ArticlePDF Available

Receptor for insulin and insulin-like growth factor-I can form hybrid dimers

Authors:

Abstract

We have demonstrated the formation of hybrid insulin/insulin-like growth factor-I(IGF-I) receptors in transfected rodent fibroblasts, which overexpress human receptors, by examining reactivity with species- and receptor-specific monoclonal antibodies. In NIH 3T3 and Rat 1 fibroblasts, endogenous IGF-I receptors were unreactive with anti-(human insulin receptor)monoclonal antibodies (47-9, 25-49, 83-14, 83-7, 18-44). However, in transfected cells expressing high levels of insulin receptors, 60-80% of high-affinity IGF-I receptors reacted with these antibodies, as assessed either by inhibition of ligand binding in intact cells or by precipitation of solubilized receptors. Conversely, endogenous insulin receptors in NIH 3T3 cells were unreactive with anti-(IGF-I receptor) antibodies alpha IR-3 and 16-13. However, approx. 50% of high-affinity insulin receptors reacted with these antibodies in cells expressing high levels of human IGF-I receptors. The hybrid receptors in transfected cells bound insulin or IGF-I with high affinity. However, responses to these ligands were asymmetrical, in that binding of IGF-I inhibited subsequent binding of insulin, but prior binding of insulin did not affect the affinity for IGF-I. The existence of hybrid receptors in normal tissues could have important implications for metabolic regulation by insulin and IGF-I.
Biochem.
J.
(1990)
270,
383-390
(Printed
in
Great
Britain)
Receptors
for
insulin
and
insulin-like
growth
factor-I
can
form
hybrid
dimers
Characterisation
of
hybrid
receptors
in
transfected
cells
Maria
A.
SOOS,*
Jonathan
WHITTAKER,t
Reiner
LAMMERS,t
Axel
ULLRICHt
and
Kenneth
SIDDLE*§
*
Department
of
Clinical
Biochemistry,
University
of
Cambridge,
Addenbrookes
Hospital,
Hills
Road,
Cambridge
CB2
2QR,
U.K.,
tDivision
of
Endocrinology,
Department
of
Medicine,
SUNY
at
Stony
Brook,
Stony
Brook,
NY
11794-8154,
U.S.A.,
and
I
Department
of
Molecular
Biology,
Max-Planck
Institut
fur
Biochemie,
8033
Martinsried,
Federal
Republic
of
Germany
We
have
demonstrated
the
formation
of
hybrid
insulin/insulin-like
growth
factor-I(IGF-I)
receptors
in
transfected
rodent
fibroblasts,
which
overexpress
human
receptors,
by
examining
reactivity
with
species-
and
receptor-specific
monoclonal
antibodies.
In
NIH
3T3
and
Rat
1
fibroblasts,
endogenous
IGF-I
receptors
were
unreactive
with
anti-(human
insulin
receptor)monoclonal
antibodies
(47-9,
25-49,
83-14,
83-7,
18-44).
However,
in
transfected
cells
expressing
high
levels
of
insulin
receptors,
60-80
%
of
high-affinity
IGF-I
receptors
reacted
with
these
antibodies,
as
assessed
either
by
inhibition
of
ligand
binding
in
intact
cells
or
by
precipitation
of
solubilized
receptors.
Conversely,
endogenous
insulin
receptors
in
NIH
3T3
cells
were
unreactive
with
anti-(IGF-I
receptor)
antibodies
aIR-3
and
16-13.
However,
approx.
50%
of
high-affinity
insulin
receptors
reacted
with
these
antibodies
in
cells
expressing
high
levels
of
human
IGF-I
receptors.
The
hybrid
receptors
in
transfected
cells
bound
insulin
or
IGF-I
with
high
affinity.
However,
responses
to
these
ligands
were
asymmetrical,
in
that
binding
of
IGF-I
inhibited
subsequent
binding
of
insulin,
but
prior
binding
of
insulin
did
not
affect
the
affinity
for
IGF-I.
The
existence
of
hybrid
receptors
in
normal
tissues
could
have
important
implications
for
metabolic
regulation
by
insulin
and
IGF-I.
INTRODUCTION
Insulin
and
insulin-like
growth
factor-I
(IGF-I)
exert
their
biological
effects
by
binding
to
specific
plasma
membrane
receptors
which
show
many
similarities
of
structure
and
function.
Each
receptor
is
synthesized
initially
as
a
proreceptor
polypeptide
which
is
processed
by
glycosylation
and
proteolytic
cleavage
to
give
a-
and
/3-subunits,
the
mature
and
functional
receptors
being
symmetrical
structures
of
two
disulphide-linked
(a,f)
units
(reviewed
in
Czech,
1985;
Rechler
&
Nissley,
1985;
Duronio
&
Jacobs,
1988;
Yarden
&
Ullrich,
1988).
The
a-subunit
is
extracellular
and
contains
the
ligand
binding
site.
The
transmembrane
fl-subunit
possesses
ligand-stimulated
tyrosine
kinase
activity
which
appears
to
be
an
essential
component
of
the
signalling
pathways
mediating
hormone
action
(reviewed
in
Rosen,
1987;
Zick,
1989).
The
cloning
and
sequencing
of
cDNAs
coding
for
the
respective
proreceptors
has
confirmed
that
receptors
for
insulin
and
IGF-I
are
the
products
of
separate
genes,
with
substantial
similarity
of
amino
acid
sequence
(Ullrich
et
al.,
1985,1986).
There
has
been
debate
as
to
the
basis
of
the
overlapping
but
apparently
distinct
biological
effects
of
insulin
and
IGF-I
(Froesch
et
al.,
1985).
These
might
be
a
consequence
of
inherently
different
signalling
capacities
of
the
respective
receptors,
or
of
differences
in
receptor
distribution
among
tissues
with
different
capacities
for
metabolic
response
to
a
given
signal.
Insulin
and
IGF-I
induce
the
phosphorylation
of
common
endogenous
substrates
for
the
receptor
tyrosine
protein
kinase
(Izumi
et
al.,
1987;
Kadowaki
et
al.,
1987).
No
differences
in
inherent
signalling
capacity
were
apparent
when
human
insulin
and
IGF-I
receptors
were
expressed
in
Chinese
hamster
ovary
cells
following
cDNA
transfection
(Steele-Perkins
et
al.,
1988).
However,
expression
of
normal
and
chimaeric
receptors
in
NIH
3T3
fibroblasts
revealed
differences
in
signalling
potential
of
the
insulin-
and
IGF-I-
receptor
cytoplasmic
domains,
at
least
for
stimulation
of
DNA
synthesis
(Lammers
et
al.,
1989).
It
has
recently
been
reported
that
in
human
tissues
and
cell
lines
expressing
both
insulin
and
IGF-I
receptors,
a
proportion
of
receptors
exists
as
hybrid
(a/Ja'/3')
structures
(Soos
&
Siddle,
1989;
Moxham
et
al.,
1989).
The
existence
of
hybrid
receptors
could
have
important
functional
consequences
if
such
structures
are
capable
of
binding
and
responding
to
both
insulin
and
IGF-I.
However,
other
work
has
suggested
the
existence
of
two
distinct
IGF-I
receptor
polypeptides
as
a
possible
basis
for
signalling
by
insulin
via
the
IGF-I
receptor
(Garofalo
&
Rosen,
1989;
Alexandrides
&
Smith,
1989).
The
availability
of
rodent
cell
lines
transfected
with
human
insulin-
or
IGF-I-receptor
cDNA
(Whittaker
et
al.,
1987;
McClain
et
al.,
1987;
Lammers
et
al.,
1989)
has
allowed
us
to
examine
the
consequences
for
receptor
assembly
when
one
type
of
receptor
is
overexpressed
relative
to
the
other.
We
have
demonstrated
conclusively
the
ability
of
insulin-
and
IGF-I-receptor
subunits
to
assemble
as
interspecies
hybrids
in
these
cells,
by
using
species-
and
receptor-
specific
monoclonal
antibodies.
We
have
also
been
able
to
Vol.
270
Abbreviations
used:
IGF-I,
insulin-like
growth
factor-I;
WGA,
wheat-germ
agglutin;
PEG,
poly(ethylene
glycol);
DMEM,
Dubecco's
modified
Eagle's
medium;
PBS,
phosphate-buffered
saline;
k.i.u.,
kallikrein-inactivating
unit;
rIR/hIGFR
and
hIR/rIGFR,
complexes
between
rodent
insulin
receptor
and
human
IGF-I
receptor,
and
human
insulin
receptor
and
rodent
IGF-I
receptor,
respectively.
§
To
whom
correspondence
should
be
addressed.
383
M.
A.
Soos
and
others
examine
some
of
the
ligand-binding
properties
of
these
hybrid
receptors,
and
to
show
that
both
insulin
and
IGF-I
are
bound
with
high
affinity.
EXPERIMENTAL
Materials
Bovine
insulin
(for
displacement
studies)
was
from
Sigma
Chemical
Co.,
Poole,
Dorset,
U.K.,
and
highly
purified
des-
amido-free
bovine
insulin
(for
iodination)
was
a
gift
from
Dr.
D.
Brandenburg,
Deutsches
Wollforschungsinstitut,
Aachen,
Germany. Recombinant
human
IGF-I
was
generously
provided
by
Ciba-Geigy,
Basle,
Switzerland.
Proteinase
inhibitors,
BSA,
methotrexate,
Sephadex
G-50
and
wheat-germ-agglutinin-
Sepharose
(WGA-Sepharose)
were
from
Sigma,
poly(ethylene
glycol)
(PEG)
6000
was
from
BDH
Chemicals,
Dagenham,
Essex,
U.K.
and
Na1251I
(IMS
30)
was
from
Amersham
Inter-
national,
Aylesbury,
Bucks.,
U.K.
Hydroxyapatite
was
pur-
chased
from
Bio-Rad
Laboratories,
Watford,
Herts.,
U.K.
All
tissue
culture
reagents
and
Geneticin
(G418
sulphate)
were
from
Gibco
Ltd.,
Paisley,
Scotland,
U.K.
Sheep
anti-(mouse
IgG)
antibodies
were
coupled
to
aminocellulose
to
obtain
immuno-
adsorbents
as
described
previously
(Soos
et
al.,
1986).
Radioiodinations
Mono-1251-insulin
with
a
specific
radioactivity
of
100-200
uCi/,#g
was
prepared
from
highly
purified
bovine
insulin
as
described
by
Linde
et
al.
(1981).
IGF-I
was
iodinated
to
a
specific
activity
of
50-150,1Ci/1sg
using
a
stoichiometric
chloramine-T
method
(Roth,
1975)
and
was
purified
by
gel
filtration
on
Sephadex
G-50
to
separate
125I-IGF-I
from
free
[125
]iodide.
Antibodies
Monoclonal
antibodies
specific
for
human
insulin
receptors
(Soos
et
al.,
1986)
were
purified
from
ascites
fluids
by
precipitation
with
(NH4)2SO4
followed
by
chromatography
on
hydroxyapatite
(Stanker
et
al.,
1985).
Monoclonal
antibody
16-13,
specific
for
human
IGF-I
receptors,
was
obtained
following
fusion
of
NSO
myeloma
cells
with
spleen
cells
from
a
mouse
immunized
with
mouse
fibroblasts
overexpressing
human
IGF-I
receptors
(IGF-I-R/3T3
cells)
using
standard
techniques
(Galfre
&
Milstein,
1981).
Monoclonal
antibody
aIR-3
(also
specific
for
human
IGF-I
receptors)
was
kindly
donated
by
Dr.
Steven
Jacobs
(Wellcome
Research
Laboratories,
Research
Triangle
Park,
NC,
U.S.A.).
Cell
culture
All
the
cell
lines
were
grown
in
medium
with
10
%
(v/v)
foetal
calf
serum,
penicillin
(100
units/ml)
and
streptomycin
(100
jug/ml).
Rat
1
cells
(untransfected)
(McClain
et
al.,
1987)
were
grown
in
Dulbecco's
modified
Eagle's
medium
(DMEM).
The
cell
lines
NIH
3T3
NEO
(mock-transfected),
NIH
3T3
HIR3.5
(Whittaker
et
al.,
1987)
and
IGF-I-R/3T3
(Lammers
et
al.,
1989)
were
routinely
grown
in
DMEM
plus
0.4
mg
of
Geneticin/ml.
HIRc-B
cells
(McClain
et
al.,
1987)
were
grown
in
F12/DMEM
(1:1,
v/v)
plus
500
nM-methotrexate
and
0.4
mg
of
Geneticin/ml.
Cultures
were
maintained
in
a
humidified
atmosphere
of
air/CO2
(19:
1)
at
37
'C.
For
the
1251-hormone
binding
studies,
cells
were
seeded
on
to
24-well
tissue
culture
plates
at
2
x
104
cells
per
well.
They
were
cultured
for
2
days
to
approximately
half
confluence
for
1251-insulin
binding
to
NIH
3T3
HIR3.5
and
HIRc-B
cells
and
for
125I-IGF-I
binding
to
IGF-I-R/3T3
cells,
and
for
3
days
to
confluence
for
all
other
binding
studies.
Receptor
preparations
Receptors
from
NIH
3T3
NEO,
NIH
3T3
HIR3.5
and
IGF-
I-R/3T3
cell
lines
were
partially
purified
from
approx.
3
x
108
cells.
After
washing
with
phosphate-buffered
saline
(PBS;
150
mM-NaCl/10
mM-sodium
phosphate,
pH
7.4),
cells
were
removed
from
flasks
by
scraping
into
PBS
containing
proteinase
inhibitors
[0.2
mM-phenylmethanesulphonyl
fluoride,
0.4
mg
of
benzamidine/ml,
1
4g
of
leupeptin/ml,
1
/g
of
pepstatin/ml,
1
4g
of
antipain/ml,
200
kallikrein-inactivating
units
(k.i.u.)
of
aprotinin/ml
and
0.2
mg
of
bacitracin/mll.
The
cell
pellet
after
centrifugation
(150
g,
7
min)
was
solubilized
for
1
h
at
4
°C
in
20
ml
of
0.05
M-Hepes,
pH
7.4,
containing
the
proteinase
in-
hibitors
and
1
%
(v/v)
Triton
X-100.
The
supernatant
after
centrifugation
(50000
g
for
30
min,
4
°C)
was
diluted
1:
1.5
with
0.05
M-Hepes,
pH
7.4,
containing
0.15
M-NaCl,
0.025
M-MgCl2
and
proteinase
inhibitors,
and
mixed
for
16-20
h
at
4
°C
with
1
ml
of
WGA-Sepharose.
The
WGA-Sepharose
was
then
washed
with
10-20
ml
of
0.05
M-Hepes,
pH
7.4,
containing
0.15
M-NaCl
and
0.1
%
Triton
X-100
before
elution
with
0.5
M-N-acetyl-
glucosamine
in
the
same
buffer.
The
single
protein
peak
was
pooled
for
use
in
assays.
Solubilized
Rat
1
and
HIRc-B
receptors
were
prepared
by
solubilizing
approx.
107
cells
in
1.5
ml
of
0.05
M-Hepes,-pH
7.4,
containing
the
proteinase
inhibitors
and
1
%
Triton
X-100
as
described
above.
This
Triton
X-100
extract
was
used
without
further
purification.
1251-Hormone
binding
assays
Cells
were
washed
twice
with
PBS
before
addition
of
insulin,
IGF-I
or
antibody
in
0.25
ml
of
modified
DMEM
(containing
25
mM-Hepes,
pH
7.8,
and
7
mM-sodium
bicarbonate)
supplemented
with
1
mg
of
BSA/ml
and
250
k.i.u.
of
aprotinin/ml.
After
30
min
at
4
°C,
0.05
ml
of
1251-insulin
or
125I.
IGF-I
(approx.
30000
d.p.m.)
was
added.
After
a
further
4
h,
cells
were
washed
twice
with
ice-cold
PBS
and
solubilized
with
0.03
%
SDS
for
determination
of
radioactivity.
Non-specific
binding
was
determined
in
the
presence
of
I
#sM-insulin
or
0.1
zM-
IGF-I
as
appropriate.
Binding
to
solubilized
receptors
was
performed
as
described
previously
(Soos
&
Siddle,
1989),
except
that
receptors
were
incubated
with
1251I-insulin
or
1251I-IGF-I
(approx.
30000
d.p.m.
in
a
total
volume
of
0.25
ml),
together
with
unlabelled
hormone,
for
18
h
at
4
'C.
Receptor-bound
radioactivity
was
determined
by
precipitation
with
PEG
6000
(Baron
&
Sonksen,
1982).
Non-
specific
binding
was
determined
in
the
presence
of
1
aM-insulin
or
0.1
,M-IGF-I
as
appropriate.
The
concentration
of
Triton
X-100
in
all
soluble
binding
assays
was
maintained
at
0.05
%.
Co-precipitation
of
receptor-1251-hormone
complexes
Assays
were
performed
as
described
in
Soos
et
al.
(1986)
by
preincubating
solubilized
receptors
with
1251-insulin
or
125I-IGF-I
(approx.
30000
d.p.m.
in
a
total
volume
of
0.1
ml)
for 18
h
at
4
'C
before
addition
of
0.1
ml
of
antibody
for
a
further
6
h.
To
establish
the
specificity
of
binding
of
labelled
hormones,
unlabelled
hormones
were
included
in
the
first
incubation.
Antibody-bound
radioactivity
was
determined
using
a
sheep
anti-(mouse
IgG)
adsorbent
as
previously
described
(Soos
et
al.,
1986).
Total
receptor-bound
radioactivity
was
measured
by
precipitation
with
PEG
6000
(Baron
&
Sonksen,
1982).
RESULTS
Relative
levels
of
insulin
and
IGF-I
receptors
in
cell
lines
Estimates
of
numbers
of
receptors
in
the
cell
lines
studied
are
summarized
in
Table
1.
These
values
take
no
account
of
whether
ligand
binding
is
to
homomeric
or
hybrid
receptors.
The
NIH
1990
384
Insulin/insulin-like
growth
factor-I
receptor
hybrids
Table
1.
Numbers
of
insulin
and
IGF-I-
receptors
in
transfected
cell
lines
Values
indicate
receptor
numbers
per
cell.
Data
are
taken
from
*Hoffman
et
al.
(1989),
t
McClain
et
al.
(1987),
Maegawa
et
al.
(1988)
and
t
Lammers
et
al.
(1989).
Rodent
receptors
were
deter-
mined
in
corresponding
untransfected
cells,
but
levels
were
very
similar
in
transfected
cells
where
measured.
l0-'
x
Receptor
no.
Rodent
receptors
Human
receptors
(untransfected
cells)
(transfected
cells)
Cell
line
Insulin
IGF-I
Insulin
IGF-I
NIH
3T3
HIR3.5*
<
3
180
3000
-
HIRc-Bt
1.7
120
1250
-
IGF-I-R/3T3T
5.4
3.4
-
1311
3T3
and
Rat
1
fibroblasts
transfected
with
insulin
receptor
cDNA
possess
a
moderately
high
number
of
endogenous
rodent
IGF-I
receptors
but
relatively
few
insulin
receptors.
The
NIH
3T3
subline
used
for
transfection
with
IGF-I
receptor
cDNA
clearly
differs
from
that
used
for
insulin
receptor
transfection
in
having
low
levels
of
endogenous
receptors
for
both
insulin
and
IGF-I.
The
levels
of
human
receptors
in
transfected
cells
greatly
exceeded
those
of
endogenous
receptors
in
all
cases.
The
NIH
3T3
HIR3.5
cell
line
(Ebina
et
al.,
1985;
Whittaker
et
al.,
1987)
was
transfected
with
a
cDNA
sequence
encoding
a
receptor
with
an
additional
12-amino-acid
segment
compared
with
that
ex-
pressed
in
the
Rat
1
HIRc-B
cells
(Ullrich
et
al.,
1985;
McClain
et
al.,
1987).
There
is
no
evidence
in
any
of
these
cells
that
type
II
IGF
receptors
(Froesch
et
al.,
1985)
contribute
significantly
to
125I-IGF-I
binding.
Thus
125I-IGF-I
binding
was
effectively
displaced
by
insulin,
albeit
at
high
concentrations,
as
expected
for
reactivity
with
'classical'
IGF-I
receptors.
Further,
type
II
receptors,
because
of
their
totally
different
structure
compared
with
insulin-
and
IGF-I-receptors,
would
not
be
expected
to
react
with
any
of
the
antibodies
used
in
this
study.
Binding
studies
with
intact
cells
overexpressing
human
insulin
receptors
Monoclonal
anti-(insulin
receptor)
antibodies
47-9
and
25-49
were
tested
for
effects
on
ligand
binding
to
normal
fibroblasts
(NIH
3T3,
Rat
1)
and
to
cells
transfected
with
human
insulin
receptor
cDNA.
These
antibodies
recognize
distinct
epitopes
on
the
human
insulin
receptor
and
inhibit
insulin
binding
(Soos
et
al.,
1986).
They
do
not
react
with rodent
insulin
receptors
(Soos
et
al.,
1986)
nor
with
human
IGF-I
receptors
in
IGF-I-R/3T3
cells
(results
not
shown).
As
expected,
therefore,
these
antibodies
had
no
effect
on
the
binding
of
IGF-I
(Fig.
1)
or
insulin
(results
not
shown)
to
mock-transfected
NIH
3T3
NEO
or
untransfected
Rat
1
cells.
The
IGF-I
binding
sites
behaved
as
a
homogeneous
population,
in
showing
inhibition
of
tracer
binding
by
low
concentrations
of
unlabelled
IGF-I
[concn.
giving
50
%
inhibition
of
binding
(IC50)
0.5-0.6
nM],
but
only
by
high
concentrations
of
insulin
(IC50
400-1000
nm,
Fig.
1).
The
level
of
IGF-I
binding
to
NIH
3T3
HIR3.5
and
HIRc-B
cells
was
similar
to
that
in
the
corresponding
untransfected
cell
lines.
However,
in
the
cells
expressing
human
insulin
receptors,
antibodies
47-9
and
25-49
inhibited
not
only
the
binding
of
insulin
(Figs.
2c
and
3c)
but
also
the
binding
of
IGF-I
(Figs.
2d
and
3d).
Maximum
inhibition
of
insulin
binding
was
95-98
%
and
of
IGF-I
binding
it
was
70-800%.
The
antibody
concentration-dependence
of
binding
inhibition
was
similar
for
both
ligands
with
antibody
25-49,
but
antibody
47-9
was
consistently
approximately
2-fold
less
potent
with
IGF-I
than
-
0
0
0
C
._
c
\
~~b
10-11
10-10
10-9
10-8
10-7
10-6
100-
80-
60-
40-
20-
10-11
10-10
10-9
10-8
10-i
10-6
Concentration
(M)
Fig.
1.
Inhibition
of
125I-IGF-I
binding
to
NIH
3T3
NEO
cells
Binding
of
labelled
IGF-I
to
cells
was
measured
in
the
presence
of
the
indicated
concentrations
of
unlabelled
insulin
(O),
IGF-I
(0),
47-9
(A\)
or
25-49
(A)
as
described
in
the
Experimental
section.
Data
points
are
the
means
of
duplicate
incubations
within
a
representative
experiment.
Specific
binding
is
expressed
as
a
per-
centage
of
that
in
the
absence
of
unlabelled
ligand.
Total
cell-bound
'25I-IGF-I
was
2.7
%
and
non-specific
binding
was
0.2
%
of
the
total
radioactivity.
Very
similar
results
were
obtained
with
Rat
1
fibroblasts.
'25I-IGF-I
binding
was
inhibited
by
low
concentrations
of
unlabelled
IGF-I
(IC50
0.5
nM)
and
high
concentrations
of
insulin
(IC50
400
nM),
whereas
antibodies
47-9
and
25-49
were
without
effect.
with
insulin.
Monovalent
Fab
fragments
of
antibodies
47-9
and
25-49
(10-100
nM)
inhibited
binding
of
both
IGF-I
and
insulin
to
the
same
maximum
extent
as
bivalent
antibodies.
Anti-(insulin
receptor)
antibodies
which
did
not
inhibit
insulin
binding
(e.g.
83-7)
were
without
effect
on
IGF-I
binding
(results
not
shown).
In
both
transfected
cell
types
the
binding
of
'25I-IGF-I
appeared
to
be
heterogeneous
in
terms
of
inhibition
by
unlabelled
insulin
(Figs.
2b
and
3b).
Approx.
75
%
of
IGF-I
binding
was
inhibited
by
low
concentrations
of
IGF-I
(IC50
0.5-1
nM)
but
only
by
high
concentrations
of
insulin
(IC50
approx.
800
nM).
Thus
most
of
the
IGF-I
binding
sites
still
showed
the
properties
expected
of
high-affinity
IGF-I
receptors,
as
in
the
untransfected
cells.
However,
a
fraction
(approx.
25
%)
of
IGF-I
binding
was
inhibited
by
low
insulin
concentrations
(IC50
approx.
0.2
nM)
and
was
assumed
to
reflect
low-affinity
binding
of
IGF-I
to
the
large
excess
of
insulin
receptors.
There
was
a
significant
difference
in
the
reactivity
of
IGF-I
with
insulin
receptors
in
the
two
different
transfected
cell
lines.
Under
the
conditions
of
these
experiments,
unlabelled
IGF-I
was
considerably
more
potent
at
inhibiting
tracer
binding
in
HIRc-B
cells
(IC50
approx.
20
nm,
Fig.
3a)
than
in
NIH
3T3
HIR3.5
cells
(IC50
>
100
nm,
Fig.
2a),
although
displacement
by
unlabelled
Vol.
270
385
M.
A.
Soos
and
others
(a)
0
b
0
0
%.I
--%
10-9
O
-___
100'
80'-
60-
40-
20
-
10-8
10-7
l'o-6
(c)
100
80
-
60-
40-
20-
0O
(b)
b
10-7
10-6
(d)
10-11
10-11
Concentration
(M)
Fig.
2.
Inhibition
of
'25I-hormone
binding
to
NIH
3T3
HIR
3.5
cells
Binding
of
labelled
insulin
(a,c)
and
IGF-I
(b,d)
to
cells
was
measured
in
the
presence
of
the
indicated
concentrations
of
unlabelled
insulin
(0),
IGF-I
(0),
47-9
(A)
or
25-49
(A)
as
described
in
the
Experimental
section.
The
means
+
S.E.M.
of
three
independent
experiments,
performed
in
duplicate,
are
shown.
Specific
binding
is
expressed
as
a
percentage
of
that
in
the
absence
of
unlabelled
ligand.
Total
cell-bound
radioactivity
was
36
+
2
%
('25I-insulin)
and
4.1
+
0.4
%
(125I-IGF-I),
and
non-specific
binding
was
0.23
+
0.13
%
(.251-insulin)
and
0.33
+
0.04
%
('25I-IGF-I)
of
the
total
radioactivity.
101
8
6
4
2
10-7
10-6
(c)
100
80
I
z
%%60
\t\
I-
8
\
40
I
-11
10-1°
10-9 10-8 10-7
16
20
10-11
-10
0
9
10-8
0-7
100
0
~~~~~~~~~~~(b)
0
-~~~~~~
50,
.
.
X
0
~~~~~~~~~%0
0o-
10-11
10-10
10-9
10-8
10-7
(d)
10-11
10-1Q
10-9
10-8
10-7
10-6
Concentration
(M)
Fig.
3.
Inhibition
of
l25l-hormone
binding
to
HIRc-B
cells
Binding
of
labelled
insulin
(a,c)
and
IGF-I
(b,d)
to
cells
was
measured
in
the
presence
of
the
indicated
concentrations
of
unlabelled
insulin
(0),
IGF-I
(-),
47-9
(A)
or
25-49
(A)
as
described
in
the
Experimental
section.
The
means
+
S.E.M.
of
three
independent
experiments
performed
in
duplicate
are
shown.
Specific
binding
is
expressed
as
a
percentage
of
that
in
the
absence
of
unlabelled
ligand.
Total
cell-bound
radioactivity
was
34+7.9%
(.251-insulin)
and
10.9
+
2.1
%
(125I-IGF-I),
and
non-specific
binding
was
0.16
+
0.07
%
(.251-insulin)
and
0.45
+
0.0
%
(125I-IGF-I)
of
the
total
radioactivity.
1990
100-
80
!
60
-
40
-
20-
0-1l
10-10
-
0
0
0)
~Ro
C
._
c
100-
80-
60-
40-
20-
10-11
100
0
a
U
0
4-
.n
100
c
80
60
40
20
60
10-6
386
"h
I
t
'A
-
----v
0i
6-9
i
.
8
i
.-7
10-6
i
;-lo
i
;-g
i
.-8
-1.-7
1
-6
10-11
10-10
10-9
10-8
Insulin/insulin-like
growth
factor-I
receptor
hybrids
(a)
0
-
-0
O
5
100
-
80
-
0
0
"S
'o
i
.
.
I
r
I
10-11
1
0-l°
1
0-9
1
0-8
10-7
10-6
60
-
40
-
20
-
(b)
0""
\
-
0O
_
I
Concentration
(M)
Fig.
4.
Inhibition
of
'l25l-ormone
binding
to
partially
purified
receptors
Binding
of
.25I-IGF-I
to
WGA-Sepharose-purified
NIH
3T3
HIR
3.5
receptors
(a)
and
of
.25I-insulin
to
WGA-Sepharose-purified
IGF-I-R/3T3
receptors
(b)
was
measured
in
the
presence
of
the
indicated
concentrations
of
unlabelled
insulin
(0)
or
IGF-I
(@)
as
described
in
the
Experimental
section.
Data
points
are
the
means
of
duplicate
incubations
within
a
representative
experiment.
Specific
binding
is
expressed
as
a
percentage
of
that
in
the
absence
of
unlabelled
ligand.
Total
receptor-bound
radioactivity
was
9.4
%
(a)
and
7.1
%
(b),
and
non-specific
binding
was
4
%
(a)
and
0.9%
(b).
insulin
was
very
similar
in
both
cell
types.
The
basis
of
the
apparently
higher
cross-reaction
of
IGF-I
with
human
insulin
receptors
expressed
in
Rat
1
compared
with
NIH
3T3
HIR3.
cells
is
unknown.
This
might
reflect
the
differences
in
recepto
sequence
or
differences
in
glycosylation
which
influence
binding
affinity
for
IGF-I
but
not
insulin.
The
conclusion
from
these
experiments
is
that
approx.
60-70
%
of
endogenous
rodent
IGF-I
receptors
became
reactive
with
anti-
(insulin
receptor)
specific
antibodies
when
a
10-20-fold
excess
of
human
insulin
receptors
was
co-expressed.
This
is
consistent
with
the
idea
that
a
large
fraction
of
IGF-I
binding
sites
were
in
hybrid
receptors
(a6z'fl')
in
which
binding
of
ligand
to
the
a'-
subunit
was
inhibited
by
binding
of
antibody
or
Fab
to
particular
epitopes
on
a.
In
other
respects,
including
affinity
for
IGF-I
and
insulin,
these
IGF-I
binding
sites
were
very
similar
to
IGF-I
receptors
in
untransfected
cells.
Binding
studies
with
solubilized
receptors
from
cells
overexpressing
human
insulin
receptors
Receptors
solubilized
and
partially
purified
from
transfected
cells
were
studied
in
order
to
permit
the
investigation
of
reactivity
with
antibodies
which
did
not
necessarily
inhibit
ligand
binding.
In
these
experiments,
125I-IGF-I
or
l25l-insulin
was
preincubated
with
receptor
before
determination
of
the
fraction
of
receptor-ligand
complexes
which
was
immunoprecipitable.
Solubilized
receptors
from
NIH
3T3
HIR3.5
and
HIRc-B
cells
behaved
similarly
to
those
from
the
intact
cells
in
terms
of
inhibition
of
1251I-ligand
binding
by
unlabelled
insulin
and
IGF-I.
The
different
potency
of
cross-reaction
of
IGF-I
with
insulin
receptors
from
the
two
cell
lines
was
still
apparent
after
solubilization.
However,
the
minor
component
of
1251I-IGF-I
binding
which
was
inhibitable
by
low
concentrations
of
insulin
in
intact
cells
was
not
apparent
with
solubilized
receptors
(Fig.
4a).
This
probably
reflects
the
fact
that
conditions
for
separation
of
bound
and
free
ligand
in
the
soluble
receptor
assays
(PEG
precipitation)
do
not
detect
the
low-affinity
component
of
IGF-I
binding
because
of
the
dissociation
during
precipitation
and
washing.
Binding
of
125I-IGF-I
in
this
assay
therefore
reflects
only
high-affinity
IGF-I
receptors
(IC50
0.4
nm
for
IGF-I
and
40-150
nm
for
insulin;
Fig.
4a).
Antibodies
for
three
different
insulin
receptor
epitopes
(83-7
and
83-14,
a-subunit;
18-44,
fl-subunit)
precipitated
not
only
receptor-'251-insulin
complexes
but
also
the
bulk
of
receptor-'25I-
IGF-I
complexes
from
solubilized
NIH
3T3
HIR3.5
and
HIRC-
B
cells
(Table
2).
None
of
the
antibodies
reacted
at
all
with
Table
2.
Reaction
of
antibodies
with
receptor-'251-hormone
complexes
WGA-Sepharose-purified
receptors
(NIH
3T3
HIR3.5
and
IGF-I-R/3T3
cells)
or
Triton
X-
100-solubilized
receptors
(HIRc-B)
were
preincubated
with
'25I-insulin
or
125I-IGF-I
before
addition
of
10
nM-antibody.
Total
and
immunoreactive
receptor-bound
radio-
activity
was
determined
as
described
in
the
Experimental
section.
In
the
absence
of
antibody,
total
specific
receptor-bound
'25I-insulin
was
2483
c.p.m.
(NIH
3T3
HIR3.5),
2521
c.p.m.
(HIRc-B)
and
1564
c.p.m.
(IGF-I-R/3T3),
and
total
receptor-bound
1251I-IGF-I
was
1492
c.p.m.
(NIH
3T3
HIR3.5),
1673
c.p.m.
(HIRc-B)
and
2370
c.p.m.
(IGF-I-R/3T3).
Values
are
the
means
of
duplicate
incubations
within
representative
experiments.
None
of
the
anti-
bodies
precipitated
receptors
from
control
cells
(NIH
3T3,
Rat
1)
which
do
not
express
human
insulin
or
IGF-I
receptors.
N.D.,
not
determined.
Immunoreactive
receptors
(%)
NIH
3T3
HIR3.5
HIRc-B
IGF-I-R/3T3
Antibody
Insulin
IGF-I
Insulin
IGF-I
Insulin
IGF-I
83-7
96
82
70
67
0
0
83-14
94
100
86
76
0
1
18-44
102
83
93
71
0
1
aoIR-3
0
0
0
2
44
85
16-13
0
N.D.
0
2
50
77
receptors
from
untransfected
cells
or
with
human
IGF-I
receptors
in
IGF-1-R/3T3
cells.
As
expected,
antibodies
acIR-3
and
16-13,
specific
for
distinct
epitopes
on
the
human
IGF-I
receptor,
did
not
react
with
rodent
IGF-I
receptors
in
either
untragsfected
or
transfected
cells
(Table
2).
It
was
concluded
that
a
substantial
fraction
of
rodent
IGF-I
receptors
in
transfected
cells
was
incorporated
into
hybrid
structures
which
were
then
reactive
with
the
whole
panel
of
antibodies
specific
for
human
insulin
receptors.
Binding
studies
with
solubilized
receptors
from
ceUs
overexpressing
human
IGF-I
receptors
The
level
of
specific
"25I-insulin
binding
to
intact
IGF-I-R/3T3
cells
was
too
low
to
permit
quantitative
studies.
Moreover,
a
significant
fraction
of
this
binding
appeared
to
reflect
sites
of
low
affinity,
in
terims
of
competition
by
unlabelled
insulin.
Solubilized
receptor
preparations
from
IGF-I-R/3T3
cells
could
be
studied
Vol.
270
100
C
0
0
0
4-
c
0.
m
C
80.
60
40
20
L4L---
-
-
I
387
10-11
10-10
10-9
10-8
10-7
10-6
M.
A.
Soos
and
others
more
readily
than
receptors
in
intact
cells
because
it
was
possible
to
achieve
greater
specific
binding
of
insulin,
both
in
absolute
terms
and
relative
to
non-specific
background.
Binding
of
'251-insulin
to
solubilized
receptors
was
inhibited
by
low
concentrations
of
unlabelled
insulin
(IC50
approx.
1
nM;
Fig.
4b),
whereas
binding
of
1251I-IGF-I
was
inhibited
only
at
much
higher
concentrations
(IC50
approx.
500
nM;
results
not
shown).
It
was
concluded
that
binding
of
1251-insulin
reflected
a
high-affinity
interaction
with
rodent
insulin
receptors,
and
not
low-affinity
binding
to
human
IGF-I
receptors.
Nevertheless,
binding
of
1251_
insulin
was
strikingly
inhibited
by
low
concentrations
of
IGF-I,
almost
identical
with
those
which
inhibited
1251I-IGF-I
binding
(IC50
0.1-0.2
nM,
Fig.
4b).
High-affinity
insulin
binding
in
extracts
of
mock-transfected
NIH
3T3
NEO
cells
was
inhibited
only
by
high
concentrations
of
IGF-1
(IC50>
100
nM;
results
not
shown).
The
immunoreactivity
of
receptors
solubilized
from
IGF-I-R/
3T3
cells
was
investigated
with
two
antibodies
specific
for
human
IGF-I
receptors:
aIR-3,
which
inhibits
insulin
binding
(Kull
et
al.,
1983),
and
16-13,
which
does
not
inhibit
binding.
Both
antibodies
precipitated
the
bulk
of
IGF-I
receptors
in
IGF-1-R/3T3
extracts,
as
expected.
However,
these
antibodies
also
precipitated
approx.
50
%
of
the
rodent
insulin
receptors
in
these
extracts
(Table
2),
although
they
were
unreactive
with
insulin
receptors
in
untransfected
cells.
Total
(PEG-precipitable)
1251-insulin
binding
under
the
conditions
of
these
experiments
was
decreased
by
antibody
aIR-3
and
increased
by
antibody
16-13
(700
c.p.m.
and
2339
c.p.m.
respectively
compared
with
the
control
value
of
1564
c.p.m.).
This
suggests
that.
aIR-3
accelerates
the
dissociation
of
receptor-bound
insulin,
whereas
16-13
increases
the
binding
affinity.
It
is
concluded
that
rodent
insulin
receptors
in
IGF-I-R/3T3
cells
were
substantially
incorporated
into
hybrid
structures
with
human
IGF-I
receptors.
These
hybrids
differed
from
normal
insulin
receptors
in
that
binding
of
1251-insulin
was
inhibited
by
low
concentrations
of
both
insulin
and
IGF-I.
DISCUSSION
Receptors
for
insulin
(/3aal/)
and
IGF-I
(/J'x'a'/J')
have
gen-
erally
been
considered
to
be
distinct
but
structurally
similar
symmetrical
heterotetramers
(Czech,
1985;
Rechler
&
Nissley,
1985;
Yarden
&
Ullrich,
1988).
Various
observations
have
indicated
that
there
may
be
subtypes
of
both
receptors
(Jonas,
1988),
but
the
structural
basis
and
functional
significance
of
this
heterogeneity
is
unclear.
Two
distinct
hypotheses
have
been
advanced.
We
have
proposed
that
a
significant
fraction
of
IGF-I
receptors
in
human
placenta
occurs
as
hybrids
with
insulin
receptors
(/aa'/J'),
based
on
reactivity
with
a
panel
of
anti-receptor
antibodies
(Soos
&
Siddle,
1989).
The
existence
of
hybrid
structures
was
suggested
independently
as
a
result
of
studies
of
IGF-I-induced
receptor
phosphorylation
in
Hep
G2
cells
(Moxham
et
al.,
1989).
However,
similar
autophosphoryl-
ation
woik
in
brain
(Garofalo
&
Rosen,
1989)
and
muscle
(Alexandrides
&
Smith,
1989)
was
interpreted
as
evidence
for
two
distinct
IGF-I
receptor
polypeptides
differing
in
primary
sequence
and/or
glycosylation
(and
therefore
in
immunological
recognition),
as well
in
developmental
regulation.
We
now
provide
conclusive
evidence
for
the
formation
in
intact
cells
of
hybrids
which
incorporate
subunits
of
receptors
for
both
insulin
and
IGF-I.
When
human
insulin
receptors
(hIR)
were
overexpressed
in
rat
or
mouse
fibroblasts
by
transfection
with
cloned
cDNA,
rodent
IGF-I
receptors
(rIGFR)
were
largely
incorporated
into
hybrid
structures
which
were
detected
by
their
reaction
with
multiple
anti-(human
insulin
receptor)
specific
monoclonal
antibodies
(Figs.
2
and
3;
Table
2).
Conversely,
when
human
IGF-I
receptors
(hIGFR)
were
overexpressed
in
similar
cells,
rodent
insulin
receptors
were
incorporated
into
hybrids
which
then
reacted
with
human
IGF-I-receptor-specific
antibodies
(Table
2).
In
NIH
3T3
HIR3.5
and
HIRc-B
cells,
hybrids
were
clearly
demonstrated
in
intact
as
well
as
solubilized
cells
by
the
criterion
of
inhibition
of
high-affinity
IGF-I
binding
by
monoclonal
antibodies
specific
for
insulin
receptors
(Figs.
2
and
3).
In
IGF-I-R/3T3
cells,
hybrids
were
demonstrated
by
immuno-
precipitation
of
solubilized
preparations
(Table
2),
although
little
high-affinity
insulin
binding
was
detected
in
intact
cells.
Hybrid
receptors
clearly
pre-exist
in
intact
cells
but
the
possibility
cannot
be
ruled
out
that
the
binding
specificity,
or
even
the
formation,
of
hybrids
is
influenced
by
solubilization.
In
previous
studies
with
placental
microsomal
membranes,
the
proportion
of
IGF-
I
receptors
appearing
as
hybrids
appeared
higher
in
solubilized
than
in
particulate
preparations
(Soos
&
Siddle,
1989).
It
has
recently
been
demonstrated
that
hybrid
insulin/IGF-I
receptors
can
be
assembled
in
vitro
from
the
respective
a,z
receptor
halves,
although
this
required
the
presence
of
the
respective
ligands
or
of
Mg
ATP
(Treadway
et
al.,
1989).
Hybrid
receptors
displayed
a
high
affinity
for
both
insulin
and
IGF-I,
as
studied
with
the
rIR/hIGFR
and
rIGFR/hIR
hybrids
respectively
(Fig.
4).
However,
comparison
of
ligand
binding
to
these
two
hybrids
revealed
a
striking
asymmetry
in
properties.
The
rIGFR/hIR
hybrids
behaved
similarly
to
homomeric
IGF-I
receptors,
in
that
binding
of
1251-IGF-I
was
inhibited
by
low
concentrations
of
IGF-I
but
only
by
high
concentrations
of
insulin.
In
contrast,
binding
of
251I-insulin
to
rIR/hIGFR
hybrids
was
inhibited
by
low
concentrations
of
both
insulin
and
IGF-I
(Fig.
4).
It
is
unknown
in
either
case
whether
the
unlabelled
ligands
remain
bound
together
with
251I-labelled
ligand
and
therefore
whether
hybrid
receptors
can
bind
both
insulin
and
IGF-I
simultaneously.
It
is
possible
that
the
different
properties
of
rIR/hIGFR
compared
with
hIR/rIGFR
reflects
the
species
asymmetry.
However,
it
is
tempting
to
speculate
that
the
differences
in
the
properties
of
the
two
hybrids
are
a
consequence
of
asymmetry
of
ligand-receptor
interactions
common
to
both
structures.
This
would
imply
that
the
insulin-IR/IGFR
complex
retained
a
high
affinity
for
IGF-I,
whereas
the
IR/IGFR-IGF-I
complex
no
longer
had
a
high
affinity
for
insulin.
It
has
been
shown
that
the
homomeric
insulin
receptor
binds
only
one
molecule
of
insulin
with
high
affinity,
this
interaction
converting
the
unoccupied
site
to
a
low-affinity
state
(Pang
&
Shafer,
1984;
Boni-Schnetzler
et
al.,
1987;
Sweet
et
al.,
1987).
The
homomeric
IGF-I
receptor
may
behave
differently
in
binding
two
molecules
of
ligand
with
high
affinity
(Feltz
et
al.,
1988),
although
this
is
not
certain
(Tollefsen
&
Thompson,
1988).
A
model
can
be
proposed
in
which
the
unoccupied
half
of
a
hybrid
receptor
displays
the
same
kinetic
properties
as
in
the
corresponding
homomeric
receptor
(Fig.
5).
It
therefore
appears
that
the
interaction
between
heterologous
receptor
halves
is
sufficient
not
only
to
allow
their
covalent
association
but
also
to
permit
inter-
subunit
conformational
transitions.
Further
evidence
for
inter-subunit
interactions
is
provided
by
the
observation
that
the
anti-(IGF-I
receptor)
antibody
aIR-3
accelerated
dissociation
of
insulin
from
hybrid
receptors,
whereas
the
anti-(insulin
receptor)
antibody
25-49
accelerated
dissociation
of
IGF-I
(M.
A.
Soos
&
K.
Siddle,
unpublished
work).
Antibody
47-9
consistently
showed
a
lower
affinity
for
hybrid
receptors
than
for
homomeric
receptors
in
transfected
cells
(Figs.
2
and
3),
as
in
placenta
(Soos
&
Siddle,
1989).
This
suggests
that
the
conformation
of
the
insulin
receptor
a-subunit
may
differ
somewhat
in
the
two
types
of
receptor.
It
is
perhaps
surprising
that
three
distinct
insulin
receptor
antibodies
47-9,
25-49
and
(results
not
shown)
83-14
all
inhibited
binding
of
IGF-I
to
hybrids
not
only
as
bivalent
antibodies
but
also
as
Fab
1990
388
Insulin/insulin-like
growth
factor-I
receptor
hybrids
H
H
H
H
H H
H
H
Empty
QOOQW
IR
IR
IGFR
IR
IR
IGFR
IGFR
IGFR
LL
HH
Filled
W
oOlc1
IR
IR
IGFR
IR
IR
IGFR
IGFR
IGFR
Fig.
5.
Schematic
representation
of
the
affinity
of
ligand
binding
sites
Homomeric
(IR.IR,
IGFR.IGFR)
and
hybrid
(IGFR.IR,
IR.IGFR)
receptors
are
represented
in
the
empty
state
and
with
one
site
occupied
by
insulin
(0)
or
IGF-I
(-).
Affinities
of
the
empty
sites
are
represented
as
high
(H)
or
low
(L).
homomeric
human
receptors.
However,
in
Hep
G2
and
NIH
3T3
cells
it
appeared
that
low
concentrations
of
IGF-I
stimulated
autophosphorylation
of
both
fl-subunits
within
putative
hybrids,
although
insulin
was
less
potent
(Moxham
et
al.,
1989).
Interestingly,
NIH
3T3
HIR3.5
cells
show
enhanced
responses
not
only
to
insulin
but
also
to
IGF-I,
concomitant
with
overexpression
of
insulin
receptors
(Hofmann
et
al.,
1989).
This
suggests
that
some
effects
of
IGF-I
might
be
signalled
more
efficiently
via
hybrid
receptors
than
via
homomeric
IGF-I
receptors.
However,
the
full
physiological
implications
of
the
finding
that
insulin
and
IGF-I
receptors
behave
as
isoenzymes
which
can
combine
to
form
hybrid
as
well
as
homomeric
structures
require
further
investigation.
Studies
with
cells
co-
transfected
with
cDNAs
for
both
insulin
and
IGF-I
receptors
may
shed
light
on
this
issue,
and
on
the
relative
efficiency
of
formation
of
hybrid
and
homomeric
structures.
fragments.
It
is
unclear
whether
steric
factors
alone
could
account
for
this
inhibition,
as
the
relationships
of
epitopes
and
potential
ligand
binding
sites
in
the
heterotetrameric
receptor
is
unknown.
However,
this
observation
might
also
reflect
an
ability
of
antibodies
bound
at
one
a-subunit
to
influence
the
ligand
affinity
for
the
other
a-subunit
within
a
hybrid
molecule
by
conformational
effects.
Several
reports
have
previously
described
receptors
with
anomalous
properties,
which
might
now
be
attributed
to
the
presence
of
hybrids.
In
the
rat
L6
myocyte
line
(Burant
et
al.,
1987)
and
in
bovine
neural
retina
(Waldbillig
&
Chader,
1988)
binding
of
1251-insulin
was
observed
which
was
competed
by
IGF-I
with
greater
potency
than
by
insulin
itself.
This
could
be
explained
if
most
of
the
insulin
receptors
exist
as
hybrids
like
the
rIR/hIGFR
species
of
the
IGF-I-R/3T3
cells.
The
converse
situation
of
125I-IGF-II
binding
which
is
competed
by
insulin
at
unexpectedly
high
potency
has
been
described
in
IM-9
cells
and
placenta
(Hintz
et
al.,
1984;
Misra
et
al.,
1986;
Jonas
et
al.,
1986).
These
properties
have
been
attributed
to
'atypical'
insulin
receptors.
However,
we
did
not
observe
such
behaviour
when
using
125I1IGF-I
as
tracer
either
in
previous
studies
with
IM-9
cells
and
placenta
(Soos
&
Siddle,
1989)
or
with
rIGFR/hIR
hybrids
in
the
present
study
(Figs.
2
and
3).
It
is
possible,
though
unlikely,
that
binding
of
insulin
to
hybrid
receptors
differentially
affects
their
affinity
for
IGF-I
and
IGF-II.
However
'atypical'
receptors
in
placenta
reacted
with
anti-(insulin
receptor)
anti-
bodies
but
not
with
the
anti-(IGF-I
receptor)
antibody
aIR-3,
which
would
not
be
consistent
with
a
hybrid
species
(Jonas
et
al.,
1989).
The
presence
of
hybrid
receptors
in
some
tissues
does
not
of
course
preclude
the
existence
of
receptor
heterogeneity,
dependent
perhaps
on
differences
in
primary
sequence
(reflecting
products
of
distinct
genes
or
alternative
RNA
splicing)
and/or
differential
glycosylation.
It
appears
that
hybrids
as
well
as
homomeric
receptors
occur
in
normal
human
tissues
in
which
insulin
and
IGF-I
receptors
are
co-expressed
(Soos
&
Siddle,
1989),
and
this
could
have
important
implications
for
metabolic
regulation
by
the
respective
ligands.
The
tyrosine
kinase
activity
of
these
receptors
appears
to
be
an
essential
component
of
their
signalling
mechanism
(Rosen,
1987;
Zick,
1989).
Ligand-induced
activation
of
the
insulin
receptor
kinase
occurs
through
an
intramolecular
auto-
phosphorylation
reaction
within
intact
heterotetramers
(O'Hare
&
Pilch,
1988;
Morrison
et
al.,
1988;
Wilden
et
al.,
1989).
The
observation
that
both
insulin
and
IGF-I
bind
to
hybrid
receptors
with
high
affinity
raises
the
question
of
whether
both
ligands
can
activate
both
receptor
kinases
within
hybrids.
This
is
difficult
to
answer
in
transfected
cells
because
of
the
very
large
excess
of
We
are
grateful
to
Dr.
Steven
Jacobs
for
the
gift
of
antibody
a.IR-3.
We
thank
the
Wellcome
Trust,
the
Medical
Research
Council
and
Serono
Diagnostics
Ltd.
for
financial
support,
and
Jill
Stigter
for
technical
assistance.
REFERENCES
Alexandrides,
T.
K.
&
Smith,
R.
J.
(1989)
J.
Biol.
Chem.
264,
12922-12930
Baron,
M.
D.
&
Sonksen,
P.
H.
(1982)
Biosci.
Rep.
2,
785-793
Boni-Schnetzler,
M.,
Scott,
W.,
Waugh,
S.
M.,
DiBella,
E.
&
Pilch,
P.
(1987)
J.
Biol.
Chem.
262,
8395-8401
Burant,
C.
F.,
Treutelaar,
M.
K.,
Allen,
K.
D.,
Sens,
D.
A.
&
Buse,
M.
G.
(1987)
Biochem.
Biophys.
Res.
Commun.
147,
100-107
Czech,
M.
P.
(1985)
Annu.
Rev.
Physiol.
47,
357-381
Duronio,
V.
&
Jacobs,
S.
(1988)
Recept.
Biochem.
Methodol.
12B,
19-36
Ebina,
Y.,
Ellis,
L.,
Jarnagin,
K.,
Edery,
M.,
Graf,
L.,
Clauser,
E.,
Ou,
J.-H.,
Masiarz,
F.,
Kan,
Y.
W.,
Goldfine,
I.
D.,
Roth,
R.
A.
&
Rutter,
W.
J.
(1985)
Cell
40,
747-758
Feltz,
S.
M.,
Swanson,
M.
L.,
Wemmie,
J.
A.
&
Pessin,
J.
E.
(1988)
Biochemistry
27,
3234-3242
Froesch,
E.
R.,
Schmid,
C.,
Schwander,
J.
&
Zapf,
J.
(1985)
Annu.
Rev.
Physiol.
47,
443-467
Galfre,
G.
&
Milstein,
C.
(1981)
Methods
Enzymol.
73,
3-46
Garofalo,
R.
S.
&
Rosen,
0.
M.
(1989)
Mol.
Cell.
Biol.
9,
2806-2817
Hintz,
R.
L.,
Thorsson,
A.
V.,
Enberg,
G.
&
Hall,
K.
(1984)
Biochem.
Biophys.
Res.
Commun.
118,
774-782
Hofmann,
C.,
Goldfine,
I.
D.
&
Whittaker,
J.
(1989)
J.
Biol.
Chem.
264,
8606-8611
Izumi,
T.,
White,
M.
F.,
Kadowaki,
T.,
Takaku,
F.,
Akanuma,
Y.
&
Kasuga,
M.
(1987)
J.
Biol.
Chem.
262,
1282-1287
Jonas,
H.
A.
(1988)
Recept.
Biochem.
Methodol.
12B,
19-36
Jonas,
H.
A.,
Newman,
J.
D.
&
Harrison,
L.
C.
(1986)
Proc.
Natl.
Acad.
Sci.
U.S.A.
83,
4124-4128
Jonas,
H.
A.,
Cox,
A.
J.
&
Harrison,
L.
C.
(1989)
Biochem.
J.
257,
101-107
Kadowaki,
T.,
Koyasu,
S.,
Nishida,
E.,
Tobe,
K.,
Izumi,
T.,
Takaku,
F.,
Sakai,
H.,
Yahara,
I.
&
Kasuga,
M.
(1987)
J.
Biol.
Chem.
262,
7342-7350
Kull,
F.
C.,
Jacobs,
S.,
Su,
Y.-F.,
Svoboda,
M.
E.,
Van
Wyk,
J. J.
&
Cuatrecasas,
P.
(1983)
J.
Biol.
Chem.
258,
6561-6566
Lammers,
R.,
Gray,
A.,
Schlessinger,
J.
&
Ullrich,
A.
(1989)
EMBO
J.
8,
1369-1375
Linde,
S.,
Hansen,
B.,
Sonne,
O.,
Holst,
J.
J.
&
Glieman,
J.
(1981)
Diabetes
30,
1-8
Maegawa,
H.,
Olefsky,
J.
M.,
Thies,
S.,
Boyd,
D.,
Ullrich,
A.
&
McClain,
D.
(1988)
J.
Biol.
Chem.
265,
12629-12737
McClain,
D.
A.,
Maegawa,
H.,
Lee,
J.,
Dull,
T.
J.,
Ullrich,
A.
&
Olefsky,
J.
M.
(1987)
J.
Biol.
Chem.
262,
14663-14671
Misra,
P.,
Hintz,
R.
L.
&
Rosenfeld,
R.
G.
(1986)
J.
Clin.
Endocrinol.
Metab.
63,
1400-1405
Morrison,
B.
D.,
Swanson,
M.
L.,
Sweet,
L.
J.
&
Pessin,
J.
E.
(1988)
J.
Biol.
Chem.
263,
7806-7813
Moxham,
C.
P.,
Duronio,
V.
&
Jacobs,
S.
(1989)
J.
Biol.
Chem.
264,
13238-13244
Vol.
270
389
M.
A.
Soos
and
others
O'Hare,
T.
&
Pilch,
P.
(1988)
Biochemistry
27,
5693-5700
Pang,
D.
T.
&
Shafer,
J.
A.
(1984)
J.
Biol.
Chem.
259,
8589-8596
Rechler,
M.
M.
&
Nissley,
S.
P.
(1985)
Annu.
Rev.
Physiol.
47,
425-442
Rosen,
0.
M.
(1987)
Science
237,
1452-1458
Roth,
J.
(1975)
Methods
Enzymol.
37,
223-233
Soos,
M.
A.
&
Siddle,
K.
(1989)
Biochem.
J.
263,
553-563
Soos,
M.
A.,
Siddle,
K.,
Baron,
M.
D.,
Heward,
J.
M.,
Luzio,
J.
P.,
Bellatin,
J.
&
Lennox,
E.
S.
(1986)
Biochem.
J.
235,
199-208
Stanker,
L.
H.,
Vanderlaan,
H.
&
Juarez-Salinas,
H.
(1985)
J.
Immunol.
Methods
76,
157-169
Steele-Perkins,
G.,
Turner,
J.,
Edman,
J.
C.,
Hari,
J.,
Pierce,
S.
B.,
Stover,
C.,
Rutter,
W.
J.
&
Roth,
R.
A.
(1988)
J.
Biol.
Chem.
263,
11486-11492
Sweet,
L.
J.,
Morrison,
B.
D.
&
Pessin,
J.
E.
(1987)
J.
Biol.
Chem.
262,
6939-6942
Treadway,
J.
L.,
Morrison,
B.
D.,
Goldfine,
I.
D.
&
Pessin,
J.
E.
(1989)
J.
Biol.
Chem.
264,
21450-21453
Tollefsen,
S.
E.
&
Thompson,
K
(1988)
J.
Biol.
Chem.
263,
16267-16273
Ullrich,
A.,
Bell,
J.
R.,
Chen,
E.
Y.,
Herrera,
R.,
Petruzzelli,
L.
M.,
Dull,
T.
J.,
Gray,
A.,
Coussens,
L.,
Liao,
Y.-C.,
Tsubokawa,
M.,
Mason,
A.,
Seeburg,
P.
H.,
Grunfield,
C.,
Rosen,
0.
M.
&
Ramachandran,
J.
(1985)
Nature
(London)
313,
756-761
Ullrich,
A.,
Gray,
A.,
Tam,
A.
W.,
Yang-Fen,
T.,
Tsubokawa,
M.,
Collins,
C.,
Henzel,
W.,
Le-Bon,
T.,
Kathuria,
S.,
Chen,
E.,
Jacobs,
S.,
Francke,
U.,
Ramachndran,
J.
&
Fujita-Yamaguchi,
Y.
(1986)
EMBO
J.
5,
2503-2512
Waldbillig,
R.
J.
&
Chader,
G.
J.
(1988)
Biochem.
Biophys.
Res.
Commun.
151,
1105-1112
Whittaker,
J.,
Okamoto,
A.
K.,
Thys,
R.,
Bell,
G.
I.,
Steiner,
D.
F.
&
Hofmann,
C.
A.
(1987)
Proc.
Natl.
Acad.
Sci.
U.S.A.
84,
5237-5241
Wilden,
P.
A.,
Morrison,
B.
D.
&
Pessin,
J.
E.
(1989)
Biochemistry
28,
785-792
Yarden,
Y
&
Ullrich,
A.
(1988)
Annu.
Rev.
Biochem.
57,
443-478
Zick,
Y.
(1989)
Crit.
Rev.
Biochem.
Mol.
Biol.
24,
217-269
Received
19
February
1990/27
April
1990;
accepted
10
May
1990
1990
390
... Dans les cellules qui expriment des récepteurs insuliniques et des récepteurs de l'IGF1, des récepteurs hybrides se forment, qui contiennent un monomère ab de chaque récepteur (Moxham et al., 1989 ;Soos & Siddle, 1989 ;Soos et al., 1990 ;Treadway et al., 1990 ;Frattali & Pessin, 1993). Leur fonction est mal connue. ...
... Récepteurs hybrides insuline/IGF-I. Moxham et al., 1989 ;Soos & Siddle, 1989 ;Bailyes et al., 1997 ;Soos et al., 1990Soos et al., , 1993Treadway et al., 1990 ;Frattali & Pessin, 1993. Clonage de l'ADNc de l'IRS-1. ...
... Dans les cellules qui expriment des récepteurs insuliniques et des récepteurs de l'IGF1, des récepteurs hybrides se forment, qui contiennent un monomère ab de chaque récepteur (Moxham et al., 1989 ;Soos & Siddle, 1989 ;Soos et al., 1990 ;Treadway et al., 1990 ;Frattali & Pessin, 1993). Leur fonction est mal connue. ...
... Récepteurs hybrides insuline/IGF-I. Moxham et al., 1989 ;Soos & Siddle, 1989 ;Bailyes et al., 1997 ;Soos et al., 1990Soos et al., , 1993Treadway et al., 1990 ;Frattali & Pessin, 1993. Clonage de l'ADNc de l'IRS-1. ...
Article
L’isolement de l’insuline du pancréas et sa purification à un degré suffisant pour permettre son administration à des patients atteints de diabète de type 1 furent accomplis il y a 100 ans à l’Université de Toronto par Banting, Best, Collip et McLeod et représentent sans conteste une des plus grandes révolutions thérapeutiques en médecine, reconnue par l’attribution du Prix Nobel de Physiologie ou Médecine en 1923 à Banting et McLeod. Les retombées cliniques furent rapides ainsi que l’internationalisation de sa production commerciale. Les retombées en matière de recherche fondamentale furent beaucoup plus lentes, en particulier en ce qui concerne les mécanismes moléculaires d’action de l’insuline sur ses cellules cibles. Presque un demi-siècle s’écoula avant la détermination de la structure tri-dimensionnelle de l’insuline en 1969 et la caractérisation de son récepteur cellulaire en 1970–1971. Le fait que le récepteur de l’insuline soit une enzyme appelée tyrosine kinase ne fut démontré que dans les années 1982–1985, et la structure cristallographique du domaine kinase intracellulaire fut déterminée dix ans plus tard. La structure cristallographique du premier substrat intracellulaire de la kinase (IRS-1) en 1991 ouvrira la voie à l’élucidation des voies de signalisation intracellulaires. Il faudra 15 ans de plus avant l’obtention de la structure cristallographique du domaine extracellulaire du récepteur (en l’absence d’insuline) en 2006. Depuis, la détermination de la structure du complexe insuline-récepteur dans les états inactif et activé a fait d’énormes progrès, en particulier grâce aux améliorations récentes dans les pouvoirs de résolution de la cryo-microscopie électronique. Je passerai ici en revue les étapes du développement du concept de récepteur hormonal, et de nos connaissances sur la structure et le mécanisme moléculaire d’activation du récepteur de l’insuline.
... Intriguingly, IR and IGF-1R ab monomers are capable of forming signaling-competent hybrid IR/IGF-1R heterodimers in tissues that express both receptors. Within these hybrids, one ab monomer derives from IR and the other from IGF-1R (Soos and Siddle, 1989;Moxham et al., 1989;Soos et al., 1990;Bailyes et al., 1997). Evidence exists that the ratio of homodimeric to heterodimeric receptors in certain tissues is determined stochastically as a function of the level of expression of the individual receptors within a particular cell, with the efficiency of ab heterodimerization being equivalent to that of homodimerization (Bailyes et al., 1997). ...
... Purification of HybZip from conditioned media was achieved by sequential antibody affinity chromatography using monoclonal antibody (mAb) 9E10 resin directed against a c-myc tag attached to IGF-1Rzip (Hilpert et al., 2001;Xu et al., 2020) followed by mAb 18-44 resin directed against a linear epitope within the N-terminal region of the IR-B b chain (Soos et al., 1986;Prigent et al., 1990). mAb 18-44 does not cross-react with IGF-1R (Soos et al., 1990;Zhang and Roth, 1991). Western-blot analysis using mAb 83-7 (specific for IR; Soos et al., 1986) and mAb 24-60 (specific for IGF-1R; Soos et al., 1992) confirmed the presence of both receptor species in the affinity-purified protein product ( Figure S2A). ...
Article
Full-text available
Monomers of the insulin receptor and type 1 insulin-like growth factor receptor (IGF-1R) can combine stochastically to form heterodimeric hybrid receptors. These hybrid receptors display ligand binding and signaling properties that differ from those of the homodimeric receptors. Here, we describe the cryoelectron microscopy structure of such a hybrid receptor in complex with insulin-like growth factor I (IGF-I). The structure (ca. 3.7 Å resolution) displays a single IGF-I ligand, bound in a similar fashion to that seen for IGFs in complex with IGF-1R. The IGF-I ligand engages the first leucine-rich-repeat domain and cysteine-rich region of the IGF-1R monomer (rather than those of the insulin receptor monomer), consistent with the determinants for IGF binding residing in the IGF-1R cysteine-rich region. The structure broadens our understanding of this receptor family and assists in delineating the key structural motifs involved in binding their respective ligands.
... The IR and IGF-1R are similar in their structure (Ullrich et al., 1986). In addition, there are insulin/IGF-1 hybrid receptors in most organs (Soos et al., 1990;Seely et al., 1995;Bailyes et al., 1997). These hybrid receptors have the capacity to dimerize and bind either insulin or IGF-1. ...
Article
Full-text available
Insulin affects metabolic processes in different organs, including the skin. The sebaceous gland (SG) is an important appendage in the skin, which responds to insulin-mediated signals, either directly or through the insulin growth factor 1 (IGF-1) axis. Insulin cues are differently translated into the activation of metabolic processes depending on several factors, including glucose levels, receptor sensitivity, and sebocyte differentiation. The effects of diet on both the physiological function and pathological conditions of the SG have been linked to pathways activated by insulin and IGF-1. Experimental evidence and theoretical speculations support the association of insulin resistance with acne vulgaris, which is a major disorder of the SG. In this review, we examined the effects of insulin on the SG function and their implications in the pathogenesis of acne.
... Insulin-like growth factor-1 (IGF-I) is vital for the regulation of mammalian development, metabolism, and growth [35]. IGF-I receptor (IGF-IR) is a membrane-spanning tyrosine kinase receptor that shares structural identity with the insulin receptor, which comprises two α subunits and two β subunits and can form heterodimers with the insulin receptor [36,37]. Recent studies have found that the expression levels of IGF-I and IGF-IR in orbital fibroblasts of TAO were significantly increased, and IGF-IR was considered to be potentially involved in the pathogenesis of TAO [38][39][40]. ...
Article
Full-text available
Thyroid-associated ophthalmopathy (TAO), also known as thyroid eye disease (TED) or Graves’ orbitopathy (GO), is a complex autoimmune condition causing visual impairment, disfigurement, and harm to patients’ physical and mental health. The pathogenesis of TAO has not been fully elucidated, and the mainstream view is that coantigens shared by the thyroid and orbit trigger remodeling of extraocular muscles and orbital connective tissues through an inflammatory response. In recent years, cytokines and the immune responses they mediate have been crucial in disease progression, and currently, common evidence has shown that drugs targeting cytokines, such as tocilizumab, infliximab, and adalimumab, may be novel targets for therapy. In this review, we summarize the research development of different cytokines in TAO pathogenesis in the hope of discovering new therapeutic targets.
... Insulin and IGF1 bind to IR and IGF1R in the form of homodimers or heterodimers with a very high a nity [12][13][14]. The binding of insulin and IGF1 to both receptors activates intrinsic receptor tyrosine kinase and downstream signaling cascades, which in turn regulate many cellular functions, including gene transcription, nutrient metabolism (glucose, lipids, and proteins), and cell growth and differentiation [15][16][17]. However, the aberrant expression and activation of IR and IGF1R are strongly associated with a greater risk of breast cancer [18][19]. ...
Preprint
Full-text available
Much higher risk of cancer is observed in patients with diabetes. Insulin receptor (IR) and insulin-like growth factor-1 receptor (IGF1R) are well-known targets in cancer research as well as diabetes treatment. Interestingly, a recent study proposed that the IR/IGF1R ratio is an important factor in breast cancer prognosis. Women with a higher IR/IGF1R ratio showed poor breast cancer prognosis and hyperinsulinemia. Here, we propose a novel mechanism by which the oncogenic protein TRIP-Br1 renders breast cancer cells to have a higher IR/IGF1R ratio by positively and negatively regulating IR and IGF1R expression at the protein level, respectively. TRIP-Br1 plays various cellular roles, one of which is as an adaptor protein. Our data revealed that TRIP-Br1 suppresses ubiquitin/proteasome-mediated IR degradation without directly interacting with IR. Meanwhile, TRIP-Br1 directly interacts with both IGF1R and NEDD4-1 E3 ubiquitin ligase, and TRIP-Br1/NEDD4-1 degrades IGF1R via the ubiquitin/proteasome system. Animal experiments indicated that TRIP-Br1 enhanced tumor progression, where a high IR/IGF1R ratio was detected. Furthermore, IR silencing elevates IGF1R expression, resulting in a lower IR/IGF1R ratio. Our extended study showed a similar effect of TRIP-Br1 on the IR/IGF1R ratio in insulin-deficient mice mimicking patients with diabetes, confirming the strong relationship between breast cancer and diabetes. In conclusion, this study provides invaluable information on the regulatory mechanism of how breast cancer cells acquire a higher IR/IGF1R ratio.
... Insulin and IGF1 bind to IR and IGF1R in the form of homodimers or heterodimers with a very high a nity [12][13][14]. The binding of insulin and IGF1 to both receptors activates intrinsic receptor tyrosine kinase and downstream signaling cascades, which in turn regulate many cellular functions, including gene transcription, nutrient metabolism (glucose, lipids, and proteins), and cell growth and differentiation [15][16][17]. However, the aberrant expression and activation of IR and IGF1R are strongly associated with a greater risk of breast cancer [18][19]. ...
Preprint
Full-text available
Much higher risk of cancer is observed in patients with diabetes. Insulin receptor (IR) and insulin-like growth factor-1 receptor (IGF1R) are well-known targets in cancer research as well as diabetes treatment. Interestingly, a recent study proposed that the IR/IGF1R ratio is an important factor in breast cancer prognosis. Women with a higher IR/IGF1R ratio showed poor breast cancer prognosis and hyperinsulinemia. Here, we propose a novel mechanism by which the oncogenic protein TRIP-Br1 renders breast cancer cells to have a higher IR/IGF1R ratio by positively and negatively regulating IR and IGF1R expression at the protein level, respectively. TRIP-Br1 plays various cellular roles, one of which is as an adaptor protein. Our data revealed that TRIP-Br1 suppresses ubiquitin/proteasome-mediated IR degradation without directly interacting with IR. Meanwhile, TRIP-Br1 directly interacts with both IGF1R and NEDD4-1 E3 ubiquitin ligase, and TRIP-Br1/NEDD4-1 degrades IGF1R via the ubiquitin/proteasome system. Animal experiments indicated that TRIP-Br1 enhanced tumor progression, where a high IR/IGF1R ratio was detected. Furthermore, IR silencing elevates IGF1R expression, resulting in a lower IR/IGF1R ratio. Our extended study showed a similar effect of TRIP-Br1 on the IR/IGF1R ratio in insulin-deficient mice mimicking patients with diabetes, confirming the strong relationship between breast cancer and diabetes. In conclusion, this study provides invaluable information on the regulatory mechanism of how breast cancer cells acquire a higher IR/IGF1R ratio.
... Insulin/insulin-like growth factor (IGF) system consists of three kinds of ligands (insulin, IGF-1, and IGF-2), four types of receptors (insulin receptor, IGF-1 receptor, hybrid IGF-1/insulin receptor, and IGF-2 receptor), IGF binding proteins (IGFBP1-IGFBP6), IGFBP protease, and insulin receptor substrate (IRS1-6). Due to structural similarities among insulin receptor, IGF-1 receptor, and hybrid IGF-1/insulin receptor, insulin was shown to bind with insulin receptor, IGF-1R, and possibly hybrid IGF-1/insulin receptor [18,19] and result in activation of intracellular signal pathways including phosphatidylinositol 3-kinase (PI3K)/mammalian target of rapamycin (mTOR) and mitogen-activated protein kinase (MAPK)/extracellular signal-regulated kinase (ERK) pathways, leading to promotion of cell proliferation and inhibition of apoptosis of cancerous colon epithelial cells [20,21]. Specifically, the PI3K/ mTOR pathway leads to cancer cell survival and migration, whereas the MAPK/ERK pathway promotes cancer cell metabolism and proliferation. ...
Article
Full-text available
Several epidemiological studies have identified diabetes as a risk factor for colorectal cancer (CRC). The potential pathophysiological mechanisms of this association include hyperinsulinemia, insulin-like growth factor (IGF) axis, hyperglycemia, inflammation induced by adipose tissue dysfunction, gastrointestinal motility disorder, and impaired immunological surveillance. Several studies have shown that underlying diabetes adversely affects the prognosis of patients with CRC. This review explores the novel anticancer agents targeting IGF-1R and receptor for advanced glycation end products (RAGE), both of which play a vital role in diabetes-induced colorectal tumorigenesis. Inhibitors of IGF-1R and RAGE are expected to become promising therapeutic choices, particularly for CRC patients with diabetes. Furthermore, hypoglycemic therapy is associated with the incidence of CRC. Selection of appropriate hypoglycemic agents, which can reduce the risk of CRC in diabetic patients, is an unmet issue. Therefore, this review mainly summarizes the current studies concerning the connections among diabetes, hypoglycemic therapy, and CRC as well as provides a synthesis of the underlying pathophysiological mechanisms. Our synthesis provides a theoretical basis for rational use of hypoglycemic therapies and early diagnosis and treatment of diabetes-related CRC.
Article
The insulin receptor (IR) is a type II receptor tyrosine kinase that plays essential roles in metabolism, growth, and proliferation. Dysregulation of IR signaling is linked to many human diseases, such as diabetes and cancers. The resolution revolution in cryo–electron microscopy has led to the determination of several structures of IR with different numbers of bound insulin molecules in recent years, which have tremendously improved our understanding of how IR is activated by insulin. Here, we review the insulin-induced activation mechanism of IR, including ( a) the detailed binding modes and functions of insulin at site 1 and site 2 and ( b) the insulin-induced structural transitions that are required for IR activation. We highlight several other key aspects of the activation and regulation of IR signaling and discuss the remaining gaps in our understanding of the IR activation mechanism and potential avenues of future research. Expected final online publication date for the Annual Review of Biochemistry, Volume 92 is June 2023. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Article
Full-text available
Insulin was discovered over 100 years ago. Whilst the first half century defined many of the physiological effects of insulin, the second emphasised the mechanisms by which it elicits these effects, implicating a vast array of G proteins and their regulators, lipid and protein kinases and counteracting phosphatases, and more. Potential growth-promoting and protective effects of insulin on the heart emerged from studies of carbohydrate metabolism in the 1960s, but the insulin receptor (and the related receptor for insulin-like growth factors 1 and 2) were not defined until the 1980s. A related third receptor, the insulin receptor-related receptor (INSRR) remained an orphan receptor for many years until it was identified as an alkali-sensor. The mechanisms by which these receptors and the plethora of downstream signalling molecules confer cardioprotection remain elusive. Here, we review important aspects of the effects of the three insulin receptor family members in the heart. Metabolic studies are set in the context of what is now known of insulin receptor family signalling and the role of protein kinase B (PKB, or Akt), and the relationship between this and cardiomyocyte survival versus death is discussed. PKB/Akt phosphorylates numerous substrates with potential for cardioprotection in the contractile cardiomyocytes and cardiac non-myocytes. Our overall conclusion is that the effects of insulin on glucose metabolism that were initially identified remain highly pertinent in managing cardiomyocyte energetics and preservation of function. This alone provides a high level of cardioprotection in the face of pathophysiological stressors such as ischaemia and myocardial infarction.
Article
Full-text available
Monoclonal antibodies for the human insulin receptor were produced following immunization of mice with IM-9 lymphocytes and/or purified placental receptor. Four separate fusions yielded 28 antibodies, all of which reacted with receptor from human placenta, liver and IM-9 cells. Some antibodies cross-reacted to varying degrees with receptor from rabbit, cow, pig and sheep, but none reacted with rat receptor. At least 10 distinct epitopes were recognized as indicated by species specificity and binding competition experiments. All of these epitopes appeared to be on extracellular domains of the receptor as shown by binding of antibodies to intact cells. In some cases the epitopes were further localized to alpha or beta subunits by immunoblotting. Several antibodies inhibited binding of 125I-insulin to the receptor, some had no effect on binding, and others enhanced the binding of 125I-insulin. It is concluded that these antibodies will be valuable probes of receptor structure and function.
Article
Full-text available
The type I insulin-like growth factor (IGF) receptor, like the insulin receptor, contains a ligand-stimulated protein-tyrosine kinase activity in its beta-subunit. However, in vivo, no substrates have been identified. We used anti-phosphotyrosine antibodies to identify phosphotyrosine-containing proteins which occur during IGF-I stimulation of normal rat kidney and Madin-Darby canine kidney cells labeled with ortho[32P]phosphate. Both cells provide a good system to study the function of the type I IGF receptors because they contain high concentrations of these receptors but no insulin receptors. In addition, physiological levels of IGF-I, but not insulin, stimulated DNA synthesis in growth-arrested cells. IGF-I stimulated within 1 min of tyrosine phosphorylation of two proteins. One of them, with a molecular mass between 97 and 102 kDa, was supposed to be the beta-subunit of the type I IGF receptor previously identified. The other protein had an approximate molecular mass of 185 kDa, which resembled, by several criteria, pp 185, originally identified during the initial response of Fao cells to insulin binding (White, M. F., Maron, R., and Kahn, C. R. (1985) Nature 318, 183-186). These data suggest that tyrosine phosphorylation of pp 185 may occur during activation of both the type I IGF receptor and the insulin receptor, and it could be a common substrate that transmits important metabolic signals during ligand binding.
Article
Full-text available
KB cells respond to insulin and insulin-like growth factor I (IGF-I) in a closely similar way (induction of membrane ruffling, stimulation of pinocytosis, and amino acid transport) but respond to epidermal growth factors (EGF) in a similar but distinct way. In the KB cells, using phosphotyrosine-specific antibody we have found that: the receptors for insulin (beta subunit), IGF-I (beta subunit), and EGF undergo tyrosine phosphorylation as early as 10 s after addition of their respective ligands; a 185-kDa protein is rapidly (less than 10 s) tyrosine phosphorylated by insulin and IGF-I through their respective receptor kinases but not EGF; tyrosine phosphorylation of a 190-kDa glycoprotein is rapidly (less than 10 s) induced by EGF through EGF receptor kinase; and tyrosine phosphorylation of a 240-kDa protein is stimulated within 30 s by all three growth factors. These patterns of tyrosine phosphorylation could be causally related to biological responses induced by the three growth factors.
Article
Full-text available
Insulin-like growth factor (IGF)-binding sites copurifying with human placental insulin receptors during insulin-affinity chromatography consist of two immunologically distinct populations. One reacts with monoclonal antibody alpha IR-3, but not with antibodies to the insulin receptor, and represents Type I IGF receptors; the other reacts only with antibodies to the insulin receptor and is precipitated with a polyclonal receptor antibody (B-10) after labelling with 125I-multiplication-stimulating activity (MSA, rat IGF-II). The latter is a unique sub-population of atypical insulin receptors which differ from classical insulin receptors by their unusually high affinity for MSA (Ka = 2 x 10(9) M-1 compared with 5 x 10(7) M-1) and relative potencies for insulin, MSA and IGF-I (40:5:1 compared with 150:4:1). They represent 10-20% of the total insulin receptor population and account for 25-50% of the 125I-MSA binding activity in Triton-solubilized placental membranes. Although atypical and classical insulin receptors are distinct, their immunological properties are very similar, as are their binding properties in response to dithiothreitol, storage at -20 degrees C and neuraminidase digestion. We conclude that atypical insulin receptors with moderately high affinity for IGFs co-exist with classical insulin receptors and Type I IGF receptors in human placenta. They provide an explanation for the unusual IGF-II binding properties of human placental membranes and may have a specific role in placental growth and/or function.
Article
Full-text available
Promising progress in understanding the molecular basis of insulin action has been achieved by demonstrating that the insulin receptor is an insulin-sensitive tyrosine kinase. Here we discuss the structure of this receptor kinase and compare it with receptors for related growth factors. We review the known modes to regulate the receptor kinase activity, either through its autophosphorylation (on tyrosine residues) or through its phosphorylation by other kinases (on serine and threonine residues). We discuss the role of the receptor kinase activity in hormone signal transduction in light of results indicating a reduced kinase activity in insulin-resistant states. Finally, studies to identify natural substrates for the insulin receptor kinase are presented. The possible physiological role of these phosphorylated substrates in mediating insulin action is evaluated.
Article
The development of sensitive radioimmunoassays for peptide hormones was accelerated by the introduction of excellent methods for labeling these hormones with iodine at high specific radioactivity under conditions such that the labeled hormone retained reactivity with the antihormone antibody and was relatively stable for several days in the assay. This chapter deals with the theoretical and practical problems of preparing radioactive hormones suitable for such studies. In theory, radioactive atoms such as 14C, 3H, and 35S that are substituted for stable atoms of the peptide hormone molecule should be preferable to atoms such as iodine, which are added to the molecule. Not only are the substituted forms more natural, but it has also been purported that the methods for producing them are milder and less damaging to the hormone. In practice, iodinated peptides, largely 125I and to a lesser extent 131I, have been most successfully used for receptor studies. They can be produced easily at much higher levels of specific radioactivity than peptides labeled with substituted atoms. This article describes methods for the preparation of 125I-labeled peptide hormones at high specific radioactivity for use in radioreceptor assays or other studies of hormone interactions with target cell receptors. These preparations are also excellent for radioimmunoassay.
Article
Three recent advances pertinent to the mechanism of insulin action include (i) the discovery that the insulin receptor is an insulin-dependent protein tyrosine kinase, functionally related to certain growth factor receptors and oncogene-encoded proteins, (ii) the molecular cloning of the insulin proreceptor complementary DNA, and (iii) evidence that the protein tyrosine kinase activity of the receptor is essential for insulin action. Efforts are now focusing on the physiological substrates for the receptor kinase. Experience to date suggests that they will be rare proteins whose phosphorylation in intact cells may be transient. The advantages of attempting to dissect the initial biochemical pathway of insulin action include the wealth of information about the metabolic consequences of insulin action and the potential for genetic analysis in Drosophila and in man.
Article
Insulin and insulinlike growth factor 1 (IGF-1) receptors are present in brain, yet their function remains obscure. Expression of these tyrosine kinase-bearing growth factor receptors during rat brain development was examined by using three antipeptide antibodies directed against epitopes in the beta subunits (AbP2, AbP4, and AbP5). All three antibodies recognized both insulin and IGF-1 receptors. Membranes were prepared from fetal brains (14 to 21 days of gestation), neonatal brain (postnatal day 1), and adult brain. Immunoblot analyses using AbP4 and AbP5 revealed a 92-kilodalton (kDa) protein that corresponded to the beta subunit of the insulin and IGF-1 receptors. Densitometric scanning of immunoblots indicated that receptor proteins were 4- to 10-fold more abundant in fetal brain membranes than in membranes from adult brain. Expression was highest during 16 to 18 days of gestation and declined thereafter to the relatively low level found in adult brain. Immunoblot analyses with AbP2 as well as ligand-activated receptor autophosphorylation revealed an additional protein of 97 kDa. This protein was phosphorylated in response to IGF-1 and was not directly recognized by AbP4 or AbP5. The covalent association of the 97-kDa protein with the 92-kDa beta subunit was indicated by the ability of AbP4 and AbP5 to immunoprecipitate both proteins under nonreducing conditions but only the 92-kDa protein after reduction. In contrast, AbP2 immunoprecipitated both proteins regardless of their association. This immunospecificity remained unchanged after deglycosylation of the isolated proteins. Two-dimensional tryptic phosphopeptide analysis showed that the 92- and 97-kDa subunits of the IGF-1 receptor are related but distinct proteins. Taken together, the data suggest that the 92- and 97-kDa subunits differ in primary amino acid sequence. Thus, two distinct beta subunits may be present in a single IGF-1 receptor in brain. These subunits have in common an epitope recognized by an antibody to the tyrosine kinase domain (AbP2) but differ in regions thought to be important in receptor kinase regulation and signal transduction.
Article
The receptors for insulin and insulin-like growth factor-I (IGF-I) are closely related in primary sequence and overall structure. We have examined the immunological relationships between these receptors by testing the reactivity of anti-(insulin receptor) monoclonal antibodies with IGF-I receptors in various tissues and cell lines. Antibodies for six distinct epitopes reacted with a subfraction of IGF-I receptors, as shown by inhibition of 125I-IGF-I binding, precipitation of 125I-IGF-I-receptor complexes or immunodepletion of receptor from tissue extracts before binding assays. Both immunoreactive and non-immunoreactive subfractions displayed the expected properties of 'classical' IGF-I receptors, in terms of relative affinities for IGF-I and insulin. The proportion of total IGF-I receptors which was immunoreactive varied in different cell types, being approx. 40% in Hep G2 cells, 35-40% in placental membranes and 75-85% in IM-9 cells. The immunoreactive fraction was somewhat higher in solubilized receptors than in the corresponding intact cells or membranes. A previously described monoclonal antibody, alpha-IR-3, specific for IGF-I receptors, inhibited IGF-I binding by more than 80% in all preparations. When solubilized placental receptors were pretreated with dithiothreitol (DTT) under conditions reported to reduce intramolecular (class I) disulphide bonds, the immunoreactivity of IGF-I receptors was abolished although total IGF-I binding was little affected. Under the same conditions insulin receptors remained fully immunoreactive. When solubilized receptor preparations were fractionated by gel filtration, both IGF-I and insulin receptors ran as symmetrical peaks of identical mobility. After DTT treatment, the IGF-I receptor was partially converted to a lower molecular mass form which was not immunoreactive. The insulin receptor peak showed a much less pronounced skewing and remained fully immunoreactive in all fractions. It is concluded that the anti- (insulin receptor) antibodies do not react directly with IGF-I receptor polypeptide, and that the apparent immunoreactivity of a subfraction of IGF-I receptors reflects their physical association with insulin receptors, both in cell extracts and in intact cells. The most likely basis for this association appears to be a 'hybrid' receptor containing one half (alpha beta) of insulin receptor polypeptide and the other (alpha' beta') of IGF-I receptor polypeptide within the native (alpha beta beta' alpha') heterotetrameric structure.
Article
The purified human placenta alpha 2 beta 2 heterotetrameric insulin receptor was reduced and dissociated into a functional alpha beta heterodimeric complex by a combination of alkaline pH and dithiothreitol treatment. In the presence of Mn/MgATP, insulin binding to the isolated alpha beta heterodimeric insulin receptor was found to induce the formation of a covalent disulfide-linked alpha 2 beta 2 heterotetrameric complex. In the absence of insulin, a noncovalent association of the alpha beta heterodimeric insulin receptor complex into an alpha 2 beta 2 heterotetrameric state required the continuous presence of both a divalent metal ion (Mn or Mg) and an adenine nucleotide (ATP, ADP, or AMPPCP). Thus, Mn/MgATP binding and not insulin receptor autophosphorylation was responsible for the noncovalent association into the alpha 2 beta 2 heterotetrameric state. However, the divalent metal ions or NaATP separately was ineffective in inducing the noncovalent association between the alpha beta heterodimers. The specific sulfhydryl agent iodoacetamide (IAN) was observed to inhibit the insulin-dependent covalent association of the alpha beta heterodimers without affecting the Mn/MgATP-induced noncovalent association into the alpha 2 beta 2 heterotetrameric state. Insulin treatment of the isolated alpha beta heterodimeric complex in the presence of IAN demonstrated that the Mn/MgATP-induce noncovalent association into the alpha 2 beta 2 heterotetrameric state was sufficient for insulin stimulation of beta-subunit autophosphorylation and exogenous substrate protein kinase activity. These data indicate that although interaction between the individual insulin receptor alpha beta heterodimers is necessary for insulin stimulation of protein kinase activity it does not require covalent disulfide bond formation.