ArticlePDF Available

The post-translational processing and intracellular sorting of PC2 in the islets of Langerhans

Authors:

Abstract

Proinsulin conversion in the insulin secretory granule is mediated by two sequence-specific endoproteases related to the Kex2 homologues, PC2 and PC3 (Bennett, D. L., Bailyes, E. M., Nielsen, E., Guest, P. C., Rutherford, N. G., Arden, S. D., and Hutton, J. C. (1992) J. Biol. Chem. 267, 15229-15236; Bailyes, E. M., Bennett, D. L., and Hutton, J. C. (1992) Enzyme, in press). Radiolabeling studies using isolated rat islets showed that PC2 was synthesized initially as a 76-kDa glycoprotein which was converted by limited proteolysis to the mature 64-66-kDa form. Conversion was initiated approximately 1 h after synthesis and proceeded via intermediates of 71, 68, and 66 kDa with a t1/2 of 140 min. Release of only the 66- and 64-66-kDa radiolabeled forms of PC2 was induced by glucose and then only at times more than 2 h following synthesis. Proinsulin conversion, by contrast, was more rapid (delay = 30 min, t1/2 = 60 min), and release commenced as soon as 1 h after synthesis with the secreted material being comprised of the precursor, intermediate, and mature forms of insulin. Ultrastructural analysis of islet beta cells showed that PC2 was concentrated in secretory granules. Subcellular fractionation combined with immunoblot analysis showed that insulinoma secretory granules contained only the mature 64-66-kDa form of PC2, whereas fractions enriched in Golgi and endoplasmic reticulum contained a mixture of the 76- and 66-kDa forms of the enzyme. These results indicate that post-translational proteolysis of PC2 is initiated before sorting into the regulated pathway of secretion and that the relative proportions of proinsulin and PC2 packaged into secretory granules will change with physiological conditions.
THE
JOURNAL
OF
BIOLOGICAL CHEMISTRY
(0
1992 by The American Society
for
Biochemistry and Malecular
Biology,
Inc
VOl.
267,
No.
31, Issue
of
November
5,
pp.
22401-22406,1992
Printed
in
U.S.A.
The Post-translational Processing and Intracellular Sorting
of
PC2
in
the Islets
of
Langerhans*
(Received for publication, June
15,
1992)
Paul C. Guest$, Susan D. Arden, Deborah
L.
Bennett, Anne Clark§, Nicholas G. Rutherford, and
John C. Hutton
From the University of Cambridge, Department of Clinical Biochemistry, Addenbrookes Hospital, Hills Road,
Cambridge, CB2
2QR
United Kingdom and the §University of Oxford, Nuffield Department of Clinical Medicine, Diabetes
Research Laboratories, Radcliffe Infirmary, Woodstock Road, Oxford,
OX2
6HE
United Kingdom
Proinsulin conversion in the insulin secretory gran-
ule
is
mediated by two sequence-specific endoproteases
related to the Kex2 homologues, PC2 and PC3 (Ben-
nett, D.
L.,
Bailyes,
E.
M.,
Nielsen,
E.,
Guest,
P.
C.,
Rutherford,
N.
G.,
Arden,
S.
D., and Hutton,
J.
C.
(1992)
J.
Biol.
Chem.
267, 15229-15236; Bailyes,
E.
M.,
Bennett, D.
L.,
and Hutton,
J.
C. (1992)
Enzyme,
in press). Radiolabeling studies using isolated rat islets
showed that PC2 was synthesized initially
as a
76-kDa
glycoprotein which was converted by limited proteol-
ysis
to the mature 64-66-kDa form. Conversion was
initiated approximately
1
h after synthesis and pro-
ceeded via intermediates of
71,68,
and
66
kDa with a
tIl2
of
140
min. Release
of
only the
66-
and 64-66-kDa
radiolabeled forms of PC2 was induced by glucose and
then only
at
times more than 2 h following synthesis.
Proinsulin conversion, by contrast, was more rapid
(delay
=
30
min,
tllz
=
60
min), and release commenced
as
soon as
1
h after synthesis with the secreted material
being comprised of the precursor, intermediate, and
mature forms of insulin. Ultrastructural analysis of
islet
6
cells showed that PC2 was concentrated in
se-
cretory granules. Subcellular fractionation combined
with immunoblot analysis showed that insulinoma
se-
cretory granules contained only the mature 64-66-
kDa form
of
PC2, whereas fractions enriched in Golgi
and endoplasmic reticulum contained
a
mixture of the
76-
and 66-kDa forms of the enzyme. These results
indicate that post-translational proteolysis of PC2
is
initiated before sorting into the regulated pathway of
secretion and that the relative proportions
of
proinsu-
lin and PC2 packaged into secretory granules will
change with physiological conditions.
Enzymological studies have shown that proinsulin conver-
sion requires two distinct endopeptidase activities (designated
type
1
and type
2)
which cleave on the C-terminal side of
Ar2’-Arg2 and Lyss4-ArgG5 (Davidson
et
al.,
1988) and an
exopeptidase (carboxypeptidase H) which removes the ex-
posed basic residues (Davidson and Hutton, 1987). All three
enzymes have a pH optimum of
5.5
which coincides with the
intragranular pH (Hutton, 1982). In addition, the endopepti-
dases have an absolute requirement for Ca2+ which would be
~ ~~ ~ ~ ~~ ~ ~~ ~~
*This work was supported by grants from the British Diabetic
Association, the Medical Research Council of Great Britain and
Wellcome Trust. The costs of publication of this article were defrayed
in part by the payment of page charges. This article must therefore
be hereby marked “aduertisement” in accordance with
18
U.S.C.
Section
1734
solely to indicate this fact.
$
To
whom correspondence should be sent.
met by the high Ca2+ concentration prevailing in mature
secretory granules (Hutton
et
al.,
1983).
Recent immunological and structural studies have identi-
fied the type
2
endopeptidase as PC2 (Bennett
et
al.,
1992), a
member of the mammalian Kex2-related family of proteases
(for review see Barr, 1991). Other members
of
the family
include the Golgi-localized furin (Breshnahan
et
al.,
1990),
PACE 4 (Kiefer
et
al.,
1991), PC3 (also known as PC1; Seidah
et
al.,
1991; Smeekens
et
aL,
1991), and PC4 (Nakayama
et
al.,
1992). Like the Kex2 gene product, all of the enzymes
appear to be involved in the post-translational processing of
proproteins in the secretory pathway and induce limited pro-
teolysis at sites preceded by basic amino acid sequences such
as Lys-Arg, Arg-Arg,
or
Arg-X-X-Arg. PC2 immunoreactivity
is present in pancreatic islets, the adrenal medulla, and the
anterior and neurointermediate lobes of the pituitary (Christie
et
al.,
1991; Kirchmair
et
al.,
1992; Bennett
et
al.,
19921,
suggesting that it has
a
general function in the conversion of
polypeptide hormone and neuropeptide precursors in cells of
the diffuse neuroendocrine system.
PC2 itself is subjected to post-translational proteolytic
processing during the course of its segregation to the site of
proinsulin conversion in secretory granules. This has been
shown by the presence of two molecular variants of the
enzyme in insulin granules which correspond to proteins with
N
termini commencing at amino acids 109 and 112
of
rat PC2
(Bennett
et
al.,
1992).
It
is conceivable that the mature
peptides are produced by autoproteolysis
or
through cleavage
by another Kex2-related enzyme such as furin.
We report here an investigation of the localization and
biosynthesis of PC2 in rat pancreatic endocrine tissue. The
results show that PC2 is proteolytically processed and sorted
into the regulated pathway of secretion and that the cellular
site of post-translational modification and kinetics of proc-
essing and transport differ substantially from the insulin-
related peptides with which the enzyme is co-secreted.
EXPERIMENTAL PROCEDURES
Chemicals and Reagents-[”S]Methionine
(1,400
Ci/mmol) was
purchased from Amersham International (Little Chalfont, Bucking-
hamshire,
U.
K.),
N-Glycanase was from NSB Biologicals (Hatfield,
Hertfordshire,
U.
K.).
Dulbecco’s modified Eagle’s medium was ob-
tained from
Flow
Laboratories (Rickmansworth, Hertfordshire). Per-
oxidase-conjugated rabbit anti-guinea pig and swine anti-rabbit sera
were from Dakopaks (Copenhagen, Denmark). All other analytical
grade biochemicals were obtained from Sigma or BDH Chemicals
(both of Poole, Dorset,
U.
K.)
unless specified otherwise.
Antibodies-A mouse monoclonal antibody (3B7) raised
to
human
proinsulin (Gray et
al.,
1987) was partially purified by
(NH,),SO,
precipitation and coupled to CNBr-activated Sepharose by the
method
of
Axen et al. (1967). The resulting immunoadsorbent had a
binding capacity of approximately
100
pg of insulin/ml of swollen
22401
22402
PC2
Biosynthesis
gel. Antiserum to a glutathione S-transferase fusion protein incor-
porating amino acids 158-391 of the rat PC2 sequence was raised in
rabbits, and antibodies reacting with glutathione S-transferase were
removed by affinity chromatography
as
described previously (Bennett
et
aL,
1992).
Tissues-Islets of Langerhans were isolated from the pancreata of
10-12-week-old New England Deaconess Hospital rats by collagenase
digestion, as described previously (Guest
et
al.,
1989a). Islets were
collected into an incubation medium consisting of modified Krebs
bicarbonate buffer (120 mM NaC1, 5 mM KCI,
1
mM MgS04, 2.5 mM
CaC12, 24 mM NaHC03) containing
20
mM Hepes (pH 7.4), 0.1%
BSA,' and 16.7 mM glucose. Insulinoma tissue was propagated in
New England Deaconess Hospital rats as described previously (Sop-
with
et
al.,
1981), and subcellular fractions were prepared by Percoll
density gradient centrifugation according to the method of Hutton et
al.
(1982).
Radioisotopic Labeling-Batches of 100-200 islets were preincu-
bated in 0.2 ml of incubation medium for 45 min at 37 "C under
O,/
CO,
(19:l) in 1.5 ml-capacity microcentrifuge tubes (Alpha; Eastleigh,
Hampshire,
U.
K.).
Islets were recovered by centrifugation for
10
s
at
800
X
g
(Heraeus Sepatech microcentrifuge; Kalkberg, Germany) and
resuspended in the same prewarmed (37 "C) medium containing
0.2
mCi
of
["'S]methionine. Islets were recovered
as
above after
1
h and
incubated for various times in nonradioactive medium under the
conditions described in the figure legends. Incubations were termi-
nated by addition of
1
ml of ice-cold Krebs incubation medium
followed by centrifugation for
10
s
at
2,000
X
g.
Islets were washed
by two further cycles of resuspension and centrifugation and then
sonicated for 15 s at
3
microns (MSE Sonifier) in 0.4 ml of a lysis
buffer consisting of 25 mM Na2B4O7 (pH 9), 3% (w/v) BSA,
1%
(w/
v)
Tween 20,
1
mM phenylmethylsulfonyl fluoride, 0.1 mM E-64
(trans-epoxysuccinyl-~-leucyl-amido-(4-guanidino)butane),
1
mM
EDTA, and 0.1% NaN:1. The lysates were centrifuged for
3
min at
10,000
x
g
and the supernatants retained. Where release of radiola-
beled proteins into the medium was monitored, media were removed
from islets, clarified by centrifugation for 3 min
at
10,000
X
g,
and
the supernatants combined with 2
X
concentrated lysis buffer for
subsequent immunoprecipitation. Incorporation of [3sS]methionine
into islet lysate and medium proteins was determined by trichloroa-
cetic acid precipitation as described previously (Guest
et
al.,
1989a).
Immunoprecipitation
of
Proinsulin-related Peptides-Islet lysates
and media samples were immunoprecipitated with
100
p1 (packed gel)
of 3B7 immunoadsorbent as described previously (Guest
et
al.,
1989a).
The immunoadsorbent was recovered by centrifugation for
10
s
at
10,000
X
g
and the supernatant used for the immunoisolation of the
PC2-related peptides (see below). The immunoadsorbent was washed
and the bound protein eluted and subjected to alkaline urea-polyacryl-
amide gel electrophoresis (PAGE), and fluorographic analysis
as
described previously (Guest
et
al.,
1989a). Incorporation of radioac-
tivity into specific protein bands was determined by densitometric
scanning of suitably exposed fluorographs using a Chromoscan
111
densitometer (Joyce-Loebl, Gateshead, U.K.).
Immunoprecipitation
of
PC2-related Peptides-The supernatants
obtained after precipitation of proinsulin-like peptides were incubated
overnight at 4 "C with 10 pl of anti-PC2 serum and 50 pl (packed gel)
of preswollen protein A-Sepharose. The protein A-Sepharose was
recovered by centrifugation and washed as above. Bound proteins
were eluted with 200
p1
of 20 mM HCI, freeze-dried, and reconstituted
in 50 pl of SDS sample loading buffer (125 mM Tris/HCl (pH 6.8)
containing
2%
(w/v) SDS, 0.25
M
sucrose, 5 mM EDTA, 65 mM
dithiothreitol, and 0.005% bromphenol blue). The samples were
heated for 5 min at 100 'C and subjected
to
SDS-PAGE on slab gels
polymerized from
10%
(w/v) acrylamide, 0.065% N,N'-methylenebis-
acrylamide, by using the discontinuous buffer system of Laemmli
(1970), and fluorography was performed
as
above. Molecular size
calibration was achieved with I4C-labeled lysozyme, 6-lactoglobulin,
cr-chymotrypsin, ovalbumin, BSA, and phosphorylase
b
(Bethesda
Research Laboratories, Paisley, Scotland).
N-Clycanase Treatment-Islets were sonicated for 15
s
at 3 microns
in 50
pl
of 100 mM Tris (pH 8.0) containing 0.5% SDS, 50 mM
P-
mercaptoethanol,
1
mM phenylmethylsulfonyl fluoride, and
1
mM
EDTA. The lysates were centrifuged for
3
min at
10,000
X
g
and the
supernatants heated for 5 min at 100 "C and then cooled for 5 min
on ice. The samples were incubated for
18
h at 37 "C in
a
final volume
of
150
pl containing 1.7% (w/v) Nonidet
P-40
and 1.5 units of
N-
'
The abbreviations used are: BSA, bovine serum albumin; PAGE,
polyacrylamide gel electrophoresis;
ER,
endoplasmic reticulum.
glycanase. The samples were then immunoprecipitated with anti-PC2
serum and the eluted proteins subjected to SDS-PAGE and fluorog-
raphy as above.
Ultrastructural Analysis-Ultrathin sections
of
human pancreatic
islets were fixed in 2.5% glutaraldehyde (pH 7.2) and embedded in
Spurr's resin. Sections were incubated for 2 h
at
room temperature
with a
1:lO
dilution of anti-PC2 serum in phosphate-buffered saline
(150 mM NaCl, 16 mM NaHzP04, 4 mM NazHP04) containing 1%
(w/v) ovalbumin and then exposed to protein A-gold (10 nm colloidal
gold) for
1
h at room temperature
as
described previously (Pow and
Clark, 1990). Ultrastructural details were visualized by staining for
10
min with saturated uranyl acetate and then
for
10 min with lead
nitrate. Sections were viewed with
a
Joel electron microscope. Control
experiments were performed by incubating sections with preabsorbed
antiserum (4 mg/ml PCZ/glutathione S-transferase fusion protein,
overnight at 4 "C).
Immunoblot Analysis-Insulinoma subcellular fractions were sus-
pended in SDS sample loading buffer, sonicated for 15
s
at
3
microns,
and heated for 5 min
at
100 "C. The samples were subjected
to
SDS-
PAGE either
as
described above (PC2-related peptides)
or
by using a
modification of the method of Schagger and von Jagow (1987) (proin-
sulin-related peptides) as described in Hutton et
al.
(1990). Gels were
subsequently equilibrated for
1
h at room temperature in 25% (v/v)
acetic acid, 10% (v/v) isopropyl alcohol and then for
1
h at room
temperature in 48 mM Tris, 39 mM glycine (pH 9.2) containing 0.1%
SDS and 20% (v/v) methanol. The electrophoresed proteins were
then transferred electrophoretically onto nitrocellulose paper and
subjected to immunoblot analysis as described previously (Guest
et
al.,
1991). Molecular size calibration was achieved with lysozyme,
0-
lactoglobulin, carbonic anhydrase, ovalbumin, BSA, and phosphoryl-
ase b (Bio-Rad).
Assays-Protein was determined by a dye-binding technique
(Bradford, 1976) with BSA as standard. Carboxypeptidase
H
activity
was determined by
a
radiometric assay
as
described previously (Guest
et
al.,
1989b).
P-N-Acetylglucosaminidase
was determined by the
fluorometric method of Barrett and Heath (1977) and galactosyl
transferase from the incorporation of [U-14C]UDP galactose into
soybean trypsin inhibitor (Bretz and Staubli, 1977). NADPH cyto-
chrome c reductase and alkaline phosphatase were determined by the
spectrophotometric methods of Sottocasa
et
al.
(1967) and Bowers
and McComb (1975), respectively.
RESULTS
Molecular Composition
of
35S-Labeled Proinsulin and PC2
Immunoprecipitates
in
Puke-Chase-labeled
Islets-Islets in-
cubated for
1
h in the presence of 16.7 mM glucose incorpo-
rated
1.2%
of the [35S]methionine from the labeling media
into trichloroacetic acid-precipitable material. Approximately
8%
of the incorporated radioactivity appeared in proinsulin-
related immunoprecipitates and
0.1%
in that of PC2. Under
these conditions only rat proinsulin I1 and not proinsulin
I
was labeled since the latter does not contain a methionine
residue (Smith, 1966).
In agreement with previous studies (see Docherty and Stei-
ner, 1982; Hutton
et
al.,
1987; Davidson
et al.,
1988) proinsulin
to insulin conversion commenced approximately 30 min after
synthesis via intermediates which co-migrated with the des-
31,32- and des-64,65-proinsulins and proceeded with apparent
first order kinetics equivalent to a
loss
of 0.83%
of
the initial
proinsulin/min
(tl,z
=
60 min) (Figs.
1A
and
2).
In contrast,
PC2 processing occurred more slowly. By completion
of
the
1-h labeling period, approximately 95%
of
the immunoprecip-
itable PCB-related radioactivity appeared as a protein of 76
kDa (Fig. 1B). After this time, the 76-kDa component de-
creased over 4-5 h with a
loss
of
0.35%/min
(tl/z
=
140
min)
(Figs. 1B and 2). Proteins of 71 and 68 kDa appeared by the
end of the
1st
h of chase incubation and together comprised
23% of the total immunoprecipitable radioactivity. These
forms disappeared between
1
and 3 h of the chase period to
be replaced
by
a 66-kDa protein. This in turn was converted
between
3
and 7 h to a diffuse 64-66-kDa form which ac-
PC2
Biosynthesis
22403
IIIII
6
98
-
43
-
FIG.
1.
Molecular composition
of
35S-labeled proinsulin and
PC2 immunoprecipitates in pulse-chase-labeled islets.
Islets
were labeled for
1
h in Krebs incubation medium containing 16.7 mM
glucose and 0.2 mCi ['"Slmethionine and then resuspended in non-
radioactive Dulhecco's modified Eagle's medium containing
4.5
mM
glucose and
10%
fetal calf serum. The cellular forms of
A,
proinsulin
and
H,
PC2 were immunoprecipitated upon termination of the incu-
hations at the indicated times and subjected to PAGE and fluorog-
raphy as described under "Experimental Procedures." The migration
of
molecular size standards, proinsulin
(PI),
proinsulin conversion
intermediates
(IN?'),
and insulin
(INS)
is
indicated. Similar results
were obtained on three separate occasions.
02468'
time
(h)
FIG. 2.
Time course of proinsulin and PC2 conversion.
The
extent of conversion
of
proinsulin
(0)
and the 76-kDa form of PC2
(0)
was derived from Fig.
1
by densitometric scanning of the fluoro-
graphs. The results are the percentage of precursor remaining and
are expressed relative to the total immunoprecipitable radioactivity
at the indicated times (h).
counted for approximately 91% of the radioactivity immuno-
precipitated at 17 h.
N-Glycanase Treatment of "S-Labeled PC2-N-Glycanase
treatment resulted in a molecular size reduction of 7 kDa for
both the 76- and 64-66-kDa forms of PC2, producing peptides
of
69 and 57-59 kDa, respectively (Fig.
3).
A similar reduction
in size has been reported previously for the native insulin
secretory granule enzyme following N-glycanase treatment
(Bennett et
al.,
1992) and is consistent with the presence of
three consensus sequences for N-linked glycosylation in rat
PC2 (Hakes et
al.,
1991).
Secretion of Proinsulin- and PC2-related Peptides from
"S-
Labeled Islets-Islets were labeled for
1
h and the proinsulin-
and PCB-related peptides immunoprecipitated from media
collected over eight successive intervals
(1
h) of chase incu-
bation in the presence of 16.7 mM glucose. The proinsulin-
12
34
IIII
98
-
h
0
2
66-
v
c
s
m
.
1
0
."
c
&
43
-
N-glycanase
-
+-
+
FIG.
3.
N-Glycanase treatment of '%-labeled PC2.
Islets
were pulse-chase labeled as in the legend for Fig. 1, lysed, and either
mock-treated
or
suhjected to N-glycanase treatment as described
under "Experimental Procedures." PC2 immunoprecipitates are from
a
1-h pulse
(lanes
I
and
2)
and
a
1-h pulse plus 17-h chase
(lanes
3
and
4).
Similar results were obtained on two separate occasions.
123.I567X
A
IIIIIIII
PI-.."
8u88a
S
B
98
-
-
2
66-
x
r
e
-
8.
43
-
FIG.
4.
Secretion of proinsulin- and PC2-related peptides
from pulse-chase-labeled islets.
Islets were labeled for 60 min in
Krebs medium containing 16.7 mM glucose and 0.2 mCi of
[""S]
methionine and then resuspended in nonradioactive Dulbecco's mod-
ified Eagle's medium containing 16.7 mM glucose and 10% fetal calf
serum. The medium was again replaced
at
1-h intervals
for
up to
8
h
of chase incubation, and the molecular forms of
A,
proinsulin and
R,
PC2 were immunoprecipitated and subjected to PAGE and fluorog-
raphy. Similar results were obtained on four separate occasions. The
abbreviations are
as
in Fig.
1.
related peptides appeared initially in the medium within 60
min of chase incubation and were comprised of a mixture of
proinsulin, intermediates, and insulin with a shift to increas-
ing proportions of insulin at later time intervals (Fig.
4A).
Approximately 90% of the initial islet proinsulin-related im-
munoreactivity was released over the 8-h chase period with a
tIl2 of
3.3
h (Fig.
5).
By contrast, release of the PCB-related
peptides was not detected until the 2nd h of chase incubation.
After this time the quantity of PC2-related material secreted
(approximately 70% of the cellular content) and the rate of
release
(
t1r2
=
3.2
h) were comparable to that of insulin (Figs.
4B
and
5).
Also in contrast to the proinsulin-related peptides,
the precursor form of PC2 was not released (Fig. 4B). The
secreted material was comprised initially of only the 66-kDa
intermediate and then increasing proportions of the 64-66-
kDa form at later intervals. Secretion of both the proinsulin-
22404
PC2
Biosynthesis
0
2
4
6
8
Time
(h)
FIG.
5.
Time course
of
secretion
of
labeled proinsulin- and
PC2-related peptides.
The amount of released radioactivity asso-
ciated with proinsulin-related
(0)
and PC2-related
(0)
peptides was
derived from Fig.
4
by densitometric scanning of the fluorographs.
The results are cumulative radioactivities released hy the indicated
times and are expressed
as
a percentage of the initial islet content at
the end of the labeling period,
as
determined in
parallel
incubations.
and PCP-related peptides occurred by regulated release of
secretory granule contents and was not caused by cell damage
or constitutive secretion, as neither of the proteins was de-
tected in significant quantity in the medium of control islets
incubated in the presence of a nonstimulatory concentration
of glucose
(3.3
mM) (data not shown).
Ultrastructural Analysis-Ultrastructural labeling of pan-
creatic
/3
cells by the protein A-gold technique showed that
PC2 immunoreactivity was localized predominantly in secre-
tory granules (26 gold particles/pm2) with the majority of
labeling occurring over the granule core (Fig. 6).
A
low level
of
protein A binding was also associated with smaller electron-
lucent vesicles and cytoplasmic elements (2 gold particles/
pm’) but not with lysozomes and mitochondria.
No
labeling
of
sections was observed using antiserum absorbed previously
with 4 mg/ml of the PCZ/glutathione S-transferase fusion
protein (data not shown).
Molecular Characterization
of
Proinsulin and PC2-related
Peptides in Insulinoma Subcellular Fractions-Analysis of the
molecular forms of proinsulin and PC2 in subcellular com-
partments of the secretory pathway was facilitated by using a
transplantable rat insulinoma. This tissue is available in gram
quantities (Chick et al., 1977) and synthesizes and stores
insulin (Gold et al., 1984) and other granule proteins such as
betagranin (Hutton et al., 1987) and PC22 with kinetics similar
to those observed in rat islets. Fractionation of the insulinoma
by Percoll density gradient centrifugation produced six frac-
tions designated 1-6 in order of increasing buoyant density.
Marker protein analysis showed that ER and Golgi elements
were concentrated in fractions
1
and 2, secretory granules in
fractions
3-5,
and lysozomes in fraction
6,
whereas plasma
membranes were distributed across the gradient with the
highest levels observed in fraction
1
(Table
I).
Immunoblot analysis of the fractions showed that insulin
and PC2 immunoreactivities were most abundant in secretory
granules with lower levels in Golgi/ER and lysozomes. By
adjusting sample loadings to give immunoblots of similar
intensity, it became apparent that the molecular forms of the
insulin- and PCB-immunoreactive peptides present in each
fraction varied (Fig. 7). Golgi/ER-enriched fractions con-
tained proteins of equivalent size to the precursor (76-kDa)
and mature (64-66-kDa forms of PC2 but only the precursor
form of insulin (proinsulin). Secretory granules, by contrast,
P. C. Guest,
S.
D.
Arden,
D.
L.
Bennett, A. Clarke,
N.
G.
Rutherford, and
.J.
C. Hutton, unpublished findings.
FIG.
6.
Ultrastructural labeling
of
an islet
@
cell.
An ultrathin
human islet section containing
a
B
cell was labeled by the protein
A-
gold technique
as
described under “Experimental Procedures.” Insulin
granules
(I),
mitochondria
(M),
and lysozomes
(L)
are indicated.
Bar
=
0.5
pm. Similar results were obtained on at least three separate
occasions.
contained only the mature form
of
PC2 and yet significant
amounts of proinsulin along with insulin.
DISCUSSION
The post-translational processing of many proteins enter-
ing the secretory pathway appears to be achieved by a highly
conserved mechanism involving initial cleavage after se-
quences containing basic amino acids by members of a family
of endoproteases related to the bacterial subtilisins (see Doch-
erty and Steiner, 1982; Thomas et al., 1988; Hutton, 1990;
Lindberg and Hutton, 1991). The majority of proproteins
which are secreted in a regulated manner appear to undergo
relatively slow conversion in dense core secretory vesicles,
whereas constitutively secreted proteins and membrane-
bound proreceptors are cleaved rapidly during brief transit
through the trans-Golgi network. Effective regulation of the
converting activities might be achieved by a combination of
compartmentalization of the enzymes and kinetic mecha-
nisms related to substrate concentration, structural features
of the cleavage site, or regulation by the prevailing ionic
conditions (e.g. Ca’+ and pH) of the compartment where
activity is expressed. Further, the processing enzymes them-
selves are likely to be produced as proproteins inasmuch as
all of those identified to date contain cleavage site consensus
sequences. Synthesis as a pro-form may afford protection of
the enzyme from denaturation in early segments of the secre-
tory pathway or provide a mechanism for limiting activity or
targeting the enzyme to specific intracellular compartments.
PC2
Biosynthesis
22405
TARLE
I
Charactwization of insulinoma subcellular fractions
Suhcellular fractions were prepared from rat insulinoma, and protein markers for secretory granules (carboxypeptidase
H),
lysozomes
(8-
N-acetylglucosaminidase),
ER
(NADPH-cytochrome c reductase), Golgi (galactosyl transferase), and plasma membranes (alkaline phospha-
tase)
were assaved
as
described under “Exaerimental Procedures.” ND. not detected.
Protein Carboxypeptidase S-N-Acetyl- NADPH-
glucosamini- cytochrome
c
dase reductase
H
Galactosyl Alkaline
transferase phosphatase
m:: nmol/min/mg nrbitrary units nmol/min/mg nmol/min/mg nmol/min/rng
Homogenate 620 0.20 7.5 4.1 ND 0.37
Fraction
1
33
0.57
7.2 16.2 0.60 1.30
Fraction 2 39 0.48
8.5
11.9 0.34 0.29
Fraction
:3
63
2.54 26.0 6.2 0.13 0.68
Fraction 4
7.5
4.15 38.4 ND
0.05
0.64
Fraction
5
8.0 5.90 83.2 ND ND 0.35
Fraction
G
5.5
1.72 100 ND ND 0.21
I:r:laion
A
123456
Illlll
PI
-“
ISS
-
v-
B
9x
-
z
66-
-
-
t
r
,.
..
43
-
FIG.
7.
Molecular composition of proinsulin- and
PC2-re-
lated peptides in insulinoma subcellular fractions.
Aliquots of
insulinoma subcellular fractions were electrophoresed and transferred
to
nitrocellulose paper, which was reacted with either
A,
guinea pig
anti-bovine insulin serum (1:500) (Guest
et al.,
1991)
or
R,
anti-PC2
serum (1:500),
as
described under “Experimental Procedures.” Load-
ings were
as
follows: fractions
1
and 2,
250
pg;
fractions 3-6,
50
pg.
Similar results were obtained on three separate occasions. The
ab-
hreviations
are
as
in Fig.
1.
The present studies show that the proinsulin-converting
enzyme, PC2, is synthesized initially as
a
76-kDa precursor
which undergoes limited post-translational processing to gen-
erate the mature 64-66-kDa form of the protein. N-Glycanase
treatment showed that both the precursor and mature forms
of
PC2 contained approximately 7 kDa contributed by
N-
linked carbohydrate units, consistent with the view that the
difference in size between the forms is
a
result of proteolysis.
The mobility of the mature form on SDS-PAGE was indistin-
guishable from that of the native insulin granule enzyme
which has been shown to be comprised of two molecular
variants (Bennett
et
al.,
1992). One of these commences at a
position equivalent to amino acid 109 of the deduced rat
sequence (Hakes
et
al.,
1991) which implicates cleavage after
the tetrabasic sequence
Arg’05-Lys’”6-Lys1”’-Arg’oR.
The sec-
ond has an
N
terminus beginning at residue 112 which might
be the result of endoproteolytic cleavage after Arg’”8-Gly1”9-
Tyr’”’-Arg”’. Both of these sequences fit the Arg-X-X-Arg
minimal motif thought to be recognized by membrane-asso-
ciated Golgi-resident proteases such as the proalbumin-con-
verting enzyme (Brennan and Peach, 1991) and furin (Molloy
et
al.,
1992). Cleavage after either sequence would result in
removal of a nonglycosylated N-terminal pro-sequence of 9-
10 kDa, which approximates the size difference observed
between the precursor and mature forms in the present pulse-
chase radiolabeling studies. The transient appearance of the
71-, 68-, and 66-kDa immunoreactive peptides during conver-
sion suggests that these may have been intermediates in the
overall process generated by cleavage after other basic sites
which occur within the pro-sequence of the precursor.
The ultrastructural localization of PC2 to secretory gran-
ules and the finding that the newly synthesized enzyme is
secreted from islets in response to glucose stimulation dem-
onstrate that it is segregated into the regulated pathway of
secretion. The time course of its synthesis and packaging into
granules however was significantly slower than that of insulin.
Proinsulin to insulin conversion commenced following an
initial delay of
30
min and thereafter proceeded with a
tlr2
of
approximately 60 min. Regulated release of the newly synthe-
sized proinsulin-related peptides (proinsulin, conversion in-
termediates, and insulin) began after
1
h and proceeded with
a
tllP
of
3.3
h. The finding that the precursor and intermediate
forms of insulin were secreted is consistent with previous
studies which have shown that proinsulin conversion occurs
mainly within secretory granules (Orci
et
al.,
1985, 1987;
Davidson
et
al.,
1988). Further evidence for this was provided
by the present immunoblot studies which showed that insu-
linoma fractions enriched in Golgi/ER elements contained
only proinsulin, whereas both proinsulin and insulin were
present in secretory granules.
Proteolytic cleavage of the PC2 precursor was significantly
slower than that of insulin with an initial delay of approxi-
mately
1
h and a
tlrz
of 140 min. In addition, glucose activated
the release of only the 66- and 64-66-kDa forms of PC2 and
then only
at
times more than 2 h after synthesis. After this
time it was released with a
tl12
similar to that of insulin. This
indicated that delivery of PC2 to secretory granules is delayed
by approximately
1
h compared with insulin but that, once
sorted, PC2 and insulin are released in response to secreta-
gogue stimulation with similar kinetics. There was no evi-
dence for secretion of PC2 via the constitutive pathway as it
was not released in the presence of a substimulatory concen-
tration of secretagogue. The finding that only the 66- and 64-
66-kDa forms of the enzyme were released suggested that
proteolysis was initiated in a pre-granule compartment. This
is consistent with the observation that insulinoma Golgi/ER
fractions contained
a
mixture of the 76- and 66-kDa forms of
PC2, whereas secretory granules contained only the mature
enzyme.
The present studies suggest that the delay in transport of
PC2 to secretory granules is caused by retention of the un-
processed precursor in
a
pre-granule compartment such as the
ER. Such retention might be a result of interactions with ER-
22406
PC2
Biosynthesis
resident proteins such as binding protein, which is thought to
promote protein assembly, and protein disulfide isomerase,
which catalyzes thiol oxidation and disulfide exchange reac-
tions (see Pelham, 1989). Retention in the ER might also
result from slow removal of glucosyl residues from N-linked
oligosaccharides, which has been shown in the case of some
secretory proteins to be necessary for transport to the Golgi
complex (Lodish and Kong, 1984).
The observation that the transfer of proinsulin and
PC2
from the ER to secretory granules occurs with different ki-
netics has important implications for secretory granule func-
tion. The results suggest that
at
the point of secretory granule
assembly, the
PC2
which is incorporated is synthesized ap-
proximately
1
h earlier than the condensing prohormone.
Since insulin mRNA translation undergoes rapid and marked
changes in response to ambient glucose concentrations, it
follows that the ratio of
PC2
to proinsulin in the nascent
granule will vary. This will occur irrespective of any effects
glucose may have on
PC2
biosynthesis. Since the
V,,,
of the
proinsulin endoproteases appear to be rate limiting, it follows
that the relative proportions of precursor, processing inter-
mediates, and insulin released might respond to changes in
glucose concentration. Other contributory factors will include
the time required
for
newly formed granules to become com-
petent for exocytosis, the pool size of stored granules, and the
intensity and duration of the stimulus. Reports that glucose
can accelerate proinsulin conversion (Nagamatsu
et
al.,
1987)
and promote preferential release of the newly synthesized
hormone (Sando
et
al.,
1972; Rhodes and Halban, 1987; Arvan
et
al.,
1991) are
of
interest in this context and warrant further
investigation. The abnormalities in circulating levels of proin-
sulin and processing intermediate which occur in persons with
glucose intolerance, noninsulin-dependent diabetes,
or
insu-
linomas are conceivably related to such phenomena (Heaton
et
al.,
1987; Ward
et
al.,
1987).
Acknowledgment-We thank
Dr.
E.
M.
Bailyes for helpful discus-
sion
and
editing
of
the manuscript.
REFERENCES
Arvan, P., Kuliawat, R., Prabakaran, D., Zavacki, A,, Elahi, D., Wang,
S.,
and
Pilkev. D.
(1991) J.
Bid.
Chem.
266.
14171-14174
Axen,
R.;
Porath, J., and Ernback,
S.
(1967)
Nature
215, 1302-1304
Bailyes,
E.
M., Bennett, D. L., and Hutton,
J.
C.
(1992)
Enzyme,
in press
Barr, P.
J.
(1991)
Cell
66, 1-3
Barrett, A. J., and Heath,
M.
F.
(1977)
Lysozomes:
A
Laboratory Handbook
(Dinele.
J.
T..
ed)
DD. 9-145.
North Holland Publishine Co., Amsterdam
Benne$,
D.
L.,’B&&es, E.
M.,
Nielsen, E., Guest,
P.
c.,
Rutherford, N. G.,
Arden.
S.
D.. and Hutton.
J.
C.
(1992) J. Biol.
Chem.
267. 15229-15236
Bowers,
b.
N.,’Jr, and McComb,
R.
B.
(1975) Clin.
Chem.
2’1, 1988-1995
Brennan,
S.
O., and Peach, R.
J.
(1991)
J.
Biol.
Chem.
266,21504-21508
Bradford, M.
M.
(1976)
Anal. Biochem.
72, 248-254
Breshnahan,
P.
A,, Leduc, R., Thomas, L., Thorner, J., Gibson, H. L., Brake,
Bretz, R., and Staubli,
W.
(1977)
Eur.
J.
Biochem.
77,
181-192
Chick, W. L., Warren,
S.,
Chute, R. N., Like, A. A,, Lauris, V., and Kitchen,
Christie, D. L., Batchelor, D. C., and Palmer, D.
J.
(1991)
J.
Biol.
Chem.
266,
Davidson, H.
W.,
and Hutton,
J.
C.
(1987)
Biochem.
J.
245, 575-582
Davidson, H. W., Rhodes,
C.
J.,
and Hutton,
J.
C.
(1988)
Nature
333, 93-96
Docherty,
K.,
and Steiner, D. F.
(1982)
Annu.
Reu.
Physiol.
44,625-638
Gold, G., Gishizky, M. L., Chick,
W.
L., and Grodsky,
G.
M.
(1984)
Diabetes
Gray,
I.
P., Siddle, K., Frank, B. H., and Hales, C. N.
(1987)
Diabetes
36,684-
Guest, P. C., Rhodes, C. J., and Hutton,
J.
C.
(1989a)
Biochem.
J. 257, 431-
Guest,
P.
C., Pipeleers, D., Rossier, J., Rhodes, C.
J.,
and Hutton,
J.
C.
(1989b)
Guest,
P.
C.,
Ravazzola, M., Davidson, H.
W.,
Orci, L., and Hutton,
J.
C.
(1991)
Hakes. D.
J..
Birch. N. P.. Mezev. A,. and Dixon.
J.
E.
(1991)
Endocrinolon
A.
J., Barr,
P.
J., and Thomas, G.
(1990) J.
Cell
Biol.
11 1, 2851-2859
K. C.
(1977)
Proc. Natl. Acad. Sei.
U.
S.
A.
74, 628-632
15679-15683
33,556-561
688
437
Biochem.
J.
264,503-508
Endocrinology
129, 734-740
129,305313063
and Leslie. R. D. G.
(1987)
Br.
Med.
J.
294,
145-146
_I I
..
Heaton, D. A,, Millward, B. A,, Gray,
I.
P.,
Tun,
Y.,
Hales, C. N., Pyke, D.
A.,
Hutton,
J.
C.
(1982)
Biochem.
J.
204, 171-178
Hutton,
J.
C.
(1990)
Curr.
Opinion
Cell
Biol.
2, 1131-1142
Hutton,
J.
q.,
Penn,
E.
J., and Peshavaria,
M.
(1982)
Diabetologia
23, 365-373
,.,
Penn, E. J., and Peshavaria, M.
(1983)
Biochem.
J.
210, 297-
Hutton,
J.
C.,
Davidson, H.
W.,
Grimaldi, K. A,, and Peshavaria, M.
(1987)
Hutton,
J.
C., Bailyes, E. M., Rhodes,
C.
J., and Guest,
P.
C.
(1990)
in
Peptide
Biochem.
J.
244,449-456
Hormone Secretion: A Practical Approach
(Hutton,
J.
C., and Siddle, K., eds)
pp.
309-336.
Oxford University Press, Oxford,
U.
K.
Kiefer, M. C., Tucker,
J.
E., Joh, R., Landsberg, K. E., Saltman, D., and Barr,
P.
J.
(1991)
DNA Cell
Biol.
10, 757-769
Kirchmair, R., Gee, P., Hogue-Angeletti, R., Laslop, A,, Fischer-Colbrie, R.,
and Winkler, H.
(1992)
FEBS Lett.
297,302-305
Laemmli, U. K.
(1970)
Nature
227, 680-685
Lindberg,
I.,
and Hutton,
J.
C.
(1991)
in
Peptide Biosynthesis and Processing
Lodisb, H. F., and Kong, N.
(1984)
J. Cell
Biol.
98, 1720-1729
(Fricker, L. D., ed) pp.
141-173,
CRC Press, Boca Raton, FL
Molloy,
S. S.,
Breshnahan,
P.
A,, Leppla,
S.
H., Klimpel, K. R., and Thomas,
Nagamatsu,
S.,
Bolaffi,
J.
L., and Grodsky, G.
M.
(1987)
Endocrinology
120,
Nakayama, K., Kim, W.-S., Torii,
S.,
Hosaka,
M.,
Nakagawa, T., Ikemizu, J.,
Ora.
L..
Ravazzola. M.. Amherdt. M.. Madsen.
0..
Vassalll, J.-D.. and Perrelet,
G.
(1992)
Proc. Natl. Acad. Sci.
U.
S.
A,,
in press
1225-1231
Baba, T., and Murakami, K.
(1992)
J.
Biol. Chem.
267!
5897-5900
A.’fi965)
Cell
42,671-681
Perrelet, A.
(1987)
Cell
49,
865-868
.. ..
Orci, L., Ravazzola, M., Storch, M.-J., Anderson, R. G. W., Vassalli, J.-D., and
Pelham, H. R. B.
(1989)
Annu.
Reu.
Cell
Biol.
5, 1-23
Pow,
D.
V., and Clark, A.
(1990)
in
Peptide Hormone Secretion: A Practical
Amroach
(Hutton.
J.
C., and Siddle, K., eds)
pp.
189-210,
Oxford University
Pi&, Oxford,
U.
K.
~~
Rhodes, C. J., and Halban,
P.
A.
(1987)
J.
Cell
Biol.
105, 145-153
Sando, H., Borg, J., and Steiner, D. F.
(1972) J.
Clin.
Inuest.
51, 1476-1485
Schagger, H., and von Jagow, G.
(1987)
Anal. Biochem.
166,368-379
Seidah,
N.
G., Marcinkiewicz, M., Benjannet,
S.,
Gaspar,
L.,
Beaubien, G.,
Mattei,
M.
G.,
Lazure, C., Mbikay,
M.,
and Chretien,
M.
(1991)
Mol. Endo-
crinol.
5, 111-122
Smeekens,
S.
P., Avruch, A.
S.,
LaMendola,
J.,
Chan,
S.
J., and Steiner,
D.
F.
(1991)
Proc.
Natl. Acad. Sci.
U.
S.
A.
88,340-344
Smith,
L.
F.
(1966)
Am.
J.
Med.
40,662-666
Sopwith, A: M., Hutton,
J.
C., Naber,
S.
P.,
Chick,
W.
L., and Hales, C. N.
Sottocasa,
G.
L., Kuylenstierna, B., Ernster, L., and Bergstrand, A.
(1967)
J.
Thomas.
G.,
Thorne, B. A,, and Hruby,
D.
E.
(1988)
Annu.
Reu.
Physiol.
50,
(1981)
Dlabetologia
21, 224-229
Cell
Biol.
32, 415-438
Ward, W.
K.,
LaCava, E. C., Paquette,
T.
L.,
Beard,
J.
C., Wallum,
B.
J., and
323-332
Porte, D., Jr.
(1987)
Diabetologra
30, 698-702
... Within the neuroendocrine cell, the bulk of proPC2 appears to reside in the ER [67,145,308,309]; mature PC2, however, is predominantly localized within regulated secretory granules [68,308]. This distribution is unusual among convertases, though PC7 seems to exhibit similar storage in the ER [310]. ...
... Within the neuroendocrine cell, the bulk of proPC2 appears to reside in the ER [67,145,308,309]; mature PC2, however, is predominantly localized within regulated secretory granules [68,308]. This distribution is unusual among convertases, though PC7 seems to exhibit similar storage in the ER [310]. ...
... Figure 9 depicts the maturation of proPC2 to its active form, in association with its binding partner 7B2. ProPC2 is initially synthesized as an Nglycosylated proform [308] which is later tyrosine sulfated [281] and sialylated; however, not all of its sugar chains can mature to sialylated forms since mature PC2 contains both endoglycosidase Hresistant and sensitive sugars [67,145,277,308,309]. We have found that deletion of sugar sites by mutagenesis results in loss of its ability to exit the ER, suggesting that glycosylation is required for stability ( J. Hwang and I. Lindberg, un published observations). ...
Article
The prohormone convertases (PC) 1/3 and 2 are calcium-activated eukaryotic subtilisins with low pH optima which accomplish the limited proteolysis of peptide hormone precursors within neurons and endocrine cells. In this lecture, we review the biochemistry, regulation, and roles of PC1/3 and 2 in disease, with an emphasis on the work published in the last 10 years. In the 20 years since their discovery, a great deal has been learned about their localization and cellular functions. Both PCs share the same four domains: the propeptides perform important roles in controlling activation and targeting; the catalytic domains confer specificity, with PC1/3 possessing a more restricted binding pocket than that of PC2; the P domain is required for expression and contributes to enzymatic properties; and the C-terminal tail assists in proper routing to granules. PC1/3, but not PC2, exists in full-length and C-terminally truncated forms that exhibit different biochemical properties. Both enzymes associate with binding proteins; proSAAS is thought to modulate precursor cleavage by PC1/3, while co-expression of 7B2 is obligatory for the formation of active PC2. Finally, new studies have revealed an increasingly important role for PC1/3 polymorphisms and mutations in glucose homeostasis and obesity. Table of Contents: General Introduction to the Prohormone Convertases / Prohormone Convertase 1/3 / Prohormone Convertase 2 / Summary and Future Directions / References / Author Biography
Article
Full-text available
AtT-20 mouse corticotrope tumor cell lines overexpressing the prohormone convertases PC1 or PC2 were established and used to examine prohormone and prohormone convertase biosynthetic processing. On a molar basis, wild-type AtT-20 cells synthesize about 20% as much PC1 as the endogenous prohormone, proopiomelanocortin (POMC). Kinetic, oligosaccharide, and temperature blockade analyses established that proPC1 is converted to PC1 in the endoplasmic reticulum at a rate independent of the level of PC1 or PC2 expression. In contrast, proPC2 is converted to PC2 primarily in a post-trans-Golgi compartment. PC1 is further shortened from its COOH-terminal end in a post-trans-Golgi compartment in a step that is accelerated at higher levels of PC1 expression, but unaltered by PC2 overexpression. The initial steps in POMC processing are speeded up by overexpression of PC1, and overexpression of PC1 leads to more extensive cleavage of POMC to smaller products. However, even when the rate of PC1 synthesis exceeds that for POMC by 2-fold, PC1 does not cleave the Lys-Lys or Arg-Lys bonds cleaved upon overexpression of PC2.
Chapter
The secretory granules of pancreatic beta cells are specialized organelles responsible for the packaging, storage and secretion of the vital hormone insulin. The insulin secretory granules also contain more than 100 other proteins including the proteases involved in proinsulin-to insulin conversion, other precursor proteins, minor co-secreted peptides, membrane proteins involved in cell trafficking and ion translocation proteins essential for regulation of the intragranular environment. The synthesis, transport and packaging of these proteins into nascent granules must be carried out in a co-ordinated manner to ensure correct functioning of the granule. The process is regulated by many circulating nutrients such as glucose and can change under different physiological states. This chapter discusses the various processes involved in insulin granule biogenesis with a focus on the granule composition in health and disease.
Chapter
Pulse radiolabelling of cells with radioactive amino acids is a common method for studying the biosynthesis of proteins. The labelled proteins can then be immunoprecipitated and analysed by electrophoresis and imaging techniques. This chapter presents a protocol for the biosynthetic labelling and immunoprecipitation of pancreatic islet proteins which are known to be affected in psychiatric disorders such as schizophrenia.
Chapter
Pulse radiolabeling of cells with radioactive amino acids is a common method for tracking the biosynthesis of proteins. Specific proteins can then be immunoprecipitated and analyzed by electrophoresis and imaging techniques. This chapter presents a protocol for the biosynthetic labeling of pancreatic islets with³⁵S-methionine, followed by multiplex sequential immunoprecipitation of insulin and three other secretory granule accessory proteins. This provided a means of distinguishing those pancreatic islet proteins with different biosynthetic rates in response to the media glucose concentrations.
Chapter
The biosynthesis of the three endothelin isoforms (ET-1, ET-2 and ET-3) follows a similar pattern with initial synthesis as precursor proteins (≈200 amino acid residues) which undergo selective proteolysis to yield the biologically inactive intermediates called big endothelins (Bloch et al. 1989a,b; Ohkubo et al. 1990; Yanagisawa et al. 1988). The human ET-1 gene encodes a 212 amino acid precursor, preproendothelin-1 (Bloch et al. 1989b). Removal of the signal sequence generates proendothelin-1 (proET-1,195 a.a.) which is processed at double basic amino acid residues to release the intermediate referred to as big ET-1 (human form, 38 a.a.) (Fig. 1). The active endothelins are generated by hydrolysis of the Trp21-Val22 bond in big ET-1 and big ET-2, or Trp21-Ile22 in big ET-3 (Fig. 2). This cleavage is unique to the endothelins and hence the role of a specific endothelin-converting enzyme (ECE) was proposed (Yanagisawa et al. 1988). Most studies have concluded that proendothelin-1 is processed intracellularly to ET-1 prior to its release (Corder et al. 1995b; Harrison et al. 1993, 1995; Russell and Davenport 1999; Xu et al. 1994). This is also the case for ET-2 (Lambert et al. 2000), but studies of an ET-3 secreting cell line have yet to be described. For ET-1 there is evidence of polar secretion from the abluminal surface of endothelial cells (Wagner et al. 1992), which indicates that ET-1 functions primarily as a paracrine modulator of vascular function. Secretion of the endothelins is mainly via constitutive secretory pathways, hence the most important factor affecting their synthesis is the level of gene expression.
Chapter
Knowledge of the gene structure of secreted proteins and analysis of their biosynthesis has revealed that many are initially produced as larger biologically inactive precursors that are subjected to limited proteolysis at sites marked by clusters of basic amino acids in characteristic linear sequences. Peptide hormones and neurotransmitters that are secreted via the regulated secretory pathway in neuroendocrine tissues are generally processed at pairs of basic amino acids, typically Lys Arg and Arg Arg and much less frequently at Lys Lys or Arg Lys sites. Growth factors and receptor molecules that follow the constitutive pathway are processed at sites marked by a more complex array of basic amino acids, typically a pair of basic amino acids with a further basic amino acid in the P4 position (for review, see ref. 7). In the majority of cases where the most C-terminal amino acid in the cluster is an arginine residue, cleavage of the amide bond occurs on the C-terminal side of this arginine. When it is a lysine residue, cleavage often occurs on the N-terminal side and probably involves a different enzymatic process in many cases. Basic amino acids exposed at the C-termini after endoproteolytic cleavage are removed by carboxypeptidase H (E) (CPH) within the regulated pathway of secretion. Whether a similar activity is present in the constitutive pathway is not clear.
Article
The biosynthesis and processing of proinsulin was investigated in the diabetic Goto-Kakizaki (GK) rat. Immunofluorescence microscopy comparing GK and Wistar control rat pancreata revealed marked changes in the distribution of alpha-cells and pronounced beta-cell heterogeneity in the expression patterns of insulin, prohormone convertases PC1, PC2, carboxypeptidase E (CPE) and the PC-binding proteins 7B2 and ProSAAS. Western blot analyses of isolated islets revealed little difference in PC1 and CPE expression but PC2 immunoreactivity was markedly lower in the GK islets. The processing of the PC2-dependent substrate chromogranin A was reduced as evidenced by the appearance of intermediates. No differences were seen in the biosynthesis and post-translational modification of PC1, PC2 or CPE following incubation of islets in 16.7 mM glucose, but incubation in 3.3 mM glucose resulted in decreased PC2 biosynthesis in the GK islets. The rates of biosynthesis, processing and secretion of newly synthesized (pro)insulin were comparable. Circulating insulin immunoreactivity in both Wistar and GK rats was predominantly insulin 1 and 2 in the expected ratios with no (pro)insulin evident. Thus, the marked changes in islet morphology and PC2 expression did not impact the rate or extent of proinsulin processing either in vitro or in vivo in this experimental model.
Article
Insulin, like most secreted proteins, enters the secretory pathway at the level of the endoplasmic reticulum (ER), being directed there by the hydrophobic leader (signal) sequence corresponding to the first 16 amino acids of the preproinsulin structure. Once in the lumen of the ER, the three disulphide bonds are formed and the leader sequence is removed by signal peptidases to form proinsulin. Some of the fundamentals of the regulation of the expression of these proteins have been elucidated, both in the case of proinsulin conversion and for other prohormones that may undergo more complex differential processing. Nascent secretory granules are characteristically membrane-bounded spherical organelles enveloping an electron dense core. Proteins travel between compartments via vesicular intermediates that bud from the donor compartment and fuse with the acceptor compartment. Formation of vesicles is controlled by a complex molecular machinery, many aspects of which are common to numerous intracellular transport steps. Major advances in identifying and characterizing the components involved have come from two complementary approaches; the development of cell-free transport assays, on the one hand, and yeast genetics, on the other. The finding that a number of genes defining secretory (sec) mutants in yeast represent proteins involved in constitutive and regulated secretion in mammalian cells illustrates the high degree of conservation of many underlying principles of vesicular transport.
Article
Full-text available
A transplantable insulinoma was developed in inbred albino rats of the NEDH strain. The original tumor, 1 cm in diameter, was removed from the pancreas of a male parabiont 566 days folowing 1000 rads (10J/kg) of total body x-irradiation. The time required for implanted fragments to grow to 0.5-1.5 cm in diameter decreased from 5-8 months in the first generation to 2-5 months in the seventh generation. Successful transplantation in male animals followed for 4 or more months after transplantation was significantly greater than in female animals followed for a similar period of time (96% versus 69%). Light and electron microscopy revealed that the tumors consisted predominantly of well-granulated beta cells. Ultrastructural studies also showed small numbers of D-cells. Tumor extracts contained an average of 223 units of immunoreactive insulin and 25.9 mug of immunoreactive somato-statin per gram wet weight of tissue. Tumors generally produced increasingly profound hypoglycemia within 2-4 months following transplantation, with plasma glucose levels frequently falling to 40 mg/100 ml or lower prior to death. Removal of tumors from chronically hypoglycemic animals resulted in transient rebound hyperglycemia with plasma glucose levels above 300mg/100 ml within the first 24 hr and a gradual decline to normal levels of 129 mg/100ml in 2-4 days. These observations correlated with findings of marked atropy and degranulation of the beta cells in the pancreata of tumor-bearing animals, and with gradual return of normal light microscopic morphology following tumor removal.
Article
Full-text available
We used the polymerase chain reaction to identify a mouse testis cDNA that represented another member of a growing class of mammalian endoproteases involved in the processing of precursor proteins. This cDNA encoded a 655-residue protein, designated PC4, containing a bacterial subtilisin-like catalytic domain closely related to those of the recently characterized precursor-processing endoproteases, furin, PC1/PC3, PC2, and Kex2. Within this domain, the amino acid sequence of PC4 was 70, 58, 55, and 45% identical with those of mouse furin, mouse PC1/PC3, mouse PC2, and yeast Kex2, respectively. Northern blot analysis indicated that the PC4 mRNA was detectable only in the testes after the 20th day of postnatal development. Moreover, this message was mainly expressed in the round spermatids. These data suggest that PC4 represents a prime candidate for a precursor-processing endoprotease in the testicular germ cells and that its gene expression is regulated during spermatogenesis.
Article
Full-text available
Enzymological studies have implicated two Ca(2+)-dependent endopeptidases in the conversion of proinsulin to insulin; a type 1 activity which cleaves on the C-terminal side of Arg31-Arg32 and a type 2 activity which cleaves C-terminally to Lys64-Arg65 in the proinsulin sequence. The possibility that these enzymes are related to the recently discovered family of mammalian subtilisin-like gene products (furin, PC2, and PC3) and the yeast propheromone-converting enzyme (KEX-2), was investigated. Degenerate oligonucleotide primers flanking the putative catalytic domain within this gene family were used in a polymerase chain reaction to amplify related sequences from rat insulinoma cDNA. One major product of 700 base pairs was obtained which was greater than 99% identical to the corresponding rat PC2 sequence. This cDNA was subcloned into the bacterial expression vector pGEX-3X to generate a recombinant protein for antibody production. Western blot analysis showed the immunoreactivity was prominent in neuroendocrine tissues as a 65-kDa protein. It was concentrated in secretory granule-enriched fractions of insulinoma tissue, where it was present as a readily solubilized monomeric protein. Deglycosylation studies using endoglycosidase H and N-glycanase showed that the 65-kDa protein was comprised of approximately 9% carbohydrate, consistent with the presence of three consensus sequences for N-linked glycosylation in rat PC2. The immunoreactivity co-eluted with the type 2 proinsulin endopeptidase on gel filtration and ion-exchange chromatography and the antisera specifically immunoprecipitated type 2 activity from insulin granule extracts. N-terminal sequence analysis of the immunoreactive protein gave two sequences which corresponded to residues 109-112 and 112-119 of rat PC2. This indicated that posttranslational processing of PC2 itself occurs C-terminally to basic amino acids to produce the mature enzyme. It is concluded that PC2 is the type 2 endopeptidase involved in proinsulin conversion. Localization of PC2 immunoreactivity to other tissues of the diffuse neuroendocrine system suggests that the type 2 endopeptidase also functions in the processing of precursor forms of other prohormones and polypeptide neurotransmitters.
Article
A protein determination method which involves the binding of Coomassie Brilliant Blue G-250 to protein is described. The binding of the dye to protein causes a shift in the absorption maximum of the dye from 465 to 595 nm, and it is the increase in absorption at 595 nm which is monitored. This assay is very reproducible and rapid with the dye binding process virtually complete in approximately 2 min with good color stability for 1 hr. There is little or no interference from cations such as sodium or potassium nor from carbohydrates such as sucrose. A small amount of color is developed in the presence of strongly alkaline buffering agents, but the assay may be run accurately by the use of proper buffer controls. The only components found to give excessive interfering color in the assay are relatively large amounts of detergents such as sodium dodecyl sulfate, Triton X-100, and commercial glassware detergents. Interference by small amounts of detergent may be eliminated by the use of proper controls.
Article
Specific antisera against synthetic Fragments of the endoproteases, PC1 and PC2, were used to characterize these proteins. In one-dimensional immunoblots these antisera labelled components of 85 kDa for PC1 and of 70 kDa for PC2 in purified bovine chromaffin granules and anterior and posterior pituitary of ox and rat. In membranes of bovine chromaffin granules glycoprotein H was identified as the major PC2 immunoreactive spot. A major part of these endoproteases appeared membrane bound.
Article
The methodology for the measurement of total alkaline phosphatase activity in human serum is discussed and is offered for criticism by the world community of users. The method is not regarded as necessarily being the final version. Detailed specifications of the substrate and buffer preparations are given, with the object to minimize differences in reagents from batch to batch. The method has been designed specifically to provide reproducible final reaction conditions over long periods of time: Buffer: 2 amino 2 methyl 1 propanol, 0.8 M, pH 10.30 ± 0.02 at 30°C; Substrate: p nitrophenylphosphate, 15 mM; Temperature: 30.00 ± 0.10°C; Magnesium: 100 mM; Sample dilution: 1/30 (one volume of sample is added to 27 volumes of buffer, and the reaction is initiated with two volumes of substrate to give a final 30 fold dilution); and Wavelength: 402.5 nm. The final reaction conditions were determined by using the AP of a twice frozen filtered pool of human serum. The major iso enzymes of this pool originated from liver and/or bone. For the manual procedure the coefficient of variation (CV) for nine separate pools, each measured for at least 120 days, ranged from 3.8 to 5.2%. The mean CV was 4.6% at activities between 65 to 150 U/l at an average outlier frequency of 0.2%. Reference values (normal limits) for healthy persons are given. Sera from 258 adults (blood donors, 20 to 60 yr old) had AP activities ranging from 7 to 127 U/l. Sera from young people are two to four fold as active as that of adults. Some details are given concerning the changes in AP activity which occur during collecting, handling and storage of the specimen. (Hindriks - Groningen)
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.
Article
A protein determination method which involves the binding of Coomassie Brilliant Blue G-250 to protein is described. The binding of the dye to protein causes a shift in the absorption maximum of the dye from 465 to 595 nm, and it is the increase in absorption at 595 nm which is monitored. This assay is very reproducible and rapid with the dye binding process virtually complete in approximately 2 min with good color stability for 1 hr. There is little or no interference from cations such as sodium or potassium nor from carbohydrates such as sucrose. A small amount of color is developed in the presence of strongly alkaline buffering agents, but the assay may be run accurately by the use of proper buffer controls. The only components found to give excessive interfering color in the assay are relatively large amounts of detergents such as sodium dodecyl sulfate, Triton X-100, and commercial glassware detergents. Interference by small amounts of detergent may be eliminated by the use of proper controls.
Article
A cDNA encoding a novel human subtilisin-like protease was identified by a polymerase chain reaction (PCR) methodology. PCR primers were designed to be specific for the subfamily of eukaryotic subtilisin-like proteases with specificity for paried basic amino acid residue processing motifs. The gene encoding this protease, designated PACE4, also encoded a smaller subtilisin-related polypeptide derived by alternate mRNA splicing. The deduced PACE4 protein sequence contained a number of interesting features not present in other family members, including an extended signal peptide region, and a relatively large carboxyl-terminal cysteine-rich region with no obvious membrane anchor sequence. As with the fur gene product, the tissue distribution of PACE4 was widespread, with comparatively higher levels in the liver. An additional relationship to the fur gene product was shown by chromosomal localization studies. The close proximity of the fur and PACE4 genes on chromosome 15 suggests that these genes probably evolved from a common ancestor by gene duplication.
Article
The intracellular distribution and molecular heterogeneity of carboxypeptidase H was studied in rat insulinoma tissue and isolated islets of Langerhans by a combination of immunohistochemical, ultrastructural, subcellular fractionation, and immunoblotting analyses. Immunofluorescence microscopy of islets demonstrated the presence of carboxypeptidase H in both insulin-containing B cells and glucagon-containing A cells. Quantitative ultrastructural analyses of islet B cells indicated that the enzyme was concentrated in mature insulin secretory granules, clathrin-coated condensing granules, and to a lesser extent the Golgi apparatus. Carboxypeptidase H activity was localized principally to secretory granule subfractions of insulinoma tissue, where it was present for the major part (70%) as a form which is readily solubilizable at pH values prevailing in the granule interior (5.5). This species migrated as a diffuse band of 53-57 kilodaltons (kDa) on immunoblot analysis using antisera raised against the purified native enzyme. In contrast, the insoluble form which was associated with the granule membrane at pH 5.5, migrated as a relatively compact band of 55-57 kDa. Carboxypeptidase H activity was also present in subcellular fractions which contained Golgi membranes together with elements of the endoplasmic reticulum, and in a low density secretory granule fraction which may represent immature granules. The enzyme in these compartments, like the granule membrane species, migrated as a compact 55-57 kDa band on immunoblots. Two-dimensional electrophoretic immunoblot analysis of secretory granules suggested that both membrane and soluble forms of the enzyme were glycoproteins and that the terminal glycosylation was similar in both instances. Antiserum raised against the deduced C-terminal 11 amino acids of the cloned carboxypeptidase H sequence recognized the 55-57 kDa membrane component in granules but did not react with the 53-57 kDa soluble species. A major difference between the soluble and membrane forms therefore appears to be a structural modification or proteolytic removal of the C-terminal domain in the trans-Golgi or early secretory granule compartment. The concept that proteolysis is involved is further supported by the observation that the relative proportion of the high and low mol wt forms of the enzyme in different subcellular fractions correlated with that of proinsulin and insulin, respectively. The membrane association of the 55-57 kDa form of carboxypeptidase H is disrupted at pH values of 9 and is dependent on ionic strength. This further suggests that the C-terminus of the protein may have an important role in the sorting or concentration of the enzyme in vesicular elements of the regulated pathway of secretion.