ArticlePDF Available

Chemokine regulation of human megakaryocy-topoiesis

Authors:

Abstract

We have previously shown that platelet factor 4 (PF4), a platelet-specific CXC chemokine, can directly and specifically inhibit human megakaryocyte colony formation. We therefore hypothesized that PF4 might function as a negative autocrine regulator of megakaryocytopoiesis. Herein we present additional studies characterizing the inhibitory effect of CXC chemokines on human megakaryocyte development. We first corroborated our initial studies by showing that recombinant human (rH) PF4, like the native protein, inhibited megakaryocytopoiesis. We then examined the inhibitory properties of other CXC family members. Neutrophil activating peptide-2 (NAP-2), a naturally occurring N-terminally cleaved beta TG peptide, was found to inhibit megakaryocytopoiesis with two to three orders of magnitude greater potency than PF4. Structure function studies showed that an N-terminal mutation, which eliminated NAP-2's neutrophil activating properties (NAP-2E2-->A), also abrogated its ability to inhibit megakaryocyte development. Further investigations of this type demonstrated that a chimeric PF4 protein (AELR/PF4) in which PF4's N-terminus was replaced with the first four amino acids of NAP-2 was also a potent inhibitor of megakaryocytopoiesis. Interleukin (IL)-8, another CXC chemokine, and three CC chemokines (macrophage inhibitory protein-1 alpha [MIP-1 alpha], MIP-1 beta, and C10) also specifically inhibited megakaryocyte colony formation at NAP-2 equivalent doses. CXC and CC chemokine inhibition was additive suggesting that the effects might be mediated through a common pathway. The inhibitory effects of NAP-2 and MIP-1 alpha could not be overcome by adding physiologically relevant amounts of recombinant human megakaryocyte growth and development factor (MGDR) (50 ng/mL) to the cultures. Using Northern blot and reverse transcriptase-polymerase chain reaction (RT-PCR) based analyses, we documented mRNA expression of IL-8 receptor isoforms alpha and beta in total platelet RNA and in normal human megakaryocytes, respectively. Based on these results, we hypothesize that chemokines play a physiologic role in regulating megakaryocytopoiesis. Because chemokines are elaborated by ancillary marrow cells, both autocrine and paracrine growth control is suggested, the effects of which might be exerted, in part, through alpha and beta IL-8 receptors.
1995 86: 2559-2567
AM Gewirtz, J Zhang, J Ratajczak, M Ratajczak, KS Park, C Li, Z Yan and M Poncz
Chemokine regulation of human megakaryocytopoiesis
http://bloodjournal.hematologylibrary.org/site/misc/rights.xhtml#repub_requests
Information about reproducing this article in parts or in its entirety may be found online at:
http://bloodjournal.hematologylibrary.org/site/misc/rights.xhtml#reprints
Information about ordering reprints may be found online at:
http://bloodjournal.hematologylibrary.org/site/subscriptions/index.xhtml
Information about subscriptions and ASH membership may be found online at:
reserved.
Copyright 2011 by The American Society of Hematology; all rights
900, Washington DC 20036.
weekly by the American Society of Hematology, 2021 L St, NW, Suite
Blood (print ISSN 0006-4971, online ISSN 1528-0020), is published
For personal use only. by guest on July 13, 2011. bloodjournal.hematologylibrary.orgFrom
Chemokine Regulation
of
Human Megakaryocytopoiesis
By
Alan
M.
Gewirtz, Jin Zhang, Janina Ratajczak, Mariusz Ratajczak, Kwang Sook Park,
Changqing Li, Zhanqing Yan, and Mortirner Poncz
We have previously shown that platelet factor 4 (PF4). a
platelet-specific CXC chemokine, can directly and specifically
inhibit
human megakaryocyte colony formation. We there-
fore hypothesized that PF4 might function as a negative au-
tocrine regulator of megakaryocytopoiesis. Herein we pre-
sent additional studies characterizing the inhibitory effect
of CXC chemokines on human megakaryocyte development.
We
first
corroborated our initial studies by showing that
recombinant human (rH1 PF4, like the native protein,
inhib-
ited megakaryocytopoiesis. We then examined the
inhibi-
tory properties of other CXC family members. Neutrophil
activating peptide-2 (NAP-2). a naturally occurring N-termi-
nally cleaved PTG peptide, was found to inhibit megakaryo-
cytopoiesis
with
two
to three orders of magnitude greater
potency than PF4. Structure function studies showed that an
N-terminal mutation, which eliminated NAP-2's neutrophil
activating properties (NAP-2mA), also abrogated its ability
to
inhibit
megakaryocyte development. Further investiga-
tions of this type demonstrated that a chimeric PF4 protein
(AELR/PF4)
in
which PF4's N-terminus was replaced with the
first four amino acids of NAP-P was also a potent inhibitor of
UMAN MEGAKARYOCYTOPOIESIS and platelet
production are complex processes whose regulation
remains incompletely understood. The identification and
cloning of the c-mpl ligand, now known as thrombopoietin
(TPO) or MGDR, represents
an
important advance in this
area because this protein appears to be the major regulator
of megakaryocyte development. MGDR has been shown,
for example, to increase the number, size, and ploidy of
megakaryocytes, and in vivo administration markedly in-
creases platelet count in recipient anirnal~."~ Nevertheless,
MGDR is clearly not the sole regulator of the megakaryo-
cytelplatelet axis, as innumerable cytokine experiments5"'
and knockout experiments of the MGDR receptor, c-Mpl,"
have shown.
Besides positive effectors, it is possible that megakaryocy-
topoiesis is regulated by inhibitory influences as well. In
this regard, it has been observed that megakaryocyte colony
growth is inferior in serum than in platelet-poor plasma sug-
gesting that a platelet released product inhibits megakaryo-
cyte gro~th.'~"~ Megakaryocytes
also
appear to release prod-
ucts that inhibit their own development. We, and others,
have shown that one of the platelet-specific a-granule chem-
okines, platelet factor 4 (PF4), can inhibit megakaryocyte
colony formation in a lineage-specific fashion.I6"' Inhibition
is manifested by a decrease in the number and size of mega-
karyocyte colonies. In our hands,16 the related platelet-spe-
cific a-granule chemokine, PTG, does not exert similar ef-
fects on cultured cells, though conflicting findings have been
reported by others." In aggregate, these studies suggest that
megakaryocytopoiesis and thrombopoiesis may be con-
trolled to an as yet undetermined extent by a negative auto-
crine growth loop.
One caveat in attributing a physiologic role to PF4 in
the regulation of human megakaryocyte development is the
amount of material required to observe the negative growth
effect. Though some markers of megakaryocyte maturation,
H
Blood,
Vol
86,
No
7 (October
l),
1995:
pp
2559-2567
megakaryocytopoiesis. lnterleukin (IL)-8, another CXC
chemokine, and three CC chemokines (macrophage
inhibi-
tory protein-l& [MIP-la], MIP-1/3, and C101 also specifically
inhibited megakaryocyte colony formation at NAP-2 equiva-
lent doses. CXC and CC chemokine inhibition was additive
suggesting that the effects might be mediated through a
common pathway. The inhibitory effects of NAP-2 and MIP-
la
could not be overcome by adding physiologically relevant
amounts of recombinant human megakaryocyte growth and
development factor (MGDR) (50 ng/mL) to the cultures.
Us-
ing Northern blot and reverse transcriptase-polymerase
chain reaction (RT-PCR) based analyses, we documented
mRNA expression of IL-8 receptor isoforms
a
and
/3
in
total
platelet RNA and
in
normal human megakaryocytes, respec-
tively. Based on these results, we hypothesize that chemo-
kines play a physiologic role
in
regulating megakaryocyto-
poiesis. Because chemokines are elaborated by ancillary
marrow cells, both autocrine and paracrine growth control
is suggested, the
effects
of which might be exerted,
in
part,
through
a
and
/3
IL-8 receptors.
0
7995
by
The American Society
of
Hematology.
for example factor
V
mRNA expression, could be inhibited
by nanogram quantities of PF4, colony inhibition occurs in
the range of
-25
pg/mL.I6 Though such concentrations are
routinely observed in serum, whether marrow concentrations
of this magnitude can really be achieved in vivo is debatable.
Accordingly, we entertained the possibility that other mem-
bers of the chemokine family to which PF4 and PTG be-
longed might possess similar, or perhaps even more potent,
inhibitory activity against the megakaryocyte lineage.I9-*'
Chemokines are
70
to
100
amino acids long and contain
four cysteine resid~es.l~-~~ The family has diverged into two
main subgroups. PF4 and PTG are members of the
CXC
or
a
chemokine family subgroup. In this group the first two of
the four conserved cysteine residues are separated from each
other by one additional amino acid residue. This subfamily
also includes interleukin-8
(IL-8),
a potent neutrophil acti-
vating agent. PF4 and
PTG
are poor neutrophil activators,
but a cleavage product of PTG, neutrophil activating peptide-
From the Departments of Pathology, Medicine and Pediatrics,
University of Pennsylvania School
of
Medicine, Philadelphia; The
Children's Hospital of Philadelphia, Philadelphia, PA; and the De-
partment of Microbiology, Korea University School of Medicine,
Seoul, Korea.
Submitted January 9, 1995; accepted June 2, 1995.
Supported by Grants No. CA36896 to A.M.G. and HL.37419 to
M.P. from the National Institutes of Health and the Tobacco Re-
search Council Grant
No.
3152 to M.P.
Address reprint requests
to
Mortimer Poncz, MD, The Children
'S
Hospital of Philadelphia, 34th St and Civic Center Blvd, Philadel-
phia, PA 19104.
The publication costs of this article were defrayed
in
part by page
charge payment. This article must therefore be hereby marked
"advertisement"
in accordance with
I8
U.S.C. section 1734
solely
to
indicate this fact.
0
1995 by The American Society of Hematology.
0006-4971~.5/8607-0019$3.00/0
2559
For personal use only. by guest on July 13, 2011. bloodjournal.hematologylibrary.orgFrom
2560
GEWIRTZ
ET
AL
2
(NAP-2),
is
a
potent neutrophil activat~r.~~”~ Of note, two
different
IL-8
receptors
(IL-8R)
are
expressed by neutro-
phils. One is designated the
a
or
type
I
IL-8R
and binds
IL-8
efficiently, but
NAP-2
poorly.
A
second
/?
or type
2
IL-8R
has also been described, which binds
IL-8
well, and
NAP-2
with moderate affinity.20.26,27
The
CC or
p
chemo-
kines, in which the first
two
cysteine residues
are
immedi-
ately adjacent
to
each
other,
represent the other branch
of
this family. CC chemokines such
as
macrophage inhibitory
protein
(MIP)-la**
appear to be important
monocyte
stimu-
lating agents and
may
also selectively suppress human
mega-
karyocyte development.
A
CC chemokine receptor has been
defined.*’
The
PF4
and
PTG
studies described above suggested that
neutrophil activating potency and megakaryocyte colony in-
hibitory activity paralleled each other. To test this hypothe-
sis,
we
examined
the effect of the neutrophil activator
NAP-
2
on human megakaryocyte development in vitro.
We
also
sought
to
determine
the
megakaryocyte colony inhibitory
activity of other
members
of this chemokine family. Herein,
we present data showing that
a
number
of
CXC
and
CC
chemokines
are
potent inhibitors of human megakaryocyto-
poiesis and that megakaryocytes
express
a
number of chemo-
kine receptors. These data suggest that chemokines, perhaps
elaborated by
ancillary
cells in the marrow microenviron-
ment,
may
play
a
physiologically significant role in regulat-
ing human megakaryocyte development
by
both
autoctine
and paracrine mechanisms.
MATERIALS
AND
METHODS
Cells
Light-density bone marrow mononuclear cells (MNC) were ob-
tained from normal remunerated consenting donors and depleted of
adherent cells and T lymphocytes (A-T-MNC) as described.3o
CD34’ were enriched from the A-T-MNC population by incubation
with anti-human progenitor cell antigen (HPCA)-1 murine mono-
clonal antibodies (Becton Dickinson, San Jose, CA) and subsequent
immunoselection of antibody-labeled cells with magnetic beads ac-
cording to the manufacturer’s protocol (Dynal,
Oslo,
Norway) as
described.” Purity of CD34’ cells selected in this manner exceeded
85%.
Hematopoietic Colony Assays
All
assays were performed with either peripheral blood or bone
marrow obtained from normal consenting volunteers. Hematopoietic
colonies were grown and identified
as
previously In
brief, colony assays were performed with either unseparated light-
density marrow nuclear cells (MNC)
or
with MNC depleted of
monocyte-macrophages and T-lymphocytes as previously de-
scribed.” Final plated cell concentrations were
2
X
10’lmL. Cultures
were supplemented with
30%
vollvol of aplastic anemia serum, ex-
cept when supplemented with recombinant human MGDF (gener-
ously provided by Dr Pamela Hunt, Amgen Corp, Thousand Oaks,
CA).4 Megakaryocyte and granulocyte colonies were supplemented
with
rH
IL-3
(-29
U/mL) and rH granulocyte-macrophage colony-
stimulating factor (GM-CSF) (-5 ng/mL) (Genetics Institute, Cam-
bridge, MA), except for the megakaryocyte studies that tested MGDF
effect. Erythroid colonies were supplemented with recombinant
erythropoietin (5 U/mL) (Amgen Corp). Recombinant human chem-
okines were added just before plating.
Megakaryocyte colonies were enumerated after
12
days in culture
by indirect immunofluorescence using either a highly-specific rabbit
antihuman platelet membrane glycoprotein antiserum’h~3”.31 or :I
monoclonal anti-GPIIbflIIa complex antibody
A2AY.”
Binding
of
the probe antibody was detected with a species appropriate Ruores-
cein-conjugated secondary antibody (Meloy, Springfield, VA). A
cluster of three
or
more intensely fluorescent cells were defined as
one colony. Plates were read by two separate individuals, including
one blinded to plate designation. Results were in agreement between
both readers within
5%
to 10%. Results
are
reported
as
the mean
L
standard error (SE)
of
colonies enumerated.
Colony forming units-erythroid (CFU-E) were cultured
for
7
days
and then enumerated after staining with
I
%
benzidine and hematoxy-
lin as previously described.’” Colony-forming unit granulocyte-mac-
rophage (CFU-GM) were cultured and identified as previously de-
scribed.3”
In
Vitro Synthesis
of
Recombinant Human Chemokines
The construction of the T,-promoter expression vectors for PF4,
NAP-2, AELRPF4, in which the N-terminal amino acid sequence
of
PF4 preceding the first cysteine residue is replaced with the amino
acids A-E-L-R, and NAP-2“2’A in which the second amino acid
residue of NAP-2 is mutated from glutamic acid to alanine has
been previously described.” These vectors were used to express
recombinant proteins in
Escherichia coli
BL21(DE3)pLys
S
(Nova-
gen. Madison,
WI),
which were allowed
to
grow to an optical density
(OD),, of -0.9, which was followed by an additional
3
hours of
growth in
1
mmol/L
isopropyl-thiogalactopyranoside
that induced
recombinant protein expression.
The recombinant proteins were processed as previously
de-
~cribed.’~.~~ Bacteria were centrifuged at
3,OOO
rpm
in
a Sorvall
GSA
rotor
for
10
minutes and then resuspended in
%,,th
the volume
of
TED
(50
mmollL Tris HCI, pH
8.0;
I
mmol/L EDTA; and
I
mmotl
L dithiothreitol
[D=]).
The cells were recentrifuged and resus-
pended in the same volume of TED with 0.1 mg/mL of lysozyme
added for
30
minutes. The lysate was sonicated at 4°C three times
with a Branson Soniiier (Branson Ultrasonics, Danbury, CT) using
a
microtip at a power of 6 for
1
minute each cycle. The samples
were centrifuged
for
10 minutes in a Sorvall GSA rotor for l0
minutes at
10,000
rpm, and the supernatants were collected and
stored.
Recombinant proteins were purified from the supernatants of bac-
terial lysates described above by a two-step procedure. Initially, they
were applied to a heparin agarose column (Sigma Chemical CO,
St
Louis, MO), washed with TE buffer (50 mmoln Tris HCl, pH 8.0.
and
1
mmoa EDTA) containing 0.15 moln NaCl and eluted with
TE buffer containing 0.5 mollL NaCl for all the NAP-2 proteins and
1.5
moUL NaCl for all the PF4 proteins. The eluted proteins were
concentrated using YM3 Amico ultrafiltration filters (W.R. Grace
CO,
Beverly, MA), and the buffer was switched to
0.1
%
trifluoroace-
tic acid. The concentrated samples were further fractionated on re-
verse phase high performance liquid chromatography (HPLC).
All purified proteins were run on
20%
precast sodium dodecyl
sulfate-polyacrylamide gel electrophoresis (SDS-PAGE) under re-
duced condition followed by Coomassie blue staining. Further identi-
fication was done by enzyme-linked immunosorbent assay (ELBA1
as previously described.” Recombinant protein concentrations were
determined using a Coomassie Protein Assay Reagent Kit with bo-
vine serum albumin as a standard (Pierce, Rockford, IL).
Recombinant 72 amino acid IL-8 protein was purchased (R
&
D
Inc, Minneapolis, MN), and MIP-
la
and
0
were generous gifts from
Drs Barbara Sherry and Anthony Cerami (Rockefeller University,
New York, NY).” Medium containing the CC chemokine C-l0 and
control medium were generously provided by Dr Mark Berger
(Uni-
versity
of
Pennsylvania, Philadelphia).
For personal use only. by guest on July 13, 2011. bloodjournal.hematologylibrary.orgFrom
CHEMOKINE REGULATION
OF
HUMAN MEGAKARYOCYTOPOIESIS
2561
IL-8Ra and
IL-8RP
Platelet Northern Blot
The IL-8Ra and IL-8RP cDNAs were kindly provided by Dr
Ingrid
U.
SchraufsWer (Scripps Institute, La Jolla, CA) in the ex-
pression vector ~sFFV.neo.~~ The
-
1.2-kb coding regions for both
the IL-8Ra and IL-8RP cDNAs, the 3.3-kb platelet glycoprotein
IIb
(GPIlb), and the 0.4-kb PG cDNA inserts”-38 were released from
their respective vectors by
EcoRI
digestion.
All
of the above cDNA
inserts were purified from an agarose gel using GENECLEAN
(BiolOl, Vista, CA). These inserts were then labeled using (Y-~~P-
dCTP, random primers, and Klenow to
-lo*
CPM/pg DNA.
Total platelet RNA was prepared
from
platelet rich plasma (PRP)
obtained from
100
mL
of sodium citrate anticoagulated blood. Only
the top two-thirds
of
the PRP was used. After spinning down the
platelets, platelet RNA was prepared using the guanidinium thiocya-
natdacid phenol single-step
RNA
isolation te~hnique.’~ The amount
of RNA applied per lane was equivalent to -25% of the total yield.
The RNA was fractionated on gels containing formaldehyde fol-
lowed by transfer to Genescreen Plus membrane (DuPont CO,
Wil-
mington, DE) and hybridization to the labeled probe as previously
described.40
Reverse Transcriptase-Polymerase Chain Reaction
of
Purijied Megakaryocyte
mRNA
Human megakaryocytes were isolated
from
the marrow of remu-
nerated, normal addt volunteers. The isolation
of
greater than
99%
pure megakaryocytes was accomplished as previously described
us-
ing counterflow centrifugal elutriation (Beckman J2-21M; Standard
elutriation motor; Beckman Instruments; Mountainview, CA) to ob-
tain an initial megakaryocyte enrichment:’ followed by isolation
of
morphologically recognizable mature megakaryocytes with a micro-
manipulator. RNA was isolated from
-50
megakaryocytes in less
than 3 hours’ time.
mRNA was isolated from these cells using the Quick-Pre mRNA
Purification Kit (Pharmacia, Piscataway, NJ). The final mRNA pellet
was washed with 75% ethanol and resuspended in
10
pL of triple
distilled autoclaved water. Reverse transcription
of
the megakaryo-
cyte mRNA was performed using
4
pL of the original sample heated
to 65°C for
10
minutes and then cooled on ice for 3 minutes. A total
of 100
U
of Moloney murine leukemia reverse transcriptase
(RT)
(GIBCO BRL, Gaithersburg, MD), 50 ng of random primers (Boeh-
ringer Mannheim, Indianapolis, IN), 40
U
of RNAzin (Promega,
Madison,
WI),
and dNTPs (50 pmoVL each) were added to the tube
and incubated for
1
hour at 37°C. Specific
oligo
primer pairs
used
for
the polymerase chain reaction
(PCR)
amplification with the mega-
karyocyte random cDNA were as follows: II-8Ra: 5“ATGTCA-
AATATTACAGATCC-3’ (sense oligonucleotide;
1
to
20
base,
be-
ginning at the transcriptional start site) and 5”AGATTCATAGAC-
AGTCCCCA-3’ (antisense,
500
to 481 base); and IL-8Rp: 5’-
GAGGACCCAGGTGATCCAGG-3’ (sense, 816 to 835 base) and
5‘-GAGAGTAGTGGAAGTGTGCC-3‘
(antisense, 1065 to 1046
The anticipated PCR products are, therefore, 500 bp and
249 bp for IL-8Ra and
P,
respectively. Ten microliters of the reverse
transcription reaction
or
100
ng of the appropriate IL-8R cDNA or
water was used in a 100-pL PCR reaction using
30
ng of both sense
and antisense primers for each receptor and 2.5
U
Taq polymerase
(Promega) using manufacturer’s supplied buffer and 2.5 pmoVL
MgClz. PCR conditions were melting at 95°C
for
1 minute, annealing
at 60°C, and extension at 72°C for 30 rounds. A total of
10
pL
of
each final PCR product was size-fractionated on a 1.2% agarose gel.
RESULTS
Hematopoietic Effects
of
Recombinant PF4
We first determined whether rH PF4 made in a prokaryotic
system would manifest effects on megakaryocytopoiesis
similar to those we previously reported with highly purified,
serum-derived material. Mature
rH
PF4 protein was synthe-
sized in
Escherichia coli
as described in Materials and Meth-
ods. Its amino acid sequence was identical to the native
protein, except that the recombinant material retained
an
initiating methionine residue.34 Biologic integrity of the engi-
neered protein was demonstrated in chemotactic studies
where the rH PF4 was as effective as serum-derived PF4.34
In a plasma clot assay, both recombinant and native material
inhibited megakaryocyte colony formation in an identical
manner throughout the dose range tested (Fig
1A).
In
agreement with our previously reported results, inhibition
was dose-dependent and most significant at concentrations
2
10
pg/mL.I6 Also
in
agreement with our previous results,
the numbers of cells/colony (Fig 1B) and the size of cells
comprising the colonies (Fig 1C) were also diminished in the
presence of exogenous PF4. Finally, inhibition was mega-
karyocyte lineage specific (data not shown).
NAP-2, AELWPF4 and Mutation Proteins
We have previously shown that PTG does not inhibit in
vitro megakaryocytopoiesis.’6 However, it is now known that
a neutrophil cathepsin G N-terminal cleavage product
of
PTG, a 70-amino acid protein termed NAP-2,
is
a biologi-
cally active form of this protein. NAP-2 is a potent activator
of neutrophils, binding to the IL-8RP on neutrophils.26 To
extend our structure/function studies and to begin to under-
stand the mechanism by which chemokines inhibit megakary-
ocyte development, we tested whether NAP-2, as opposed
to PTG, would inhibit megakaryocytopoiesis. As shown in
Fig 2A, megakaryocyte colony formation was much more
sensitive to the inhibitory effects of NAP-2 than PF4. Where
PF4
inhibited in the
pg/rnL
range, half maximal inhibitory
concentrations of NAP-2 were in the
10
to
100
ng/mL range.
These concentrations were comparable to those needed for
NAP-2 to activate
neutrophil^.^^
As was the case with PF4,
NAP-2’s inhibitory effect was lineage-specific. It has pre-
viously been reported that at PF4 concentrations
2
10 pg/mL
inhibition of CFU-E and Cm-GM might
be
~bserved.”.’~
However, we did not observe such effects
in
our cultures
and
no
definitive changes in CFU-E or CFU-GM colony
formation were observed with increasing doses of NAP-2.
We also sought to determine if NAP-2 inhibited mega-
karyocyte development directly,
or
indirectly via a secondary
effect on accessory marrow cells.
To
address this question,
marrow mononuclear cells were depleted of monocyte-mac-
rophages and T-lymphocytes and then enriched for
CD34+
cells using immunomagnetic beads before exposure to NAP-
2.
Purity of the CD34+ cells in this population consistently
exceeded
85%.
At
50
pg/mL, PF4 decreased the number of
megakaryocyte colonies
to
8%
of the control, and at
750
ngl
mL, NAP-2 decreased expression to
10%
of the control.
These results support the hypothesis that the chemokines
tested exerted a direct inhibitory effect on megakaryocyte
progenitor cells.
Because NAP-2 is thought to activate neutrophils through
IL-8R0, we tested other NAP-2 and PF4 mutant constructs
whose interactions with the neutrophil IL-8 receptors have
been previously defined.33
For
example, alanine substitutions
For personal use only. by guest on July 13, 2011. bloodjournal.hematologylibrary.orgFrom
2562
GEWIRTZ
ET
AL
I I
I I
I
I
I
I
0
5 10 15 20 25 30 35
40
PF4
@g/ml)
no
PF4
supplement
0
5
10
15 20 25 30 35
40
PF4
(pg/ml)
PF4
supplement
Fig
1.
Inhibition of megakaryocytopoiesis by rPF4.
(A)
Percent decrease
in
the number of colonies seen per
lo5
marrow cells plated at
increasing concentrations
of
PF4.
(01,
rH PF4;
(0).
native PF4.
(B)
The total numbers
of
cells seen per colony
with
increasing concentrations
of rPF4 are shown.
(C)
Immunofluorescence of typical megakaryocyte colonies with (bottom) and without rH PF4 (top), demonstrating the
decrease
in
the size
of
the colony and
of
the megakaryocytes within the colony. Data
in
(A)
and
(B)
represent analysis of
two
or more separate
experiments with four separate plates for each data point. The number of megakaryocyte colonies seen without added chemokine ranged
from
56
to
107
per plate.
at the N-terminus of NAP-2 markedly inhibit its ability
to
activate neutrophils. Interestingly, NAP-2""*, in which the
second amino acid
is
mutated from glutamic acid to alanine
(Table
l),
also
loses its ability to inhibit megakaryocyto-
poiesis (Fig 2B). In contrast, AELRPF4,
in
which PF4's
first eight amino acids proximal
to
the first cysteine residue
are replaced with the four NAP-2 N-amino acid residues
preceding its first cysteine residue, is equivalent
to
NAP-2
in terms of its ability
to
activate neutrophils and to inhibit
megakaryocyte colony formation (Fig 2B).
Inhibitory Effects
of
Other Chemokines
We tested additional chemokines to see whether they too
could inhibit megakaryocytopoiesis.
IL-8
was almost as ef-
fective
as
NAP-2
in
inhibiting megakaryocytopoiesis
in
the
5
to
500
ng/mL concentration range (Fig 3A). Surprisingly,
the
CC
chemokine MIP-la was equally effective (Fig 3A).
Inhibition with both of these chemokines was again noted
to be lineage specific (data not shown). Two additional
CC
chemokines were tested and
also
inhibited megakaryocyto-
poiesis. MIP-
10
decreased megakaryocyte colony formation
to
52%
of the untreated control value at
500
ng/mL, and
a
culture supernatant containing
C-IO,
another member of the
CC
chemokine family, decreased megakaryocyte colonies to
35% of control values at
a
concentration of 2% (vol/vol).
We then sought to determine whether the inhibitory effects
of the
CXC
and
CC
chemokines might be additive
or
syner-
gistic. Progenitor cells were therefore cultured with the indi-
vidual chemokines
at
varying doses,
or
with the two added
together. As shown in Fig 3B, the combination of NAP-2 and
MIP-la had greater inhibitory effect than either chemokine
individually. There does not appear
to
be
a
synergistic effect.
For personal use only. by guest on July 13, 2011. bloodjournal.hematologylibrary.orgFrom
CHEMOKINE REGULATION
OF
HUMAN MEGAKARYOCYTOPOIESIS
2563
"-1
B
'"7
0'
d
;OO
1'01
1'02
1'0s
1'04
1'0s
1'06
1'07
NAP-2
(ng/ml)
Fig 2. inhibition
of
m.gakaryocytopoi.rh by NAP-2 and
related
constructs. (A) Percant
decr.rw
in
the
number
of
megakaryocyte
colonies aeon par
IC
marrow caih plated at incroasing concentra-
tions of NAP-2.
(01,
CFU-Meg colonies;
(m),
CFU-GM colonies; and
(AI,
CFU-E colonin. PM studlr
at
25 pglmL were
also
done.
(0).
CFU-Mag colonies;
(U),
CFU-GM colonies; and
(A),
CFU-E colonies.
Error bar
nfm
to
OM
standard devlatlon for rmpln studied four
or
mora
times.
IBI
Analyois of CFU-Meg colony numh for PF4
(01.
NAP-2
(01,
NAP-2- (X),
and
AEWPF4 (t).
Data
In (AI and
(B)
analysis of
two
or
mora
.rpernte
exporimento
wlth
four
soparota plates for rch
doto point. The numbor of megakaryocyte
colonin mn without added chemokine ranged
from
51
to
128
per
pinta.
Rather, the combination of
NAP-2
and
MIP-la
better ap-
proximates an additive effect. If the inhibition observed
is
additive, CXC and CC chemokines may inhibit megakaryo-
cyte development by signaling through a common pathway.
It should also
be
noted that even at the highest combined
chemokine dose, megakaryocyte colony formation was not
completely extinguished. When compared with an untreated
control group,
-30%
of maximal colony formation was still
observed even at the highest combined dose level. The nature
of the difference@) between
this
resistant subpopulation and
those progenitor cells that were sensitive to the chemokine
inhibitory effects remains to be determined.
Finally, we also determined whether the chemokine inhib-
itory effect could be abrogated by MGDF stimulation. We
first established that, in our culture system, megakaryocyte
colony growth was maximal at MGDF doses -20 ng/mL.
At such doses, the number of colonies observed in the culture
dishes was comparable to the number observed when optimal
concentrations of aplastic anemia serum plus
L-3
and
E-6
were used as stimulators. We then cultured progenitor cells
in the presence of
50
ng/mL of MGDF and varying doses
of either NAP-2 or MIP-la. As shown in Fig
4,
in compari-
son to the numbers of colonies obtained in the presence of
MGDF alone, NAP-2 at
25
ng/mL and MIF"1a at
>50
ng/
mL
significantly inhibited colony formation. Therefore, even
in the presence of a significant physiologic stimulator, used
at maximally effective concentrations, these chemokines ap-
peared capable of blunting megakaryocyte colony growth.
Demonstration
of
the Expression
of
IL-8Ra
and
IL-8Rp
by
Megakaryocytes
The above studies suggested that chemokines exert a di-
rect effect on megakaryocyte development. If
our
interpreta-
tion of the experiments was correct, it follows that megakary-
ocytes should express the receptors that bind these ligands.
To
provide these important data, we pursued two indepen-
dent, but complementary approaches. First, we isolated total
platelet RNA and looked for
E-8Ra
and
IL-8R@
-A.
The presence of these messages in the total RNA pool would
provide evidence that these receptors might, in fact, be ex-
pressed by megakaryocytes. We also performed RT-PCR for
these same messages on an essentially pure population
of
normal human megakaryocytes.
Platelet
RNA
was extracted
from
the top two-thirds
of
platelet-rich plasma of low-speed centrifuged blood. White
cell contamination was approximately
1
cell per
5,000
plate-
lets. Duplicate lanes of total RNA were hybridized to cDNAs
for the
L-8Ra
and
IL-8RP
and platelet GPIIb and PG as
platelet-specific positive controls. As seen in Fig 5A, there
were single detectable bands
of
the expected size of
-3
kb
for both of the L-8 receptors and
3.3
kb for GPIIb.38.42 An
intense signal was detected with pTG at
0.8
kb with addi-
tional bands seen at
-
1.2 and 1.8 kb.
Total
peripheral blood
neutrophils detect
two
major L-8R bands of
2.4
and
3.0
kb,
Table
1.
Recombinant
PF4
and NAP-2 Chemoklnes
Name Amino Acid Sequence
NAP-2 (wild
type)
AELRCMC
.
.
.
THCN
.
.
.
DGRKICLDPDAPRIKKIVQKLAGDESAD
PF4 (wild
type)
EAEEDGDLQCLC
. .
.
PHCP
. .
.
NGRKICLDLQAPLYKKIIKKLLES
AELWPF4
eCLC.
.
.
PHCP
. .
.NGRKICLDLQAPLYKKIIKLLES
NAP-2EW
+RCMC.
. .
THCN.
.
.
DGRKICLDPDAPRIKKIVQKLAGDESAD
Amino acid differences from wild
type
are underlined.
For personal use only. by guest on July 13, 2011. bloodjournal.hematologylibrary.orgFrom
2564
GEWIRTZ
ET
AL
while HL60 cell lines only express the
3.0
kb band, similar
to the single band we detected
in
platelet RNA.4'
Even though the blots depicted
in
Fig
5
were hybridized
under high stringency conditions,
it
is possible that we were
only detecting a single IL-8R species because of the
-70%
homology
in
the coding regions of the IL-8R cDNAs. There-
fore, to provide additional proof that we were,
in
fact, de-
tecting both IL-8Ra and IL-gRP, we performed RT-PCR
reactions on mRNA isolated from a small number of
100%
2
-
00
00
-
g g
pure normal human megakaryocytes!3 Primers chosen were
om
33
Oa
Oa
0
7
\a
m=
unique for the two IL-8 receptors, as shown by the fact that
cDNAs (data not shown). These RT-PCR studies demon-
cytes (Fig
5B).
m
0-
2;.a2\azYz
\a
gY
the primers do not yield cross-amplify using IL-8 receptor
5
-
"
d
2.5
IL-8
MIP-1
a
NAP-2
strate that both IL-8 receptors are expressed
in
megakaryo-
okTLXO
Chemokine added (ng/ml)
Fig
3.
Inhibitory
effects
of
CXC
and
CC
chemokines on megakary-
ocytopoiesis. (AI The inhibitory effect of 11-8 and MIP-la on mega-
karyocyte formation relative to an untreated control. NAP2 data
done simultaneously are also included. Data represent the average
of
two
experiments with four plates read for each point
in
each exper-
iment. Error bars refer to one SD. At every concentration shown, the
inhibitory effect was significant at P
c
.003.
(B)
Effect of combined
treatment with NAP-2 and MIP-la. Megakaryocyte colony formation
was tested with NAP-2
(W,
MIP-la
(01,
or both
(XI.
Error bar refers
to
1
SD
for samples studied four or more times. Data
in
(A) and
(El
represent analysis of
two
or more separate experiments with four
separate plates for each data point.
100461
T
0.04
0.009
T
0.y02
S
5'0
500
NAP-2
(ng/ml)
10
DISCUSSION
We have previously shown that PF4, a platelet-specific
CXC chemokine, can directly inhibit
in
vitro megakaryocy-
topoiesis. We now show that other CXC chemokines, NAP-
2,
IL-8, and even the more distantly related CC chemokines,
MIP-la and
P,
and C10, have a similar direct inhibitory
effect
on
in
vitro megakaryocyte development as manifested
by the appearance of fewer colonies composed of smaller
numbers of less mature cells. Inhibition is observed when
chemokines are added to progenitor cell cultures at concen-
trations equivalent to those at which they activate neutrophils
and monocytes,-" suggesting that the inhibitory signals they
generate may be of physiologic significance. Studies per-
formed with mutated NAP-2 and PF4 proteins also support
this hypothesis as the ability
to
activate neutrophils and to
inhibit megakaryocytopoiesis closely parallel each other.
Another highly suggestive finding is that megakaryocytes
express both
a
and isoforms of the IL-8 receptor. Never-
theless, although the presence of these receptors provides a
mechanism for the observed inhibitory effects of some of
the CXC chemokines, further studies will be needed to define
the
full
repertoire of megakaryocyte chemokine receptors
and,
in
particular, whether CC chemokine receptors also
exist on these cells.
0
.L8
5
5'0
560
MIP-la
(ng/rnl)
Fig
4.
Interaction of MGDF and chemokines.
In-
hibitory studies with NAP-2 and MIP-la were per-
formed
in
the presence of
50
ng/mL of rH MGDF.
Data represent the average of
two
experiments with
four plates read for each point
in
each experiment.
Error bars refer to one
SD.
The Pvalue for each study
is shown above the error bar.
For personal use only. by guest on July 13, 2011. bloodjournal.hematologylibrary.orgFrom
CHEMOKINE REGULATION
OF
HUMAN MEGAKARYOCYTOPOIESIS
A
W
l-
c?!
7.5
kb
m
4.4
2.4
t
I
B
M1
2
3123
"
I
L-8
Ra
IL-8RR
Fig
5.
Expression of IL-8Ra and
f3
RNA in megakaryocytes. (A)
Northern blot analysis of total platelet RNA probed with the
cDNAs for pTG, GPllb, IL-8Rrr, and IL-8Rf3.
(B)
Agarose gel of the
RT-PCR products of IL-8Ra and
p.
Lane
1,
mRNA from isolated
megakaryocytes, Lane
2,
the cDNA for appropriate receptor, and
Lane
3,
water control. Size markers are indicated at the side of
both (A) and
(B).
2565
Although many stimulatory cytokines have been described,
relatively few inhibitory proteins have been elucidated. The
best-studied examples of these include the interferons
(INFs),
tumor necrosis factor-a
(TNF-a),
transforming growth factor-
p
(TGF-p), and MP-la.""' The physiologic role of these
cytokines
in
the regulation of human hematopoiesis remains
speculative.44 This is,
in
part, because the biologic effects of
these cytokines are context dependent, a situation that has
tended to generate apparently contradictory reports on their
activity. For example, MP-la appears to specifically inhibit
the growth of cycling hematopoietic cells,46 but when admixed
with IL-3
or
GM-CSF appears to promote colony
growth
in
a
synergistic manner:' In addition,
TNF-a
and the
INFs
appear
to be general suppressers of hematopoietic cell growth, while
the activity of TGF-p is similar to MIP-la
in
that it appears
to block proliferation of growth factor stimulated progenitor
cells, but alone may stimulate CFU-GM and
CFU-E.
These
observations have prompted speculation that some of these
inhibitors,
in
particular TGF-p and MP-la, may play a role
in
maintaining stem cells
in
a
Go
state when hematopoiesis is
otherwise stimulated." In support of this hypothesis,
it
has
been
reported that inhibition of TGF-p expression with antisense
DNA
increases the apparent number of assayable multilineage
progenitor cells
in
cord blood?R These apparently discordant
results may also be explained by the observation that the inhibi-
tory effects of MP-la and TGF-p depend on the maturation
of the progenitor that has been exposed to this chemo-
Many investigators have reported that MP-la
either has no effect
on,
or
actually stimulates the growth
of
more mature progenitors, while it suppresses the growth of
more primitive
C~IIS~~~~~
The fact that the chemokines we have
investigated have no apparent effect on CFU-GM derived col-
ony formation is
in
accord with these
results.
Why the appar-
ently mature CFU-Meg are inhibited by these chemokines when
CFU-GM are not, remains unknown.
Whether the findings we report are of physiologic signifi-
cance remains unclear. The fact that members of this family
can inhibit megakaryocyte development at concentrations
three orders
of
magnitude lower than PF4 is suggestive
of
physiologic relevance because concentrations in this range
are likely achievable
in
vivo. Further, the ability to have
significant inhibition despite maximal levels of
TPO
also
supports a potential in vivo role. Nevertheless, establishing
whether there is a correlation between megakaryocyte/plate-
let mass and chemokine concentrations, either in plasma
or
in
the marrow, will have to be investigated. This will be
particularly important
in
patients with inflammatory states
who may be expected to have relatively high circulating
chemokine levels, and perhaps paradoxically, reactive
thrombocytosis. Measurement of MGDF levels will
be
of
equal importance then, as we have already speculated that
stimulatory growth factors may overdrive the effects of ap-
parently weaker negative regulators.'" Indeed, we have also
hypothesized that negative regulatory loops, whether auto-
crine
or
paracrine
in
nature, are more likely to play a role
in
regulating basal,
or
nonstimulated, megakaryocyte pro-
duction." If shown to be of physiologic significance, these
studies may represent a significant advance
in
the develop-
ment of new pharmacologic strategies to regulate disordered
or
inappropriate thrombopoiesis.
kine.4".49.%1
For personal use only. by guest on July 13, 2011. bloodjournal.hematologylibrary.orgFrom
2566
GEWIRTZ
ET
AL
ACKNOWLEDGMENT
We thank Dr Pamela Hunt at Amgen Corp (Thousand Oaks, CA)
for the MGDF, Ingrid
U.
Schraufstlitter at the Scripps Institute (La
Jolla, CA) for the E-8R cDNAs, and Dr Mark Berger at the Univer-
sity of Pennsylvania (Philadelphia, PA) for the C10-containing me-
dium.
REFERENCES
1.
De
Sauvage FJ, Hass PE, Spencer SD, Malloy BE, Gurney
AL,
Spencer SA, Darbonne WC, Hemel WJ, Wong SC, Kuang
W-J, Oles
KJ,
Hultgren B, Solberg LA,
Jr.,
Goeddel DV, Eaton DL:
Stimulation of megakaryocytopoiesis and thrombopoiesis by the c-
Mpl ligand. Nature 369:533, 1994
2. Luk
S,
Kaushansky K, Holly RD, Kuijper JL, Lofton-Day CE,
Oort PJ, Grant FJ, Heipel MD, Burkhead
SK,
Kramer JM, Bell LA,
Sprecher CA, Blumberg H, Johnson R, Prunkard D, Ching
AFT,
Mathewes SL, Bailey MC, Forstrom JW, Buddle
MM,
Osbom
SG,
Evans
SJ,
Sheppard PO, F’resnell
SR
Cloning and expression
of
murine thrombopoietin cDNA and stimulation of platelet production
in
vivo.
Nature 369565, 1994
3. Kaushansky
K,
Lok
S.
Holly
RD,
Broudy VC, Lin N, Bailey
MC, Forstrom
JW,
Buddle
MM,
Oort PJ, Hagen
FS,
Roth
GI,
Papa-
yannopoulou T, Foster DC: Promotion of megakaryocyte progenitor
expansion and differentiation by the c-Mpl ligand thrombopoietin.
Nature 369:568, 1994
4. Bartley
TD,
Bogenberger J,
Hunt
P, Li
Y-S,
Lu HS, Martin
F,
Chang M-S,
Samal
B, Nichol
L,
Swift
S,
Johnson MJ, Hsu
R-Y, Parker
VP,
Suggs
S,
Skrine JD, Merewether LA, Clogston C,
Hsu E, Hokom
MM,
Hornkohl A, Choi
E,
Pangelinan M, Sun Y,
Mar V: Identification and cloning of a megakaryocyte growth and
development factor that is a ligand for the cytokine receptor Mpl.
Cell 77:1117, 1994
5.
Leary
AG, Yang YC, Clark SC, Gasson JC, Golde DW, Ogawa
M: Recombinant gibbon Interleukin
3
supports formation of human
multilineage colonies and blast cell colonies in culture: Comparison
with recombinant human granulocyte-macrophage colony stimulat-
ing factor. Blood 701343, 1987
6. Warren MK, Conroy LB. Rose
JS:
The role
of
interleukin
6
and interleukin 1 in megakaryocyte development. Exp Hematol
17:1095, 1989
7.
Williams N, De Giorgio
T,
Banu
N, Withy R, Hirano T,
Kishi-
mot0 T: Recornbinant interleukin
6
stimulates immature murine
megakaryocytes. Exp Hematol 18:69, 1990
8.
Quesenberry PJ, McGrath
HE,
Williams
ME,
Robinson BE,
Deacon DH,
Clark
S,
Urdal
D, McNiece
IK
Multifactor stimulation
of
megakaryocytopoiesis: Effects
of
interleukin
6.
Exp Hematol
19:35, 1991
9.
Takahashi
T,
Tsuyuoka
R,
Ueda
Y,
Suzuki A, Ichiba
S,
Okuno
Y, Nakamura
K,
Imura
H: Megakaryocyte potentiating activity of
IL-l, IL-6 and GM-CSF
as
evaluated by their action
on
in
vitro
human megakaryocytic colonies. Br
J
Haematol78:480, 1991
10. Kaushansky K, O’Hara P, Bemer K, Segal GM, Hagen FS,
Adamson
W.
Genomic cloning, characterization
and
multilineage
growth-promoting activity of human granulocyte macrophage col-
ony-stimulating factor. Proc Natl Acad Sci USA 83:3101,
1986
11. Briddell
RA,
Bruno
E,
Cooper
RJ,
Brandt
JE,
Hoffman
R
Effect of
c-kit
ligand on in vitro human megakaryocytopoiesis. Blood
78:2854, 1991
12. Gurney
AL,
Carvermoore K, Desavage FJ, Moore
MW:
Thrombocytopenia in c-Mpl-deficient mice. Science 265:1445, 1994
13. Vainchenker W, Chapman
J,
Deschamps
JF,
Vinci
G,
Bouget
J, Titeux M, Breton-Gorius
J
Normal human serum contains a fac-
tor(s) capable of inhibiting megakaryocyte colony formation. Exp
Hematol 10650, 1982
14. Messner HA, Jamal
N,
Izaquirre
C:
The growth
of
large
megakaryocyte colonies from human bone marrow.
J
Cell Physiol
1:45,
1994
15. Kimura HS, Burstein SA, Thorning D, Powell JS,
Harker
LA, Fialkow PJ, Adamson
JW:
Human megakaryocytic progenitors
(CFU-M) assayed in methylcellulose: Physical characteristics and
requirements for growth.
J
Cell Physiol 118:87, 1984
16. Gewirtz AM, Calabretta B, Rucinski
B,
Niewiarowski
S,
Xu
WY: Inhibition of human megakaryocytopoiesis in vitro by platelet
factor 4 (PF4) and a synthetic COOH-terminal PF4 peptide.
J
Clin
Invest 83:1477, 1989
17. Han ZC, Senskbe L, Abgrall JF, Bribe J: Platelet factor 4
inhibits human megakaryocytopoiesis in vitro. Blood 751234, 1990
18. Han ZC, Bellucci
S,
Tenza D, Caen JP: Negative regulation
of
human rnegakaryocytopoiesis by human platelet factor 4 and beta
thromboglobulin: Comparative analysis in bone
mow
cultures
from normal individuals and patients with essential thrombocythae-
mia and immune thrombocytopenic purpura. Br
J
Haematol 74:395,
1990
19. Baggiolini M, Dewald B, Moser B: Interleukin-8 and related
chemotactic cytokines-CXC and CC chemokines. Adv Immunol
55:97, 1994
20. Horuk R: The interleukin-8-receptor family: From chemo-
kines to malaria. Immunol Today 15:169, 1994
21. Mantovani A, Sozzani
S:
Chemokines. Lancet 343:923, 1994
22. Walz A, Baggiolini M: A novel cleavage product of 0-throm-
boglobulin formed in cultures of stimulated mononuclear cells acti-
vates human neutrophils. Biochem Biophys Res Commun 159:969,
1989
23. Walz A, Dewald B, Tscharner
V,
Baggiolini M: Effect
of
neutrophil-activating peptide NAP-2, platelet basic protein, connec-
tive tissue-activating peptide
m,
and platelet factor 4 on human
neutrophils.
J
Exp Med 170:1745, 1989
24. Walz A, Baggiolini M: Generation
of
the neutrophil-activat-
ing peptide NAP-2 from platelet basic protein
or
connective tissue
peptide
111
through monocyte protease.
J
Exp Med 171:449, 1990
25. Holt JC, Yan Z, Lu W, Stewart GJ, Niewiarowski
S:
Isolation,
characterization, and immunological detection of neutrophil activat-
ing peptide
2:
A proteolytic degradation product of platelet basic
protein.
Proc
Soc
Exp Biology Med 199:171, 1991
26. Leonard
El,
Yoshimura
T,
Rot A, Noer
K,
Walz A, Baggiolini
M: Chemotactic activity and receptor binding of neutrophil attract-
ant/activation protein-l (NAP-l) and structurally related host de-
fense cytokines: Interaction of NAP-2 with the NAP-l receptor.
J
Leukoc Biol49:258, 1991
27. Murphy PM, Tiffany
HL:
Cloning of complementary DNA
encoding a functional human interleukin-8 receptor. Science
253:1280, 1991
28. Koch
AE,
Kunkel
SL,
Harlow
LA,
Mazarakis
DD,
Haines
GK, Burdick
MD,
Pope
RM,
Strieter
RM:
Macrophage inflammatory
protein-la. A novel chemotactic cytokine for macrophages in rheu-
matoid arthritis. J Clin Invest 93:921, 1994
29. Neote
K,
Digregorio
D,
Mak JY, Honk R, Schall
TJ:
Molecu-
lar-cloning, functional expression, and signaling characteristics of a
C-C chemokine receptor. J Immunol72415, 1994
30. Ratajczak
MZ,
Kuczynski
W,
Onodera K, Moore
J,
Ratajc-
zak J, Kregenow DA, DeRiel
K,
Gewirtz
AM.
A reappraisal of the
role
of
the insulin like
growth
factor-I (IGF-I) in the regulation of
the human hematopoiesis.
J
Clin Invest 94:320, 1994
31. Gewirtz
AM,
Xu
WY,
Mangan
KF:
Role of natural killer
cells, in comparison with
T
lymphocytes and monocytes in the regu-
lation of
normal
human megakaryocytopoiesis.
J
Immunol 139:2915,
1987
32. Bennett JS, Hoxie JA, Leitman
SF,
Vilaire
G,
Cines DB:
For personal use only. by guest on July 13, 2011. bloodjournal.hematologylibrary.orgFrom
CHEMOKINE REGULATION
OF
HUMAN MEGAKARYOCYTOPOIESIS
2567
Inhibition of fibrinogen binding to stimulated human platelets by a
monoclonal antibody. Proc Natl Acad Sci USA 80:2417, 1983
33. Yan Z, Zhang J, Holt JC, Stewart GJ, Niewiarowski
S,
Poncz
M: Structural requirements of platelet chemokines for neutrophil
activation. Blood 84:2334, 1994
34. Park KS, Rifat
S,
Eck H, Adachi K, Surrey
S,
Poncz M:
Biologic and biochemic properties of recombinant platelet factor 4
demonstrate identity with the native protein. Blood 75:1290, 1990
35. Wang
JM,
Sherry B, Fivash MJ, Kelvin DJ, Oppenheim JJ:
Human recombinant macrophage inflammatory protein-l alpha and
-beta and monocyte chemotactic and activating factor utilize com-
mon and unique receptors on human monocytes. J Immunol
150:3022, 1993
36. Schraufsttitter IU, Barritt DS, Ma M, Oades
ZG,
Cochrane
CG: Multiple sites on
IL-8
responsible for binding to
Q
and
p
IL-
8
receptors.
J
Immunol 151:6418, 1993
37. Majumdar
S,
Gonder D, Koutsis B, Poncz M: Characteriza-
tion of the human P-thromboglobulin gene. Comparison with the
gene for platelet factor 4.
J
Biol Chem 2665785, 1991
38. Poncz M, Eisman
R,
Heidenreich R, Silver SM, Vilaire
G,
Surrey
S,
Schwartz E, Bennett
JS:
Structure of the platelet membrane
glycoprotein
IIb:
Homology to the alpha subunits
of
the vitronectin
and fibronectin receptors.
J
Biol Chem 262:8476, 1987
39. Newman
PJ,
Gorski J, White
GCII,
Gidwitz
S,
Cretney CJ,
Aster RH: Enzymatic amplification of platelet-specific messenger
RNA using
the
polymerase chain reaction. J Clin Invest 82:739,
1988
40. Alwine
JC:
Hybridization analysis of specific RNAs by elec-
trophoretic separation in agarose gels and transfer
to
diazobenzyl-
oxymethyl paper. Gene Amplif
Anal
2565, 1981
41. Gewirtz
AM,
Keefer M, Doshi A, Annamali HC, Chiu C,
Colman RW: Biology of human megakaryocyte factor V. Blood
67:1639, 1986
42. Moser B, Barella L, Mattei
S,
Schumacher C, Boulay F,
Colombo MP, Baggiolini M: Expression of transcripts for two in-
terleukin
8
receptors in human phagocytes, lymphocytes and mela-
noma cells. Biochem J 294285, 1993
43. Gewirtz A, Boghosian-Sell L, Catani
L,
Ratajczak M, Shen
YM,
Schreiber AD. Expression of FcyRII and CD4 receptors by
normal human megakaryocytes. Exp Hematol20:512, 1992
44.
Ogawa M. Differentiation and proliferation of hematopoietic
stem cells. Blood 81:2844, 1993
45. Keller JR, Bartelmez SH, Sitnicka E, Ruscetti
FW,
Ortiz M,
Gooya JM, Jacobsen SEW Distinct and overlapping direct effects
of macrophage inflammatory protein-la and transforming growth
factor
p
on hematopoietic progenitor/stem cell growth. Blood
84:2175, 1994
46. Jacobsen SEW, Ruscetti
FW,
Oniz
M, Gooya
JM,
Keller JR:
The growth response of Lin-Thy-l+ hematopoietic progenitors to
cytokines is determined by the balance between synergy
of
multiple
stimulators and negative cooperation of multipIe inhibitors. Exp
Hematot 22:985, 1994
47. Mantel C, Kim YJ, Cooper
S,
Dwon B, Broxmeyer
HE.
Poly-
merization
of
murine macrophage inflammatory protein la inacti-
vates its myelosuppressive effects in vitro: The active form is a
monomer. Proc Natl Acad Sci USA 90:2232, 1993
48. Cardoso AA,
Li
ML, Batard P, Hatzfeld A, Brown EL, Lev-
esque JP, Sookdeo H, Panterne B, Sansilvestri P, Clark SC, Hatzfeld
J. Release from quiescence of CD34TD38- human umbilical cord
blood cells reveals their potentiality to engraft adults. Proc Natl
Acad Sci USA 908707, 1993
49. Broxmeyer HE, Sherry B,
Lu
L, Cooper
S,
Oh
K-0,
Tekamp-
Olson P, Kwon BS, Cerami A. Enhancing and suppressing effects
of recombinant murine macrophage inflammatory protein
la,
an
inhibitor of primitive hematopoietic cells.
Roc
Natl Acad Sci USA
90:12015, 1990
50. Mayani H, Little MT, Dragowska W, Thornbury
G,
Lansdorp
PM. Differential effects of the hematopoietic inhibitors
MP-la,
TGF-p, and
TNF-a
on cytokine-induced proliferation
of
subpopula-
tions of CD34+ cells purified from cord blood and fetal liver. Exp
Hematol 23:422, 1995
For personal use only. by guest on July 13, 2011. bloodjournal.hematologylibrary.orgFrom
Article
Full-text available
Background Ex vivo production of induced megakaryocytes (MKs) and platelets from stem cells is an alternative approach for supplying transfusible platelets. However, it is difficult to generate large numbers of MKs and platelets from hematopoietic stem cells and progenitor cells (HSPCs). Methods To optimize the differentiation efficiency of megakaryocytic cells from HSPCs, we first employed a platelet factor 4 (PF4)-promoter reporter and high-throughput screening strategy to screen for small molecules. We also investigated the effects and possible mechanisms of candidate small molecules on megakaryocytic differentiation of human HSPCs. Results The small molecule Ricolinostat remarkably promoted the expression of PF4-promoter reporter in the megakaryocytic cell line. Notably, Ricolinostat significantly enhanced the cell fate commitment of MK progenitors (MkPs) from cord blood HSPCs and promoted the proliferation of MkPs based on cell surface marker detection, colony-forming unit-MK assay, and quantitative real-time PCR analyses. MkPs generated from Ricolinostat-induced HSPCs differentiated into mature MKs and platelets. Mechanistically, we found that Ricolinostat enhanced MkP fate mainly by inhibiting the secretion of IL-8 and decreasing the expression of the IL-8 receptor CXCR2. Conclusion The addition of Ricolinostat to the culture medium promoted MkP differentiation from HSPCs and enhanced the proliferation of MkPs mainly by suppressing the IL-8/CXCR2 pathway. Our results can help the development of manufacturing protocols for the efficient generation of MKs and platelets from stem cells in vitro.
Article
Full-text available
Stromal derived factor‐1α (SDF‐1α), the high‐affinity ligand of CXC‐chemokine receptor 4 (CXCR4), was added to human CD34⁺ hematopoietic progenitor cells that can be induced to differentiate along the monocytic or megakaryocytic lineages. In control liquid cell cultures supplemented with two different cytokine cocktails: stem cell factor (SCF), interleukin‐3 (IL‐3), macrophage‐colony stimulating factor (M‐CSF), and 10% fetal calf serum (FCS), or, SCF and thrombopoietin (TPO), the expression of surface CXCR4 progressively increased in both the CD14⁺ monocytic and CD41⁺ megakaryocytic lineages. While SDF‐1α caused only modest effects on cells of the monocytic lineage, it induced profound down‐regulation of CXCR4 in megakaryocytic cells at all stages of differentiation. Moreover, while SDF‐1α initially up‐regulated the early megakaryocytic antigen CD41, at later time points (days 12–16) it induced down‐regulation of the late megakaryocytic antigen CD42b. Consistently, at day 16, the number of mature megakaryocytes was significantly decreased in cultures supplemented with SDF‐1α. These findings indicate that, besides its primary role in regulating the retention of precursor cells in hematopoietic tissues, the SDF‐1α/CXCR4 system participates in the regulation of megakaryocytic development by stimulating the formation of immature megakaryoblasts and inhibiting the formation of mature megakaryocytes. Anat Rec 260:141–147, 2000. © 2000 Wiley‐Liss, Inc.
Chapter
Current models of hematopoiesis stress the stochastic rather than the instructive nature of stem cells differentiation1. These models suggest that the differentiation of pluripotent stem cells is determined by intrinsic transcriptional factors and not by the presence of extrinsic growth factors or their receptors. Growth factors only provide proliferative and activation signals once lineage commitment has been established. Consistent with the stochastic model, it is known that early and aberrant expression of growth factor receptors on hematopoietic precursors does not alter the lineage commitment of these cells2.
Chapter
The recent identification, purification, and cloning of thrombopoietin (TPO), a lineage-specific regulator of platelet production, is the culmination of several decades of research dealing with the regulation of megakaryocytopoiesis (1–5). Although endogenous TPO plays a pivotal role in promoting the proliferation and maturation of megakaryocytc progenitor cells and megakaryocytes, a considerable amount of data exists that indicates that other growth factors are also capable of altering this finely regulated biological process (6,7). These non-TPO regulatory factors are capable not only of promoting megakaryocytc proliferation and maturation, but also of downregulating these cellular processes (6,7). These growth factors might play an important role in the physiological regulation of megakaryocytopoiesis and may be instrumental in the pathogenesis of a number of clinical syndromes (8–12). In addition, recombinant forms of these growth factors have considerable potential in the treatment of clinical disorders of thrombopoiesis (7).
Article
Anagrelide is a treatment option for patients with essential thrombocythemia. Although the clinical efficacy of anagrelide has been established, there is limited knowledge of the molecular mechanism underlying its effect. Here, we evaluated the effect of anagrelide on primary megakaryocytic progenitors from cord blood-derived CD34-positive cells. Anagrelide treatment reduced the expression of megakaryocytic markers (CD41 and CD61). Microarray analysis was performed to characterize gene profiles altered by exposure to anagrelide. The analysis demonstrated upregulation and downregulation (>2-fold) of eight and 34 genes, respectively, in anagrelide-treated megakaryocyte progenitors. This included genes encoding prototypical megakaryocytic proteins, such as PPBP, PF4, and GP6. Gene ontology analysis of genes suppressed by anagrelide treatment revealed significant enrichment of genes involved in platelet activation and degranulation. Expression levels of transcription factors involved in megakaryocyte commitment/differentiation were further evaluated by quantitative RT-PCR, demonstrating significant downregulation of FLI1 and TAL1 in anagrelide-treated megakaryocyte progenitors. Knockdown of TAL1 in primary megakaryocyte progenitors confirmed significant downregulation of FLI1 and megakaryocytic genes. Anagrelide had no significant effect on the surface expression of erythroid markers or on the expression of transcription factors involved in erythroid commitment/differentiation. In conclusion, anagrelide suppresses megakaryocytic differentiation, partly through decreasing the expression of megakaryocytic transcription factors.
Chapter
Chemokines are small proinflammatory peptides that regulate trafficking, activation, and sometimes the proliferation of myeloid, lymphoid, melanocytes, keratinocytes, and endothelial cells (1). The chemokines have been divided into four subfamilies based upon structure and function: the CXC, CX3C, CC, and C chemokines (2–4). The CXC chemokine family includes four MGSA/GRO melanoma growth stimulatory activity/ growth-related oncogene genes (α,β,γ,δ) as well as interleukin 8 (IL-8), gamma interferon-inducible gene (IP-10), monocyte induced by y-interferon (MIG), ENA-78, granulocyte chemotactic protein-2 (GCP-2), neutrophil activating peptide-2, the mitogen for B-cell progenitors known as stromal derived factor-1 (SDF-1), and others (2,5–14). The proteins encoded by these genes exhibit an NH2 terminal cysteine alignment of two cysteines separated by an intervening amino acid (CXC) (2,15–17) (see Table 1). The CXC chemokines that contain an ELR motif at the amino terminus are angiogenic (IL-8, MGSA/GRO, NAP-2, ENA-78, GCP-2), whereas those not containing this motif are angiostatic (MIG, IP-10, PF-4) (18). The murine MGSA/GRO orthologs are KC and MIP-2. The chemokine-β subfamily, noted by two adjacent cysteines (CC) at the N terminus, includes RANTES, MCP-1-3, MIP-1α and β, and numerous others (2,19). Only one γ-chemokine has been identified, lymphotactin, and this chemokine is characterized by a single conserved cysteine in the amino terminus of the protein (3). Lymphotactin is expressed in progenitor T-cells and is chemotactic for lymphocytes but not monocytes or neutrophils.
Article
Dipeptidylpeptidase4 (DPP4)/CD26 truncates certain proteins, and this post-translational modification can influence their activity. Truncated (T)-colony stimulating factors (CSFs) are decreased in potency for stimulating proliferation of hematopoietic progenitor cells (HPCs). T-CXCL12, a modified chemokine, is inactive as an HPC chemotactic, survival, and enhancing factor for replating or ex-vivo expansion of HPCs. Moreover, T-CSFs and T-CXCL12 specifically down-modulates the positively acting effects of their own full length molecule. Other chemokines have DPP4-truncation sites. In the present study, we evaluated effects of DPP4 inhibition (by Diprotin A) or gene deletion of HPC on chemokine inhibition of multi-cytokine stimulated HPC, and on chemokine enhancing effects on single CSF-stimulated HPC proliferation, as well as effects of DPP4-treatment of a number of chemokines. Myelosupressive effects of chemokines with, but not without, a DPP4-truncation site were greatly enhanced in inhibitory potency by pretreating target bone marrow cells with Diprotin A, or by assaying their activity on DPP4/CD26<sup>-/-</sup> BM cells. DPP4-treatment of myelosuppressive chemokines containing a DPP4 truncation site produced a non-myelosuppressive molecule, but one which had the capacity to block suppression by that unmodified chemokine both in vitro and in vivo . Additionally, DPP4-treatment ablated the single cytokine stimulated HPC enhancing activity of CCL3/MIP-1α and CCL4/MIP-1β, and blocked the enhancing activity of each unmodified molecule, in vitro and in vivo . These results highlight the functional post-translational modulating effects of DPP4 on chemokine activities, information offering additional biological insight into chemokine regulation of hematopoiesis.
Article
Macrophage inflammatory protein-1 alpha (MIP-1α) is an 8 kD protein which tends to form aggregates and is a member of the C-X-C chemokine subfamily, being a pro-inflammatory agent and is chemotactic for lymphocytes. MIP-1α has been shown to inhibit haemopoietic progenitor cell proliferation in vivo and in vitro and to reduce haemopoietic stem cell loss with improved neutrophil recovery after radiotherapy or chemotherapeutic agents in vivo.
Article
This randomised double-blind placebo-controlled parallel-group multicentre phase IIa study evaluated the effect of the CXCR2 antagonist AZD5069 on sputum neutrophil counts in adults with bronchiectasis.Patients were randomised 1:1 to receive AZD5069 80 mg or placebo orally twice daily for 28 days. Assessments included blood cell counts, inflammatory markers in blood, morning spontaneous sputum, lung function, safety and tolerability and patients completed daily BronkoTest diary cards. The primary outcome measure was the change in absolute sputum neutrophil count.Of 52 randomised patients, 45 completed treatment, 20 (76.9%) out of 26 receiving AZD5069 and 25 (96.2%) out of 26 receiving placebo. AZD5069 reduced the absolute neutrophil cell count in morning sputum by 69% versus placebo (p=0.004); percentage sputum neutrophil count was reduced by 36% (p=0.008). The number of infections/exacerbations was similar with AZD5069 and placebo (nine versus eight), but these led to more study discontinuations with AZD5069 (four versus zero). Sputum interleukin (IL)-6 and growth-regulated oncogene (GRO)-α and serum GRO-α, IL-1ß and IL-8 levels increased with AZD5069 versus placebo (all p<0.001), while serum high-sensitivity C-reactive protein levels did not change. AZD5069 was well tolerated.AZD5069 markedly reduced absolute sputum neutrophil counts in bronchiectasis patients, although this was not associated with improvements in clinical outcomes in this exploratory study. Copyright ©ERS 2015.
Article
Background Megakaryocytes express and store platelet factor 4 (PF4) in alpha-granules. In vivo, PF4 is a clinically-relevant, negative regulator of megakaryopoiesis and hematopoietic stem cell (HSC) replication. These findings would suggest a regulated source of free intramedullary PF4.Objectives Define the source of free intramedullary PF4 and it's intramedullary life cycle.Methods We interrogated both murine and human bone marrow-derived cells during megakaryopoiesis in vitro by confocal microscopy and ELISA. Using immunohistochemistry, we examined in vivo free PF4 in murine bone marrow before and after radiation injury and in the setting of megakaryocytopenia and thrombocytopenia.ResultsExogenously added human PF4 is internalized by murine megakaryocytes. Human megakaryocytes similarly take up murine PF4, but not the related chemokine, platelet basic protein. Confocal microscopy shows that internalized PF4 co-localizes with endogenous PF4 in alpha-granules and is available for release upon thrombin stimulation. Immunohistochemistry shows free PF4 in the marrow – but not another alpha-granule protein, von willebrand factor. Free PF4 increases with radiation injury and decreases with megakaryocytopenia. Consistent with the known role of low-density lipoprotein receptor related protein 1 (LRP1) in the negative paracrine effect of PF4 on megakaryopoiesis, PF4 internalization is at least partially LRP1-dependent.Conclusions Thus PF4 has a complex intramedullary life cycle with important implications in megakaryopoiesis and HSC replication not seen with other tested alpha-granule proteins.This article is protected by copyright. All rights reserved.
Article
Full-text available
An evaluation of the effects of a recombinant, soluble form of the c-kit ligand alone and in combination with either granulocyte-macrophage colony-stimulating factor (GM-CSF) or interleukin-3 (IL-3) on the regulation of human megakaryocytopoiesis was performed using a serum-depleted clonal assay system and a long-term bone marrow culture system. The effects of the c-kit ligand on the primitive megakaryocyte (MK) progenitor cell, the burst-forming unit-megakaryocyte (BFU-MK), and the more differentiated colony-forming unit-megakaryocyte (CFU-MK) were determined. The c-kit ligand alone had no megakaryocyte colony-stimulating activity (MK-CSA) but was capable of augmenting the MK-CSA of both GM-CSF and IL-3. The range of synergistic interactions of c-kit ligand varied with the class of MK progenitor cell assayed. In the case of the BFU-MK, the c-kit ligand synergistically augmented the numbers of colonies formed in the presence of IL-3, but not GM-CSF, but increased the size of BFU-MK-derived colonies cloned in the presence of both of these cytokines. However, at the level of the CFU-MK, c-kit ligand synergized with both GM-CSF and IL-3 by increasing both colony numbers and size. Although the c-kit ligand alone exhibited limited potential in sustaining long-term megakaryocytopoiesis in vitro, it synergistically augmented the ability of IL-3, but not GM-CSF, to promote long-term megakaryocytopoiesis. These data indicate that multiple cytokines are necessary to optimally stimulate the proliferation of both classes of MK progenitor cells and that the c-kit ligand plays a significant role in this process by amplifying the MK-CSA of both GM-CSF and IL-3.
Article
Full-text available
beta-Thromboglobulin (beta TG) is a platelet alpha-granule protein that is released in large amounts following platelet activation. This protein has approximately 70% amino acid identity with another platelet alpha granule protein, platelet factor 4 (PF4), and approximately 30% identity with other members of a family of proteins encoded by the small inducible genes (SIG) that are involved in the overlapping processes of inflammation, wound repair, and coagulation. Thus, the isolated beta TG gene is of interest in that it may further increase our understanding of beta TG's biological role, the regulation of its megakaryocytic-specific nature of expression, and the evolution and divergence of the SIG family. Using a beta TG cDNA isolated from a DAMI cell cDNA library, we cloned the corresponding beta TG gene. The gene is 1,139 base pairs (bp) long, and, like other members of the SIG family, the beta TG gene is divided into 3 exons. Two major transcriptional start sites were defined by primer extension analysis of platelet RNA, and, based on the more commonly used start site, the 5'-untranslated region is 87 bp. A TATA box is present beginning 32 bp upstream to this site. The first exon contains the 5'-untranslated region as well as the signal peptide. The second exon begins 6 bp 3' to the homologous site in PF4, and the third exon begins at a position homologous to that in PF4. Interestingly, the beta TG and PF4 genes have little detectable homology in the flanking or intronic sequences. In particular, a pyrimidine tract 5' to both the rat and human PF4 genes is not present in the upstream region of the beta TG gene. Genomic Southern blot analysis suggests that, as with the PF4 gene, there are multiple copies of the beta TG gene in the human genome. Chromosomal localization using polymerase chain reaction analysis of human/hamster somatic cell hybrids demonstrated that the beta TG gene, like the PF4 gene, is located on chromosome 4. It is, therefore, possible that the beta TG and PF4 gene form a single genomic locus on chromosome 4 whose members become coordinately activated during megakaryocyte differentiation.
Article
Available evidence indicates that qualitative changes in hematopoietic stem cells and progenitors, such as the decision of stem cells to self- renew or differentiate, or selection of lineage potentials by the multipotential progenitors during differentiation (commitment), are intrinsic properties of the progenitors and are stochastic in nature. In-contrast, proliferative kinetics of the progenitors, namely survival and expansion of the progenitors, appear to be controlled by a number of interacting cytokines. While proliferation and maturation of committed progenitors is controlled by late-acting lineage-specific factors such as Ep, M-CSF, G-CSF, and IL-5, progenitors at earlier stages of development are controlled by a group of several overlapping cytokines. IL-3, GM-CSF, and IL-4 regulate proliferation of multipotential progenitors only after they exit from G0 and begin active cell proliferation. Triggering of cycling by dormant primitive progenitors and maintenance of B-cell potential of the primitive progenitors appears to require interactions of early acting cytokines including IL-6, G-CSF, IL-11, IL-12, LIF, and SF. Currently, this simple model fits our understanding of the interactions of growth factors with hematopoietic progenitors. Naturally the model risks oversimplification of a very complex process. However, because the model is testable, it will hopefully challenge investigators to design new experiments to examine its validity.
Article
Physiological platelet synthesis is thought to require the humoral activities of meg-CSF and thrombopoietin, which respectively promote proliferation and maturation of megakaryocytic cells. A meg-CSF/thrombopoietin-like protein that is present in plasma of irradiated pigs has been purified and cloned. This protein binds to and activates the c-mpl protein, a member of the cytokine receptor superfamily. The isolated MpI ligand shares homology with erythropoietin and stimulates both megakaryocytopoiesis and thrombopoiesis.
Article
THE major regulator of circulating platelet levels is believed to be a cytokine termed thrombopoietin1,2. It is thought to be a lineage-specific cytokine affecting the proliferation and maturation of committed cells resulting in the production of megakaryocytes and platelets. Despite considerable efforts by a number of laboratories, the unequivocal identification of thrombopoietin has proven elusive. Here we report the functional cloning of a murine complementary DNA encoding a ligand for the receptor encoded by the c-mpl proto-oncogene (c-Mpl) 3–5. The encoded polypeptide has a predicted molecular mass of 35,000 (M r 35K). The protein has a novel two-domain structure with an amino-terminal domain homologous with erythropoietin and a carboxy-terminal domain rich in serine, threonine and proline residues and containing seven potential N-linked glycosylation sites. Intraperitoneal injections of mice with recombinant protein increase circulating platelet levels by greater than fourfold after 7 days. These results along with those presented in the accompanying report strongly suggest that the ligand for c-Mpl is thrombopoietin.
Article
Summary We examined whether recombinant cytokines enhance the in vitro platelet production of interleukin-3 (IL-3)-induced human megakaryocytic colonies (Meg-colony). We classified Meg-colonies into four categories based on platelet production during in situ observation on day 14: type 0, absence of cytoplasmic processes in a colony; type 1, one to three processes in at least one megakaryocyte in a colony; type 2, four to eight processes; type 3, more than nine processes or division of cytoplasm. Type 3 colonies were considered to be platelet-producing. In control cultures, type 1 Meg-colonies were dominant, followed by type 2, type 3 and type 0. Of the cytokines added at the initiation of culture, interleukin-1 alpha (IL-1α), interleukin-6 (IL-6), and granulocyte/macrophage colony stimulating factor (GM-CSF) significantly increased the number of colonies. Furthermore, these three cytokines significantly elevated the proportion of type 3 colonies. Interleukin-4 (IL-4), granulocyte-CSF, macrophage-CSF and erythropoietin did not affect the colony count or distribution of colony type. IL-1α. IL-6 and GM-CSF also significantly elevated the proportion of type 3 colonies, even when added to the culture on days 8 or 11. These results indicate that IL-1α, IL-6 and GM-CSF promote platelet production of in vitro Meg-colonies.
Article
The basic culture requirements and several physical characteristics were defined for megakaryocytic colony-forming cells (CFU-M) from normal human marrow growing in methylcellulose. Ficoll-hypaque separated mononuclear cells from human, marrow gave rise to megakaryocytic colonies in the presence of normal human plasma and phytohemagglutinin-stimulated leukocyte-conditioned medium (PHA-LCM). Their identity as megakaryocytic colonies was confirmed by immunofluorescence staining with a monoclonal antibody to human factor VIII antigen and by electron microscopy of individually harvested colonies. Demonstration of the single-cell origin of the colonies was provided by analysis of the glucose-6-phosphate dehydrogenase (G-6-PD) enzyme type of individually harvested colonies grown from a G-6-PD heterozygote. The colonies grew best in heparinized or citrated plasma as opposed to serum. Detailed studies suggested that platelet-release products were responsible for this difference. Tritiated thymidine suicide studies showed that the percentage of CFU-M in DNA synthesis was 23 ± 8% (n = 10). The modal velocity sedimentation rate of CFU-M was 4.9 ± 0.6 mm/hr (n = 4) while that of concurrently studied granulocyte/macrophage colony-forming cells (CFU-GM) was 5.7 ± 0.5 mm/hr. Examination of the PHA-LCM dose-response characteristics suggested the presence in the conditioned medium of an inhibitor to megakaryocyte colony growth which was partially removed by chromatography of the medium on Sephadex G-100. The resulting conditioned medium increased the cloning efficiency for CFU-M compared with that with crude PHA-LCM (15.3 ± 7.0 and 8.2 ± 5.3/105 marrow cells, respectively).
Article
Neutrophil-activating peptide 2 (NAP-2), corresponding to platelet basic protein fragment 25-94, was prepared by chymotryptic digestion of its precursors, low affinity platelet factor 4 or beta-thromboglobulin, followed by purification by high performance liquid chromatography. NAP-2 (0.1-1.5 microns) caused the release of human granulocyte elastase from cytochalasin B-treated neutrophils in a dose-dependent manner. In the same system, beta-thromboglobulin, human platelet factor 4, S-pyridylethyl NAP-2, and platelet basic protein C-terminal fragment (77-94) were inactive, whereas platelet basic protein fragment 22-89 had low, but significant, activity. Sensitive immunological identification of NAP-2 based on nonequilibrium isoelectric focusing and immunoblotting is described.
Article
We studied human megakaryocytes to determine if they both expressed and synthesized Fc gamma and CD4 membrane receptors. The strategy employed relied on demonstration of receptor protein and mRNA in megakaryocytes present in freshly made marrow smears, or in megakaryocytes isolated from aspirated normal bone marrow by counterflow centrifugal elutriation. Protein was detected immunochemically, whereas mRNA was detected either by in situ hybridization, or by reverse transcription, polymerase chain reaction (RT-PCR). Using these methods CD4 and Fc gamma RII protein and mRNA were detected in most megakaryocytes. Fc gamma RI and Fc gamma RIII protein was not detected in these cells. Megakaryocytes were also cultured with recombinant human granulocyte-macrophage colony-stimulating factor (rhGM-CSF) to determine the effect of this growth factor on Fc gamma RII expression. As has been noted in cells of the monocyte-macrophage lineage, exposure to rhGM-CSF resulted in a significant increase in the level of megakaryocyte Fc gamma RII mRNA and protein. These observations are significant because they provide a physiologic basis for known viral trophism displayed by megakaryocytes. They are also of interest because they suggest that alternative portals exist for entry of human immunodeficiency virus (HIV-1) into megakaryocytes and that such infection may play a role in acquired immunodeficiency syndrome (AIDS)-related thrombocytopenia.
Article
We have previously demonstrated that interleukin 3 (IL-3), granulocyte-macrophage colony-stimulating factor (GM-CSF), and granulocyte colony-stimulating factor (G-CSF) stimulate various aspects of megakaryocytopoiesis. We have investigated the capacity of interleukin 6 (IL-6) to stimulate megakaryocyte colony formation from both normal Balb/C marrow and light-density marrow extensively depleted of adherent, pre-B, B and T cells. Human recombinant IL-6 (167 ng/ml) stimulated megakaryocyte colony formation from normal marrow (8.6 +/- 1 megakaryocyte colony-forming units [CFU-meg]/10(5) cells) as compared to control (1.5 +/- 4 CFU-meg/10(5) cells) in 16 determinations (p less than 0.01). IL-6 (167 ng/ml) also stimulated CFU-meg formation from depleted marrow (control, 10.8 +/- 4 CFU-meg/10(5) cells versus IL-6, 68 +/- 19 CFU-meg/10(5) cells in 12 determinations, p less than 0.01). IL-6 synergistically augmented IL-3-induced colony formation (139% IL-3 control, 120% calculated IL-3 plus IL-6 control, n = 11, p less than 0.01) in normal marrow and showed an additive effect in depleted marrow (133% IL-3 control, p less than 0.01, 114% of IL-3 plus IL-6, value not significant [NS] at 0.05 level). Studies with recombinant murine IL-6 gave similar results. There was an increasing level of megakaryocyte colony-stimulating activity from G-CSF (16,667 U/ml, 2.47 +/- 0.6 CFU-meg/10(5) cells, n = 17), to IL-6 (167 ng/ml, 8.47 +/- 0.96 CFU-meg/10(5) cells, n = 19), to GM-CSF (52 U/ml, 23 +/- 4 CFU-meg/10(5) cells, n = 14), to IL-3 (167 U/ml, 48 +/- 5 CFU-meg/10(5) cells, n = 20) as compared to media-stimulated marrow (range 1.29-1.86 CFU-meg/10(5) cells). A similar hierarchy was seen with depleted marrow. Combinations of factors (including IL-3, GM-CSF, G-CSF, and IL-6) tested against normal unseparated murine marrow did not further augment CFU-meg numbers over IL-3 plus IL-6 but did increase colony size. These data suggest that IL-6 is an important megakaryocyte regulator, that at least four growth factors interact synergistically or additively to regulate megakaryocytopoiesis, and that combinations of growth factors, possibly in physical association, might be critical in stimulating megakaryocyte stem cells.