ArticlePDF Available

Targeting Proteins to the Lumen of Endoplasmic Reticulum Using N-terminal Domains of 11 -Hydroxysteroid Dehydrogenase and the 50-kDa Esterase

Authors:

Abstract

Previous studies identified two intrinsic endoplasmic reticulum (ER) proteins, 11beta-hydroxysteroid dehydrogenase, isozyme 1 (11beta-HSD) and the 50-kDa esterase (E3), sharing some amino acid sequence motifs in their N-terminal transmembrane (TM) domains. Both are type II membrane proteins with the C terminus projecting into the lumen of the ER. This finding implied that the N-terminal TM domains of 11beta-HSD and E3 may constitute a lumenal targeting signal (LTS). To investigate this hypothesis we created chimeric fusions using the putative targeting sequences and the reporter gene, Aequorea victoria green fluorescent protein. Transfected COS cells expressing LTS-green fluorescent protein chimeras were examined by fluorescent microscopy and electron microscopic immunogold labeling. The orientation of expressed chimeras was established by immunocytofluorescent staining of selectively permeabilized COS cells. In addition, protease protection assays of membranes in the presence and absence of detergents was used to confirm lumenal or the cytosolic orientation of the constructed chimeras. To investigate the general applicability of the proposed LTS, we fused the N terminus of E3 to the N terminus of the NADH-cytochrome b5 reductase lacking the myristoyl group and N-terminal 30-residue membrane anchor. The orientation of the cytochrome b5 reductase was reversed, from cytosolic to lumenal projection of the active domain. These observations establish that an amino acid sequence consisting of short basic or neutral residues at the N terminus, followed by a specific array of hydrophobic residues terminating with acidic residues, is sufficient for lumenal targeting of single-pass proteins that are structurally and functionally unrelated.
Targeting Proteins to the Lumen of Endoplasmic Reticulum Using
N-terminal Domains of 11
b
-Hydroxysteroid Dehydrogenase and the
50-kDa Esterase*
(Received for publication, December 31, 1998, and in revised form, February 17, 1999)
Hassan Mziaut‡, George Korza‡, Arthur R. Hand§, Craig Gerard, and Juris Ozols‡i
From the Department of Biochemistry and §Electron Microscopy, University of Connecticut Health Center,
Farmington, Connecticut 06030-3305 and the Department of Pediatrics, Children’s Hospital, Harvard Medical School,
Boston, Massachusetts 02115
Previous studies identified two intrinsic endoplasmic
reticulum (ER) proteins, 11
b
-hydroxysteroid dehydro-
genase, isozyme 1 (11
b
-HSD) and the 50-kDa esterase
(E3), sharing some amino acid sequence motifs in their
N-terminal transmembrane (TM) domains. Both are type
II membrane proteins with the C terminus projecting
into the lumen of the ER. This finding implied that the
N-terminal TM domains of 11
b
-HSD and E3 may consti-
tute a lumenal targeting signal (LTS). To investigate this
hypothesis we created chimeric fusions using the puta-
tive targeting sequences and the reporter gene, Ae-
quorea victoria green fluorescent protein. Transfected
COS cells expressing LTS-green fluorescent protein chi-
meras were examined by fluorescent microscopy and
electron microscopic immunogold labeling. The orienta-
tion of expressed chimeras was established by immuno-
cytofluorescent staining of selectively permeabilized
COS cells. In addition, protease protection assays of
membranes in the presence and absence of detergents
was used to confirm lumenal or the cytosolic orientation
of the constructed chimeras. To investigate the general
applicability of the proposed LTS, we fused the N termi-
nus of E3 to the N terminus of the NADH-cytochrome b5
reductase lacking the myristoyl group and N-terminal
30-residue membrane anchor. The orientation of the cy-
tochrome b5 reductase was reversed, from cytosolic to
lumenal projection of the active domain. These observa-
tions establish that an amino acid sequence consisting
of short basic or neutral residues at the N terminus,
followed by a specific array of hydrophobic residues
terminating with acidic residues, is sufficient for lume-
nal targeting of single-pass proteins that are structur-
ally and functionally unrelated.
A number of proteins are resident and catalytically active in
the lumen of the endoplasmic reticulum (ER).
1
The best char-
acterized motif targeting this compartment is for soluble pro-
teins bearing C-terminal sequence KDEL (1, 2). The molecular
signals responsible for the insertion and retention of mem-
brane proteins in the ER with a lumenal orientation of their
functional domains are poorly understood. Most ER proteins
are targeted for membrane insertion by a hydrophobic signal
peptide at the N terminus (3). During translation, the signal
peptide is recognized by a cytosolic RNA/protein complex
termed the signal recognition particle (SRP) (4). Upon binding
a signal peptide, the SRP arrests the protein synthesis until it
has facilitated transfer of the nascent polypeptide-ribosome
complex to the translocation channel (5). The signals control-
ling folding and eventual topology of ER lumenal proteins after
the nascent chain is transferred to the translocation channel
are unknown. One early critical event in the multistep folding
process is whether or not the signal peptide is removed. If the
signal peptide is removed, then the newly formed N terminus is
generally located in the lumen of the ER. In cases where the
signal peptide is not removed, the resulting N-terminal hydro-
phobic segment may function as a membrane anchor, directing
the transmembrane insertion of the nascent polypeptide.
Therefore, membrane proteins with uncleaved signal peptide
may have either cytosolic or lumenal orientation. Mutagenesis
experiments on a number of membrane proteins have revealed
that the charge distribution, the length of the membrane seg-
ment, as well as the charge of the segments following the
membrane segment are some of the factors that determine the
specific topology of such proteins (6). Detailed analysis of these
factors have been complicated by use of multispanning model
proteins with a lack of agreement on the topology. Moreover,
additional ambiguities are encountered in trying to overex-
press polytopic eukaryotic membrane proteins in prokaryotic
hosts. Previously, we identified the membrane binding anchor
of cytochrome b5 (7), cytochrome b5 reductase (8), cytochromes
P-450 (9–11), epoxide hydrolase (12), D9 stearyl coenzyme A
desaturase (13), and three forms of flavin-containing monooxy-
genases (14). All of these ER proteins appeared to be oriented
toward the cytosolic side of the ER membrane by single or
multispanning membrane segments. In further studies to de-
fine the proteins found or oriented in the lumenal compartment
of the ER, we identified two isoforms of 60-kDa carboxylester-
ases (15) and the lumenal NADP glucose-6-phosphate dehydro-
genase (16). The esterases and the glucose-6-phosphate dehy-
drogenase were devoid of any transmembrane segments. The
lumenal esterases carried a C-terminal segment HIEL- and
HTEL-related KDEL, the ER lumen retention motif (1). The
primary structure of the glucose-6-phosphate dehydrogenase
failed to display sequence segments or a motif that would be
responsible for its lumenal orientation or retrieval. We then
extended our studies to oligosaccharyltransferase complex (OT)
* This work was supported by Grant R01 GM-26351 from the Na-
tional Institutes of Health (to J. O.). The costs of publication of this
article were defrayed in part by the payment of page charges. This
article must therefore be hereby marked advertisement in accordance
with 18 U.S.C. Section 1734 solely to indicate this fact.
iTo whom correspondence should be addressed: Dept. of Biochemis-
try, School of Medicine of the University of Connecticut Health Center,
Farmington, CT 06030-3305. Tel.: 860-679-2211; E-mail: ozols@sun.
uchc.edu.
1
The abbreviations used are: ER, endoplasmic reticulum; 11
b
-
HSD, 11
b
-hydroxysteroid dehydrogenase; E3, liver microsomal 50-
kDa esterase/N-deacetylase; GFP, green fluorescent protein; LTS,
lumenal targeting signal; PAGE; polyacrylamide gel electrophoresis;
PBS, phosphate-buffered saline; SLO, streptolysin O; OT, oligo-
saccharyltransferase; AEBSF, 4-(2-aminoethyl)-benzenesulfonyl flu-
oride; PCR, polymerase chain reaction; BSA, bovine serum albumin.
THE JOURNAL OF BIOLOGICAL CHEMISTRY Vol. 274, No. 20, Issue of May 14, pp. 14122–14129, 1999
© 1999 by The American Society for Biochemistry and Molecular Biology, Inc. Printed in U.S.A.
This paper is available on line at http://www.jbc.org14122
by guest on December 29, 2015http://www.jbc.org/Downloaded from
(18). The OT from mammalian tissues was found to be a tri-
molecular complex consisting of ribophorins I and II and a
50-kDa protein. The predicted membrane orientation of OT
indicated that both ribophorins and the 50-kDa proteins have
large N-terminal lumenal domains and a short C-terminal
cytoplasmic domain (18). Studies on the orientation of the
11
b
-HSD (17) and the 50-kDa esterase/N-deacetylase (E3) (19)
revealed that both proteins shared a short N-terminal cytoplas-
mic domain and a large C-terminal lumenal domain.
In the present study, we investigated whether the N-termi-
nal membrane binding segment of 11
b
-HSD and E3 may act as
a targeting sequence for the lumenal orientation of proteins in
the ER. Here we show that a transmembrane segment consist-
ing of basic residues at the N terminus followed by an array of
specific 17 hydrophobic residues terminating with acidic resi-
dues constitute a lumenal targeting signal for a set of single-
membrane-spanning proteins that are otherwise structurally
and functionally unrelated.
EXPERIMENTAL PROCEDURES
Materials—Chemical products were purchased from Sigma unless
otherwise noted. COS-7 cells were from American Type Culture Collec-
tion (ATCC). Cell reagents, restriction, and modification enzymes were
from Life Technologies, Inc. The GFPS65T mutant protein in
pEGFP-N1 vector and an anti-GFP mouse monoclonal antibody were
from CLONTECH. Anti-stearyl CoA-desaturase polyclonal antibody
was raised in a rabbit by injecting keyhole limpet hemocyanin-conju-
gated 20-residue synthetic peptide corresponding to residues 338–358
of stearyl CoA-desaturase antigen (20). Avian anti-b5 and anti-reduc-
tase were raised against the polar moieties of rat cytochrome b5 and
cytochrome b5 reductase (19). Anti-calreticulin rabbit polyclonal anti-
body was from Affinity Bioreagents. Alkaline phosphatase-conjugated
anti-mouse and anti-rabbit IgG were from Sigma. Biotin-coupled goat
anti-mouse IgG was from Molecular Probes. Iodocarbocyanine (Cy3)-
streptavidin and Cy3-conjugated goat anti-rabbit IgG were from Jack-
son Immunoresearch Laboratories. Proteinase K and 4-(2-aminoethyl)-
benzenesulfonyl fluoride (AEBSF) were from Roche Molecular
Biochemicals. Gold-labeled secondary antibody was from Amersham
Pharmacia Biotech.
Construction of Plasmids—All expression vectors were driven by the
cytomegalovirus promoter-enhancer contained in the pEGFP-N1 vec-
tor. The N tag-GFP chimeras were constructed by inserting the coding
region of GFP at the C terminus of the first 23 and 34 amino acids
corresponding to lumen targeting signal (LTS) of 11-
b
HSD and esterase
3, respectively and variants thereof, as follows (see Fig. 1). First, cDNA
encoding residues 1–34 of human esterase 3 preceded by Kozak se-
quence and EcoRI site at 59end and extended by BclI site at 39was
synthesized using two long overlapping oligonucleotides (59-CTTCGA-
ATTCCCACCATGGGAAGAAAATCGCTGTACCTTCTGATTGTGGG-
GATCCTCAATTT-39as sense long oligo and 59-TGGATTGATCATTC-
TCCATGGCTCCTCAACGTTATCTGGGAGAGGCGTATAAATATAAT-
G-39as antisense long oligo) and two short oligonucleotides (59-CTTC-
GAATTCCCACCATG-39as sense short oligo and 59-TGGATTGATCA-
TTCTCCA-39as antisense short oligo) and the chain polymerase reac-
tion (PCR). Second, the coding sequence of GFP was amplified from
pEGFP-N1 by PCR using a 59-oligonucleotide, which contains a BclI
restriction site (59-ATGGTGATCAAGGGCGAGGAGCTG-39) and 39-
oligonucleotide containing NotI site (59-CCTCTACAAATGTGGTATGG-
C-39). This was done in order to remove the Kozak sequence preceding
the first ATG codon of GFP in pEGFP-N1 vector so that the initiation
codon introduced in the N-tag sequences will be used by N-tag-GFP
chimeras.
The resulting PCR products were purified and digested with EcoRI-
BclI and BclI-NotI for N-tags and GFP, respectively and inserted into
EcoRI-NotI-digested pEGFP-N1 vector in one-step ligation. All the N-
tag-GFP chimeras and their mutated or deleted variants were con-
structed using the experimental procedure described above.
The expression vector pEN1.rab E3 N-b5 red.(31–300) encodes a
fusion protein consisting of rabbit esterase 3 LTS, followed by three
novel amino acids (PPV) encoded by the restriction site for AgeI, by
amino acids 31–300 of bovine liver NADH-cytochrome b5 reductase,
and FLAG epitope TAG (LARIKRTGDGSHKSS). This fusion protein
was expressed in pEN1 vector obtained by removing the coding se-
quence of GFP from pEGFP-N1. The rab E3 N-b5 red.(31–300) was
constructed as follows. First, the sequence corresponding to amino acids
31–300 of reductase was amplified by PCR using as a template the
940-bp DNA fragment coding for entire bovine liver microsomal redu-
ctase inserted in pGEM3zf(1) (21) with a pair of primers, 59-GATCC-
ACCGGTCCCGGCCATCACGCTCGAGAAT-39matching with the
amino acids 31–37 of beef reductase preceded by AgeI site, and 59-AG-
AGTCGCGGCCGCTTTAGCTACTCTTGTGGCTCCCATCTCCAGTTC-
TCTTAATCCTGGCTAAGAAGGCGAAGCAGCGTTCTTT-39matching
with amino acids 294–300. This oligonucleotide included the desatu-
rase epitope, stop codon, and NotI site. The PCR fragment was purified,
digested with AgeI-NotI and inserted along with the EcoRI-AgeI frag-
ment corresponding to LTS of rabbit esterase3 into EcoRI-NotI-digested
pEN1 vector. The integrity of the constructs were confirmed by DNA
sequencing.
Cell Culture and Transient Transfection—COS-7 cells were cultured
in Dulbecco’s modified Eagle’s medium (DMEM) supplemented with
10% heat-inactivated fetal bovine serum, 100 units/ml penicillin, and
100 mg/ml streptomycin. The cells were seeded at approximately 30%
confluence onto 35-mm dishes at 37 °C under 5% CO
2
. After 24 h, when
the cells became 60–70% confluent, they were transfected using the
LipofectAMINE reagent according to the manufacturer’s instructions.
The cell staining was observed 48–72 h after transfection under Zeiss
LSM 410 confocal microscope.
Immunoblot Analysis—Particulate components and the soluble frac-
tion of the cells were separated as described previously (22). Insoluble
material was removed by centrifugation, samples resolved by SDS-
PAGE, and Western blot analysis was performed using anti-mouse
monoclonal antibody against GFP. Alkaline phosphatase-conjugated
antibodies were used as secondary antibodies for protein visualization.
Isolation and Protease Digestion of Microsomes from COS-7 Cells—
Microsomal membranes were prepared as described by Ref. 23. The
pellet fraction was enriched in cytochrome b5 by adding 10
m
l of 0.5 mM
pure detergent free cytochrome b5 (24). The mixture was resuspended
in a solution containing 250 mMsucrose, 10 mMpotassium-Hepes, pH
7.2, 50 mMpotassium chloride, 2 mMmagnesium chloride, and 2 mM
calcium chloride to a final volume of 0.2 ml. Protease digestions were
performed at 4 °C for 30 min with increasing concentrations of protein-
ase K in presence or absence of 0.5% Triton X-100. AEBSF (4 mM) was
used to terminate the protease digestions. After 15 min of incubation on
ice, the membranes were solubilized with SDS gel loading buffer, and
the proteins subjected to SDS-PAGE and immunoblotted as described
above.
Selective Permeabilization and Immunofluorescence Analysis—Per-
meabilization of cells using digitonin, streptolysin O, and saponin was
performed 48 h after transfection, as described previously (25). Immu-
nocytofluorescent staining was conducted by incubation of the cells with
a 1:100 dilution of anti-GFP mouse monoclonal antibody in the perme-
abilization buffers containing 0.1% BSA. The cells were then rinsed
three times and then incubated with a 1:100 dilution of biotin-coupled
goat anti-mouse immunoglobulin. After the wash, cells were incubated
with streptavidin coupled-iodocarbocyanine (Cy3) diluted 1:100 in the
permabilization buffers containing 3% BSA. The cells were rinsed sev-
eral times before mounting in Mowiol 4.88 and then viewed under Zeiss
LSM 410 confocal microscope.
Electron Microscopic Immunogold Labeling—Transfected COS-7
cells with rab E3 N-GFP were fixed with 3% paraformaldehyde plus
0.15% glutaraldehyde in 0.1 Msodium cacodylate buffer, pH 7.4. The
cells were allowed to fix for about1hatroom temperature, the fixative
was removed, and the cells stored at 4 °C in 1% paraformaldehyde in 0.1
Mcacodylate, pH 7.4. The cells were removed from the flask by scraping
and then pelleted in 0.2% BSA in PBS. The cells were resuspended in
0.1% BSA/PBS, transferred to a tube containing 3% low-gelling agarose
and pelleted again. The agarose was chilled on ice, and the tip of the
tube was cut off. The embedded cell pellet was rinsed in 0.1 Mcacodylate
buffer, pH 7.4, dehydrated in methanol, infiltrated, and embedded at
220 °C in Lowicryl K4M resin. After polymerization with ultraviolet
light (365 nm), thin sections of the embedded cells were cut with a
diamond knife and collected on Formvar-coated, 200-mesh nickel grids.
Immunogold labeling was done essentially as described elsewhere
(26). Nonspecific binding was blocked with 1% BSA plus 1% instant
milk in PBS. The sections were labeled overnight at 4 °C with anti-GFP
mouse monoclonal antibody diluted 1:50 in 1% BSA and 5% normal goat
serum in PBS. After rinsing with PBS, the bound immunoglobulins
were visualized by incubating with 10-nm diameter gold-labeled sec-
ondary antibody diluted 1:15 in PBS for1hatroom temperature. After
thorough rinsing with PBS and distilled water, the sections were
stained with uranyl acetate and lead citrate and observed in Philips
CM10 TEM.
Targeting Proteins to the ER Lumen 14123
by guest on December 29, 2015http://www.jbc.org/Downloaded from
RESULTS
The N Termini of 11
b
-HSD and Esterase 3 (E3) Determines
Their Subcellular Localization—The proposed LTS of rat, rab-
bit, and human 11
b
-HSD aligned with sequences of E3 of
rabbit and human are shown in Fig. 1A. The two protein
families share a short positively charged N termini (region I),
followed by an array of some 17 hydrophobic residues contain-
ing an aromatic cluster Ala-Tyr-Tyr-X-Tyr or Gly-Tyr-Tyr-Tyr
cluster (region II), terminating with di-glutamyl residues (re-
gion III). Sequence similarity beyond the N termini could not be
found in these two protein families (19). To ascertain whether
N terminus of 11
b
-HSD is a determinant of its subcellular
localization in mammalian cells, constructs were generated
encoding N terminus of 11
b
-HSD fused to GFP (11
b
-HSD N-
GFP) (Fig. 1B). The A. victoria GFP is widely recognized as a
powerful tool in cell biology serving as a reporter for monitoring
localization and dynamics of intracellular proteins and or-
ganelles (27). When rab 11
b
-HSD N-GFP (Construct a) was
transiently expressed in COS-7 cells and examined by fluores-
cent microscopy, a staining pattern characteristic of ER local-
ization was seen (Fig. 2A). Expression of native GFP showed
green fluorescent signals in both the cytoplasm and nucleus
(Fig. 2B) consistent with the previous studies (28). To explore
whether the N-terminal domain of E3 also imparts GFP a
membrane localization, GFP constructs hum and rab E3N-GFP
(Constructs c and e, respectively) were generated and ex-
pressed in COS-7 cells. Cells expressing Constructs c and e
showed fluorescence pattern of ER localization, which was sim-
ilar to that seen with Construct a (Fig. 2, A,C, and D). Analysis
of the membrane fractions of cells expressing the Construct e
by SDS-PAGE, followed by immunoblotting with GFP antibody
showed the localization of GFP construct in the insoluble frac-
tion. Upon SDS-PAGE, a decreased mobility of rab E3 N-GFP
as compared with untagged GFP was observed, implying that
the signal sequence like N terminus of the construct was not
removed by the signal peptidase (Fig. 2E). As anticipated,
untagged GFP expressed in cells was found only in the soluble
fraction.
The N Termini Targets ER Localization—To confirm that the
fluorescence distribution along the reticular network observed
in Fig. 2 is characteristic of ER localization, we compared the
fluorescence pattern of cells expressing rab E3N-GFP (Con-
struct e) with a known ER marker, calreticulin. The fluores-
cence pattern of Construct e (Fig. 2F) is almost identical to the
calreticulin staining (Fig. 2G). Indeed, the combined fluores-
cence revealed a yellow staining indicative of the colocalization
of the rab E3N-GFP-associated green fluorescence and the
calreticulin-associated red fluorescence as evidenced by
Fig. 2H.
Electron Microscopic Immunogold Labeling—To confirm
these results at the ultrastructural level, ultrathin cryosections
were analyzed in double-labeling experiments with the immu-
nogold technique, using anti-GFP antibodies to identify the ER.
Electron microscopic examination of thin sections of rab E3
N-GFP (Construct e) transfected cells labeled with anti-GFP
antibodies revealed the presence of gold particles over the
nuclear envelope and the ER (Fig. 3, Aand B). Clusters of
heavily labeled ER membranes were found in the perinuclear
region. The gold particles were clearly associated with periph-
ery of the ER cisternae. Although the labeling was most fre-
quently seen in association with ribosome-studded membranes,
cisternae of apparently smooth ER were also labeled (Fig. 3C).
The labeling obtained with the polyclonal and monoclonal anti-
GFP preparations was approximately equivalent. Nonspecific
background labeling was relatively low; very few particles were
found over the nuclear chromatin, mitochondria, or other
structures.
Transmembrane Topology of Rab E3N-GFP, Construct e—To
determine the orientation of Construct e in the membrane, we
performed immunofluorescent staining of selectively permeabi-
lized COS-7 cells by saponin, digitonin, and streptolysin O
(SLO). As previously reported, digitonin (20
m
g/ml) and SLO
(200 units/ml) selectively permeabilized plasma membrane
leaving internal membranes including ER intact, whereas sap-
onin permeabilized all the membranes (29). For control exper-
iments, in cells expressing untagged GFP treated with digito-
nin (Fig. 4A), or saponin (Fig. 4C), the antibodies stained the
nucleus as well as the cytoplasm as expected. Cells treated with
SLO (Fig. 4B) showed a similar staining except for the nuclear
membrane, which was not permeabilized by SLO. Cells trans-
fected with cDNA for rab E3N-GFP (Construct e) and perme-
abilized with either digitonin (Fig. 4D) or SLO (Fig. 4E) failed
to stain with the antibodies to GFP even though these cells
expressed the GFP construct as indicated by the green fluores-
FIG.1.N-terminal sequences of native proteins and GFP constructs. A, N-terminal sequences of rat, rabbit, and human 11
b
-HSD are
aligned with those of rabbit and human E3. The positions of N-terminal amino acid residues are numbered below the sequence. Conserved amino
acids are enclosed in boxes.Dashes were inserted to maintain alignment of conserved residues. The LTS is denoted by regions I, II and III as
indicated above. The Protein Identification Resource accession numbers for the rat, rabbit, and human 11
b
-HSDs are A34430, A55573, and
A41173, respectively. Rabbit and human E3 accession numbers are A58922 and A53856. B, amino acid sequences of LTS of native proteins and
the derivatives thereof with altered LTS fused to GFPs, which were prepared for the experiments described here. Amino acid residues or segments
that were altered are indicated by letters in boldface or by dashed line.
Targeting Proteins to the ER Lumen14124
by guest on December 29, 2015http://www.jbc.org/Downloaded from
cent emission. However, after permeabilization with saponin
(Fig. 4F), all the cells expressing GFP chimeras became reac-
tive to the GFP antibody. The fact that the GFP fluorescence
pattern uniformly overlapped with the GFP antibody staining
pattern only when all the cell membranes were completely
permeabilized with saponin indicates that the GFP tagged with
E3 LTS is oriented toward the lumenal side of ER (Fig. 4F).
Evidence for Lumenal Localization by Proteinase K As-
say—To confirm the lumenal topography of rab E3N-GFP (Con-
struct e), membranes from cells expressing Construct e were
subjected to proteinase K digestion in the presence and absence
of detergent. As seen in Fig. 4G, in the intact membranes
Construct e was not susceptible to proteinase K digestion.
Proteolysis of Construct e could be demonstrated when deter-
gent was added to intact membranes prior to incubation with
the protease. Taken together, these results are consistent with
the lumenal orientation of the expressed chimera. The prote-
olysis of cytochrome b5 was used as a positive control in this
proteolysis protection assay. Cytochrome b5 is an integral
membrane protein oriented toward the cytosolic side of ER, and
is readily released from the membrane upon proteolysis (19,
24). Cytochrome b5 added to a membrane preparation is spon-
taneously incorporated into the membrane. As shown in Fig.
4G, proteolysis of membrane fractions in the presence or ab-
sence of detergent results in the conversion of cytochrome b5to
a species with faster mobility upon SDS-PAGE. The latter
species lacks the C-terminal 40 residue membrane anchor, but
retains its catalytic activity (7). Cleavage of cytochrome b5 was
observed in intact microsomes of cells containing Construct e at
protease concentration where no cleavage of the expressed
protein occurred (Fig. 4G).
Mutagenesis of the Lumenal Targeting Sequence—Sequences
of 11
b
-HSD from different mammalian species contain two Lys
residues in region I. The region I of human E3 contains an
Arg-Lys sequence. In the rabbit E3 sequence, the Arg residue is
replaced by Val, implying that in region I a single lysyl residue
rather than two basic residues do not alter the orientation of E3
in the membrane. Replacement of the single Lys residue by Ile
as in rab E3N(K4/I)-GFP (Construct f) led to ER localization of
the resulting chimera as evidenced by immunofluorescence
(Fig. 5A). The fluorescence image of cells expressing Construct
f is similar to that seen for rab E3N-GFP (Construct e). The
membrane localization of Construct f was confirmed by subcel-
lular fractionation and immunoblot analysis. As seen in Fig.
FIG.2. Expression of rab11
b
-HSD N-GFP, hum E3 N-GFP, rab E3 N-GFP, and native GFP in COS-7 cells. Evidence for the
colocalization with the ER marker calreticulin. Representative fluorescence images of COS 7 cells transfected with GFP and the indicated fusion
proteins are shown. A, Rab 11
b
-HSD N-GFP (Construct a). B, GFP control. C, Hum E3 N-GFP (Construct c). D, Rab E3 N-GFP (Construct e). The
cells were cultured for 48 h after transfection and observed under Zeiss LSM 410 confocal microscope (magnification, 340). E, immunoblot analysis
of rab E3 N-GFP (Construct e) and native GFP in cellular subfractions. Cells transfected with cDNA for GFP and rab E3 N-GFP were harvested
48 h after transfection, disrupted, and subjected to centrifugation. The whole cell extract (W), soluble fraction (S), and pellet fraction (P) (50
m
gof
protein) were subjected to SDS-PAGE and immunoblotted with 5
m
g/ml of an anti-GFP mouse monoclonal antibody. F,G, and H, colocalization of
rab E3 N-GFP (green) with the ER marker calreticulin (red) in COS-7 transfected cells permeabilized with 0.2% saponin. F, fluorescence image of
COS-7 cells expressing rab E3 N-GFP (green). G, the same cell as in Fwas costained by incubation with a rabbit polyclonal antibody against the
ER-resident protein calreticulin followed by incubation with Cy3-conjugated goat anti-rabbit IgG. The cells were then observed under confocal
microscope to visualize the staining pattern of the fluorescent dye Cy3 (red). H, merged images of Fand Greveal considerable colocalization of
brightest signal for rab E3 N-GFP and calreticulin in yellow (magnification 340).
Targeting Proteins to the ER Lumen 14125
by guest on December 29, 2015http://www.jbc.org/Downloaded from
5B, the expressed protein was present in pellet and absent in
the soluble fraction. To establish the orientation of the ex-
pressed protein, cells were selectively permeabilized with sap-
onin, digitonin, SLO, and analyzed by immunofluorescence
microscopy (Figs 5, C–E). Again, the staining pattern of such
permeabilized cells was identical to that obtained with Con-
struct e (Fig. 4, D–F). This result indicates that positively
charged residues are not an essential topogenic signal for the
lumenal orientation.
When the single Gly residue in region II was replaced by Glu,
and the resulting chimera (Construct b) expressed in cells, such
mutation led to the expression of the protein with an apparent
cytosolic and nuclear localization (Fig. 6A). Deletion of six
residues in region II as in the Construct d also led to a cytosolic
and nuclear localization of the protein (Fig. 6B).
Reorientation of NADH Cytochrome b5 Reductase Conferred
by the LTS—To test the utility of LTS as an in vivo trafficking
signal for proteins other than GFP, we made a fusion protein
between the polar segment of microsomal NADH cytochrome
b5 reductase and the N terminus of rab E3. The catalytically
active polar segment of the reductase consists of some 270
residues, and lacks the hydrophobic 28-residue N-myristoy-
lated membrane-anchoring domain at the N terminus present
in the intact protein (8). The native reductase has a cytosolic
orientation. To assess whether the LTS can redirect the reduc-
tase derivative to the lumen of ER, we fused the LTS of rabbit
E3 to the N terminus of the polar segment of the reductase. In
addition, we added to the C terminus of the reductase the
highly antigenic C-terminal sequence of stearyl CoA desatu-
rase as an epitope tag. We termed the chimera rab E3-N-b5 red
(31–300) or Construct g. The orientation of Construct g in the
ER membrane was examined by proteinase K digestion. As
shown in Fig. 7, digestion of intact microsomes (lane 1–3 and 5)
revealed a protease-resistant fragment approximately of 35–40
kDa in mass, indicating that the construct was intralumenal.
When microsomal membranes were disrupted with 0.5% Triton
X-100, the protein became sensitive to proteinase K digestion
and was completely degraded (lane 4). The proteinase K pro-
tection assay results confirm our previous results of the GFP
chimera orientation and show that soluble as well as membra-
nous proteins with a cytoplasmic orientation wearing 11
b
or E3
LTS are oriented toward the lumenal side of ER.
DISCUSSION
11
b
-HSD and E3 are two unrelated ER lumenal proteins
with type II orientation (17, 19). Analysis of their covalent
structures implied that the LTS for 11
b
-HSD and E3 is encoded
in the N-terminal segment. The N-terminal amino acid se-
quence of 34 residues of this group of proteins from several
mammalian species is shown in Fig. 1A. For discussion pur-
poses, the LTS sequence is divided into regions I, II, and III. In
addition to the positive charge at the N-terminal residue, fea-
tures of region I that are shared by several species include a
single lysyl residue. A stretch of aliphatic and aromatic hydro-
phobic residues (region II) follows region I, and continues into
a segment of negatively charged residues (region III). A strik-
ing feature of region I is the variability of the first three
residues and the conservation of a single lysyl residue. Region
II is a hydrophobic segment containing a cluster of tyrosyl
residues. In region III, there is a conserved Asn-X-Glu-Glu
segment. The remaining sequence of the C-terminal ectodo-
main shared no obvious homology between the dehydrogenases
and the esterases. Despite that the bulk of the polypeptide
chain is translocated across the membrane, the N-terminal
leader peptide like sequence in this group of proteins escapes
signal peptidase cleavage and serves as the membrane anchor.
Fig. 1Bdisplays the mutations and deletions of the chimeras
constructed by fusion of N-terminal domain of 11
b
-HSD or E3
to the N terminus of GFP. These constructs were expressed in
COS 7 cells, and analysis of their cellular targeting was accom-
plished by combination of fluorescence microscopy and electron
microscopic immunogold labeling. The topology of expressed
proteins was established by immunocytofluorescent staining of
selectivity permeabilized COS cells. Protease protection assay
of membranes in the presence and absence of detergents was
also used to distinguish the membrane sidedness of the GFP
chimeras.
The subcellular localization and orientation of the con-
structed GFP chimeras is summarized in Fig. 8 (Aand B).The
most important finding of this study is that fusion of the N-
terminal sequences from both 11
b
-HSD and E3 proteins to
GFP resulted in the lumenal localization of the chimera in the
ER membrane. This finding implied that the information (LTS)
for the lumenal orientation of 11
b
-HSD and E3 is encoded in
the N-terminal segment of some 20 to 25 residues.
As displayed in the amino acid sequences of the native pro-
teins from different species, the presence of two neutral or two
basic residues in region I of LTS are not essential for the
lumenal targeting. This is based on the assumption that the
proteins from different animal species share an identical ori-
entation. The proposed essential di-arginine sequences (30) at
the cytoplasmic N terminus of type II membrane proteins are
not indispensable for the ER localization for the group of pro-
teins described here. Deletion of the single lysyl residue in
region I (Construct f) led to ER targeting of the GFP chimera
with a lumenal ER orientation (Fig. 5). Therefore, the absence
of basic residues in region I does not alter the lumenal target-
FIG.3. Electron microscopic immunogold labeling of rab E3
N-GFP transfected cells with anti-GFP antibodies. Aand B, illus-
trate clusters of ER in the perinuclear region. In A, the asterisks (*)
indicate dilated regions of ER with membrane-associated gold particles.
In B, the arrows indicate regions where labeling of the ER is particu-
larly prominent. In both panels, the arrowheads indicate gold particles
associated with the nuclear envelope. Nucleus is denoted by n.InC,
several smooth-surfaced membranes are labeled (arrows). Scale bar 5
0.25
m
m.
Targeting Proteins to the ER Lumen14126
by guest on December 29, 2015http://www.jbc.org/Downloaded from
ing of the protein. Clearly, the positively charged residues are
not required for the ER localization, but the extreme negative
charge of region III would conform to the N .C charge role for
the lumenal orientation.
In order to further characterize the essential residues of the
LTS, several chimeras of LTS fused to GFP were constructed
and their subcellular localization analyzed. The Construct b
bearing a single negative charge in the center of region II led to
the expression of protein with fluorescence pattern similar to
the control GFP (Fig. 6A). Immunoblot of the subcellular frac-
tions of cells expressing Construct b indicated its electro-
phoretic mobility upon SDS-PAGE identical to native GFP,
implying that a truncation of the N terminus of the chimera
had occurred (Fig. 6C), but the protein failed to enter the
FIG.4. Immunocytofluorescent
staining of selectively permeabilized
COS-7 cells and the protease sensitiv-
ity of the expressed chimeras. COS-7
cells transfected with cDNA coding for
GFP (A–C), and rab E3 N-GFP (D–F) were
permeabilized either with digitonin (20
m
g/ml), streptolysin O (200 units/ml), or
saponin (0.2%) before incubation with an-
ti-GFP mouse monoclonal antibody.
Bound antibody was visualized by incuba-
tion with biotin-coupled goat anti-mouse
and then with Cy3-streptavidin as de-
scribed under “Experimental Procedures”
(magnification, 340). G, protease sensi-
tivity of rab E3 N-GFP and cytochrome b5
in membranes. The microsomal mem-
branes of COS-7 cells transfected with
cDNA coding for rab E3 N-GFP fusion
protein and enriched in cytochrome b5
were incubated with the indicated concen-
tration of proteinase K for 30 min on ice in
the absence (lanes 1–3) or presence (lane 4
and 5) of 0.5% Triton X-100. The mobility
of rab E3 N-GFP and cytochrome b5 was
analyzed by SDS-PAGE and immunoblot
with 5
m
g/ml of an anti-GFP mouse mono-
clonal antibody or with 1:1,000 dilution of
an anti-cytochrome b5 chicken polyclonal
antibody.
FIG.5.Expression of rab E3 N(K4/I)-
GFP in COS-7 cells to analyze the im-
portance of positively charged resi-
due of region I in the targeting to the
ER. A, subcellular localization of rab E3
N(K4/I)-GFP (Construct f) determined by
fluorescence microscopy (magnification,
340). The (K4/I) symbol indicates the po-
sition of positively charged residue within
E3 region I in which the single lysine was
mutated to an isoleucine residue. B, im-
munoblot analysis of COS-7 cells express-
ing Construct f and fractionated as de-
scribed in Fig. 2. The fluorescence image
and the immunoblot show the same intra-
cellular localization as the constructs E3
N-GFP in Fig. 2 (Cand D). Selective per-
meabilization with digitonin (C), SLO (D),
and saponin (E) of cells expressing Con-
struct f was done as described in Fig. 4.
Targeting Proteins to the ER Lumen 14127
by guest on December 29, 2015http://www.jbc.org/Downloaded from
secretory pathway as anticipated. This implied that the signal
peptidase was not responsible for the cleavage of the N-termi-
nal signal. What may have compensated for the translocation
deficiency of Construct b? It is most likely that the introduction
of a glutamyl residue in region II of the LTS resulted in a
conformational change interpreted by the cytosolic quality con-
trol system as a misfolded protein. Proteolytic elimination of
malfolded proteins, including uncomplexed subunits of protein
assemblies, is a common defense mechanism of the cell. Mutant
forms of cystic fibrosis transmembrane conductance regulator
expressed in mammalian cells, or carboxypeptidase Y mutants
that fail to translocate across the membrane and accumulate
on the surface of ER are readily degraded by the cytoplasmic
proteosome pathway (31, 32). One reason we observed the GFP
tag in cells expressed with Construct b could be the resistance
of the GFP molecule toward proteolysis. Expression of the N
terminus of Construct b fused to a polypeptide that is more
sensitive to proteolysis than the GFP molecule most likely
would result in the degradation of the entire construct.
A striking feature of region II of the parent proteins is the
presence of a cluster of tyrosyl residues (AYYXY or GYYY).
Repeats of tyrosyl residues (AYPYYA) are found in the trans-
membrane domain of the 48-kDa subunit of microsomal OT. OT
has a short C terminus oriented toward the cytosol, and a
single transmembrane domain with the bulk of the protein
molecule positioned in the lumen of ER (18). The T-cell receptor
delta chain displays a GYYYYV sequence in its membranous
segment (33). That a tyrosine-containing motif mediates ER
retention of CD3-
e
chain has also been reported (34). To deter-
mine the topogenic importance of the tyrosyl residue cluster
GFP chimera, Construct d lacking the tyrosyl residues in re-
gion II was generated. Construct d upon its expression in COS
cells displayed fluorescence in the nucleus and cytosol (Fig.
6B). This fluorescence pattern of intracellular structures was
similar to that observed by the untagged GFP. These findings
suggest that a cluster of tyrosyl residues is essential for the
correct folding and the lumenal targeting. Further analysis will
be needed to establish whether replacement of these residues
with phenylalanines or glycines provides an appropriate con-
text for the function of the conserved tyrosyl residues. Deletion
of five to six residues in region III of the 11
b
-HSD-GFP con-
structs does not affect the lumenal orientation of the constructs
(Fig. 2A).
Presently, the 11
b
-HSD and the E3 protein are the only type
II, static lumenal ER proteins with a single N-terminal trans-
membrane segment that have been characterized. The asialo-
FIG.6.Effect of mutations in the hydrophobic region of 11
b
-HSD and E3 LTS on the localization of GFP chimera proteins. A,
fluorescence microscopy of COS-7 cells transfected with rab 11
b
-HSD N(G10/E)-GFP (Construct b) in which the glycine residue in position 10 was
mutated to glutamic acid residue. B, fluorescence image of COS-7 cells transfected with hum E3 N(D16–21)-GFP (Construct d) in which the
hydrophobic segment was shortened by deletion of the six residues AYYIYT. A strong labeling pattern of nucleus, ER, and cytoplasm is seen for
the mutant chimera proteins as compared with the ER labeling pattern seen for the wild chimera proteins in Fig. 2 (Cand D) (magnification, 340).
C, immunoblot analysis of cells expressing GFP, Construct b, and Construct a.
FIG.7. Evidence for the lumenal orientation of rab E3 N-b5
red.(31–300) in microsomal membranes of transfected cells. Mi-
crosomes containing the chimeric protein (Construct g) were incubated
with the indicated concentrations of proteinase K in the presence (lane
4) or absence (lanes 1–3 and 5) of 0.5% Triton X-100. Samples of the
proteolysis reaction mixture were subjected to SDS-PAGE analysis,
followed by immunoblot with 1:2,000 dilution of anti-FLAG polyclonal
antibody.
FIG.8. Targeting proteins to the lumen of ER with LTS. A,
summary of subcellular localization of all the constructs expressed. The
constructs are abbreviated a-g as shown in Fig. 1B. The abbreviations
for the subcellular localizations are N, ER, C, S, L, and Cyt, nucleus,
endoplasmic reticulum, cytosolic, secreted, lumen, and cytoplasmic
side, respectively, proposed membrane topology of the LTS-GFP con-
structs. Nand Crepresent the N and C termini of the native and
chimera proteins. The chimeras are displayed in the upper part, and the
native proteins in the lower part of the model. My at the N terminus of
native cytochrome b5 reductase denotes a myristoyl residue.
Targeting Proteins to the ER Lumen14128
by guest on December 29, 2015http://www.jbc.org/Downloaded from
glycoprotein receptor (35) and the influenza virus neuramidi-
nase (36) are type II plasma membrane proteins. The
glycoprotein, paramyxovirus hemagglutinin-neuroaminidase
(HN) is a type II membrane protein localized to the Golgi
cisternae (37). The predicted amino acid sequence of the simian
virus HN protein includes a 17-residue cytoplasmic tail, a
19-residue membranous segment, and a large 523-residue C-
terminal ectodomain (38). Newly synthesized HN oligomerizes
into tetramers before transport from ER to Golgi, and alter-
ations of the C-terminal ectodomain can prevent ER to Golgi
transport (39). The large family of glycosidases and glycosyl-
transferases, although type II transmembrane proteins are all
located in the Golgi cisternae. Their common features include
an N-terminal cytoplasmic tail, a transmembrane domain, and
a large catalytic domain oriented toward the lumenal side of
the membrane (40). Two mutually complementary models have
been proposed to explain the mechanism of Golgi retention of
the glycosyltransferases mediated by their transmembrane do-
mains. One model postulates the retention through oligomer-
ization, which prevents proteins from entering transport vesi-
cles (41, 42). The other model suggests that their retention
depends on the length of a membrane-spanning domain, which
can be accommodated by the specific lipid composition of Golgi
complex membrane (43, 44). It has been pointed out that nei-
ther the oligomerization nor the membrane lipid composition
alone can explain the sorting of Golgi proteins.
Having established that the N-terminal domain of 11
b
-HSD
or E3 can mediate lumenal localization of the following down-
stream GFP molecule, we sought to test the general applica-
bility of this finding to targeting of ER proteins that display a
cytosolic orientation in their native state. The flavoprotein
NADH cytochrome b5 reductase has an amphiphilic structure
in which the hydrophilic, catalytic domain of some 270 residues
is linked to a membrane-anchoring, hydrophobic domain that
serves to orient the catalytic site of the reductase at the mem-
brane-aqueous interface to permit a rapid electron transfer to
cytochrome b5 (8). The hydrophobic domain at the N terminus
of the reductase consists of some 28 residues as well as N-
myristoylation of the N-terminal glycine residue (45) (Fig. 8B).
Fusion of the LTS of rabbit E3 to the N terminus of the polar
segment of the reductase (Construct g) resulted in the lumenal
targeting of the fusion protein (Figs. 7 and 8B).
Whether the topogenic signal described here is position-in-
dependent as regards to lumenal targeting of proteins, in ad-
dition to identifying the structural features that prevent these
constructs transiting to the Golgi, are the topics for further
studies. In conclusion, a static lumenal targeting signal of
proteins reported here should add to the repertoire of tech-
niques for studies on the structure, organization, and processes
of the lumen of ER.
Acknowledgment—We thank Dr. C. S. Ramaro (University of Alabama
at Birmingham) for constructing and expressing Constructs a and b.
REFERENCES
1. Munro, S., and Pelham, H. R. B. (1987) Cell 48, 899–907
2. Scheel, A. A., and Pelham, H. R. (1996) Biochemistry 35, 10203–10209
3. Blobel, G. (1980) Proc. Natl. Acad. Sci. U. S. A. 77, 1496–1500
4. Schatz, G., and Dobberstein, B. (1996) Science 271, 1519–1526
5. Hegde, R. S., Voigt, S., Rapoport, T. A., and Lingappa, V. R. (1998) Cell 92,
621–631
6. Gafvelin, G., Sakaguchi, M., Andersson, H., and von Heijne, G. (1997) J. Biol.
Chem. 272, 6119–6127
7. Ozols, J., and Gerard, C. (1977) Proc. Natl. Acad. Sci. U. S. A. 74, 3725–3729
8. Ozols, J., Korza, G., Heinemann, F. S., Hediger, M. A., and Strittmatter, P.
(1985) J. Biol. Chem. 260, 11953–11961
9. Ozols, J., Heinemann, F. S., and Johnson, E. F. (1985) J. Biol. Chem. 260,
5427–5434
10. Ozols, J. (1986) J. Biol. Chem. 261, 3965–3979
11. Heinemann, F. S., and Ozols, J. (1982) J. Biol. Chem. 257, 14988–14999
12. Heinemann, F. S., and Ozols, J. (1984) J. Biol. Chem. 259, 797–804
13. Thiede, M. A., Ozols, J., and Strittmatter, P. (1986) J. Biol. Chem. 261,
13230–13235
14. Ozols, J. (1994) Biochemistry 33, 3751–3757
15. Ozols, J. (1989) J. Biol. Chem. 264, 12533–12545
16. Ozols, J. (1993) Proc. Natl. Acad. Sci. U. S. A. 90, 5302–5306
17. Ozols, J. (1995) J. Biol. Chem. 270, 2305–2312
18. Kumar, V., Heinemann, F. S., and Ozols, J. (1994) J. Biol. Chem. 269,
13451–13457
19. Ozols, J. (1998) Biochemistry 37, 10336–10344
20. Heinemann, F. S., and Ozols, J. (1998) Mol. Biol. Cell 9, 3445–3453
21. Strittmatter, P., Kittler, J. M., Coghill, J. E., and Ozols, J. (1992) J. Biol.
Chem. 267, 2519–2523
22. Mori, M., Morita, T., Ikeda, F., Amaya, Y., Tatibana, M., and Cohen, P. P.
(1981) Proc. Natl. Acad. Sci. U. S. A. 78, 6056–6060
23. Haynes, R. L., Zheng, T., and Nicchitta, C. V. (1997) J. Biol. Chem. 272,
17126–17133
24. Ozols, J. (1989) Biochim. Biophys. Acta 997, 121–130
25. Otto, J. C., and Smith, W. L. (1994) J. Biol. Chem. 269, 19868–19875
26. Hand, A. R. (1995) in Introduction to Biophysical Methods for Protein and
Nucleic Acid Research (Glasel, J. A., and Deutscher, M., eds) pp. 205–260,
Academic Press, New York
27. Chalfie, M., Tu, Y., Euskirchen, G., Ward, W. W., and Prasher, D. C. (1994)
Science 263, 802–805
28. Yano, M., Kanazawa, M., Terada, K., Namchai, C., Yamaizumi, M., Hanson,
B., Hoogenraad, N., and Mori, M. (1997) J. Biol. Chem. 272, 8459–8465
29. Schulz, I. (1990) Methods Enzymol. 192, 280–300
30. Schutze, M. P., Peterson, P. A., and Jackson, M. R. (1994) EMBO J. 13,
1696–1705
31. Kopito, R. R. (1997) Cell 88, 427–430
32. Hiller, M. M., Finger, A., Schweiger, M., and Wolf, D. H. (1996) Science 273,
1725–1728
33. Hein, W. R., and Dudler, L. (1993) EMBO J. 12, 715–724
34. Mallabiabarrena, A., Jimenez, M. A., Rico, M., and Alarcon, B. (1995) EMBO
J. 14, 2257–2268
35. Spiess, M., and Lodish, H. F. (1986) Cell 44, 177–185
36. Bos, T. J., Davis, A. R., and Nayak, D. P. (1984) Proc. Natl. Acad. Sci. U. S. A.
81, 2327–2331
37. Parks, G. D. (1996) J. Biol. Chem. 271, 7187–7195
38. Hiebert, S. W., Paterson, R. G., and Lamb, R. A. (1985) J. Virol. 54, 1–6
39. Ng, D. T., Hiebert, S. W., and Lamb, R. A. (1990) Mol. Cell. Biol. 10, 1989–2001
40. Paulson, J. C., and Colley, K. J. (1989) J. Biol. Chem. 264, 17615–17628
41. Colley, K. J. (1997) Glycobiology 7, 1–13
42. Dahdal, R. Y., and Colley, K. J. (1993) J. Biol. Chem. 268, 26310–26319
43. Pelham, H. R. B., and Munro, S. (1993) Cell 75, 603–605
44. Munro, S. (1995) Biochem. Soc. Trans. 23, 527–530
45. Ozols, J., Carr, S. A., and Strittmatter, P. (1984) J. Biol. Chem. 259,
13349–13354
Targeting Proteins to the ER Lumen 14129
by guest on December 29, 2015http://www.jbc.org/Downloaded from
Hand, Craig Gerard and Juris Ozols
Hassan Mziaut, George Korza, Arthur R.
Dehydrogenase and the 50-kDa Esterase
-HydroxysteroidβDomains of 11
Endoplasmic Reticulum Using N-terminal
Targeting Proteins to the Lumen of
CELL BIOLOGY AND METABOLISM:
doi: 10.1074/jbc.274.20.14122
1999, 274:14122-14129.J. Biol. Chem.
http://www.jbc.org/content/274/20/14122Access the most updated version of this article at
.JBC Affinity SitesFind articles, minireviews, Reflections and Classics on similar topics on the
Alerts:
When a correction for this article is posted When this article is cited
to choose from all of JBC's e-mail alertsClick here
http://www.jbc.org/content/274/20/14122.full.html#ref-list-1
This article cites 44 references, 31 of which can be accessed free at
by guest on December 29, 2015http://www.jbc.org/Downloaded from
... Here, as proof of principle, we constructed fusion of roGFP2 with the two microsomal enzymes 11β-HSD1 and 11β-HSD2, which have been shown to face the luminal and cytosolic compartment, respectively [27][28][29][30]. We then implemented this method to unravel the intracellular localization and membrane topology of enzymes belonging to the 17βhydroxysteroid dehydrogenase (17β-HSD) family [31,32]. ...
... As proof of principle, we applied the roGFP2 fusion method and confirmed the topology of 11β-HSD1 and 11β-HSD2, two enzymes with known ER membrane topology [27][28][29][30] that essentially catalyze the local availability of active glucocorticoids to corticosteroid receptors [60]. In a next step, we determined the topology of enzymes of the 17β-HSD family (results summarized in Fig. 7). ...
Article
Endoplasmic reticulum Redox sensor Green-fluorescent protein Live-imaging Topology 17β-hydroxysteroid dehydrogenase Membrane proteins of the endoplasmic reticulum (ER) are involved in a wide array of essential cellular functions. Identification of the topology of membrane proteins can provide significant insight into their mechanisms of action and biological roles. This is particularly important for membrane enzymes, since their topology determines the subcellular site where a biochemical reaction takes place and the dependence on luminal or cytosolic co-factor pools and substrates. The methods currently available for the determination of topology of proteins are rather laborious and require post-lysis or post-fixation manipulation of cells. In this work, we have developed a simple method for defining intracellular localization and topology of ER membrane proteins in living cells, based on the fusion of the respective protein with redox-sensitive green-fluorescent protein (roGFP). We validated the method and demonstrated that roGFP fusion proteins constitute a reliable tool for the study of ER membrane protein topology, using as control microsomal 11β-hydroxysteroid dehydrogenase (11β-HSD) proteins whose topology has been resolved, and comparing with an independent approach. We then implemented this method to determine the membrane topology of six microsomal members of the 17β-hydroxysteroid dehydro-genase (17β-HSD) family. The results revealed a luminal orientation of the catalytic site for three enzymes, i.e. 17β-HSD6, 7 and 12. Knowledge of the intracellular location of the catalytic site of these enzymes will enable future studies on their biological functions and on the role of the luminal co-factor pool.
... The HSD11B1 dimer (PDB ID 1XSE) has similar catalytic cores (RMSD 2.00 Å for 350 Cα) and helix-turn-helix motifs. It also has the N-terminal membrane anchoring helices, but at their N-termini there are basic residues (K4/K5), critical for interacting the ER bilayer 36 , which are absent in HSD17B13. Although the construct used in the published HSD17B11 structure (PDB ID 1YB1) lacked both the N-terminal helices and the helix-turn-helix motifs, the catalytic cores of HSD17B13 and HSD17B11 are similar (RMSD 0.6Å for 348 Cα). ...
Article
Full-text available
Hydroxysteroid 17-beta-dehydrogenase 13 (HSD17B13) is a hepatic lipid droplet-associated enzyme that is upregulated in patients with non-alcoholic fatty liver disease. Recently, there have been several reports that predicted loss of function variants in HSD17B13 protect against the progression of steatosis to non-alcoholic steatohepatitis with fibrosis and hepatocellular carcinoma. Here we report crystal structures of full length HSD17B13 in complex with its NAD ⁺ cofactor, and with lipid/detergent molecules and small molecule inhibitors from two distinct series in the ligand binding pocket. These structures provide insights into a mechanism for lipid droplet-associated proteins anchoring to membranes as well as a basis for HSD17B13 variants disrupting function. Two series of inhibitors interact with the active site residues and the bound cofactor similarly, yet they occupy different paths leading to the active site. These structures provide ideas for structure-based design of inhibitors that may be used in the treatment of liver disease.
... 11β-HSD1 amplification of intracellular active GC production requires a high [NADPH]/[NADP+] ratio, which can be crucially provided by the enzyme hexose-6phosphate dehydrogenase (H6PDH) [9,10]. H6PDH is an enzyme localized to the ER lumen that converts glucose-6phosphate (G6P) and NADP to produce NADPH, thereby driving 11β-HSD1 reductase activity and consequently intracellular GC production [11,12]. ...
Article
Full-text available
Background: Visceral fat accumulation increases the risk of developing type 2 diabetes and metabolic syndrome, and is associated with excessive glucocorticoids (GCs). Fat depot-specific GC action is tightly controlled by 11ß-hydroxysteroid dehydrogenase (11ß-HSD1) coupled with the enzyme hexose-6-phosphate dehydrogenase (H6PDH). Mice with inactivation or activation of H6PDH genes show altered adipose 11ß-HSD1 activity and lipid storage. We hypothesized that adipose tissue H6PDH activation is a leading cause for the visceral obesity and insulin resistance. Here, we explored the role and possible mechanism of enhancing adipose H6PDH in the development of visceral adiposity in vivo. Methods: We investigated the potential contribution of adipose H6PDH activation to the accumulation of visceral fat by characterization of visceral fat obese gene expression profiles, fat distribution, adipocyte metabolic molecules, and abdominal fat-specific GC signaling mechanisms underlying the diet-induced visceral obesity and insulin resistance in H6PDH transgenic mice fed a standard of high-fat diet (HFD). Results: Transgenic H6PDH mice display increased abdominal fat accumulation, which is paralleled by elevated lipid synthesis associated with induction of lipogenic transcriptor C/EBPα and PPARγ mRNA levels within adipose tissue. Transgenic H6PDH mice fed a high-fat diet (HFD) gained more abdominal visceral fat mass coupled with activation of GSK3β and induction of XBP1/IRE1α, but reduced pThr308Akt/PKB content and browning gene CD137 and GLUT4 mRNA levels within the visceral adipose tissue than WT controls. HFD-fed H6PDH transgenic mice also had impaired insulin sensitivity and exhibited elevated levels of intra-adipose GCs with induction of adipose 11ß-HSD1. Conclusion: These data provide the first in vivo mechanistic evidence for the adverse metabolic effects of adipose H6PDH activation on visceral fat distribution, fat metabolism, and adipocyte function through enhancing 11ß-HSD1-driven intra-adipose GC action.
... Kinetic studies of 11b-HSD1 expressed in cultured cells using recombinant Vaccinia virus suggested that cofactor availability might determine the reaction direction of 11b-HSD1 [9]. The determination of the intracellular localization then revealed that 11b-HSD1 is an integral membrane enzyme of the ER with a lumenal orientation of its active site [11][12][13]. The subsequent identification of the luminal enzyme hexose-6-phosphate dehydrogenase (H6PDH) as a supplier of NADPH [14,15] provided an explanation for the reaction direction of 11b-HSD1 in intact cells. ...
... Jamieson, Chapman et al. 1995;Kotelevtsev, Holmes et al. 1997;Jamieson, Walker et al. 2000). 11β-HSD1 is localized to the endoplasmic reticulum (ER) lumen with its N-terminus anchored to the ER membrane and the C-terminus inside the ER lumen (Ozols 1998;Mziaut, Korza et al. 1999;Odermatt, Arnold et al. 1999). 11β-HSD1 and 11β-HSD2 use nicotinamide cofactors to transfer hydride to interconvert active GC (cortisol in humans and corticosterone in rodents), able to bind to receptors, and inactive GC (cortisone in humans and 11-dehydrocorticosterone in rodents), which show negligible binding to receptors. ...
Article
Glucocorticoids are steroid hormones that have potent anti-inflammatory actions. Endogenous glucocorticoid action is modulated by 11β-hydroxysteroid dehydrogenase (11β-HSD) which catalyses the interconversion of active glucocorticoids (cortisol, corticosterone) and intrinsically inert forms (cortisone, 11-dehydrocorticosterone). There are 2 isozymes; 11β-HSD type 1 regenerates active glucocorticoids in vivo whereas 11β-HSD type 2 inactivates glucocorticoids. Although 11β-HSD1 is highly expressed in the lung, its role there has been little explored. In this study, the expression and localization of 11β-HSD1 mRNA in lung was confirmed by in situ hybridization. Immunohistochemical staining of mouse lung localized 11β-HSD1 to the cytoplasm of fusiform cells in alveolar walls, in a multivesicular pattern characteristic of interstitial fibroblasts. A lung fibrosis model of inflammation was used to test the role and regulation of 11β-HSD1. The results suggest that levels of 11β-HSD1 mRNA and enzyme were not changed during bleomycin-induced lung inflammation. However, 11β-HSD1-deficient mice showed a more severe inflammatory response than congenic wild-type controls, with greater inflammatory cell infiltration into the lung, and increased levels of HO-1 and iNOS mRNA 14 days following bleomycin installation into lung. Picrosirius red staining of lung sections suggested more collagen deposition in 11β-HSD1-deficient mice than in wild-type controls during the course of the lung inflammatory response. Moreover, whereas naïve 11β-HSD1-deficient mice had significantly lower collagen content in lung (84% of WT levels, p
... However, tissuespecific GC action not only depends on circulating GCs levels but can also be regulated by an intracellular endoplasmic reticulum (ER) lumen resident enzyme, 11␤-hydroxysteroid dehydrogenase type 1 (11␤-HSD1), that converts inactive cortisone (11-dehydrocorticosterone in rodents) to active cortisol (corticosterone) and therefore amplifies intracellular GC action, particularly in adipose tissue (6,21,24,41). 11␤-HSD1 is regulated by its cofactor NADPH, which is provided by the enzyme hexose-6-phosphate dehydrogenase (H6PDH) (32,34). In the ER lumen, H6PDH can metabolize glucose 6-phosphate and NADP to generate NADPH, thus ensuring 11␤-HSD1 activity. ...
Article
Full-text available
Long-term glucocorticoid exposure increases the risk of developing type 2 diabetes. Pre-receptor activation of glucocorticoid availability in target tissue by 11ß-hydroxysteroid dehydrogenase type 1 (11ß-HSD1) coupled with hexose-6-phosphate dehydrogenase (H6PDH) are important mediators of the metabolic syndrome. We explored whether the tissue-specific modulation of 11ß-HSD1 and H6PDH in adipose tissue mediates glucocorticoid-induced insulin resistance and lipolysis and analyzed the effects of 11β-HSD1 inhibition on the key lipid metabolism genes and insulin signaling cascade. We observed that corticosterone (CORT) treatment increased expression of 11β-HSD1 and H6PDH and induced lipase HSL and ATGL with suppression of pThr(172)AMPK in adipose tissue of C57BL/6J mice. In contrast, CORT induced adipose insulin resistance, as reflected by a marked decrease in IR and IRS-1 gene expression with reduction in pThr(308) Akt/PKB. Furthermore, 11β-HSD1 shRNA attenuated CORT-induced 11β-HSD1 and lipase expression and improved insulin sensitivity with a concomitant stimulation of pThr(308) Akt/PKB and pThr(172)AMPK within adipose tissue. Addition of CORT to 3T3-L1 adipocytes enhanced 11ß-HSD1 and H6PDH and impaired pThr(308)Akt/PKB leading to lipolysis. Knockdown of 11ß-HSD1 by shRNA attenuated CORT-induced lipolysis and reversed CORT-mediated inhibition of pThr(172)AMPK accompanied by a parallel improvement of insulin signaling response in these cells. These findings suggest that elevated adipose 11β-HSD1 expression may contribute to glucocorticoid-induced insulin resistance and adipolysis.
Chapter
11β-Hydroxysteroid dehydrogenase type 1 (11β-HSD1) converts inactive 11-keto-glucocorticoids to their active 11β-hydroxylated forms. It also catalyzes the oxoreduction of other endogenous and exogenous substrates. The ubiquitously expressed 11β-HSD1 shows high levels in liver and other metabolically active tissues such as brain and adipose tissue. Pharmacological inhibition of 11β-HSD1 was found to ameliorate adverse metabolic effects of elevated glucocorticoids in rodents and humans, improve wound healing and delay skin aging, and enhance memory and cognition in rodent Alzheimer's disease models. Thus, there is an interest to develop 11β-HSD1 inhibitors for therapeutic purposes. This chapter describes in vitro methods to assess 11β-HSD1 enzyme activity for different purposes, be it in disease models, for the assessment of the kinetics of novel substrates or for the screening and characterization of inhibitors. 11β-HSD1 protein expression and preparations of the different biological samples are discussed first, followed by a description of a well-established and easily adaptable 11β-HSD1 enzyme activity assay. Finally, different readout methods are shortly described. This chapter should provide the reader with a toolbox of methods to assess 11β-HSD1 activity with instructions in the form of a decision tree for the choice and implementation of an appropriate enzyme activity assay.
Article
This review briefly addresses the history of the discovery and elucidation of the three cloned 11β-hydroxysteroid dehydrogenase (11βHSD) enzymes in the human, 11βHSD1, 11βHSD2 and 11βHSD3, an NADP⁺-dependent dehydrogenase also called the 11βHSD1-like dehydrogenase (11βHSD1L), as well as evidence for yet identified 11βHSDs. Attention is devoted to more recently described aspects of this multi-functional family. The importance of 11βHSD substrates other than glucocorticoids including bile acids, 7-keto sterols, neurosteroids, and xenobiotics is discussed, along with examples of pathology when functions of these multi-tasking enzymes are disrupted. 11βHSDs modulate the intracellular concentration of glucocorticoids, thereby regulating the activation of the glucocorticoid and mineralocorticoid receptors, and 7β-27-hydroxycholesterol, an agonist of the retinoid-related orphan receptor gamma (RORγ). Key functions of this nuclear transcription factor include regulation of immune cell differentiation, cytokine production and inflammation at the cell level. 11βHSD1 expression and/or glucocorticoid reductase activity are inappropriately increased with age and in obesity and metabolic syndrome (MetS). Potential causes for disappointing results of the clinical trials of selective inhibitors of 11βHSD1 in the treatment of these disorders are discussed, as well as the potential for more targeted use of inhibitors of 11βHSD1 and 11βHSD2.
Article
Membrane proteins of the endoplasmic reticulum (ER) are involved in a wide array of essential cellular functions. Identification of the topology of membrane proteins can provide significant insight into their mechanisms of action and biological roles. This is particularly important for membrane enzymes, since their topology determines the subcellular site where a biochemical reaction takes place and the dependence on luminal or cytosolic co-factor pools and substrates. The methods currently available for the determination of topology of proteins are rather laborious and require post-lysis or post-fixation manipulation of cells. In this work, we have developed a simple method for defining intracellular localization and topology of ER membrane proteins in living cells, based on the fusion of the respective protein with redox-sensitive green-fluorescent protein (roGFP). We validated the method and demonstrated that roGFP fusion proteins constitute a reliable tool for the study of ER membrane protein topology, using as control microsomal 11β-hydroxysteroid dehydrogenase (11β-HSD) proteins whose topology has been resolved, and comparing with an independent approach. We then implemented this method to determine the membrane topology of six microsomal members of the 17β-hydroxysteroid dehydrogenase (17β-HSD) family. The results revealed a luminal orientation of the catalytic site for three enzymes, i.e. 17β-HSD6, 7 and 12. Knowledge of the intracellular location of the catalytic site of these enzymes will enable future studies on their biological functions and on the role of the luminal co-factor pool. Copyright © 2015. Published by Elsevier B.V.
Article
Full-text available
An expression vector for bovine NADH-cytochrome b5 reductase was constructed from two DNA fragments that were derived from beef liver poly(A+) RNA using the polymerase chain reaction. Site-directed mutagenesis of the 3 lysine residues of the reductase, previously implicated in the formation of active-site charge pairs with carboxylate residues of cytochrome b5, was then used to obtain the purified catalytic domains of flavoproteins modified at each of these sites. The observed marked decreases in catalytic efficiencies of substitutions of a negative charge at the normally positively charged residues with the catalytic domain of cytochrome b5 are consistent with their participation in the formation of charge pairs with carboxylate groups of the hemeprotein to optimize rapid electron transfer from the reductase flavin to the heme of the cytochrome.
Article
Full-text available
The role of N-linked glycosylation in protein maturation and transport has been studied by using the simian virus 5 hemagglutinin-neuraminidase (HN) protein, a model class II integral membrane glycoprotein. The sites of N-linked glycosylation on HN were identified by eliminating each of the potential sites for N-linked glycosylation by oligonucleotide-directed mutagenesis on a cDNA clone. Expression of the mutant HN proteins in eucaryotic cells indicated that four sites are used in the HN glycoprotein for the addition of N-linked oligosaccharide chains. These functional glycosylation sites were systematically eliminated in various combinations from HN to form a panel of mutants in which the roles of individual carbohydrate chains and groups of carbohydrate chains could be analyzed. Alterations in the normal glycosylation pattern resulted in the impairment of HN protein folding and assembly which, in turn, affected the intracellular transport of HN. The severity of the consequences on HN maturation depended on both the number of deleted carbohydrate sites and their position in the HN molecule. Analysis of the reactivity pattern of HN conformation-specific monoclonal antibodies with the mutant HN proteins indicated that one specific carbohydrate chain plays a major role in promoting the correct folding of HN. Another carbohydrate chain, which is not essential for the initial folding of HN was found to play a role in preventing the aggregation of HN oligomers. The HN molecules which were misfolded, owing to their altered glycosylation pattern, were retained in the endoplasmic reticulum. Double-label immunofluorescence experiments indicate that misfolded HN and folded HN are segregated in the same cell. Misfolded HN forms disulfide-linked aggregates and is stably associated with the resident endoplasmic reticulum protein, GRP78-BiP, whereas wild-type HN forms a specific and transient complex with GRP78-BiP during its folding process.
Article
Full-text available
Hepatic poly(A+) RNA from rats induced for stearyl-CoA desaturase was used for primer-extension of cDNA coding for stearyl-CoA desaturase. Previously, Northern blot analysis showed that translatable desaturase mRNA is 4,900 nucleotides in length (Thiede, M. A., and Strittmatter, P. (1985) J. Biol. Chem. 260, 14459-14463). Six overlapping cDNAs, ranging from 850 to 1450 bases, were used to compile the 4,689-nucleotide sequence. The cDNA includes a 1,074-base open reading frame coding for 358 amino acids, corresponding to a molecular mass of 41,400 daltons. Positive identification of this open reading frame was accomplished by matching the amino acid sequence of both amino-terminal and cyanogen bromide peptides of the purified enzyme with regions of the sequence deduced from the cDNA. Amino acid composition data from the cDNA compares well with that from the desaturase. The protein contains 62% hydrophobic amino acids. An interesting feature of this mRNA is the 3,500-base 3' noncoding region, which has been localized on a single 3' exon by Southern blot analysis.
Article
Full-text available
A second form (form 2) of glycosylated esterase was isolated from liver microsomal membranes and characterized. The subunit molecular weight of form 2 is identical to that of the 60-kDa protein previously reported (Ozols, J. (1987) J. Biol. Chem. 262, 15316–15321). The NH2 terminus of the form 2 enzyme is blocked. Digestion of form 2 with pyroglutamyl aminopeptidase, followed by electroblotting and sequence analysis of the blotted protein, indicated that a pyroglutamyl residue was located at the NH2 terminus. Sequence analysis of the deblocked protein as well as characterization of the peptides obtained from enzymatic and chemical cleavages of the intact protein led to the elucidation of its complete amino acid sequence. The protein is a single polypeptide consisting of 532 residues. Carbohydrate is attached at asparaginyl residue 249. The sequence of form 2 esterase is 50% identical to the sequence of form 1 enzyme. The amino acid sequence of the first 26 residues of form 1 enzyme from human liver microsomes shows that 23 residues are identical to that of rabbit form 1, but only 8 residues that are identical to form 2. Treatment of the forms 1 and 2 isozymes with N-glycosidase F or endo-N-acetylglucosaminidase H resulted in a decrease of their subunit molecular weights, indicating that the carbohydrate attached is of the high mannose type. To determine whether the 60-kDa proteins are located on the cytoplasmic or luminal side of the endoplasmic membrane, microsomes were treated with proteolytic enzymes and the two 60-kDa isozymes were isolated and characterized. Sequence analysis of both proteins indicated that their NH2 termini were unaffected by proteolysis. Form 1 isozyme isolated from trypsin-treated microsomes, however, lacked the COOH-terminal heptapeptide (residues 533–539). These results, in addition to the finding of an N-linked carbohydrate, suggest that the two 60-kDa proteins are oriented on the luminal side of the endoplasmic membrane.
Chapter
This chapter illustrates the principles and fundamentals of electron microscopy. Knowledge of cell and molecular structure and function has made tremendous advances in the 50 years that electron microscopes have been used in the biological sciences. Electron microscopy spawned the field of cell biology and provided the structural framework necessary for correlating events at the molecular level with the physiological and pathological changes exhibited by the organism. Electron microscopic investigation of biological samples can provide unique information. Certain types of information can be obtained more rapidly and easier than with other methods. The 1000-fold greater resolution of the electron microscope compared to the light microscope can give details of structure in the 1 nm range. Identification of subcellular structures in sections and in isolated preparations is greatly facilitated. Electron microscopy, in combination with the use of specific antibodies or nucleotide probes, can yield data on the distribution and localization of biologically relevant antigens and nucleic acid sequences. Finally, detection and analysis of the interactions of electrons with atoms of the specimen can provide information on its composition.
Article
Translocational pausing is a mechanism used by certain specialized secretory proteins whereby discrete domains of a nascent chain destined for the endoplasmic reticulum lumen are transiently exposed to the cytosol. Proteoliposomes reconstituted from total endoplasmic reticulum proteins properly assemble translocationally paused intermediates. The capacity of the translocon to correctly pause the nascent chain is dependent on a glycoprotein fraction whose active component is TRAM. In the absence of TRAM, the normally sealed ribosome-membrane junction still opens in response to a pause transfer sequence. However, nascent chain domains that are not exposed to the cytosol in the presence of TRAM are so exposed in its absence. Thus, TRAM regulates which domains of the nascent chain are visible to the cytosol during a translocational pause.
Article
The primary structure of the membranous segment of porcine liver microsomal cytochrome b5 has been determined. This polypeptide is at the COOH terminus of the cytochrome molecule and consists of 43 amino acids. It is essential for the insertion of the cytochrome into the endoplasmic reticular membrane. Automated sequence analysis of tryptic and cyanogen bromide/anhydrous heptafluorobutyric acid peptides provided data from which the following unique amino acid sequence was deduced: Ile-Ala-Lys-Pro-Ser-Glu-Thr-Leu-Ile-Thr-Thr-Val-Glu-Ser-Asn-Ser-Ser-Trp-Trp-Thr-Asn-Trp-Val-Ile-Pro-Ala-Ile-Ser-Ala-Leu-Val-Val-Ser-Leu-Met-Tyr-His-Phe-Tyr-Thr-Ser-Glu-Asn. A prediction of alpha-helices, beta-structures, and beta-turns basedon the sequence of this polypeptide is also presented.
Article
The techniques described allow controlled permeabilization of plasma membranes from different types of cells for gaining access to the cell interior and enables one to control intracellular events. Most common techniques are electropermeabilization, permeabilization with mild non-ionic detergents such as saponin and digitonin and by pore-forming toxins, such as alpha-toxin and streptolysin O. Whereas electropermeabilization and alpha-toxin create small pores of approximately 2 nm, digitonin, saponin, and streptolysin O form bigger holes and therefore also allow the introduction of large molecules, such as enzymes and immunoglobulins. A disadvantage of the latter methods is the loss of cytosolic constituents which might be necessary for signal-transduction pathways in the cell. In secretory cells the main requirement for exocytosis appears to be Ca2+, which brings about the full response comparable to hormone effects in some cells (platelets), adrenal medullary cells, but not in all cells (pancreatic acinar cells). The nucleotide, anion, and cation requirements are different for different cell types and are probably intimately related to the cell-specific mechanisms involved in exocytosis such as regulation of ion channels and ion carriers, or the involvement of nucleotide-binding proteins. Since permeabilized cells are preparations intermediate between intact cells and isolated organelles, they offer great opportunities for the advancement of our understanding of the mechanisms involved in stimulus-response coupling.