER (23, 34 –36). In this
ture form. Compared with other polytopic membrane proteins, model, ER proteins are directly dislocated from the ER mem-
the hERG channel has intermediate maturation efficiency. For brane into the membrane-bound proteasome for degradation.
example, the maturation efficiency of the Shaker potassium The 19 S cap ATPases may provide the driving force for the
channel is near 100%, whereas less than 40% of newly synthe- membrane extraction. However, some ERAD substrates have
sized wild type cystic fibrosis transmembrane conductance and been reported to accumulate in the cytosol when proteasome
delta opioid receptor are processed to the mature forms, and activity is inhibited (18 –23). This suggests that proteasome
the remaining immature forms are degraded by the ubiquitin- activity is not required for the dislocation of these ERAD sub-
proteasome pathway (18, 30, 31). Our data also suggest that a strates from the ER to the cytosol. Recently, it has been shown
fraction of wild type hERG protein that fails to mature is that the cdc48/p97䡠Ufd1䡠Npl4 complex is involved in the dislo-
degraded by the ubiquitin-proteasome pathway. In the pres- cation of ERAD substrates from the ER to the cytosol (37, 38).
ence of proteasome inhibitors, there is an increase in the im- It is proposed that the ATPase activity of cdc48/p97 is involved
mature form of wild type hERG protein. In addition, protea- in extracting proteins from the ER membrane (39). In our
some inhibition results in the accumulation of ubiquitinated experiments, a fraction of the hERG channel protein accumu-
19424 Proteasomal Degradation of Mutant hERG Channels
lates in the cytosol following inhibition of the proteasome. Defective trafficking of mutant channels to the plasma mem-
However, the fact that the rate at which the hERG channel brane has been increasingly recognized as an important mech-
protein accumulates in the cytosol is much slower than the rate anism of hERG channel dysfunction in LQT2. Trafficking-de-
of degradation in the absence of proteasome inhibitors indi- fective LQT2 mutations lead to misfolding, ER retention, and
cates that the efficiency of dislocation is reduced in the pres- degradation of mutant hERG channels. Our present study sug-
ence of proteasome inhibitors. A similar reduction in disloca- gests that the ubiquitin-proteasome pathway plays an impor-
tion efficiency by proteasome inhibitors has been reported for tant role in the ER retention and degradation of LQT2 mutant
other ERAD substrates (18, 20, 23). Thus, although proteasome channels. Because hERG belongs to a large superfamily of
activity is not obligatory for the dislocation of hERG channel, voltage-gated potassium channels and defective trafficking of
inhibition of the proteasome affects the efficiency of hERG disease-causing mutations has been reported in several volt-
channel dislocation. age-gated potassium channels such as Kv1.1 and KvLQT1 (50 –
Our present results show that mannose trimming by ER 52), elucidating the mechanisms underlying the degradation of
mannosidase I is required for the degradation of Y611H mu- LQT2 mutant channels will increase our knowledge of the
tant channels. We have previously shown that the hERG chan- pathogenesis of a variety of human diseases involving voltage-
nel undergoes N-linked glycosylation at residue Asp-598 in the gated potassium channels.

Downloaded from http://www.jbc.org/ at FUDAN UNIVERSITY SCHOOL OF PHARMACY on May 24, 2018
extracellular domain between transmembrane segments 5 and
Acknowledgments—We thank Drs. Linda S. Musil and
6 (24). N-Linked glycosylation begins with the en bloc transfer William R. Skach for helpful advice and discussion of the manuscript.
of a preassembled oligosaccharide chain, Glu3Man9GluNA2, to
REFERENCES
asparagine residues of glycoproteins in the ER (40). The oligo-
1. Curran, M. E., Splawski, I., Timothy, K. W., Vincent, G. M., Green, E. D., and
saccharide chain is subject to the trimming of glucose and Keating, M. T. (1995) Cell 80, 795– 803
mannose residues by a number of glycosidases including glu- 2. Sanguinetti, M. C., Jiang, C., Curran, M. E., and Keating, M. T. (1995) Cell 81,
299 –307
cosidases I and II and ER mannosidases I and II. Several 3. Trudeau, M. C., Warmke, J. W., Ganetzky, B., and Robertson, G. A. (1995)
reports have indicated the prerequisite of mannose trimming Science 269, 92–95
by ER mannosidase I in proteasomal degradation of glycopro- 4. January, C. T., Gong, Q., and Zhou, Z. (2000) J. Cardiovasc. Electrophysiol. 11,
1413–1418
teins (25–29). In our experiments, the ER mannosidase I in- 5. Zhou, Z., Gong, Q., Epstein, M. L., and January, C. T. (1998) J. Biol. Chem.
hibitor kifunensine suppresses the degradation of Y611H mu- 273, 21061–21066
6. Brodsky, J. L., and McCracken, A. A. (1999) Semin. Cell Dev. Biol. 10,
tant channels. Unlike proteasome inhibitor treatment, in 507–513
which a fraction of hERG protein accumulates in the cytosol, 7. Ellgaard, L, Molinari, M., and Helenius, A. (1999) Science 286, 1882–1888
8. Ellgaard, L., and Helenius, A. (2003) Nat. Rev. Mol. Cell. Biol. 4, 181–191
kifunensine treatment does not result in the cytosolic accumu- 9. Molinari, M., Calanca, V., Galli, C., Lucca, P., and Paganetti, P. (2003) Science
lation of hERG protein. The results suggest that mannose 299, 1397–1400
trimming contributes to the initial targeting process that is 10. Oda, Y., Hosokawa, N., Wada, I., and Nagata, K. (2003) Science 299,
1394 –1397
upstream of the dislocation of the hERG protein in the ubiq- 11. Ward, C. L., Omura, S., and Kopito, R. R. (1995) Cell 83, 121–127
uitin-proteasome pathway. This process may involve the inter- 12. Qu, D., Teckman, J. H., Omura, S., and Perlmutter, D. H. (1996) J. Biol. Chem.
271, 22791–22795
action of the mannose-trimmed hERG channel with EDEM 13. Delisle, B. P., Anson, B. D., Rajamani, S., and January, C. T. (2004) Circ. Res.
because down-regulation of EDEM by RNA-mediated interfer- 94, 1418 –1428
ence suppresses the degradation of Y611H mutant channels. 14. Gong, Q., Keeney, D. R., Robinson, J. C., and Zhou, Z. (2004) J. Mol. Cell.
Cardiol. 37, 1225–1233
Our results support the proposal that EDEM functions as a 15. Ficker, E., Dennis, A. T., Wang, L., and Brown, A. M. (2003) Circ. Res. 92,
lectin that recognizes the mannose-trimmed misfolded gly- 87–100
16. Kagan, A., Yu, Z., Fishman, G. I., and McDonald, T. V. (2000) J. Biol. Chem.
coproteins and promotes their degradation by the cytosolic 275, 11241–11248
proteasome (9, 10, 41). 17. Zhou, Z., Gong, Q., Ye, B., Fan, Z., Makielski, J. C., Robertson, G. A., and
January, C. T. (1998) Biophys. J. 74, 230 –241
The present experiments show that the hERG channel is mod- 18. Petaja-Repo, U. E., Hogue, M., Laperriere, A., Bhalla, S., Walker, P., and
ified by ubiquitination. The requirement of ubiquitination for Bouvier, M. (2001) J. Biol. Chem. 276, 4416 – 4423
proteasomal degradation has been shown in many ERAD sub- 19. Bebok, Z., Mazzochi, C., King, S. A., Hong, J. S., and Sorscher, E. J. (1998)
J. Biol. Chem. 273, 29873–29878
strates (42). Recent studies also suggest that attachment of 20. VanSlyke, J. K., and Musil, L. S. (2001) J. Cell Biol. 157, 381–394
polyubiquitin chains to ERAD substrates is required for their 21. Wiertz, E. J., Jones, T. R., Sun, L., Bogyo, M., Geuze, H. J., and Ploegh, H. L.
(1996) Cell 84, 769 –779
dislocation from the ER to the cytosol (43– 45). In our experi- 22. Huppa, J. B., and Ploegh, H. L. (1997) Immunity 7, 113–122
ments, ubiquitinated hERG channels were recovered from both 23. Mancini, R., Fagioli, C., Fra, A. M., Maggioni, C., and Sitia, R. (2000) FASEB
J. 14, 769 –778
membrane and cytosolic fractions, suggesting that ubiquitination 24. Gong, Q., Anderson, C. L., January, C. T., and Zhou, Z. (2002) Am. J. Physiol.
of hERG channels starts prior to their dislocation from the ER to 283, H77–H84
the cytosol. Thus, it is possible that ubiquitin modification may 25. Liu, Y., Choudhury, P., Cabral, C. M., and Sifers, R. N. (1997) J. Biol. Chem.
272, 7946 –7951
contribute to dislocation as well as proteasomal degradation of 26. Jakob, C. A., Burda, P., Roth, J., and Aebi, M. (1998) J. Cell Biol. 142,
hERG channels. In addition to ubiquitin modification, hERG 1223–1233
27. Tokunaga, F., Brostrom, C., Koide, T., and Arvan, P. (2000) J. Biol. Chem. 275,
channel accumulated in the cytosol was also found to be degly- 40757– 40764
cosylated. The presence of deglycosylated intermediates in the 28. Fagioli, C., and Sitia, R. (2001) J. Biol. Chem. 276, 12885–12892
cytosol has been reported for other glycosylated ERAD substrates 29. Molinari M, Galli C, Piccaluga V, Pieren M, and Paganetti P. (2002) J. Cell
Biol. 158, 247–257
(18, 21–23). These intermediates are believed to be the result of 30. Khanna, R., Myers, M. P., Laine, M., and Papazian, D. M. (2001) J. Biol. Chem.
deglycosylation of ERAD substrates by a cytoplasmic PNGase 276, 34028 –34034
31. Ward, C. L., and Kopito, R. R. (1994) J. Biol. Chem. 269, 25710 –25718
prior to their degradation by the proteasome (21, 22). Recently, 32. Pilon, M., Schekman, R., and Romisch, K. (1997) EMBO J. 16, 4540 – 4548
genes encoding yeast cytoplasmic PNGase PNG1 and its mouse 33. Plemper, R. K., Bohmler, S., Bordallo, J. Sommer, T., and Wolf, D. H. (1997)
Nature 388, 891– 895
homologue mPNG1 have been identified (46). Both Png1p and 34. Mayer, T. U., Braun, T., and Jentsch, S. (1998) EMBO J. 17, 3251–3257
mPng1p display N-glycanase activity toward intact glycoproteins 35. Plemper, R. K., Egner, R., Kuchler, K., and Wolf D. H. (1998) J. Biol. Chem.
and are believed to be responsible for the deglycosylation of 273, 32848 –32856
36. Xiong, X., Chong, E., and Skach, W. R. (1999) J. Biol. Chem. 274, 2616 –2624
ERAD substrates in the cytosol (47). The cytoplasmic PNGase 37. Jarosch, E., Taxis, C., Volkwein, C., Bordallo, J., Finley, D., Wolf, D. H., and
has been shown to interact with the proteasome and may play an Sommer, T. (2002) Nat. Cell Biol. 4, 134 –139
38. Ye, Y., Meyer, H. H., and Rapoport, T. A. (2001) Nature 414, 652– 656
important role in proteasome-mediated degradation of misfolded 39. Tsai, B., Ye, Y., and Rapoport, T. A. (2002) Nat. Rev. Mol. Cell. Biol. 3, 246 –255
glycoproteins (48, 49). 40. Kornfeld, R., and Kornfeld, S. (1985) Annu. Rev. Biochem. 54, 631– 664
Proteasomal Degradation of Mutant hERG Channels 19425
41. Hosokawa, N., Wada, I., Hasegawa, K., Yorihuzi, T., Tremblay, L. O., Hersco- 48. Hirsch, C., Blom, D., and Ploegh, H. L. (2003) EMBO J. 22, 1036 –1046
vics, A., and Nagata, K. (2001) EMBO Rep. 2, 415– 422 49. Blom, D., Hirsch, C., Stern, P., Tortorella, D., and Ploegh, H. L. (2004) EMBO
42. Hershko, A., and Ciechanover, A. (1998) Annu. Rev. Biochem. 67, 425– 479 J. 23, 650 – 658
43. de Virgilio, M., Weninger, H., and Ivessa, N. E. (1998) J. Biol. Chem. 273, 50. Manganas, L. N., Akhtar, S., Antonucci, D. E., Campomanes, C. R., Dolly,
9734 –9743 J. O., and Trimmer, J. S. (2001) J. Biol. Chem. 276, 49427– 49434
44. Yu, H., and Kopito, R. R. (1999) J. Biol. Chem. 274, 36852–36858 51. Yamashita, F., Horie, M., Kubota, T., Yoshida, H., Yumoto, Y., Kobori, A.,
45. Shamu, C. E., Flierman, D., Ploegh, H. L., Rapoport, T. A., and Chau, V. (2001) Ninomiya, T., Kono, Y., Haruna, T., Tsuji, K., Washizuka, T., Takano, M.,
Mol. Biol. Cell 12, 2546 –2555 Otani, H., Sasayama, S., and Aizawa, Y. (2001) J. Mol. Cell Cardiol. 33,
46. Suzuki, T., Park, H., Hollingsworth, N. M., Sternglanz, R., and Lennarz, W. J. 197–207
(2000) J. Cell Biol. 149, 1039 –1052 52. Gouas, L., Bellocq, C., Berthet, M., Potet, F., Demolombe, S., Forhan, A.,
47. Park, H., Suzuki, T., and Lennarz, W. J. (2001) Proc. Natl. Acad. Sci. U. S. A. Lescasse, R., Simon, F., Balkau, B., Denjoy, I., Hainque, B., Baro, I., and
98, 11163–11168 Guicheney, P. (2004) Cardiovasc. Res. 63, 60 – 68

Downloaded from http://www.jbc.org/ at FUDAN UNIVERSITY SCHOOL OF PHARMACY on May 24, 2018
Degradation of Trafficking-defective Long QT Syndrome Type II Mutant Channels
by the Ubiquitin-Proteasome Pathway
Qiuming Gong, David R. Keeney, Maurizio Molinari and Zhengfeng Zhou
J. Biol. Chem. 2005, 280:19419-19425.
doi: 10.1074/jbc.M502327200 originally published online March 10, 2005

Access the most updated version of this article at doi: 10.1074/jbc.M502327200

Downloaded from http://www.jbc.org/ at FUDAN UNIVERSITY SCHOOL OF PHARMACY on May 24, 2018
Alerts:
• When this article is cited
• When a correction for this article is posted

Click here to choose from all of JBC's e-mail alerts

This article cites 52 references, 31 of which can be accessed free at


http://www.jbc.org/content/280/19/19419.full.html#ref-list-1

You might also like