ArticlePDF Available

The Role of the Propeptide for Processing and Sorting of Human Myeloperoxidase

Authors:

Abstract

Myeloperoxidase (MPO), stored in azurophil granules of neutrophils, is critical for an optimal oxygen-dependent microbicidal activity of these cells. Pro-MPO goes through a stepwise proteolytic trimming with elimination of an amino-terminal propeptide to yield one heavy and one light polypeptide chain. The propeptide of MPO may have a role in retention and folding of the nascent protein into its tertiary structure or in targeting of pro-MPO for processing and storage in granules. A propeptide-deleted pro-MPO mutant (MPOΔpro) was constructed to determine if deletion of the propeptide interferes with processing and targeting after transfection to the myeloid 32D cell line. Transfection of full-length cDNA for human MPO results in normal processing and targeting of MPO to cytoplasmic dense organelles. Although the efficiency of incorporation was lower for MPOΔpro, both pro-MPO and MPOΔpro showed heme incorporation indicating that the propeptide is not critical for this process. Deletion of the propeptide results in synthesis of a protein that lacks processing into mature two-chain forms but rather is degraded intracellularly or secreted. The finding of continued degradation of MPOΔpro in the presence of lysosomotrophic agents or brefeldin A rules out that the observed degradation takes place after transfer to granules. Intracellular pro-MPO has high mannose oligosaccharide side chains, whereas stored mature MPO was found to have both high mannose and complex oligosaccharide side chains as judged by only partial sensitivity to endoglycosidase H. The propeptide may normally interfere with the generation of certain complex oligosaccharide chain(s) supported by the finding of high mannose side chains in secreted pro-MPO and lack of them in MPOΔpro that contained complex oligosaccharide side chains only. In conclusion, elimination of the propeptide of pro-MPO blocks the maturation process and abolishes accumulation of the final product in granules suggesting a critical role of the propeptide for late processing of pro-MPO and targeting for storage in granules.
The Role of the Propeptide for Processing and Sorting of Human
Myeloperoxidase*
(Received for publication, June 6, 1997, and in revised form, December 4, 1997)
Elinor Andersson‡, Lars Hellman§, Urban Gullberg‡, and Inge Olsson‡
From the Department of Hematology, Research Department 2, E-blocket, University Hospital, S-221 85 Lund
and the §Department of Medical Immunology and Microbiology, University of Uppsala Biomedical Center,
Box 582, S-751 23 Uppsala, Sweden
Myeloperoxidase (MPO), stored in azurophil granules
of neutrophils, is critical for an optimal oxygen-depend-
ent microbicidal activity of these cells. Pro-MPO goes
through a stepwise proteolytic trimming with elimina-
tion of an amino-terminal propeptide to yield one heavy
and one light polypeptide chain. The propeptide of MPO
may have a role in retention and folding of the nascent
protein into its tertiary structure or in targeting of pro-
MPO for processing and storage in granules. A propep-
tide-deleted pro-MPO mutant (MPODpro) was con-
structed to determine if deletion of the propeptide
interferes with processing and targeting after transfec-
tion to the myeloid 32D cell line. Transfection of full-
length cDNA for human MPO results in normal process-
ing and targeting of MPO to cytoplasmic dense
organelles. Although the efficiency of incorporation was
lower for MPODpro, both pro-MPO and MPODpro
showed heme incorporation indicating that the propep-
tide is not critical for this process. Deletion of the
propeptide results in synthesis of a protein that lacks
processing into mature two-chain forms but rather is
degraded intracellularly or secreted. The finding of con-
tinued degradation of MPODpro in the presence of lyso-
somotrophic agents or brefeldin A rules out that the
observed degradation takes place after transfer to gran-
ules. Intracellular pro-MPO has high mannose oligosac-
charide side chains, whereas stored mature MPO was
found to have both high mannose and complex oligosac-
charide side chains as judged by only partial sensitivity
to endoglycosidase H. The propeptide may normally in-
terfere with the generation of certain complex oligosac-
charide chain(s) supported by the finding of high man-
nose side chains in secreted pro-MPO and lack of them
in MPODpro that contained complex oligosaccharide
side chains only. In conclusion, elimination of the
propeptide of pro-MPO blocks the maturation process
and abolishes accumulation of the final product in gran-
ules suggesting a critical role of the propeptide for late
processing of pro-MPO and targeting for storage in
granules.
Neutrophil granulocytes are specialized for a role in host
defense. A regulated pathway targets enzymes and antibiotic
proteins to a storage compartment in these cells consisting of
cytoplasmic azurophil, specific, and gelatinase granules formed
sequentially, whereas a constitutive pathway exports proteins
to the cell surface (1). Azurophil granules are thought to be
specialized lysosomes, and their protein constituents are often
subject to posttranslational glycosylation and proteolytic trim-
ming similar to that of lysosomal enzymes (2). A retention
mechanism may be necessary to avoid constitutive secretion of
granule proteins, and a condensation mechanism is necessary
for efficient packaging. Signals for targeting storage in gran-
ules have been sought within the structure of neutrophil gran-
ule proteins. For instance, a pro-region segment is necessary
for targeting to granules of neutrophil defensins (3), but car-
boxyl-terminal prodomains or asparagine-linked carbohy-
drates of hematopoietic serine proteases are not required in
targeting storage in granules (4, 5). Myeloperoxidase (MPO)
1
of
azurophil granules plays a major role in the oxygen-dependent
killing of microorganisms after release into phagolysosomes by
amplifying the effects of oxygen derivatives formed during the
respiratory burst (6). Pro-MPO undergoes extensive process-
ing, including the removal of an amino-terminal propeptide not
found in mature MPO. Therefore, in this work we have inves-
tigated whether the propeptide of MPO has a role in intracel-
lular trafficking and targeting to granules.
The processing steps for MPO are shown in Fig. 1. Mature
MPO is a 150-kDa tetramer composed of two glycosylated 59
64-kDa heavy subunits and two unglycosylated 14-kDa light
subunits as a pair of protomers linked together by a disulfide
bond (7). Each heavy subunit carries a covalently bound heme
prosthetic group (8), although the crystal structure of canine
MPO suggests that heme of the intact molecule associates with
both subunits (9). The primary translation product undergoes
cotranslational glycosylation with production of 89-kDa heme-
free apopro-MPO followed by incorporation of heme and con-
version into enzymatically active pro-MPO (7). Processing and
maturation of pro-MPO is a slow process (10) that can be
accomplished only after acquisition of heme (11–13). Calreticu-
lin, a calcium-binding protein that resides in the ER, has been
suggested to function as a molecular chaperone and facilitate
the critical folding of apopro-MPO to allow insertion of heme
followed by conversion to pro-MPO (14). The stepwise process-
ing of pro-MPO has been investigated in myeloid cells (10,
15–20), and the results obtained are consistent with those later
deduced from cDNA sequence data. Thus, during subsequent
processing of pro-MPO the amino-terminal propeptide, a small
peptide between the light and heavy chains, and a single serine
residue at the carboxyl-terminal are removed (21). Intermedi-
* This work was supported by the Swedish Cancer Foundation, the
Swedish Medical Research Council Project No. 11546, the Alfred O
¨
ster-
lund Foundation, and the Greta and Johan Kocks Foundation. The
costs of publication of this article were defrayed in part by the payment
of page charges. This article must therefore be hereby marked adver-
tisement in accordance with 18 U.S.C. Section 1734 solely to indicate
this fact.
To whom correspondence should be addressed: Research Dept. 2,
E-blocket, University Hospital, S-221 85 Lund, Sweden. Tel.: 46-46-
173533; Fax: 46-46-184493; E-mail: inge.olsson@hematologi.lu.se.
1
The abbreviations used are: MPO, myeloperoxidase; [4-
14
C]ALA,
d
-[4-
14
C]aminolevulinic acid hydrochloride; ER, endoplasmic reticulum;
Endo-H, endoglycosidase H; N-glycanase, N-glycosidase F; PCR, polym-
erase chain reaction; BFA, brefeldin A.
THE JOURNAL OF BIOLOGICAL CHEMISTRY Vol. 273, No. 8, Issue of February 20, pp. 4747–4753, 1998
© 1998 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.org 4747
by guest on July 10, 2015http://www.jbc.org/Downloaded from
ate processing forms have been observed with molecular
masses of 81 and 74 kDa (10, 18, 19, 22) of which the smaller
can be converted directly into mature MPO after cleavage
between the heavy and the light subunit (19, 22). This finding
suggests that the amino-terminal propeptide, which does not
seem to be part of the 74-kDa form, is removed during an
intermediate step before final processing.
One can envision a role for the amino-terminal propeptide of
MPO in retention and folding of the nascent protein into its
tertiary structure or in targeting pro-MPO to pregranule struc-
tures for further processing and storage in granules. A propep-
tide-deleted pro-MPO mutant (MPODpro) was constructed to
determine if propeptide deletion interferes with processing and
targeting. In this work, we describe the consequences of these
manipulations for posttranslational processing, intracellular
sorting, and constitutive secretion after transfection of the
cDNA for MPO and MPODpro into the murine myeloid 32D
clone 3 cell line.
EXPERIMENTAL PROCEDURES
Materials—The eucaryotic expression vector pCDNA 3 was from
Invitrogen, British Biotechnology, Oxon, UK. The vector provides a
cytomegalovirus promoter-driven expression of introduced cDNA. The
plasmid also confers resistance to geneticin, allowing selection of re-
combinant cells. [
35
S]Methionine/[
35
S]cysteine (cell labeling grade) was
from Amersham International (Buckinghamshire, UK).
d
-[4-
14
C]Ami-
nolevulinic acid hydrochloride ([4-
14
C]ALA) was from DuPont, Belgium.
Prior to use, [4-
14
C]ALA was concentrated 10-fold in a vacuum centri-
fuge and pH adjusted with NaOH. Percoll and protein A-Sepharose
CL-4B were from Pharmacia (Uppsala, Sweden). Protein G-Sepharose
was from Sigma. Geneticin, N-glycosidase F (N-glycanase) and endogly-
cosidase H (Endo-H) were from Boehringer Mannheim (Mannheim,
Federal Republic of Germany). Brefeldin A (BFA), a gift from Sandoz
AB, was dissolved in methanol and stored at 220 °C.
cDNA, Mutagenesis, and Construction of Expression Vector—A par-
tial cDNA clone encoding approximately 80% of human prepro-MPO
was obtained from the American Type Culture Collection (clone
pMP503, ATCC 57694). This clone lacks the coding region for the
amino-terminal part of the protein. To obtain a full-length clone for
transfection studies, an approximate 500-base pair fragment, originat-
ing from the 59 end of the mRNA, was isolated by PCR amplification.
This fragment was isolated by using one primer directed against a
region starting approximately 80 base pairs upstream of the start codon
and a second primer directed against a region just downstream of a
single XbaI site present in the 59 end of the clone pMP503 (in the coding
region of the clone). The PCR fragment was cloned as an RsaI/XbaI
fragment, and the entire nucleotide sequence was determined for two
separate clones. One of the clones was found to have an identical
sequence to that for MPO. To obtain the full-length MPO clone, two
separate fragments were ligated into the vector pcDNAIneo, one XbaI/
EcoRI fragment originating from the pMP503 clone and one HindIII/
XbaI fragment from the PCR clone. After sequence analysis of 59 and 39
ends of the resulting clone and restriction mapping for a panel of
internal sites, this clone was used for the subsequent transfection
studies and as starting material for the construction of the MPODpro.
Construction of cDNA of MPO Lacking the Propeptide (MPODpro)—For
site-directed mutagenesis cDNA of human MPO (pcDNA1neo/MPO) was
used as template in a two-step “spliced overhang extension” polymerase
chain reaction in the following way. In the first reaction two separate
amplifications with 100 ng of DNA template in a 20-cycle PCR produced two
fragments of myeloperoxidase positioned amino-terminally and carboxyl-
terminally of the propeptide (Pro
43
-Gly
164
), respectively (Fig. 2). By design of
the primers, the “Kozak” consensus leader sequence for maximum transla-
tional efficiency was introduced 59 to the ATG initiation codon, and the
flanking restriction enzyme sites HindIII and BamHI were included for
subsequent cloning into plasmid. The PCR primers in the two amplifications
were upstream 59-GACTTCAAGCTTGCCACCATGGGGGTTCCCTTCTT-
CTCT-39 (primer 1) plus downstream 59-CGGGCAAGTCACCCCCACGTC-
GGGCTGGGGCGTGGCCAGAAT-39 (primer 2), and upstream 59-GACGT-
GGGGGTGACTTGCCCG-39 (primer 3) plus downstream 59-CTTCAGGGA-
TCCCTAGGAGGCTTCCCTCCAGGA-39 (primer 4), respectively (start and
stop codons in boldface and restriction enzyme sites underlined). The PCR
products were isolated on agarose gel, mixed, and subjected to a second
20-cycle splicing PCR amplification with primers 1 and 4, thus creating
MPO lacking the propeptide (MPODpro). The resulting PCR product was
digested by HindIII and BamHI, followed by isolation on agarose gel and
cloning into plasmid (pcDNA3) to create the expression vector
pcDNA3/MPODpro.
All PCRs were performed in a Perkin-Elmer 480 Thermal Cycler
using Pfu polymerase (Stratagene, La Jolla, CA) according to the man-
ufacturer’s instructions.
Cell Culture—32D clone 3 cells (23, 24), kindly provided by G. Rovera
(Philadelphia, PA) were grown in complete medium consisting of
Iscove’s modified Dulbecco’s medium supplemented with 10% heat-
inactivated fetal bovine serum and 30% WEHI-conditioned medium as
a source of interleukin 3 (25). The cell cultures were kept in 5% CO
2
at
37 °C in a fully humidified atmosphere. Exponentially growing cells
were used in all experiments.
Transfection Procedure—32D cells were transfected with
pcDNA1neo/MPO and pcDNA3/MPODpro using the Bio-Rad Electropo-
ration Apparatus (Bio-Rad) with electrical settings of 960 microfarads
and 300 V as described previously (5). Forty-eight hours after electro-
FIG.1. The processing steps of MPO. The primary translation
product undergoes cotranslational cleavage of the signal peptide fol-
lowed by N-linked glycosylation to generate apopro-MPO. Initial proc-
essing also includes acquisition of heme, which yields enzymatically
active pro-MPO. The stepwise processing into mature dimeric MPO
includes removal of the propeptide.
FIG.2.Schematic view of MPO and of the MPO deletion mu-
tant (MPODpro). PCR primers used for construction as described
under “Experimental Procedures” are indicated with arrows. Amino
acids are indicated by three-letter symbols. SP, signal peptide; PRO
propeptide; LIGHT, light chain of mature MPO; HEAVY, heavy chain of
mature MPO.
Myeloperoxidase Processing4748
by guest on July 10, 2015http://www.jbc.org/Downloaded from
poration, geneticin (1 mg/ml) was added to select for recombinant clones
expressing the geneticin resistance of pcDNA 3. Individual clones grow-
ing in the presence of antibiotic were isolated, expanded into mass
cultures, and screened by biosynthetic radiolabeling for expression of
the protein encoded by the transfected cDNA. Clones with the most
pronounced expression were chosen for further experiments.
Biosynthetic Radiolabeling—Biosynthetic radiolabeling of newly
synthesized proteins was performed as described (26). Briefly, cells
were starved for 30 min in methionine/cysteine-free medium, followed
by radiolabeling with 15 or 30
m
Ci/ml [
35
S]methionine/[
35
S]cysteine for
30 or 60 min. In experiments with [4-
14
C]ALA labeling, cells were
incubated with 25
m
Ci/ml for 3 h for radiolabeling. In chase experi-
ments, following radiolabeling, cells were resuspended in complete me-
dium. At timed intervals, cells were withdrawn and lysed or homoge-
nized for subcellular fractionation.
Subcellular Fractionation—Subcellular fractionation was performed
as described (26). Briefly, the postnuclear cell homogenate was fraction-
ated in a Percoll density gradient, after which nine fractions were
collected with all cytosol in fraction 9. The distribution of lysosomes and
Golgi elements in the density gradient was determined by assaying
b
-hexosaminidase and
b
-galactosyltransferase as described elsewhere
(27, 28). Peak activities of
b
-hexosaminidase and galactosyltransferase
were localized in fractions 2 and 6, respectively (data not shown).
Immunoprecipitation—For immunoprecipitation, whole cells or Per-
coll-containing subcellular fractions were solubilized, and biosyntheti-
cally radiolabeled MPO or MPODpro was precipitated with polyclonal
anti-MPO (29) and subjected to electrophoretic analysis followed by
fluorography as described previously (26, 30).
Digestion with Endo-H and N-Glycanase—The susceptibility of MPO
and MPODpro to digestion with Endo-H and N-glycanase was deter-
mined as described (26, 30).
RESULTS
Construction of Full-length and Mutated MPO and Estab-
lishment of Stable Transfectants—To determine whether the
propeptide of pro-MPO carries a targeting signal for granules,
a mutant form of MPO lacking the propeptide (MPODpro) was
constructed by polymerase chain reactions as described under
“Experimental Procedures.” The sequence encoding 122 amino
acids from Pro
43
to Gly
164
(numbered from the methionine
constituting the translation initiation site) was deleted from
MPO cDNA leaving the first three residues of the propeptide
and the entire signal peptide intact (Fig. 2). If the propeptide
plays a role for sorting, MPODpro-protein would, unlike intact
pro-MPO, not be targeted to granules. Likewise, if the propep-
tide plays a role for folding of nascent protein, the mutant
protein might be misfolded and retained in the ER.
Wild type MPO and MPODpro were transfected to 32D cells,
and stable cell clones were established. Clones with synthesis
of protein from transfected cDNA were chosen for further ex-
periments in which the consequences of the MPO mutation
were investigated. The murine origin of the cell line facilitates
the detection of expression of transfected human proteins by
biosynthetic radiolabeling followed by immunoprecipitation.
No endogenous synthesis of MPO in 32D cells is detected with
the antiserum used (data not shown). 32D cells have cytoplas-
mic granule-like vacuoles that have been shown to be able to
accumulate human neutrophil granule constituents such as
defensins expressed from DNA transfected into these cells (3).
In addition, the human hematopoietic serine proteases cathep-
sin G and proteinase 3 have been successfully transfected to
32D cells and targeted to the granule-like vacuoles (5, 32).
These cells were also found to have the machinery for process-
ing of MPO (see below).
Human Wild Type MPO in 32D Cells Is Processed and Tar-
geted to Granules—Stable 32D cells transfected with wild type
cDNA of human MPO show a biosynthesis and processing
pattern of MPO similar to that of human myeloid cells express-
ing MPO. The initially detectable protein is a proform of mo-
lecular mass 89 kDa (pro-MPO) (Fig. 3). As observed earlier in
promyelocytic HL-60 (9, 22, 33) and in PLB 985 cells (11),
processing of pro-MPO into the mature form is slow. A slow
processing of pro-MPO is also observed in transfected 32D cells.
Thus, a 64-kDa heavy chain and a 15-kDa light chain, repre-
senting mature MPO, begin to occur between 6 and 24 h of
chase of the radiolabel (Fig. 3). Additional MPO species with
molecular masses of approximately 45 kDa, precipitated with
anti-MPO, increase with chase of the radiolabel. These pep-
tides are known to be the result of autolytic cleavage of the
heavy subunit (34). Similar to the behavior of endogenous MPO
in myeloid cells, constitutive secretion of pro-MPO to medium
proceeds continually from 32D cells during chase of the
radiolabel (Fig. 3).
MPO is normally targeted to azurophil granules of the neu-
trophil series for storage. To investigate targeting in MPO-
transfected 32D cells, pulse-chase radiolabeling experiments
followed by subcellular fractionation were performed (Fig. 4).
Radiolabeled MPO was found to be slowly translocated to dense
fractions containing the granule-like vacuoles, where it is
clearly visible after 22 h of chase. Translocated (granule-asso-
ciated) MPO is present almost exclusively in fully mature form
consisting of a 64-kDa heavy and 15-kDa light chain. These
results are consistent with earlier data from investigations on
the processing of endogenous MPO in myeloid cells (10, 15–20)
and demonstrate that 32D cells can process human pro-MPO
into mature MPO that is at least partially targeted to a dense
subcellular fraction containing granules.
Both Wild Type MPO and Propeptide-deleted MPO (MPODpro)
Incorporate Heme—Proteolytic processing of endogenous wild type
MPO precursor to the mature storage form in granules requires
incorporation of heme into pro-MPO (11–13). Thus, heme incorpo-
ration occurs prior to removal of the propeptide, and it is therefore
of interest to determine whether the propeptide is necessary for
incorporation of heme. Wild type MPO and propeptide-deleted
MPO (MPODpro) in transfected 32D cells were therefore compared
in this respect. Cells were radiolabeled with [4-
14
C]ALA, a precur-
sor of heme synthesis, followed by immunoprecipitation with an
anti-MPO antibody. As expected, the proform of wild type MPO
incorporates heme, indicated by labeling of the protein with
[4-
14
C]ALA (Fig. 5), confirming earlier results (9). Incorporation of
heme into the propeptide-deleted MPO (MPODpro) is also seen
FIG.3. Processing of MPO in 32D cells. Cells transfected with
cDNA for human MPO were incubated with [
35
S]methionine/[
35
S]cys-
teine for 30 min followed by chase of the label for up to 24 h. At depicted
time points, 20 3 10
6
cells were removed and, after lysis, subjected to
immunoprecipitation with anti-MPO. In addition, MPO was also pre-
cipitated from the incubation medium at each chase time point. The
immunoprecipitates were analyzed by SDS-polyacrylamide gel electro-
phoresis in a 5–20% gradient gel followed by fluorography. The fluoro-
gram was exposed for 3 days. The positions of the pro-MPO (pro), the
heavy (
a
) and the light (
b
) subunits are indicated to the right with
arrows. Numbers to the left in this and subsequent figures are the
values of molecular mass standards.
Myeloperoxidase Processing 4749
by guest on July 10, 2015http://www.jbc.org/Downloaded from
(Fig. 5), and this form has a molecular mass of 76 kDa. Thus, the
presence of the propeptide is not necessary for incorporation of
heme into the proform of MPO. On the other hand, the relative
efficiency of insertion of heme seemed to be lower for MPODpro
compared with the normal proform. This comparison was possible
to make as control radiolabeling with [
35
S]methionine/[
35
S]cysteine
showed immunoprecipitates with similar density for both MPO and
MPODpro also when visualized through shorter exposure time of
the fluorogram than in Fig. 5 (not shown).
Lack of Processing and Targeting to Granules of Propeptide-
deleted MPO—32D cells expressing MPODpro show an abnor-
mal biosynthesis and processing pattern of MPO (Fig. 6). As
expected, a 76-kDa polypeptide is synthesized that may corre-
spond to the size of a pro-MPO that lacks propeptide. However,
deletion of the propeptide results in synthesis of a protein that
lacks processing into mature two-chain forms and is secreted
upon prolonged chase of the radiolabeled product or degraded
intracellularly (Fig. 6). Unlike secreted full-length pro-MPO
(Fig. 3), secreted MPODpro disappears with time (Fig. 6), sug-
gesting extracellular degradation. To investigate if MPODpro
could be transferred to granules, pulse-chase radiolabeling ex-
periments were carried out followed by subcellular fraction-
ation. Only trace amounts of radiolabeled MPODpro and deg-
radation products are visible in dense fractions containing the
granule-like vacuoles (Fig. 7).
Since only trace amounts of partially degraded MPODpro are
detected in dense fractions, the results suggest that degrada-
tion of non-secreted MPODpro preferentially takes place in a
pre-granule compartment. However, it is possible that substan-
tial degradation of MPODpro would still take place in granule-
like vacuoles but too rapidly to be detectable. Therefore, to rule
out considerable transfer to granules with degradation, exper-
iments were performed with chloroquine and NH
4
Cl, agents
that block lysosomal proteolysis. In particular, chloroquine
blocks late proteolytic processing of MPO (16). Biosynthetic
radiolabeling of MPODpro in transfected 32D cells and chase of
the radiolabel in the presence of NH
4
Cl or chloroquine does not
diminish the degradation rate of MPODpro, indicating that
lysosomes are not involved to any great extent in the degrada-
tion observed (Fig. 8). The secretion of MPODpro also occurs
FIG.4.Targeting of MPO to granules in 32D cells. Cells trans-
fected with cDNA for human MPO were incubated with [
35
S]methi-
onine/[
35
S]cysteine for 30 min followed by chase of the label for 5 and
22 h. At these time points, 100 3 10
6
cells were homogenized after
which subcellular fractionation of the postnuclear supernatant was
performed by centrifugation in Percoll followed by collection of nine
subcellular fractions, fraction 9 containing all the cytosol. The fractions
were lysed and subjected to immunoprecipitation with anti-MPO. Im-
munoprecipitates were analyzed as described in the legend to Fig. 3.
The fluorogram was exposed for 11 days. The positions of the pro-MPO
(pro), the heavy (
a
), and the light (
b
) subunits are indicated to the right
with arrows. Peak activities of
b
-hexosaminidase, fraction 2, and galac-
tosyltransferase, fraction 6, indicate the position of lysosomes and Golgi
elements, respectively.
FIG.5.Incorporation of heme into pro-MPO and propeptide-
deleted MPO (MPODpro). 32D cells (20 3 10
6
) transfected with
cDNA for MPO or MPODpro were incubated for 3 h with [4-
14
C]ALA
and for 2 h with [
35
S]methionine (
35
S-meth)/[
35
S]cysteine followed by
lysis and immunoprecipitation with anti-MPO. Immunoprecipitates
were analyzed as described in the legend to Fig. 3. The fluorogram was
exposed for 12 weeks. The positions of the proform of MPO (pro) and the
MPODpro (Dpro), respectively, are indicated with arrows.
FIG.6. Lack of processing of propeptide-deleted MPO
(MPODpro) in 32D cells. Cells transfected with cDNA for human
MPODpro were incubated with [
35
S]methionine/[
35
S]cysteine for 30 min
followed by chase of the label for up to 24 h. At depicted time points,
20 3 10
6
cells were removed and, after lysis, subjected to immunopre-
cipitation with anti-MPO. In addition, MPO was also precipitated from
the incubation medium at each chase time point. Immunoprecipitates
were analyzed as described in the legend to Fig. 3. The position of the
MPODpro (Dpro) is indicated with an arrow. The fluorogram was ex-
posed for 5 days.
Myeloperoxidase Processing4750
by guest on July 10, 2015http://www.jbc.org/Downloaded from
during incubation with NH
4
Cl and chloroquine, although it is
reduced by NH
4
Cl. Secreted MPODpro is of slightly higher
molecular weight than the protein retained intracellularly both
in the control and in the presence of NH
4
Cl or chloroquine,
indicating additional glycosylation during the secretory proc-
ess. To characterize the localization of the degradation further,
cell radiolabeling experiments were performed in the presence
of brefeldin A (BFA), which induces the disassembly of the
Golgi complex, thus blocking ER-Golgi transport (35, 36). As
expected, BFA blocks the secretion of MPODpro completely, but
total degradation is observed with time (Fig. 8), indicating that
degradation of MPODpro can occur in a pre-Golgi compart-
ment. A higher molecular weight form of MPODpro is observed
with time in the presence of BFA (Fig. 8). This form, which is
also degraded, is probably the result of aberrant glycosylation
in the presence of BFA.
Secreted MPODpro Is Resistant to Endo-H Indicating Com-
plex Oligosaccharide Side Chains—Both intracellular pro and
mature MPO are normally sensitive to digestion with Endo-H
indicating the presence of high mannose oligosaccharide side
chains (16, 17, 20). Consistent with published data, the intra-
cellular forms of pro and mature MPO in 32D cells transfected
with wild type MPO both show sensitivity to digestion with
Endo-H indicating the presence of high mannose groups (Fig.
9A). However, an additional reduction in size is observed for
the large subunit of mature MPO upon digestion with N-gly-
canase as compared with digestion with Endo-H. To ensure
that complete digestion had taken place with Endo-H and
N-glycanase, the concentrations of glycosidase were varied
(Fig. 9A). The results show that the molecular mass is reduced
by 4.5 kDa upon complete digestion with Endo-H and by 12.5
kDa upon complete digestion with N-glycanase (mean values
from two separate experiments). Thus, the large subunit con-
tains Endo-H-resistant oligosaccharides indicating the pres-
ence of complex oligosaccharides. The presence not only of high
mannose but also of complex oligosaccharides in the large
subunit of MPO has for technical reasons been overlooked in
previous studies. We observed both high mannose and complex
oligosaccharides also in the large MPO subunit of HL-60 cells
that normally produce MPO (data not shown). Also the secreted
pro-MPO shows partial Endo-H resistance both in the 32D cells
transfected with MPO (Fig. 9A) and in HL-60 cells (data not
shown). This indicates that the secreted pro-MPO achieves
some complex mannose groups during passage through the
Golgi compartment during constitutive secretion. The secreted
pro-MPO shows heterogeneity, and removal of all oligosaccha-
rides with N-glycanase reveals at least two distinct protein
forms that differ in molecular mass (Fig. 9A). The smaller
protein form is resistant and the larger is sensitive to Endo-H.
Two distinct protein forms are also observed for secreted pro-
MPO in the HL-60 cell line (data not shown).
The glycosylation pattern for MPODpro is shown in Fig. 9B.
Intracellular MPODpro is highly sensitive to digestion with
Endo-H. In contrast, the secreted MPODpro is homogeneous
and Endo-H-resistant corresponding to the presence of complex
oligosaccharide side chains. As judged by results from digestion
with N-glycanase, the secreted form contains slightly more
carbohydrate than the intracellular form consistent with proc-
essing into complex forms.
DISCUSSION
Neutrophils carry at least the following three types of gran-
ules: azurophil, specific, and gelatinase granules (37). Unique
constituents of azurophil granules such as MPO and serine
proteases are stored in enzymatically active forms, whereas
proteases of specific and gelatinase granules are stored in
FIG.7. Lack of targeting of propeptide-deleted MPO
(MPODpro) in 32D cells. Cells transfected with cDNA for human
MPODpro were incubated with [
35
S]methionine/[
35
S]cysteine for 1 h
followed by chase of the label for 3 and 12 h. At these time points, 100 3
10
6
cells were homogenized after which subcellular fractionation of the
postnuclear supernatant was performed by centrifugation in Percoll
followed by collection of nine subcellular fractions, fraction 9 containing
all the cytosol. The fractions were lysed and subjected to immunopre-
cipitation with anti-MPO. Immunoprecipitates were analyzed as de-
scribed in the legend to Fig. 3. The position of MPODpro (Dpro)is
indicated with an arrow. The fluorogram was exposed for 5 days. Peak
activities of
b
-hexosaminidase, fraction 2, and galactosyltransferase,
fraction 6, indicate the position of lysosomes and Golgi elements,
respectively.
FIG.8.The effect of NH
4
Cl, chloroquine, and brefeldin A on the
processing of propeptide-deleted MPO (MPODpro) in 32D cells.
Cells transfected with cDNA for human MPODpro were incubated with
[
35
S]methionine/[
35
S]cysteine for 30 min followed by chase of the label
for up to 24 h. Separate experiments were carried out with 10 mmol/
liter NH
4
Cl, 1 mmol/liter chloroquine, or 5 mg/ml brefeldin A; cells were
preincubated with these agents for 30 min, and the agents were also
present during pulse labeling and during chase of the label. At depicted
time points, 20 3 10
6
cells were removed and, after lysis, subjected to
immunoprecipitation with anti-MPO. In addition, MPO was also pre-
cipitated from the incubation medium at each chase time point. Immu-
noprecipitates were analyzed as described in the legend to Fig. 3. The
fluorograms were exposed for 5 days. The position of MPODpro (Dpro)is
indicated with an arrow.
Myeloperoxidase Processing 4751
by guest on July 10, 2015http://www.jbc.org/Downloaded from
inactive forms to become activated first after exocytosis (1).
Thus, azurophil granule enzymes are activated prior to storage,
e.g. by removal of an activation peptide from proforms of the
serine proteases (38). The activation peptide keeps enzyme
activity latent and is removed as a late step during intracellu-
lar trafficking. Likewise, the propiece of prodefensin is re-
moved before storage in azurophil granules of mature antibi-
otic defensin, which is non-catalytic. In this case the propiece is
essential for subcellular trafficking and sorting, e.g. by inter-
action with a complementary hydrophobic part of mature de-
fensin peptide or with a chaperone protein that facilitates
transit and protects against adverse effects of the mature pep-
tide (3). MPO, on the other hand, is enzymatically active prior
to proteolytic removal of its propiece (1). Therefore, the pro-
piece of pro-MPO probably does not have a role in protection
against peroxidation during intracellular travelling unless it
interacts with other molecules for this purpose. Rather, the
propiece may play a role in conformational stability, retention,
and/or sorting. The results of the present work are viewed in
this context.
The murine myeloid 32D cell line was successfully employed
for investigation of MPO synthesis. Thus 32D cells stably
transfected with the cDNA for human MPO demonstrate the
normal characteristics of MPO synthesis. Heme is incorporated
into apopro-MPO resulting in production of pro-MPO that is
processed into mature heterodimeric protein targeted to gran-
ule-like vacuoles of 32D cells. The same cell line has previously
been utilized for the investigation of the posttranslational proc-
essing of human neutrophil defensin (3) and neutrophil serine
proteases (5, 32). Previous attempts to use Chinese hamster
ovary cells (3941), baby hamster kidney cells (42), or baculo-
virus-infected Sf9 cells (43) for expression of MPO cDNA have
not resulted in processing of the protein product. But, recently
the human erythroleukemia K562 cell line transfected with
MPO cDNA showed the typical processing seen during biosyn-
thesis of MPO in myeloid cells (44).
What do our results reveal about the role of the propiece for
post-translational processing, targeting, and secretion of MPO?
Propeptide-deleted pro-MPO (MPODpro) was found to lack
processing into mature light and heavy chain MPO and pri-
marily became secreted to the exterior or degraded in a
pregranule compartment. Therefore, the propiece is necessary
for subcellular trafficking. The finding of continued degrada-
tion of MPODpro in the presence of lysosomotrophic agents and
BFA rules out that the observed degradation should, after all,
take place upon transfer to granules but too rapidly to be
detectable. The results obtained for MPO processing can be
compared with those for the lysosomal hydrolase cathepsin D,
in which the precursor domains are indispensible for the for-
mation of a stable proenzyme (45). Thus, in the latter case the
propeptide appears to be necessary for the correct folding of the
proenzyme that is required for trafficking. However, it was not
possible to prove a direct role for the propeptide of cathepsin D
in sorting, because the propeptide when attached to a secretory
protein,
a
-lactalbumin, did not redirect it for lysosomes indi-
cating that the propeptide might not be necessary for the sort-
ing process as such (45). If a sorting machinery were to recog-
nize precursors rather than mature peptides, propiece-deleted
pro-MPO when available for sorting would be secreted instead
of being sorted for storage in granules. This seems to be con-
sistent with the finding that a large part of MPODpro is se-
creted, whereas almost none is transported to granules. How-
ever, a part of MPODpro is retained, most likely in the ER, and
degraded. Proteasomes may have a role, although unproven, in
proteolysis of MPODpro. One theoretical explanation is mis-
folding; if the propiece were required for folding, misfolding of
a propiece-deleted pro-MPO might lead to retention in the ER
because of lack of native conformation. On the other hand, the
characteristics of MPODpro do not indicate misfolding, because
MPODpro can incorporate heme, can be secreted, and can
achieve complex oligosaccharide side chains (see below) when
transferred to trans-Golgi. It is important to consider that
processing of pro-MPO is normally extremely slow, and it can
take as long as 6–15 h to chase radiolabel from the precursor
into mature MPO (10, 11). If MPODpro were degraded within
this period processing forms would not be visible. Thus, it is
suggested that undegraded MPODpro is transferred to Golgi
and secreted because the lack of propeptide prohibits targeting
to granules. In conclusion, removal of the propiece leads to a
block in the normal trafficking and maturation process of MPO.
The behavior of pro-MPO and MPODpro differs during se-
cretion. First, the molecular mass of secreted MPODpro, but
not of secreted pro-MPO, is slightly higher than that of the
corresponding intracellular forms. Second, in contrast to
MPODpro, intracellular pro-MPO in HL-60 cells (16) or trans-
fected pro-MPO of 32D cells of the present work does not
acquire detectable complex oligosaccharide side chains. There-
FIG.9. Oligosaccharide side chains of processing forms of
MPO and MPODpro. 32D cells transfected with MPO (A)orMPODpro
(B) were incubated with 30
m
Ci/ml [
35
S]methionine/[
35
S]cysteine for 30
min (pulse). Chase experiments of the radiolabel were performed for
24h(A)and6h(B), respectively. At depicted time points, 100 3 10
6
cells were removed and, after lysis, subjected to immunoprecipitation
with anti-MPO. In addition, MPO or MPODpro, respectively, was also
precipitated from the incubation medium at the end of chase. A, ali-
quoted immunoprecipitates were incubated with either Endo-H (Endo-
H1) (units 3 10
22
/ml) or N-glycanase (N-Glyc1) (units 3 10
21
/ml), or
served as controls (2). Immunoprecipitates of cells from 24-h chase
experiments were incubated with several different concentrations of
Endo-H or N-glycanase to ensure that complete digestion had taken
place, but results are given only for two concentrations of glycosidases.
B, aliquoted immunoprecipitates were incubated with either 0.1
units/ml of Endo-H (Endo-H1) or 7 units/ml of N-glycanase (N-glyc 1)
for 24 h, or served as controls (2). In both A and B SDS-polyacrylamide
gel electrophoresis and fluorography were performed, and the fluoro-
grams were exposed for 18 days.
Myeloperoxidase Processing4752
by guest on July 10, 2015http://www.jbc.org/Downloaded from
fore, the propeptide of pro-MPO may promote resistance to
mannosidases and/or glycosyltransferases whose action is re-
quired for production of complex mannose groups in a late
Golgi compartment. However, results from baby hamster kid-
ney cells transfected with MPO have shown that secreted pro-
MPO contains at least one Endo-H-resistant oligosaccharide
indicating the presence of complex mannose groups (42). Thus
the presence of the propeptide does not prevent generation of
complex oligosaccharides totally. Likewise, the present results
show that secreted pro-MPO contains complex mannose groups
which must have been added at a rapidly transient step as they
are not detectable in cellular pro-MPO. That MPODpro, in
contrast, contains complex mannose groups exclusively sug-
gests that the propiece can, when present, prevent the gener-
ation of certain complex mannose side chains.
MPODpro lacks targeting to granules and is instead con-
veyed to the secretory path with concomitant synthesis of com-
plex mannose groups during passage of trans-Golgi. The find-
ing of some complex mannose groups in secreted pro-MPO also
indicates that at least part of it has travelled the secretory
pathway through trans-Golgi. The secreted pro-MPO consists
of at least two protein forms with different molecular masses
easily seen after removal of carbohydrate with N-glycanase and
only the smaller one contains complex mannose groups. Simi-
lar extracellular pro-MPO forms were observed in superna-
tants from HL-60 cells (data not shown) indicating that their
occurrence may be a general phenomenon. It is possible that
the two forms have arrived at the cell surface through separate
routes. The higher molecular mass component might have
come through a secretory path excluding trans-Golgi and lack-
ing complex mannose groups, whereas the lower molecular
mass form might have come through another path. We specu-
late that the latter path is that for processing and storage of
MPO but that it is linked to the secretory pathway at a distal
point. Thus, the secreted lower molecular mass species could
represent an intermediate MPO processing form that is in part
released to the secretory pathway from an acidic pregranule
compartment in which intermediate processing forms have
been suggested to be produced (19, 22). Final processing occurs
later (in granules) when escape to the secretory pathway is
blocked. Intermediate MPO processing might take place in late
acidic endosomes after receiving contents, including pro-MPO,
from Golgi-derived vesicles. Because late endosomes are in-
volved in transport in and out of the cell, it is possible that some
material delivered to late endosomes escapes to the outside. An
additional unproven possibility is that secreted pro-MPO, but
not MPODpro, can re-enter the cell through receptor-mediated
uptake into the endocytic pathway with transport to late en-
dosomes and granules for processing and storage.
Acquisition of heme by heme-free apopro-MPO seems to be a
rate-limiting step in subsequent processing into mature MPO
of hematopoietic cells (11–13). The calcium-binding calreticu-
lin, present in the ER of many cells, was shown to interact
specifically with fully glycosylated apopro-MPO during a rela-
tively short period early in MPO synthesis and not with heme-
containing pro-MPO or mature MPO (14). These data suggest a
role of calreticulin as a molecular chaperone facilitating heme
insertion after which the calreticulin-MPO precursor complex
dissociates and pro-MPO can leave the ER for further process-
ing and targeting. Our results show that pro-MPO and
MPODpro both have incorporated heme although the relative
efficiency of incorporation is lower for MPODpro. In any case,
the lack of propeptide may not block the interaction between
apopro-MPO and calreticulin that is proposed to be necessary
for heme incorporation (14). The propeptide seems not to play a
major role for the initial processing of the translational product
but rather plays a role later in processing and trafficking.
Finally, our results provide novel information on MPO bio-
synthesis, processing, and targeting. Elimination of the
propeptide from pro-MPO blocks the maturation process, al-
lows secretion, but abolishes accumulation of the final product
for storage, suggesting a critical role of the propeptide for late
processing of pro-MPO.
Acknowledgments—We greatly appreciate the skilled technical as-
sistance of Eva Nilsson and Ann-Maj Persson.
REFERENCES
1. Gullberg, U., Andersson, E., Garwicz, D., Lindmark, A., and Olsson, I. (1997)
Eur. J. Haematol. 58, 137–153
2. Hasilik, A. (1992) Experientia (Basel) 48, 130–151
3. Liu, L., and Ganz, T. (1995) Blood 85, 1095–1103
4. Gullberg, U., Lindmark, A., Lindgren, G., Persson, A.-M., Nilsson, E., and
Olsson, I. (1995) J. Biol. Chem. 270, 12912–12918
5. Garwicz, D., Lindmark, A., and Gullberg, U. (1995) J. Biol. Chem. 270,
28413–28418
6. Klebanoff, S. J. (1992) in Inflammation: Basic Principles and Clinical
Correlates (Gallin, J. I., Goldstein, I. M., and Snyderman, R., eds) 2nd Ed.,
pp. 541–588, Raven Press, Ltd., New York
7. Nauseef, W., Olsson, I., and Arnljots, K. (1988) Eur. J. Haematol. 40, 97–110
8. Andrews, P. C., and Krinsky, N. I. (1981) J. Biol. Chem. 256, 4211–4218
9. Zeng, J., and Fenna, R. E. (1992) J. Mol. Biol. 226, 185–207
10. Olsson, I., Persson, A.-M., and Stro¨mberg, K. (1984) Biochem. J. 223, 911–920
11. Nauseef, W. M., McCormick, S., and Yi, H. (1992) Blood 80, 2622–2633
12. Castaneda, V. L., Parmley, R. T., Pinnix, I. B., Raju, S. G., Guzman, G. S., and
Kinkade, J. M. (1992) Exp. Hematol. 20, 916–924
13. Pinnix, I. B., Guzman, G. S., Bonkovsky, H. L., Zaki, S. R., and Kinkade, J. M.
(1994) Arch. Biochem. Biophys. 312, 447–458
14. Nauseef, W. M., McCormick, S. J., and Clark, R. A. (1995) J. Biol. Chem. 270,
4741–4747
15. Yamada, M. (1982) J. Biol. Chem. 257, 5980–5982
16. Stro¨mberg, K., Persson, A.-M., and Olsson, I. (1985) Eur. J. Cell Biol. 39,
424431
17. Hasilik, A., Pohlmann, R., Olsen, R. L., and von Figura, K. (1984) EMBO J. 3,
2671–2676
18. Koeffler, H. P., Ranyard, J., and Pertcheck, M. (1984) Blood 65, 484 491
19. Akin, D. T., and Kinkade, J. M., Jr. (1986) J. Biol. Chem. 261, 83708375
20. Nauseef, W. M. (1987) Blood 70, 1143–1150
21. Hashinaka, K., Nishio, C., Hur, S.-J., Sakiyama, F., Tsunasawa, S., and
Yamada, M. (1988) Biochemistry 27, 5906–5914
22. Akin, D. T., and Kinkade, J. M. (1987) Arch. Biochem. Biophys. 255, 428436
23. Valtieri, M., Tweardy, D. J., Caracciolo, D., Johnson, K., Mavilio, F., Altmann,
S., Santoli, D., and Rovera, G. (1987) J. Immunol. 138, 3829–3835
24. Liu, L., Oren, A., and Ganz, T. (1995) J. Immunol. Methods 181, 253–258
25. Kreider, B. L., Phillips, P. D., Prytowsky, M. B., Shirsat, N., Pierce, J. H.,
Tushinski, R., and Rovera, G. (1990) Mol. Cell. Biol. 10, 48464853
26. Gullberg, U., Lindmark, A., Nilsson, E., Persson, A.-M., and Olsson, I. (1994)
J. Biol. Chem. 269, 25219–25225
27. Hultberg, B., Lindsten, J., and Sjo¨blad, S. (1976) Biochem. J. 155, 599605
28. Bretz, R., and Sta¨ubli, W. (1977) Eur. J. Biochem. 77, 181–192
29. Olsson, I., Olofsson, T., and Odeberg, H. (1972) Scand. J. Haematol. 9,
483–491
30. Lindmark, A., Persson, A.-M., and Olsson, I. (1990) Blood 76, 2374 –2380
31. Deleted in proof
32. Garwicz, D., Lindmark, A., Hellmark, T., Gladh, M., Jo¨gi, J., and Gullberg, U.
(1997) J. Leukocyte Biol. 61, 113–123
33. Nauseef, W. M. (1986) Blood 67, 865–872
34. Taylor, K. L., Pohl, J., and Kinkade, J. M., Jr. (1992) J. Biol. Chem. 267,
25282–25288
35. Lippincott-Schwartz, J., Yuan, L. C., Bonifacino, J. S., and Klausner, R. D.
(1989) Cell 56, 801–813
36. Lippincott-Schwartz, J., Donaldson, J. G., Schweizer, A., Berger, E. G., Hauri,
H.-P., Yuan, L. C., and Klausner, R. D. (1990) Cell 60, 821–836
37. Borregaard, N., Lollike, K., Kjeldsen, L., Sengelo¨v, H., Bastholm, L., Nielsen,
M. H., and Bainton, D. F. (1993) Eur. J. Haematol. 51, 187–198
38. Salvesen, G., Enghild, J. J. (1990) Biochemistry 29, 5304–5308
39. Moguilevsky, N., Garcia-Quintana, L., Jacquet, A., Tournay, C., Fabry, L.,
Pierard, L., and Bollen, A. (1991) Eur. J. Biochem. 197, 605–614
40. Jacquet, A., Deleersnyder, V., Garcia-Quintana, L., Bollen, A., and
Moguilevsky, N. (1992) FEBS Lett. 302, 189–191
41. Jacquet, A., Deby, C., Mathy, M., Moguilevsky, N., Deby-Dupont, G., Thirion,
A., Goormaghtigh, E., Garcia-Quintana, L., Bollen, A., and Pincemail, J.
(1991) Arch. Biochem. Biophys. 291, 132–138
42. Cully, J., Harrach, B., Hauser, H., Harth, N., Robenek, H., Nagata, S., and
Hasilik, A. (1989) Exp. Cell Res. 180, 440450
43. Taylor, K. L., Uhlinger, D. J., and Kinkade, J. M. (1992) Biochem. Biophys.
Res. Commun. 187, 1572–1578
44. Nauseef, W. M., Cogley, M., and McCormick, S. (1996) J. Biol. Chem. 271,
9546–9549
45. Conner, G. E. (1992) J. Biol. Chem. 267, 21738–21745
Myeloperoxidase Processing 4753
by guest on July 10, 2015http://www.jbc.org/Downloaded from
Gullberg and Inge Olsson
Elinor Andersson, Lars Hellman, Urban
and Sorting of Human Myeloperoxidase
The Role of the Propeptide for Processing
CELL BIOLOGY AND METABOLISM:
doi: 10.1074/jbc.273.8.4747
1998, 273:4747-4753.J. Biol. Chem.
http://www.jbc.org/content/273/8/4747Access 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/273/8/4747.full.html#ref-list-1
This article cites 43 references, 21 of which can be accessed free at
by guest on July 10, 2015http://www.jbc.org/Downloaded from
... Inactive apo-pro-MPO is a peroxidase enzyme requiring the acquisition of heme group to acquire enzymatic function, heme acquisition take place before leaving the endoplasmic reticulum (ER). Pro-MPO is then trimmed from its pro-segment, a 116-amino acid peptide, which is critical for proper routing to lysosomal compartments and excision of the ASFVTG peptide between the heavy and the light chains by a cysteine protease (Andersson et al. 1998;Nauseef 2018). In lysosomes, which are matured in azurophilic granules in neutrophils, mature MPO can then homodimerize to yield catalytically active complex further stored in specialized granules upon triggered release. ...
... In the absence of prosegment, apo-pro-MPO acquire heme, but do not get stored in lysosomes neither as acquire its catalytic activity. In transfected 32D cells, which are from myeloid lineage, exogenously expressed prosegment-deleted pro-MPO suggested a role of the this propeptide in the late processing of pro-MPO and the targeting for storage in granules (Andersson et al. 1998). Based on constructs carrying deletion in HEK293 cells, it has been reported that pro-MPO's pro-segment cleavage occurs at the 128 RKLRSLWRR 136 multibasic site, likely by a serine protease from the proprotein convertase (PC) family as per the sensitivity to decanoyl-RVKR-chloromethylketone (CMK) pan-PCs inhibitor (McCormick et al. 2012). ...
... Given the high lability of neutrophils, which require fresh isolation from donor blood, the HL-60 cell line has proven itself as a convenient source to obtain granulocyte-like cells for research purposes. This explains why very few studies have been able to investigate MPO directly in neutrophils and the recurrent use of overexpression strategies instead (Olsson et al. 1984;Andersson et al. 1998;Laura et al. 2016). The HL-60 promyelocytic leukemia cell line is an extensively used model of human myeloid cells when induced toward granulocytic differentiation. ...
Article
Full-text available
Neutrophil myeloperoxidase/H2O2/chloride system is a key mechanism to control pathogen infection. This enzyme, myeloperoxidase, plays a pivotal role in the arsenal of azurophilic granules that are released through degranulation upon neutrophil activation, which trigger local hypochlorous acid production. Myeloperoxidase gene encodes a protein precursor named promyeloperoxidase that arbors a propeptide that gets cleaved later during secretory routing in post-endoplasmic reticulum compartments. Although evidence suggested that this processing event was performed by one or different enzymes from the proprotein convertases family, the identity of this enzyme was never investigated. In this work, the naturally producing myeloperoxidase promyelocytic cell line HL-60 was used to investigate promyeloperoxidase cleavage during granulocytic differentiation in response to proprotein convertase inhibitors decanoyl-RVKR-chloromethylketone and hexa-d-arginine. Stable PC knockdown of endogenously expressed proprotein convertases, furin and PC7, was achieved using lentiviral delivery of shRNAs. None of the knockdown cell line could reproduce the effect of the pan-proprotein convertases inhibitor decanoyl-RVKR-chloromethylketone that accumulated intracellular promyeloperoxidase stores in HL-60 cells, therefore illustrating that both furin and PC7 redundantly process this proprotein.
... In the current study, we demonstrated that MPO proteins are also transported to the cell surface by associating with HLA-DR. Although mature MPO consists of a heavy subunit (60 kd) and a light subunit (12 kd), the MPO protein that coimmunoprecipitated with HLA-DR was 90 kd, which is a molecular weight compatible with apopro-MPO or pro-MPO (37). Because both ends of the peptide-binding groove of HLA class II are open (38)(39)(40), it is possible (structurally) that the peptide-like structure exposed on full-length MPO proteins binds to the peptide-binding groove. ...
Article
Objective: Autoantibodies against myeloperoxidase (MPO) that are expressed in neutrophils play an important role in the pathogenesis of microscopic polyangiitis (MPA). We recently found that misfolded cellular proteins are transported to the cell surface by HLA class II molecules and are targeted by autoantibodies in rheumatoid arthritis and antiphospholipid syndrome, suggesting that HLA class II molecules play an important role in autoantibody recognition. Here, we demonstrate that MPO is expressed on the cell surface by associating with HLA class II molecules. Methods: Association of MPO with HLA-DR was analyzed using MPO and HLA-DR transfectants as well as neutrophils from healthy donors and MPA patients. Autoantibody binding to MPO/HLA-DR complex was analyzed by flow cytometry. Association of MPO with HLA-DR was assessed by immunoprecipitation. Function of MPO-ANCA was assessed using a neutrophil-like cell line expressing HLA-DR and MPO. Results: MPO protein was detected on the cell surface in the presence of HLA-DR, and MPO/HLA-DR complex was recognized by MPO-ANCA. Competitive inhibition assay suggested that MPO associated with HLA-DR express cryptic autoantibody epitopes for MPO-ANCA. Autoantibody binding to MPO/HLA-DR complex was correlated with disease susceptibility conferred by each HLA-DR allele, suggesting that MPO/HLA-DR complex is involved in pathogenicity of MPA. Indeed, MPO/HLA class II complexes were detected in neutrophils from a MPA patient as well as cytokine-stimulated neutrophils from healthy donors. Moreover, MPO-ANCA stimulated MPO/HLA-DR complex-expressing HL-60 cells. Conclusion: Our findings suggest that MPO complexed with HLA class II molecules is involved in MPA pathogenesis as a target for MPO-ANCA. This article is protected by copyright. All rights reserved.
Article
We have completed the characterization of the turbot (Scophthalmus maximus) myeloperoxidase (mpx) gene and protein, which we partially described in a previous study. The turbot mpx gene has 15 exons that encode a protein of 767 aa, with a signal peptide, propeptide and light and heavy chains, and also with haem cavities, a Ca⁺²-binding motif and several N- and O-glycosylation sites. The mature protein forms homodimers of about 150 kDa and is very abundant in turbot neutrophils. In addition to the mpx (epx2a) gene, another three peroxidase genes, named epx1, epx2b1 and epx2b2, were identified in the turbot genome. Epx1, Epx2b1 and Epx2b2 proteins also have signal peptides and many structural characteristics of mammalian MPO and eosinophil peroxidase (EPX). Mpx was strongly expressed in head kidney, while epx2b1 and epx2b2 were strongly expressed in the gills, and epx1 was not expressed in any of the tissues or organs analysed. In vitro stimulation of head kidney leucocytes with the parasite Philasterides dicentrarchi caused a decrease in mpx expression and an increase in epx2b1 expression over time. In turbot infected experimentally with P. dicentrarchi a significant increase in mpx expression in the head kidney was observed on day 7 postinfection, while the other genes were not regulated. However, mpx, epx2b1 and epx2b2 were downregulated in the gills of infected fish, and epx1 expression was not affected. These results suggest that the four genes responded differently to the same stimuli. Interestingly, BLAST analysis revealed that Epx1 and Mpx showed greater similarity to mammalian EPX than to MPO. Considering the phylogenetic and synteny data obtained, we concluded that the epx/mpx genes of Gnathostomes can be divided into three main clades: EPX1, which contains turbot epx1, EPX2, which contains turbot mpx (epx2a) and epx2b1 and epx2b2 genes, and a clade containing mammalian EPX and MPO (EPX/MPO). EPX/MPO and EPX2 clades share a common ancestor with the chondrichthyan elephant shark (Callorhinchus milii) and the coelacanth (Latimeria chalumnae) peroxidases. EPX2 was only found in fish and includes two sister groups. One of the groups includes turbot mpx and was only found in teleosts. Finally, the other group contains epx2b1 and epx2b2 genes, and epx2b1-2b2 loci share orthologous genes with other teleosts and also with holosteans, suggesting that these genes appeared earlier on than the mpx gene.
Article
Full-text available
Anti-neutrophil cytoplasmic autoantibodies (ANCAs) are directed against lysosomal components of neutrophils. ANCAs directed to proteinase 3 and myeloperoxidase (MPO) in particular are associated with distinct forms of small vessel vasculitides. MPO is an abundant neutrophil-derived heme protein that is part of the antimicrobial defense system. The protein is typically present in the azurophilic granules of neutrophils, but a large portion may also enter the extracellular space. It remains unclear why MPO is frequently the target of antibody-mediated autoimmune responses. MPO is a homodimeric glycoprotein, post-translationally modified with complex sugars at specific sites. Glycosylation can strongly influence protein function, affecting its folding, receptor interaction, and backbone accessibility. MPO potentially can be heavily modified as it harbors five putative N-glycosylation sites (ten in the mature dimer). Although considered important for MPO structure and function, the full scope and relative abundance of the glycans attached to MPO is unknown. Here, combining bottom-up glycoproteomics and native MS approaches, we structurally characterized MPO from neutrophils of healthy human donors. We quantified the relative occupancy levels of the glycans at each of the five sites and observed complex heterogeneity and site-specific glycosylation. In particular, we detected glycosylation phenotypes uncommon for glycoproteins in the extracellular space, such as a high abundance of phosphorylated high-mannose species and severely truncated small glycans having the size of paucimannose or smaller. We hypothesize that the atypical glycosylation pattern found on MPO might contribute to its specific processing and presentation as a self-antigen by antigen-presenting cells.
Article
Members of Chordata peroxidase subfamily [1] expressed in mammals, including myeloperoxidase (MPO), eosinophil peroxidase (EPO), lactoperoxidase (LPO), and thyroid peroxidase (TPO), express conserved motifs around the heme prosthetic group essential for their activity, a calcium-binding site, and at least two covalent bonds linking the heme group to the protein backbone. Although most studies of the biosynthesis of these peroxidases have focused on MPO, many of the features described occur during biosynthesis of other members of the protein subfamily. Whereas MPO biosynthesis includes events typical for proteins generated in the secretory pathway, the importance and consequences of heme insertion are events uniquely associated with peroxidases. This Review summarizes decades of work elucidating specific steps in the biosynthetic pathway of human MPO. Discussion includes cotranslational glycosylation and subsequent modifications of the N-linked carbohydrate sidechains, contributions by molecular chaperones in the endoplasmic reticulum, cleavage of the propeptide from proMPO, and proteolytic processing of protomers and dimerization to yield mature MPO. Parallels between the biosynthesis of MPO and TPO as well as the impact of inherited mutations in the MPO gene on normal biosynthesis will be summarized. Lastly, specific gaps in our knowledge revealed by this review of our current understanding will be highlighted.
Article
Phagocytes respond to stimulation with a burst of oxygen consumption, and much, if not all, of the extra oxygen consumed in the respiratory burst is converted first to the superoxide anion and then to hydrogen peroxide (H2O2). Myeloperoxidase (MPO), which is released from cytoplasmic granules of neutrophils and monocytes by a degranulation process, reacts with the H2O2 formed by the respiratory burst to form a complex that can oxidize a large variety of substances. Among the latter is chloride, which is oxidized initially to hypochlorous acid, with the subsequent formation of chlorine and chloramines. These products of the MPO-H2O2-chloride system are powerful oxidants that can have profound biological effects. The primary function of neutrophils is the phagocytosis and destruction of microorganisms, and the release of MPO and H2O2 into the phagosome containing the ingested microorganism generally leads to a rapid microbicidal effect. Neutrophils from patients with chronic granulomatous disease (CGD) have a microbicidal defect that is associated with the absence of a respiratory burst and, thus, H2O2 production. Neutrophils from patients with a hereditary MPO deficiency, who lack MPO, also have a microbicidal defect, although it is not as severe as that seen in CGD. MPO and H2O2 also can be released to the outside of the cell where a reaction with chloride can induce damage to adjacent tissue and, thus, contribute to the pathogenesis of disease. It has been suggested that pulmonary injury, renal glomerular damage, and the initiation of atherosclerotic lesions may be caused by the MPO system.
Article
Full-text available
Myeloperoxidase (MPO) is synthesized by neutrophil and monocyte precursor cells and contributes to host defense by mediating microbial killing. Although several steps in MPO biosynthesis and processing have been elucidated, many questions remain, such as the structure-function relationships of monomeric unprocessed proMPO versus the mature dimeric MPO and the functional role of the propeptide. Here we present the first and high resolution (at 1.25 A) crystal structure of proMPO and its solution structure obtained by small angle X-ray scattering. Promyeloperoxidase hosts five occupied glycosylation sites and six intrachain cystine bridges with C158 of the very flexible N-terminal propeptide being covalently linked to C319 thereby hindering homodimerization. Furthermore, the structure revealed (i) the binding site of proMPO processing proconvertase, (ii) the structural motif for subsequent cleavage to the heavy and light chains of mature MPO protomers and (iii) three covalent bonds between heme and the protein. Studies on the mutants C158A, C319A and C158A/C319A demonstrate significant differences from the wild-type protein, including diminished enzymatic activity and prevention from export to the Golgi due to prolonged association with the chaperone calnexin. These structural and functional data provide novel insights into MPO biosynthesis and processing.
Article
Most growth factors and hormones are synthesized as pre‐pro‐proteins which are processed to the biologically active mature protein. The pre‐ and prodomains are cleaved from the precursor protein in the secretory pathway or, in some cases, extracellularly. The canonical functions of these prodomains are to assist in folding and stabilization of the mature domain, to direct intra and extracellular localization, to facilitate storage, and to regulate bioavailability of their mature counterpart. Recently, exciting evidence has revealed that prodomains of certain growth factors, after cleaved from the precursor pro‐protein, can act as independent active signaling molecules. In this review, we discuss the various classical functions of prodomains, and the biological consequences of these pro‐peptides acting as ligands. We will focus our attention on the brain‐derived neurotrophic factor prodomain ( pBDNF ), which has been recently described as a novel secreted ligand influencing neuronal morphology and physiology. image
Article
Full-text available
Caveolin 1 (Cav1) is a required structural component of caveolae and its phosphorylation by Src is associated with an increase in caveolae-mediated endocytosis. Here, we demonstrate using quantitative live-cell 4D, TIRF, and FRET imaging that endocytosis and trafficking of caveolae is associated with a Cav1 Tyr14 phosphorylation-dependent conformational change which spatially separates, or loosens, Cav1 molecules within the oligomeric caveolar coat. When tracked by TIRF and spinning-disk microscopy, cells expressing phospho-mimicking Cav1 (Y14D) mutant formed vesicles which were greater in number and volume when compared with Y14F-Cav1-GFP. Furthermore, we observed in HEK cells cotransfected with wild-type, Y14D, or Y14F Cav1-CFP and -YFP constructs that FRET efficiency was greater with Y14F pairs than with Y14D, indicating pY14-Cav1 regulates the spatial organization of Cav1 molecules within the oligomer. In addition, albumin-induced Src activation or direct activation of Src using a rapamycin-inducible Src construct (RapR-Src) led to an increase in monomeric Cav1 in Western blots, as well as a simultaneous increase in vesicle number and decrease in FRET intensity, indicative of a Src-mediated conformational change in CFP/YFP-tagged WT-Cav1 pairs. We conclude that phosphorylation of Cav1 leads to separation or "spreading" of neighboring negatively-charged N-terminal phospho-tyrosine residues, promoting swelling of caveolae followed by their release from the plasma membrane.
Article
Full-text available
Ion-exchange chromatography of gel filtration demonstrated the presence of different molecular forms of nine lysosomal enzymes in cultured amniotic-fluid cells. The patterns of molecular forms were similar to those known from skin fibroblasts and liver tissue. During cultivation total enzyme activities fluctuated with the number of passages, without any consistent trend of increase or decrease, and without correlation to the dominating cell type in the culture.
Article
The processing of the neutral proteases cathepsin G and neutrophil elastase, normally synthesized in myeloid precursor cells and stored in azurophil granules, were investigated by biosynthetic labeling with 14C- leucine of the monoblastic cell line U-937. The proteases were precipitated with specific antibodies and the immunoprecipitates were analyzed by sodium dodecyl sulfate polyacrylamide gel electrophoresis (SDS-PAGE) followed by fluorography. The transfer to lysosomes of newly synthesized proteases was demonstrated in pulse-chase labeling experiments followed by centrifugation of cell homogenates in a Percoll gradient. The presence of a closely spaced polypeptide band-doublet at intermediate gradient density suggested cleavage of the specific aminoterminal pro dipeptide extension before storage in lysosomes. The molecular heterogeneity observed for cathepsin G and neutrophil elastase seemed to be due to modifications occurring after sorting into lysosomes, most likely because of C-terminal processing. Modifications of the secreted enzymes were not detectable by SDS-PAGE. In contrast to other lysosomal enzymes, no phosphorylation was demonstrated. Newly synthesized cathepsin G and neutrophil elastase rapidly became resistant to endoglycosidase H, indicating transport through the medial and trans cisternae of the Golgi complex and conversion to “complex” oligosaccharide side chains. This conversion was inhibited by an agent swainsonine, but translocation from the Golgi complex and secretion were unaffected. The processing described may play a role in activation of the proteases.
Article
Biosynthesis of myeloperoxidase (MPO), a myeloid lysosomal hemoprotein critical for the optimal oxygen-dependent microbicidal activity of human neutrophils, is incompletely understood. The primary translation product undergoes cotranslational N-linked glycosylation with subsequent insertion of the Fe-containing prosthetic group into the peptide backbone, thereby converting the enzymatically inactive, heme- free apoproMPO into the peroxidatively active precursor, proMPO. Eventually, proMPO undergoes proteolytic processing into native, lysosomal MPO, with subunits of 59 and 13.5 Kd. We studied three unanswered questions regarding MPO biosynthesis: (1) At what point during MPO biosynthesis is the heme moiety inserted into the apoenzyme? (2) What consequences does heme-insertion have on subsequent processing events? (3) What role does the mannose-6-phosphate receptor (M6PR) system play in the delivery of MPO to the lysosome? Disruption of Golgi by brefeldin A (BFA) produced two major changes in MPO biosynthesis: (1) processing of the 89-Kd precursor to mature MPO was blocked and (2) constitutive secretion of the MPO precursor was inhibited. Inhibition of heme synthesis with succinyl acetone (SA) reduced peroxidase activity and profoundly blocked processing of proMPO to mature MPO. This inhibition of processing was not a generalized effect on all lysosomal enzymes, because the maturation of a non-heme-containing lysosomal enzyme, beta-glucuronidase, was not altered. Electron microscopy showed that, although the normal peroxidase staining of endoplasmic reticulum was absent in SA-treated cells, there were MPO- related peptides in the ER. The role of the M6PR system was assessed by immunoprecipitating fractions obtained from M6PR affinity column chromatography. The 89-Kd proMPO failed to adhere to the M6PR affinity column, whereas the 59-Kd heavy subunit of mature MPO was specifically eluted from the column. We interpret these data to indicate that: (1) processing of proMPO to mature MPO occurs in a post-ER compartment that is itself BFA-sensitive or is distal to a BFA-sensitive compartment and (2) heme insertion into apoproMPO precedes and may be a prerequisite for proteolytic processing to enzymatically active mature MPO. Our analysis of the M6PR system in MPO biosynthesis led to the unanticipated finding that there were phosphomannosyl residues on mature MPO, but none on proMPO. We suggest that the bulk of proMPO at any time is not phosphorylated, but, when generated, the phosphorylated proMPO is quickly processed to the phosphorylated 59-Kd subunit of mature MPO. Thus, if the M6PR is important in the intracellular transport of MPO, it is the phosphorylated mature MPO that is directed to the lysosomal compartment by this system.(ABSTRACT TRUNCATED AT 400 WORDS)
Article
Myeloperoxidase (MPO) is a lysosomal enzyme present in the azurophilic granules of human neutrophils and monocytes and is important for optimal oxygen-dependent killing of microorganisms. The native molecule is a heterodimer composed of a pair of heavy-light protomers, each containing a 59-kDa and 13.5-kDa subunit. The intracellular processing during biosynthesis of MPO was examined in the human promyelocytic cell line HL-60. Endoglycosidase H and F digestion of immunoprecipitated pro- MPO demonstrated the presence of five N-linked--high-mannose oligosaccharide side chains and no complex mannose units. Incorporation of the threonine analogue beta-hydroxynorvaline produced species approximately 2.5 kDa and approximately 5 kDa smaller than the fully glycosylated pro-MPO, suggesting that two of the glycans were in the asparagine-X-threonine tripeptide sequence. Processing of pro-MPO occurred very rapidly, within approximately five minutes, and was best identified using glucosidase inhibitors. The presence of such inhibitors resulted in synthesis of a 92-kDa glycoprotein rather than the usually identified 89-kDa peptide. Swainsonine, a Golgi mannosidase inhibitor, did not alter the size of the earliest synthesized protein, suggesting that pro-MPO exited the endoplasmic reticulum or cis-Golgi proximal to the site of mannosidases. Intracellular transport and proteolytic maturation of MPO was retarded by weak bases (NH4Cl, chloroquine) or monensin at concentrations shown to raise intralysosomal pH. However, these agents did not qualitatively alter transport nor increase secretion. Thus, although MPO biosynthesis resembled that of other lysosomal enzymes, significant differences exist, including only limited oligosaccharide processing and intracellular transport and proteolytic maturation of pro-MPO that was only retarded by alkalinizing lysosomes without affecting the products or the fraction of pro-MPO secreted. Characterization of the determinants for targeting and of the regulatory factors in processing lysosomal enzymes in myeloid cells will provide insight into the molecular mechanisms underlying common disorders such as myeloperoxidase deficiency.
Article
Myeloperoxidase (MPO) is a major protein present in myeloid cells and is used by these cells to help kill microbes. The human promyelocytic HL-60 line can be induced to differentiate to granulocytes or macrophagelike cells. Poly (A) containing RNA was isolated from HL-60 granulocytes, HL-60 macrophages, HL-60 blasts, and normal human granulocytes. The mRNA was translated in a reticulocyte lysate system in the presence of 35S-methionine. The MPO was precipitated from the lysate with rabbit IgG antiserum to human MPO. The resulting precipitate from HL-60 blasts gave a major band of radioactivity of approximately 77,000 daltons and another band at approximately 46,000 daltons on sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE). The MPO identity of the labeled bands was confirmed by cold competition. The relative mRNA activity expressed as a percentage of radioactivity incorporated into MPO (77,000-dalton band) as compared with total trichloracetic acid (TCA) precipitable radioactivity was 0.2%. Negligible mRNA activity for MPO was present in HL-60 granulocytes, HL-60 macrophages, and normal human granulocytes. Pulse- chase experiments showed that MPO was an approximate 75,000-dalton major band and 77,000-dalton minor band of radioactivity after HL-60 blasts were labeled for 1/2 hour with 35S-methionine and the cell lysate immunoprecipitated and subjected to SDS-PAGE. The chase experiments (one to 24 hours) showed that the 77,000- and 75,000-dalton bands of radioactivity were replaced with two major bands (55,000 and 15,000 daltons) and one minor band (approximately 39,000 daltons) of radioactivity. Six-hour 35S-methionine labeling experiments showed that the relative rate of MPO synthesis compared with total TCA precipitable radioactivity was 0.5% in HL-60 blasts and almost negligible in HL-60 macrophages and granulocytes, normal human granulocytes, and B- lymphocytes. The KG-1 myeloblasts and KG-1a early myeloblasts synthesized a small amount of the 75,000-dalton MPO protein. Although HL-60 cells no longer synthesized MPO after differentiation, HL-60 granulocytes and HL-60 macrophages continued to contain MPO as measured by enzyme activity.(ABSTRACT TRUNCATED AT 400 WORDS)
Article
The biosynthesis and processing of myeloperoxidase (MPO), a cationic enzyme present in the azurophilic granules of human polymorphonuclear leukocytes (PMNs), were studied in the human promyelocytic leukemia cell line, HL-60. HL-60 cells produce large quantities of enzymatically active MPO that has the same electrophoretic behavior as MPO isolated from normal PMNs. Mature MPO is a glycoprotein of approximately 150,000 molecular weight (mol wt) composed of two heavy-light protomers (alpha 2 beta 2) with subunits of 59,000 and 13,500 mol wt, respectively, under reducing conditions. The primary translation product of MPO messenger RNA (mRNA) isolated from HL-60 cells was a single polypeptide of mol wt 80,000. In HL-60 cells labeled with [35S]-methionine, the labeled MPO isolated by immunoprecipitation had a mol wt of 89,000. Treatment of this 89-kilodalton (kDa) species with endoglycosidase H produced a 79-kDa peptide, suggesting that the 89-kDa protein contained high-mannose side chains. The 89-kDa species had no detectable peroxidase activity. During chase experiments some of the 89-kDa peptide was processed to smaller species of mol wt 39,000, 59,000, and 13,500, although a fraction of the 89-kDa peptide remained unprocessed after a chase of 100 hours. In addition, a small amount of the 89-kDa peptide appeared in the medium without any of the processed smaller peptides. These studies suggest that the primary translation product in MPO biosynthesis is an 80-kDa peptide that undergoes cotranslational cleavage of the signal peptide and glycosylation to produce an 89-kDa pro-MPO, that pro-MPO is a single polypeptide containing the alpha and beta subunits of MPO and contains endoglycosidase H-susceptible high- mannose side chains, and that posttranslational modification of pro-MPO results in targeting to the lysosome and proteolytic maturation of pro- MPO to active enzyme. In light of the previous observation that MPO- deficient and normal PMNs contain an 89-kDa protein immunochemically related to MPO, these studies on MPO biosynthesis indirectly support the hypothesis that defective posttranslation processing by pro-MPO may underlie hereditary MPO deficiency.
Article
Three clones of full-length cDNA encoding human myeloperoxidase were isolated from a human leukemia HL-60 cell cDNA library in lambda gt10 and characterized. Analysis of the nucleotide sequence of one of the cDNA clones, lambda MP-H17, indicated that the cDNA contained 3207 bp with an open reading frame of 2238 bp, a 5' noncoding region of 159 bp, a 3' noncoding region of 800 bp, and a poly(A) tail of 10 bp. cDNA of the two other clones, lambda MP-H7 and lambda MP-H14, each contained insertions with shorter sequences of 96 and 82 bp, respectively, on the open reading frame of lambda MP-H17 cDNA. A myeloperoxidase genomic clone was isolated, and the structure of its 5' region was determined and compared with the structures of these cDNAs. The comparison revealed that the three cDNAs were derived from myeloperoxidase mRNAs produced by alternative splicing from a transcript of the single gene. Nucleotide sequence analysis of the 3' region of the cDNAs of several clones indicated that the mRNAs were polyadenylated at five different sites. Amino acid sequence determination of the amino-terminal and carboxy-terminal portions of the myeloperoxidase light and heavy chains revealed that, during processing of a precursor polypeptide into the mature protein, the amino-terminal polypeptide, the small peptide between the light and heavy chains, and the carboxy-terminal amino acid were excised.
Article
Myeloperoxidase (MPO), a heme protein, is a major component of azurophilic granules of neutrophils. Optimal oxygen-dependent microbicidal activity depends on MPO as the critical enzyme for the generation of hypochlorous acid and other toxic oxygen products. MPO is synthesized during the promyelocytic stage of myeloid differentiation, the stage at which azurophilic granules are formed. Like other lysosomal enzymes, MPO is synthesized as a larger precursor which is subsequently processed and transported intracellularly to the lysosomes. The primary translation product is a single 80-kDa protein which undergoes cotranslational N-linked glycosylation to produce a 92-kDa glycoprotein. Glucosidases in the endoplasmic reticulum or early cis Golgi convert the proMPO to a 90-kDa form which is sorted into a prelysosomal compartment that undergoes final proteolytic maturation to native MPO, a pair of heavy-light protomers with subunits of 60 kDa and a 12 kDa. These events contrast with similar processes seen with other lysosomal enzymes in two ways. First, alkalinization of lysosomes with NH4+ does not alter processing or transport, in contrast to the pH dependence of these processes for other lysosomal enzymes. However, some studies indicate retardation of processing in the presence of the proton ionophore monensin. Second, intracellular transport of MPO is not apparently mediated by the mannose-6-phosphate receptor system. The gene for MPO is on the long arm of chromosome 17 (17q22, 23) near the breakpoint of the 15, 17 translocation of acute promyelocytic leukemia. The gene spans approximately 14 kb and contains 11 introns and 12 exons. The cloned full-length cDNA is approximately 2.2 kb and both normal bone marrow and cultured promyelocytic leukemia cells express two species of mRNA. Inherited MPO deficiency, a relatively common disorder, is associated with the absence of mature MPO but the presence of proMPO, consistent with a post-translational defect. Studies at the molecular level aimed at identifying the underlying genetic defect are thus far consistent with that hypothesis. In addition, the basis for the observed association between acquired MPO deficiency and some myeloid leukemias can now be studied at the molecular level using these probes.
Article
Granulocytes expose phagocytized microbes to microbicidal substances that are stored in cytoplasmic granules and delivered by fusion to the phagocytic vacuoles (phagosomes). To determine if the murine myeloid cell line 32D cl3 is suitable as a genetic model of protein translocation to granules and phagosomes, we permanently transduced 32D cl3 cells with human HNP-1 defensin cDNA, incubated them with opsonized zymosan, and immunostained them for human defensin HNP-1. Although their phagocytic rate was much slower than that of neutrophil granulocytes, 32D cl3 cells ingested zymosan into vacuoles that accumulated most of the transgenic defensin. The 32D cl3 cell line should be useful for studies of the targeting of proteins to granules and phagosomes.
Article
1. Galactosyltransferase activities in postnuclear supernatants and Golgi fractions from rat liver were assayed with two improved and simplified methods, using high‐ and low‐molecular‐weight acceptors. Transfer to N ‐acetylglucosamine was measured after the separation of the reaction product N ‐acetyllactosamine from all other radioactive molecules (including galactose) on an ion‐exchange column partially converted to the borate form. To determine the transfer of galactose to a glycoprotein acceptor we used ovomucoid, which accepts galactose without any previous chemical or enzymic modification. 2. Both enzymic activities were enriched 60–80‐fold (compared with the post‐nuclear supernatant) in Golgi fractions, which were isolated on two subsequent sucrose gradients and identified morphologically by their high contents of stacked Golgi elements. The two activities could not be resolved by isolation of the Golgi fractions or by detergent solubilization. Each acceptor inhibited the galactose transfer to the other one (up to 95%), presumably because both compete for the same enzyme. 3. The transferase activities were enhanced by the nonionic detergent Triton X‐100. The degree of activation depended directly on the amount of Triton bound to the membrane, i.e. the Triton/phospholipid ratio and not the w/v concentration of the detergent in the assay medium. This relationship persisted, regardless of the purity of the Golgi preparation: Half‐maximal activation occurred at the same Triton/phospholipid ratio in postnuclear supernatants as well as in isolated Golgi fractions. The activation could not be explained by complete solubilization, because 50% of the fully activated enzyme could still be sedimented (1 h, 100000 × g ). 4. Galactose transfer to the high‐molecular‐weight acceptor required a higher Triton/phospholipid ratio for half‐maximal activation than did the transfer to the monosaccharide N ‐acetylglucosamine (1 mg/mg compared with 0.5 mg/mg). The degree of activation maximally achieved was much higher with the protein acceptor (400%) than with the sugar (150%). Because both activities are probably due to the same enzyme, it is suggested that these differences in activation reflect properties of the membrane rather than the enzyme, e.g . the presence of a tight diffusion barrier for ovomucoid and the breakdown of this barrier by the detergent.