ArticlePDF Available

Abstract and Figures

Human cytomegalovirus (HCMV) infection in immunocompromised patients is associated with impaired immunological function. Blood monocytes, which differentiate into macrophage effector cells, are of central importance for immune reactivity. Here, we demonstrate that HCMV transiently blocks cytokine-induced differentiation of monocytes into functionally active phagocytic macrophages. In HCMV-treated cultures, the cells had classical macrophage markers but lacked the classical morphological appearance of macrophages and had impairments in migration and phagocytosis. Even at very low multiplicities of infection, macrophage differentiation was almost completely inhibited. The inhibition appeared to be mediated by a soluble factor released upon viral treatment of monocytes. Human immunodeficiency virus or measles virus had no such effects. These findings suggest that HCMV impairs immune function by blocking certain aspects of cytokine-induced differentiation of monocytes and demonstrate an efficient pathway for this virus to evade immune recognition that may have clinical implications for the generalized immunosuppression often observed in HCMV-infected patients.
Content may be subject to copyright.
10.1128/JVI.78.19.10378-10389.2004.
2004, 78(19):10378. DOI:J. Virol.
Söderberg-Nauclér
Sara Gredmark, Tamara Tilburgs and Cecilia
Differentiation
Cytokine-Induced Macrophage
Human Cytomegalovirus Inhibits
http://jvi.asm.org/content/78/19/10378
Updated information and services can be found at:
These include:
REFERENCES http://jvi.asm.org/content/78/19/10378#ref-list-1at:
This article cites 63 articles, 39 of which can be accessed free
CONTENT ALERTS more»articles cite this article),
Receive: RSS Feeds, eTOCs, free email alerts (when new
http://journals.asm.org/site/misc/reprints.xhtmlInformation about commercial reprint orders: http://journals.asm.org/site/subscriptions/To subscribe to to another ASM Journal go to:
on June 1, 2013 by guesthttp://jvi.asm.org/Downloaded from
JOURNAL OF VIROLOGY, Oct. 2004, p. 10378–10389 Vol. 78, No. 19
0022-538X/04/$08.000 DOI: 10.1128/JVI.78.19.10378–10389.2004
Copyright © 2004, American Society for Microbiology. All Rights Reserved.
Human Cytomegalovirus Inhibits Cytokine-Induced
Macrophage Differentiation
Sara Gredmark, Tamara Tilburgs, and Cecilia So¨derberg-Naucle´r*
Karolinska Systems Biomedicine Center, Department of Medicine, Center for
Molecular Medicine, Karolinska Institute, Stockholm, Sweden
Received 20 August 2003/Accepted 24 May 2004
Human cytomegalovirus (HCMV) infection in immunocompromised patients is associated with impaired
immunological function. Blood monocytes, which differentiate into macrophage effector cells, are of central
importance for immune reactivity. Here, we demonstrate that HCMV transiently blocks cytokine-induced
differentiation of monocytes into functionally active phagocytic macrophages. In HCMV-treated cultures, the
cells had classical macrophage markers but lacked the classical morphological appearance of macrophages
and had impairments in migration and phagocytosis. Even at very low multiplicities of infection, macrophage
differentiation was almost completely inhibited. The inhibition appeared to be mediated by a soluble factor
released upon viral treatment of monocytes. Human immunodeficiency virus or measles virus had no such
effects. These findings suggest that HCMV impairs immune function by blocking certain aspects of cytokine-
induced differentiation of monocytes and demonstrate an efficient pathway for this virus to evade immune
recognition that may have clinical implications for the generalized immunosuppression often observed in
HCMV-infected patients.
Human cytomegalovirus (HCMV), a member of the herpes-
virus family, is an opportunistic pathogen that causes serious
health problems in transplant recipients and in AIDS patients
(5). It is associated with atherosclerosis, restenosis after coro-
nary angioplasty, chronic rejection in organ transplant patients,
and chronic graft-versus-host disease in bone marrow trans-
plant recipients (15, 28, 51). After a primary infection, HCMV
persists in a latent form, and an estimated 60 to 100% of the
population carries the virus. In the latent phase, the viral ge-
nome exists in an episomal circular form (4) and does not
replicate, enabling the virus to avoid immune recognition. In-
creasing evidence suggests that HCMV infection adversely af-
fects both innate and adaptive immune responses. For exam-
ple, HCMV reduces the expression of HLA class I and II
molecules; inhibits antigenic processing, peptide presenta-
tion, and T-cell activation; confers resistance against natural
killer cells; and interferes with the humoral immune re-
sponse and cytokine-signaling events (reviewed in reference
26).
Neutrophils are considered the major cell type carrying the
virus during acute infection (11). However, monocytes are
thought to be responsible for dissemination of the virus and
are the predominant cell type harboring HCMV in the periph-
eral blood of seropositive individuals (54, 55). In CD14-posi-
tive monocytes, the HCMV genome is maintained at a rela-
tively low copy number (6 to 13 copies/cell) (46). HCMV
infection of monocytes is nonpermissive and restricted to early
events of gene expression (9). The absence of late gene expres-
sion and virus production are consistent with the hypothesis
that monocytes are reservoirs for latent virus. Although the
virus cannot replicate in monocytes, HCMV-infected macro-
phages expressing late viral genes have been identified in tissue
specimens from HCMV-infected patients (12).
Differentiation of monocytes into macrophages is a prereq-
uisite for productive HCMV infection (10, 18, 50, 56). It has
been shown that latent HCMV can be reactivated in differen-
tiated macrophages produced by allogeneic stimulation of pe-
ripheral blood mononuclear cells (PBMCs) (49). It has also
been shown that the inflammatory cytokines gamma interferon
(IFN-) and tumor necrosis factor alpha (TNF-) produced by
allogeneically stimulated T cells are important for the reacti-
vation of latent virus and the growth of HCMV in differenti-
ated macrophages (48). In myeloid cells, TNF-activates the
HCMV immediate-early (IE) promoter (37, 38, 40, 53). These
results suggest that immune activation and production of in-
flammatory cytokines are important for viral reactivation and
replication in HCMV-infected patients.
Since peripheral blood monocytes have a short half-life, the
reactivation of HCMV in macrophages suggests that HCMV
is maintained in a precursor population of the myeloid cell
lineage. Such cells would provide an ideal latency site for a
virus whose activation is closely linked to the immune system.
HCMV can infect CD34
pluripotent stem cells both in vitro
and in vivo (22, 29–31, 45, 62). CD33
granulocyte-macro-
phage progenitor cells derived from fetal liver can also be
infected with HCMV, and their differentiation into CD14
macrophages results in virus production (21, 23). However,
differentiation of all infected cells after viral entry would elim-
inate the virus during the infectious cycle.
We hypothesized that the establishment of latency by
HCMV in myeloid lineage cells affects cellular differentiation
signals. To test this hypothesis, we examined the effects of
HCMV on macrophage differentiation by assessing cell mor-
phology, expression of cell-surface markers, phagocytic activ-
ity, and cellular migration. Our results suggest that HCMV
* Corresponding author. Mailing address: Karolinska Institute, De-
partment of Medicine, Center for Molecular Medicine, L8:03, Karo-
linska Hospital, S-171 76 Stockholm, Sweden. Phone: 46-8-51779896.
Fax: 46-8-313147. E-mail: cecilia.soderberg.naucler@cmm.ki.se.
10378
on June 1, 2013 by guesthttp://jvi.asm.org/Downloaded from
blocks the differentiation of monocytes into functionally active
macrophages, which may provide an efcient way for HCMV
to avoid immune recognition.
MATERIALS AND METHODS
Establishment of macrophage cultures. PBMCs from healthy donors (47)
were plated (Primaria Falcon dishes; Becton Dickinson, San Jose, Calif.) at 10
10
6
to 18 10
6
cells/ml in Iscoves modied medium with 2 mM L-glutamine, 100
U of penicillin/ml, 100 g of streptomycin (Gibco BRL, Grand Island, N.Y.)/ml,
and 10% AB serum. After incubation at 37°C for 2 h, nonadherent cells were
removed, and the cultures were extensively washed and treated with supernatant
containing cytokines produced by an allogeneic reaction between T cells and
monocytes (allo-cytokines) to induce macrophage differentiation (50). Briey,
PBMCs from different donors were mixed and incubated for 24 h in Iscoves
complete medium. The supernatant was collected, cleared by centrifugation, and
used to stimulate separate monocyte cultures. In some experiments, cells were
restimulated once for 24 h at 1, 2, 6, and 18 days after treatment.
Separate monocyte cultures were also stimulated with recombinant IFN-(500
U/ml) and TNF-(10 ng/ml) (R&D Systems, Minneapolis, Minn.) either alone,
in combination, or with lipopolysaccharide (LPS) (1 g/ml; Sigma) or with
phorbol myristate acetate (PMA) (10 ng/ml). Twenty-four hours after stimula-
tion, the cells were washed with Iscoves medium and cultured in complete 60/30
medium (60% AIM-V medium, 30% Iscoves modied medium, 10% AB serum,
L-glutamine, penicillin, and streptomycin). The medium was replaced with fresh
complete 60/30 medium every 3 to 4 days.
HCMV treatment. Monocyte-enriched cultures were washed with phosphate-
buffered saline (PBS) and then mock infected or infected with HCMV at a
multiplicity of infection (MOI) of 1 to 10. The cultures were stimulated at the
time of infection and incubated for 24 h at 37°C. Three HCMV strains were used:
AD169, Towne, and a clinical isolate (PO). Some cultures were infected with
human immunodeciency virus (HIV) (viral titer, 1/10 or 1/1) or measles virus
(viral titer, 1/40 or 1/25); approximately 100% of monocytes are infected after
treatment with virus at these titers (Annika Lindhe and Anders So¨nnerborg
[Karolinska Institutet], personal communication).
Experiments were also performed with strain AD169 or Towne that had been
UV irradiated four times (Auto Cross Link, UV Stratalinker 1800; Stratagene)
or neutralized by incubation with intravenous immune globulin (IVIG) (50
mg/ml; Immuno AG, Vienna, Austria) for1hatroom temperature. Additional
experiments were performed with virus stocks that had been ltered through an
Acrodisc (0.1-m-pore-size lter; Gelman Sciences, Ann Arbor, Mich.) to re-
move 100% of the viral infectivity and with virus stock supernatant that had been
ultracentrifuged at 10,000 rpm for 16 h. The efciency of these respective treat-
ments was tested by inoculating the respective preparations with human lung
broblasts, which showed that these treatments reduced infectivity by 99%.
Experiments were also performed with virus particles that had been pelleted at
10,000 rpm for 16 h and dissolved in 50 mM Tris HCl, 50 mM NaCl, and 10 mM
MgCl
2
at an MOI of approximately 1. Cell-free virus stocks were prepared from
supernatants of infected human lung broblasts, frozen, and stored at 70°C.
Viral titers were determined by plaque assays (60).
In some experiments, supernatants from stimulated, HCMV-treated, or mock-
treated monocytic cultures were collected at 6, 12, or 24 h; cleared by centrifu-
gation; and stored at 70°C until use. These supernatants were ltered to
remove virus particles, diluted 1:10 and 1:100, and used for infection or mock
infection of cultures.
Twenty-four hours after infection or mock infection, the cultures were washed
with Iscoves medium and cultured in complete 60/30 medium. On day 7, the cells
were xed, stained, and analyzed as described below.
Treatment with recombinant proteins and antibodies. At the time of stimu-
lation, the mock-treated cultures were also treated with recombinant IFN-(0.05
to 5 ng/ml) or recombinant IFN-(0.05 to 5 ng/ml), or the HCMV-treated
cultures were treated with antibodies to either IFN-(10 to 1,000 U) or IFN-
(10 to 2,500 U) (all from Biosource International, Camarillo, Calif.). The mock-
treated cultures were also treated with recombinant interleukin-6 (IL-6) (100 to
1,000 ng/ml) or recombinant IL-10 (1 g/ml), and the HCMV-treated cultures
were treated with antibodies to either IL-6 and/or IL-6R (2.5 g/ml) or with
antibodies to either IL-10 (10 g/ml) and/or IL-10R (10 g/ml) (all from R&D
Systems).
Measurement of cytokine production. Cytokine levels in the supernatant of
mock- and HCMV-treated cultures were measured with colorimetric sandwich
enzyme-linked immunosorbent assays (ELISAs) (for IFN-and IFN-, colori-
metric sandwich ELISA; PBL Biomedical Laboratories, Piscataway, N.J.; for
IL-6, IL-10, IL-12, and IL-1, Quantikine human colorimetric sandwich ELISAs;
BD Biosciences, San Jose, Calif.).
Quantication of macrophages. Differentiated macrophages are readily dis-
tinguished from monocytic cells by size, morphology, and the presence of nu-
merous cytoplasmic vacuoles (41). All monocytic cells except histiocytes and
specialized macrophages lack -naphthyl acetate esterase enzyme activity. On
day 7 after mock treatment or HCMV treatment, cultures were xed, stained
with an -naphthyl acetate esterase staining kit (Sigma, St. Louis, Mo.), and
examined with an inverted microscope. The number of macrophages was deter-
mined by counting esterase-positive cells that exhibited a classical macrophage
appearance (histological phenotype).
At least 100 cells were counted in each well, and the percentage of monocytes
was calculated. Approximately 20% of cells in the monocyte-enriched cultures
(both HCMV-treated and mock-treated cultures) are not of monocytic origin,
but these numbers remain constant in different cultures from the same donor.
Hence, small cells can account for approximately 20% of the total cell number
(e.g., 20% of monocytes in mock-treated cultures represents complete differen-
tiation of most monocytic cells in the cultures). All experiments were performed
in duplicate with cells from at least 3 (generally 10) donors. Data are presented
as means the standard error of the mean (SEM). Statistical signicance was
determined by paired, two-tailed ttests.
Flow cytometric analysis. The expression of cellular markers was assessed with
auorescence-activated cell sorter (FACSort; Becton Dickinson). Adherent
macrophages were harvested by being scraped after a 60-min preincubation in
versene (Gibco BRL) at room temperature. The cells were stained with anti-
bodies recognizing different HLA class II molecules (HLA DR, DQ, and DP;
Becton Dickinson); HLA class I molecules (Dakopatts, Glostrup, Denmark); the
cell surface markers CD13 and CD14 (Dakopatts), CD40, CD68, and CD86; the
intracellular adhesion molecule (ICAM); or isotype controls (immunoglobulin
G1 and G2a) (Dakopatts). This was followed by treatment with appropriate
uorescein isothiocyanate (FITC)-conjugated secondary antibodies (Dakopatts).
Expression levels were measured as mean channel uorescence values compared
to the isotype control. The difference in the histogram mean channel value for
uninfected and HCMV-treated cells was calculated on a linear scale; a difference
of more than 10 channels between uninfected and HCMV-treated cells was
considered a positive or negative change, based on variations of controls.
Detection of HCMV replication. Seven days after infection, RNA from mock-
treated and HCMV-treated macrophages was prepared by adding Trizol (Gibco
BRL) to the cell cultures, and RNA was puried as previously described (47).
cDNA was synthesized with a rst-strand cDNA kit (Pharmacia LKB Biotech-
nology, Uppsala, Sweden) and used as a template for HCMV-specic primer
pairs for the major early and pp150 genes in nested reverse transcription-PCR
assays. DNA was prepared as previously described (47). As a positive control for
the detection of DNA or RNA, primers specic for the glucose-6-phosphate
dehydrogenase gene were used for each sample (47). DNA and cDNA samples
from uninfected and HCMV-infected human lung broblasts were included as
positive and negative controls. The PCR products were visualized on 2% agarose
gels.
Phagocytosis assay. Seven days after HCMV treatment, yeast particles labeled
with FITC (42) were added to the cultures (420,000 particles/well in 200 lof
60/30 medium) and incubated at 37°C. In some experiments, restimulation of
HCMV-treated cells was performed 48 h after initial infection, and the cultures
from mock, HCMV, and HCMV-restimulated cells were used after a total of 9
days in culture. As a negative control, cytochalasin D (1 g/ml; Sigma) was added
to the cultures 30 min before the yeast particles. After 10, 30, or 60 min, phago-
cytosis was arrested by moving the plates to 4°C. The medium was replaced with
PBS containing 5% trypan blue to quench uorescence from the extracellular
bound particles. Phagocytosis was quantied by uorescence microscopy. Cells
that had ingested at least one yeast particle were counted after 10 and 60 min
(42). At least 50 to 100 cells/well were assessed in six independent experiments;
each experiment was performed in duplicate.
Migration assay. Chemotaxis assays were performed with 48-well polycarbon-
ate transwell culture chambers with 8-m pores (Costar, Cambridge, Mass.) (27).
Briey, 100,000 cells in 100 l of Iscoves medium containing 10% AB serum, 2
mM L-glutamine, 100 U of penicillin/ml, and 100 g of streptomycin (Gibco
BRL)/ml were added to the upper compartment, and 600 l of serum-free
medium with either 100 ng of RANTES/ml, 100 ng of macrophage inammatory
protein 1(MIP-1)/ml, or 50 ng of monocyte chemoattractant protein 1 (MCP-
1)/ml (all from R&D Systems) was added to the lower compartment. The cells
were incubated for 24 h at 37°C. The lters were washed three times in PBS, xed
in methanol for 5 min, and washed once with PBS. The wells were stained with
Mayer hematoxylin and eosin (Histolab Products, Gothenburg, Sweden). The
cells from the upper side of the lter were removed with a cotton swab, and the
VOL. 78, 2004 HCMV INHIBITS MACROPHAGE DIFFERENTIATION 10379
on June 1, 2013 by guesthttp://jvi.asm.org/Downloaded from
cells on the lower side of each lter were counted in four representative elds by
light microscopy at 40magnication.
RESULTS
HCMV inhibits cytokine-induced macrophage differentia-
tion. HCMV inhibited macrophage differentiation induced by
allo-cytokines in a dose-dependent fashion (Fig. 1A and B). At
an MOI of 1, all three HCMV strains completely inhibited
differentiation (Fig. 1C). HCMV also inhibited macrophage
differentiation induced by recombinant IFN-and TNF-or
by PMA or LPS (Fig. 1D). HCMV-treated monocytes were not
apoptotic (not shown).
AB
0
20
40
60
80
100
1 0.5 0.25 0.05 0.03 0.01 0
Viral Titer (MOI)
% Monocytes
0
10
20
30
40
50
60
70
80
90
100
AD169 Towne PO
% Monocytes
CD
Differentiated
Macrophages
HCMV Treated Cells Unstimulated
Monocytes
0
10
20
30
40
50
60
70
80
90
100
LPS
LPS HCMV
PMA
PMA HCMV
IFN/TNF
IFN/TNF HCMV
% Monocytes
FIG. 1. HCMV inhibits the differentiation of monocytes into macrophages. (A) Dose-dependent effects of infection with AD169 (MOI, 1 to
0.01) on macrophage differentiation. Values shown are means SEM for six separate experiments performed in duplicate. Esterase-negative cells
were subtracted for the data presentation. (B) Phase-contrast microscopy photograph of mock-treated, HCMV-treated (AD169), and unstimulated
monocytes at 7 days. Original magnication, 40. (C) Effects of HCMV infection with AD169, Towne, or the clinical isolate PO at an MOI of
1. Values are means SEM for 10 separate experiments performed in duplicate. (D) Differentiation of monocytes stimulated with LPS, PMA,
or IFN-plus TNF-in the presence or absence of HCMV (AD169). Values are means SEM for three separate experiments performed in
duplicate. The differentiation was dened by esterase staining as well as by morphology.
10380 GREDMARK ET AL. J. VIROL.
on June 1, 2013 by guesthttp://jvi.asm.org/Downloaded from
As shown by immunohistochemical staining, the expression
levels of CD40, CD68, HLA class II molecules, and the ICAM
were higher on differentiated macrophages than on monocytes
but were not affected by HCMV. In HCMV-treated cultures,
the macrophage markers CD13, CD64, CD68, CD86, and
ICAM-1, as well as HLA class I and class II molecules, were
expressed at high levels (Fig. 2A). CD14 levels, however, were
signicantly lower in macrophages than in HCMV-treated or
freshly isolated monocytes (Fig. 2B and C), but varied between
donors. Therefore, the striking differences in morphology of
the cells after HCMV treatment were not accompanied by phe-
notypic differences, indicating that most, but not all, overt
aspects of monocyte differentiation were altered by the virus.
HCMV transiently inhibits differentiation. Next, we as-
sessed the time course of inhibition by treating monocytes with
HCMV at different times after stimulation with allo-cytokines.
HCMV treatment at 12 h inhibited macrophage differentiation
almost completely, but treatment at 48 or 72 h had no effect
(Fig. 3A), suggesting that the virus blocks early steps in differ-
entiation. We then assessed the effects of restimulating the cul-
tures with allo-cytokines for 24 h at different times after HCMV
treatment. In the absence of restimulation, no evidence of dif-
CD86
CD64
CD68
CD40
CD14
180
0
20
40
60
80
100
120
140
160
Monocytes
Macrophages
HCMV-treated
monocytes
***
HLA II
Mean Channel Expression
CD13
HLA I
ICAM
A
B
0
20
40
60
80
100
120
Donor 1
Donor 2
Donor 3
Donor 4
Donor 5
Mean Channel Expression
Monocytes
Macrophages
HCMV-treated
monocytes
C
HCMV
Mock
FIG. 2. HCMV-treated monocytes retain high levels of CD14 expression. (A) Expression of cellular markers by uninfected monocytes,
macrophages, and HCMV-treated monocytes after 7 days in culture as determined by ow cytometric analysis. The data represent mean channel
uorescence values SEM for ve separate experiments. ***, P0.001 (two-tailed, paired ttest; fresh monocytes versus HCMV-treated
monocytes). (B) CD14 expression of donors 1 to 5, comparing the mean channel uorescence values for fresh monocytes, macrophages, and
HCMV-treated monocytes after 7 days in culture. (C) Representative histogram of CD14 expression by HCMV-treated monocytes and macro-
phages, assessed by ow cytometry.
VOL. 78, 2004 HCMV INHIBITS MACROPHAGE DIFFERENTIATION 10381
on June 1, 2013 by guesthttp://jvi.asm.org/Downloaded from
ferentiation was seen for up to 6 weeks. However, restimula-
tion 1 to 18 days after infection induced differentiation (Fig.
3B), which indicates that the viral inhibitory effect is transient.
HCMV-infected monocytes exhibit decreased phagocytosis
and migration. The differentiation of monocytes should be
associated with increases in macrophage functions such as cel-
lular migration in response to chemoattractants and phagocytic
activity. First, we tested the ability of mock-treated macro-
phages and HCMV-treated monocytes to migrate in response
to RANTES, MIP-1, and MCP-1 in a transwell lter system
7 days after infection. Migration of the HCMV-treated cells
at 24 h was reduced by 70 to 90% compared with migration
of mock-treated macrophages (Fig. 4A and B). Restimu-
lated HCMV treated cells were able to migrate in response to
RANTES to the same extent as mock-infected cells (Fig. 4C).
Similarly, phagocytosis of FITC-labeled yeast particles 7 days
after HCMV treatment or mock treatment was about 60%
lower in HCMV-treated monocytes than in mocked-treated
macrophages 60 min after addition of the yeast. Restimulation
of HCMV-treated cells resulted in phagocytosis at levels sim-
ilar to those of uninfected cells (Fig. 5). Cells pretreated with
cytochalasin D for 30 min did not phagocytose any yeast par-
ticles.
Differentiation is inhibited by a component of HCMV and
does not require viral replication. Since viruses other than
HCMV can impair immunological functions, we assessed the
effects of treatment with HIV or measles virus on cellular
differentiation. Neither virus inhibited macrophage differenti-
ation (Fig. 6A). Similarly, virus-free supernatants prepared by
ltration or ultracentrifugation of the medium from HCMV-
infected and mock-infected broblast cultures did not inhibit
differentiation. Concentrated virus particles, however, inhibit-
ed differentiation by 100% (Fig. 6B). Interestingly, pretreat-
ment of the virus with IVIG reduced infectivity by more than
99% (as tested on broblasts; data not shown) but did not affect
HCMVs ability to block macrophage differentiation (Fig. 6C).
To determine if active HCMV replication was required to
block macrophage differentiation, monocytic cultures were
exposed to UV-inactivated, replication-decient HCMV. The
cultures were not infected, but macrophage differentiation was
inhibited to levels similar to those obtained with untreated
virus (Fig. 6C).
A
0
20
40
60
80
100
120
0 3 6 12 24 48 72
Time after Stimulation (h)
% Monocytes
FIG. 3. HCMVs ability to inhibit macrophage differentiation de-
clines with time after stimulation and is reversible. (A) Monocyte-
enriched cultures were stimulated with allo-cytokines; treated with
HCMV 0, 3, 6, 12, 24, 48, or 72 h after stimulation; and cultured for 7
days. Values are the means SEM for six separate experiments
performed in duplicate. Esterase-negative cells were subtracted for the
data presentation. (B) Monocyte-enriched cultures were stimulated
with allo-cytokines, treated with HCMV, and restimulated once for
24h1to18days after infection. Phase-contrast microscopy photo-
graphs are of mock-treated monocyte-derived macrophages, HCMV-
treated monocytes, HCMV-treated monocytes restimulated 6 days af-
ter infection, and HCMV-treated monocytes restimulated 18 days after
infection. The photographs were taken 7 days after restimulation.
Original magnication, 40. The differentiation was dened by ester-
ase staining as well as by morphology.
B
HCMV + restim at 18 days
Mock-treated HCMV-treated HCMV + restim at 6 days
10382 GREDMARK ET AL. J. VIROL.
on June 1, 2013 by guesthttp://jvi.asm.org/Downloaded from
Next, we analyzed RNA and DNA samples from mock-treated
and AD169-treated cells 1 and 6 weeks after infection. In a nested
PCR or reverse transcription-PCR assay with primers specic for
the HCMV major IE and pp150 genes, all HCMV-treated cells
were positive for HCMV DNA at both time points, but HCMV
RNA was not detected (data not shown), consistent with a latent
infection. Control samples from uninfected macrophage cultures
were negative for HCMV DNA and RNA (data not shown).
FIG. 4. HCMV-treated monocytes exhibit decreased migration in response to chemoattractants. (A) Cells were mock treated or treated with
HCMV and cultured for 7 days, and migration in response to RANTES, MIP-1, and MCP-1 was assessed with a transwell culture system. Cells
that migrated were counted by light microscopy in four representative elds at 40 magnication. Values are the means SEM for three separate
experiments performed in duplicate. (B) Phase-contrast microscopy photographs of mock-treated macrophages or HCMV-treated monocytes that
migrated through the lter pores. Original magnication, 40. Arrows are pointing at cells to exemplify migrated cells. (C) Cells were mock
treated, treated with HCMV, or restimulated 1 day postinfection after HCMV treatment. Migration in response to RANTES was assessed after
7 days in a transwell culture system. Values are the means SEM for three separate experiments performed in duplicate.
FIG. 5. HCMV-treated monocytes exhibit impaired phagocytosis. (A) After 7 days in culture, mock-treated and HCMV-treated monocytes
were tested for their ability to phagocytose FITC-labeled yeast particles. Cells that had phagocytosed yeast particles were quantied after 10 and
60 min. Values are means SEM for six separate experiments performed in duplicate. (B) Fluorescence microscopy photographs showing
phagocytosed yeast particles in mock-treated and HCMV-treated monocytes at 30 min. Original magnication, 40. Arrows are pointing at cells
to exemplify phagocytosed FITC-labeled yeast particles. (C) Cells were mock treated, treated with HCMV, or restimulated 1 day postinfection
after HCMV treatment. Phagocytosis of FITC-labeled yeast particles was assessed after 7 days. Cells that had phagocytosed yeast particles were
quantied after 30 min. Values are the means SEM for three separate experiments performed in duplicate.
VOL. 78, 2004 HCMV INHIBITS MACROPHAGE DIFFERENTIATION 10383
on June 1, 2013 by guesthttp://jvi.asm.org/Downloaded from
Monocytes treated with HCMV release a soluble factor that
inhibits differentiation. HCMV infection can alter cytokine
production proles, which could explain why monocytes chal-
lenged with HCMV failed to differentiate into macrophages.
To assess this possibility, we treated the cells with supernatants
collected at different times after HCMV treatment or mock
treatment. After ltration to remove virus particles, these su-
pernatants inhibited macrophage differentiation in a dose-de-
pendent manner, but those from mock-treated cultures did not
(Fig. 7A and B). These observations suggest that a soluble
factor(s) is released from monocytes upon HCMV treatment.
To identify such putative soluble factor that inhibited mac-
rophage differentiation, we tested the supernatants for IFN-,
IFN-, IL-1, IL-6, IL-10, and IL-12 by ELISAs. Only IFN-,
IL-6, and IL-10 were present at higher levels in supernatants
from HCMV-treated cultures than in those from mock-treated
cultures (Fig. 7C to E). Cytokine levels in the virus inocula
were very low or undetectable, and these results thus could not
explain the increased levels in the supernatants from HCMV-
treated cultures (data not shown).
We further assessed the individual effect on macrophage
differentiation of the different cytokines that were increased
upon HCMV treatment. While addition of increasing concen-
trations of recombinant IFN-and IFN-resulted in an inhi-
bition of macrophage differentiation, treatment of cells with
high doses of IL-6 did not (Fig. 8). High doses of recombinant
IL-10 inhibited macrophage differentiation by approximately
20% (Fig. 8B). Furthermore, we performed neutralization ex-
periments and found that antibodies against IFN-inhibited
the viral effect on macrophage differentiation by 25%, whereas
antibodies against IFN-or sheep serum did not affect HCMVs
ability to inhibit macrophage differentiation (Fig. 8A). We were
not able to reverse the effect by HCMV on macrophage dif-
ferentiation by using antibodies specic for IL-10 and/or the
IL-10 receptor (Fig. 8B).
DISCUSSION
This study shows that HCMV blocks cytokine-induced mac-
rophage differentiation. HCMV-treated monocytes remained
small and morphologically distinct from differentiated macro-
phages and exhibited impairments in phagocytosis and migra-
tion. These ndings are consistent with increasing evidence
that immune activation resulting in the production of inam-
matory cytokines leads to macrophage differentiation and re-
activation of latent virus in HCMV-infected myeloid cells (2, 8,
25). Blocking the differentiation of monocytes into functionally
active macrophages may provide an efcient way for HCMV to
evade immune recognition.
The cell surface differentiation marker CD14 was expressed
at high levels by HCMV-treated cells, but its expression de-
creased during the differentiation of mock-treated cells. CD14
was thought to be a marker for myeloid lineage cells at differ-
ent stages of differentiation; however, its reactivity on mono-
cytes and macrophages varies depending on the specicity of
the antibodies used (20, 64). Early progenitor cells are CD14
negative; the majority of primary blood monocytes express high
levels of CD14, while alveolar macrophages are only weakly
immunopositive for CD14 (3, 35). Thus, our ndings imply that
monocytic cells exposed to HCMV are arrested at a stage of
differentiation preceding maturation into macrophages.
The HCMV-induced inhibition of macrophage differentia-
tion appeared to be mediated by a component of the virus
particle and not by soluble factors produced by HCMV-in-
BC
0
20
40
60
80
100
120
Mock
HCMV
HIV 1/10
HIV 1/1
MV 1/25
MV 1/40
% Monocytes
A
0
20
40
60
80
100
120
Mock
HCMV
HCMV
UV
HCMV
IVIG
% Monocytes
0
20
40
60
80
100
120
Mock
HCMV
HCMV-
sup
UC-
HCMV
HL-sup
% Monocytes
FIG. 6. Inhibition of macrophage differentiation is not a general event after virus exposure and is mediated by a structural component of
HCMV. Macrophage differentiation after treatment of monocytic cultures with HIV or measles virus (MV) (A), ultracentrifuged virus-free
supernatants (HCMV-sup), concentrated virus particles (UC-HCMV), or supernatants from mock-infected cultures (HL-sup) (B), or HCMV
inactivated by treatment with UV or IVIG (HCMV UV and HCMV IVIG) (C). Values are means SEM for three separate experiments
performed in duplicate. The differentiation was dened by esterase staining as well as by morphology.
10384 GREDMARK ET AL. J. VIROL.
on June 1, 2013 by guesthttp://jvi.asm.org/Downloaded from
fected broblasts in the virus stocks. Ultracentrifuged virus
inocula did not inhibit differentiation, whereas concentrated
HCMV particle preparations that had been cleared of soluble
factors and cytokines produced by the infected broblasts in-
hibited macrophage differentiation completely at an MOI of 1.
These observations suggest that the HCMV-induced inhibition
is directly dependent upon a protein component in the virus
particle.
UV inactivation did not prevent the virus from inhibiting
differentiation, indicating that viral replication was not re-
quired for the inhibitory effect. However, it is possible that
defective particles and UV-inactivated particles that cannot
produce infective virus particles support transcription from
specic areas of the viral genome. Alternatively, defective
HCMV particles might act as helper viruses and complement
replication, as suggested by higher titers of defective particles
than of infectious virus in HCMV preparations (57). Although
IVIG treatment reduced viral infectivity by 99%, it did not
affect the ability of the virus to inhibit macrophage differenti-
ation. Even at a neutralization rate of 99%, however, one
would expect to see at least a small reduction in the ability of
the virus to inhibit differentiation. That no reduction was seen
AB
0
10
20
30
40
50
60
70
80
90
100
% Monocytes
Mock
HCMV
6 h 12 h 24 hControl
0
10
20
30
40
50
60
70
80
90
100
Mock
HCMV
24 h Mock 1:10
24 h HCMV 1:10
24 h Mock 1:100
24 h HCMV 1:100
% Monocytes
CD
0
100
200
300
400
500
600
IFN-pg/ml
Mock
HCMV
6 h 12 h 24 h
0
2000
4000
6000
8000
10000
12000
14000
16000
18000
IL-6 pg/ml
Mock
HCMV
6 h 12 h 24 h
0
50
100
150
200
250
300
IL-10 pg/ml
Mock
HCMV
24 h12 h6 h
E
FIG. 7. HCMV-treated monocytes release a soluble factor that blocks macrophage differentiation. (A) Effects of supernatants on macrophage
differentiation of monocytic cultures stimulated with allo-cytokines. Supernatants from HCMV-treated and mock-treated monocytic cultures were
collected 6, 12, and 24 h after HCMV treatment, cleared by centrifugation, ltered to remove infectious virus, and diluted 1:10. (B) Effects of
supernatants on macrophage differentiation of monocytic cultures stimulated with allo-cytokines. Supernatants from HCMV-treated and mock-
treated monocytic cultures were collected 24 h after HCMV treatment, cleared by centrifugation, ltered to remove infectious virus, and diluted
1:10 and 1:100. Values are means SEM for three separate experiments performed in duplicate. The differentiation was dened by esterase
staining as well as by morphology. (C to E) Levels of IFN-(undetectable IFN-levels in mock-treated cultures) (C), IL-6 (D), and IL-10 (E) as
detected by ELISA in the supernatants obtained from mock- and HCMV-infected cultures, respectively.
VOL. 78, 2004 HCMV INHIBITS MACROPHAGE DIFFERENTIATION 10385
on June 1, 2013 by guesthttp://jvi.asm.org/Downloaded from
suggests that the inhibitory effect is mediated by nonimmuno-
genic components of the viral envelope or that determinants of
the viral glycoproteins involved in inducing inhibition of mac-
rophage differentiation were not blocked by antibodies in the
IVIG preparations.
HCMV treatment of monocytes inhibited differentiation
even at very low MOIs. This nding may be explained by the
fact that only a small percentage of the cells need to be ex-
posed to the responsible viral protein. Experiments with virus-
free supernatants obtained from monocytic cultures at differ-
ent times after HCMV treatment showed 50% inhibition of
differentiation by the 6-h supernatant and 80% inhibition by
the 12- and 24-h supernatants. Supernatants from mock-
treated cultures had no effect on differentiation. These results
FIG. 8. IFN-affects macrophage differentiation. (A) Recombinant IFN-and IFN-were added to monocytic cultures at the same time as
cytokine stimulation (IFN concentrations, 0.05 to 5 ng/ml). Antibodies against IFN-(10 to 1,000 U) and IFN-(10 to 2,500 U) and a control sheep
serum were added to the cultures at the same time point as HCMV treatment. (B) Recombinant IL-6 (100 to 1,000 ng/ml) or recombinant IL-10
(1,000 ng/ml) was added to the cultures at the same time as cytokine stimulation. Antibodies against IL-6 and/or IL-6 receptor or antibodies against
IL-10 and/or the IL-10-receptor were added at the same time point as HCMV treatment. The differentiation was dened by esterase staining as
well as by morphology.
10386 GREDMARK ET AL. J. VIROL.
on June 1, 2013 by guesthttp://jvi.asm.org/Downloaded from
imply that HCMV treatment of monocytes leads to release of
a soluble factor that inhibits surrounding cells from differen-
tiating into macrophages.
Supernatants from the HCMV-treated monocytic cultures
contained higher levels of IL-6, IL-10, and IFN-, suggesting
that HCMV induces the secretion of one or more cytokines
that can inhibit differentiation. The most likely candidate is
IFN-, which was not present in the viral inoculum but was
detected at increasing levels in the supernatants from virus-
treated monocytes. In support of this hypothesis, PBMCs ex-
posed to HCMV in vitro produced IFN-within 4 to 10 h (34),
which largely accounted for the observed immunosuppressive
effects, including reduced morphological differentiation of
monocytes and decreased oxidative activity. We found here
that exogenously added IFN-and IFN-were both able to
inhibit the differentiation of macrophages. Recombinant IL-10
and a viral IL-10 homologue (24) inhibit the proliferation of
mitogen-stimulated PBMCs (52), and IL-10 prevents the dif-
ferentiation of monocytes into dendritic cells and instead pro-
motes their maturation into macrophages (1, 16). Similarly,
IL-6 upregulates the expression of functional receptors for
macrophage colony-stimulating factor on monocytes, which
alters their differentiation from a dendritic cell phenotype to a
macrophage phenotype (7). We found that high doses of re-
combinant IL-10, but not IL-6, were able to inhibit macro-
phage differentiation by approximately 20%. Interestingly, only
antibodies to IFN-, but not to IL-6 or IL-10, were able to
partly reverse the viral inhibition of macrophage differentia-
tion. However, we were not able to completely reverse the
inhibition of macrophage differentiation in this experimental
system; the neutralizing antibodies to IFN-only reversed
25% of the inhibitory effect, which suggests that multiple cy-
tokines may be involved in this process. Alternatively, released
IFN-may be rapidly internalized by the cells in the culture
and hence not affected by exogenous antibodies.
How does the virus benet from blocking cytokine-induced
macrophage differentiation? One possibility is that blocking
enables the virus to avoid immune recognition in the early
phase of infection. Another is that HCMV infection in undif-
ferentiated myeloid cells results in latency, consistent with the
presence of HCMV DNA, but not RNA, in the cells 6 weeks
after exposure to HCMV, when the monocytes remained small
and did not exhibit the classical morphological appearance of
macrophages. HCMV did not induce apoptosis in the virus-
treated cells, and its inhibitory effect was reversible. After
several weeks in culture, the monocytes differentiated into
macrophages upon restimulation with allo-cytokines, and the
restimulated cells had the same ability to migrate and phago-
cytose as mock-treated macrophages. This is important be-
cause latently infected cells must undergo differentiation be-
fore latent virus can be reactivated. We found that HCMV
transiently blocks cytokine-induced differentiation of mono-
cytes into macrophages. Thus, latent virus might be activated
when monocytes encounter an inammatory response, enter
tissues, and differentiate into macrophages.
Cellular differentiation is a prerequisite for HCMV replica-
tion in cells that normally undergo differentiation. For exam-
ple, HCMV infection in monocytes occurs only at a low fre-
quency, is abortive, or is restricted to expression of IE genes
(18, 44). In contrast, when monocytes differentiate into mac-
rophages, a high proportion of cells becomes productively in-
fected by HCMV (10, 18, 50). Cellular differentiation is also
important for productive HCMV infection of human terato-
carcinoma cells, which become permissive after differentiation
is induced by retinoic acid (13). Since both adsorption and
penetration of virus occur to the same extent in monocytes and
teratocarcinoma cells (13, 18), the inability of the virus to
replicate in undifferentiated cells is not caused by a virus entry
block. The virus may be inactive in monocytes early in the
infectious process, as a result of a direct inactivating effect of
HCMV on the infected cells. Hence, the virus-induced inhibi-
tion of cellular differentiation may facilitate the establishment
of latency. However, the viral and cellular mechanisms for
maintaining HCMV latency are largely unknown.
Transcripts associated with HCMV latency have been iden-
tied in HCMV-infected, fetal liver-derived CD33
granulo-
cyte-macrophage progenitor cells in vitro and in HCMV-sero-
positive individuals and may play a critical role in the control of
latency (17). These transcripts are represented by sense (open
reading frame 94 [ORF94]) and anti-sense (ORF152 and
ORF154) transcripts expressed from a region of the genome
(UL122-UL123) normally involved in the expression of the
transcriptional activators (IE1 and IE2) that participate in lytic
replication (23). Recently, Goodrum et al. developed a model
for HCMV latency in hematopoietic progenitor cells, in which
CD34
cells were infected with HCMV and cultured (14).
These cells did not produce infectious virus during the culture
period, but virus reactivation could be induced by coculture
with human broblasts. As shown by microarray analysis, the
patterns of viral gene expression were distinctly different dur-
ing latent, productive, and nonproductive infections (14).
In our study, HCMV-treated monocyte cultures examined 1
and 6 weeks after infection contained viral DNA but no viral
RNA, indicating that the cells were neither abortively nor
productively infected. We did not detect latency-associated
transcripts by our HCMV IE-specic primers, but we cannot
exclude the existence of gene expression from a latency-asso-
ciated region of the HCMV genome that could not be detected
with these primers. Recent observations also suggest that in-
activation of ORF94 does not affect reactivation of latent
HCMV in CD33
granulocyte-macrophage progenitor cells
derived from fetal liver (61). In addition, a previous study did
not detect these transcripts in allogeneically stimulated mac-
rophages (49). Hence, it is unclear whether these transcripts
are critical for maintaining latent virus in undifferentiated
monocytes. However, immune activation with concomitant
production of proinammatory cytokines appears to be essen-
tial for the reactivation and replication of HCMV in infected
patients (2, 8, 25).
Our ndings may have important clinical implications.
HCMV predisposes infected subjects to other opportunistic
infections and is associated with bacterial and fungal infections
in transplant patients (32, 36, 59), possibly by altering the
function of monocyte-derived macrophages and their progen-
itor cells during infection (6, 33, 39, 43, 58). HCMV also
impairs monocyte functions, for example, by suppressing the
production of IL-1 and IL-2 in PBMCs and by decreasing the
proliferative response of these cells to mitogens in vitro (19).
HCMV infection also inhibits monocyte differentiation by in-
ducing IFN-from nonadherent PBMCs (34). In our study,
VOL. 78, 2004 HCMV INHIBITS MACROPHAGE DIFFERENTIATION 10387
on June 1, 2013 by guesthttp://jvi.asm.org/Downloaded from
HCMV-infected monocytes had a decreased ability to migrate
in response to chemokines and to phagocytose yeast particles.
As a result of their decreased ability to present peptides in the
context of major histocompatibility complex class II molecules,
HCMV-infected monocytes may be impaired in their ability to
clear other infections, resulting in immunosuppression.
Other viruses also cause immunosuppression. HIV induces
immunosuppression mainly by targeting CD4
T cells, and
measles virus both inhibits T-cell proliferation and induces
functional abnormalities in actively infected monocytes (33,
63). Cells infected with measles virus are also impaired in their
ability to present antigens, despite induced expression of major
histocompatibility complex molecules. In our study, neither
HIV nor measles virus inhibited macrophage differentiation,
which suggests that HCMVs effect on cellular differentiation is
not a general event after viral exposure.
In conclusion, our ndings provide new tools to examine
cellular differentiation response pathways and may help in the
development of new strategies to improve immunological func-
tions in HCMV-infected patients.
ACKNOWLEDGMENTS
We thank Mireille Vossen for technical assistance, Annika Linde for
measles virus, Anders So¨nnerborg for HIV and access to P3 facilities,
and Erna Mo¨ller, William B. Britt, Daniel Streblow, and Lena Ser-
rander for helpful discussions.
This work was supported by grants from the Swedish Medical Re-
search Council (K98-06X-12615-01A and K2001-16X-12615-04A to
C.S.-N. and K202-06X-00793-37C to E.M.), the Tobias Foundation
(1313/98, 20/01, and 33/02), the Swedish Childrens Cancer Research
Foundation (1998/065 and 01/046), the Heart-Lung Foundation
(199941305 and 200241138), the Swedish Society for Medicine (1999-
02-0347), the Goljes Foundation (325, 520), and the Emil and Wera
Cornells Foundation. C.S.-N. is a fellow of the Wenner-Gren Foun-
dation, Sweden.
ADDENDUM IN PROOF
Interestingly, Smith et al. recently published data which sug-
gest that HCMV instead could induce differentiation of mono-
cytes into macrophages (M. S. Smith, G. L. Bentz, J. S. Alex-
ander, and A. D. Yurochko, J. Virol., 78:44444453, 2004).
Collagen-coated plates were used in their experimental system,
which may explain the different results obtained, and, impor-
tantly, these differences may also reect differences in the in
vivo situation in which monocytes in the peripheral blood or
monocytes entering tissues as macrophages will become
HCMV infected.
REFERENCES
1. Allavena, P., L. Piemonti, D. Longoni, S. Bernasconi, A. Stoppacciaro, L.
Ruco, and A. Mantovani. 1998. IL-10 prevents the differentiation of mono-
cytes to dendritic cells but promotes their maturation to macrophages. Eur.
J. Immunol. 28:359369.
2. Asadullah, K., S. Prosch, H. Audring, I. Buttnerova, H. D. Volk, W. Sterry,
and W. D. Docke. 1999. A high prevalence of cytomegalovirus antigenaemia
in patients with moderate to severe chronic plaque psoriasis: an association
with systemic tumour necrosis factor alpha overexpression. Br. J. Dermatol.
141:94102.
3. Biondi, A., T. H. Rossing, J. Bennett, and R. F. Todd III. 1984. Surface
membrane heterogeneity among human mononuclear phagocytes. J. Immu-
nol. 132:12371243.
4. Bolovan-Fritts, C. A., E. S. Mocarski, and J. A. Wiedeman. 1999. Peripheral
blood CD14() cells from healthy subjects carry a circular conformation of
latent cytomegalovirus genome. Blood 93:394398.
5. Britt, W. J., and C. A. Alford. 1996. Cytomegalovirus, p. 24932523. In B. N.
Fields, D. M. Knipe, and P. M. Howley (ed.), Fields virology, vol. 2, 3rd ed.
Lippincott-Raven Publishers, Philadelphia, Pa.
6. Bron, D., L. Lagneaux, A. Delforge, C. Otte, R. Snoeck, and P. Stryckmans.
1991. Prevention of CMV-induced myelosuppression by anti-CMV antibod-
ies: an in vitro model. Exp. Hematol. 19:132135.
7. Chomarat, P., J. Banchereau, J. Davoust, and A. K. Palucka. 2000. IL-6
switches the differentiation of monocytes from dendritic cells to macro-
phages. Nat. Immunol. 1:510514.
8. Docke, W. D., S. Prosch, E. Fietze, V. Kimel, H. Zuckermann, C. Klug, U.
Syrbe, D. H. Kruger, R. von Baehr, and H. D. Volk. 1994. Cytomegalovirus
reactivation and tumour necrosis factor. Lancet 343:268269.
9. Einhorn, L., and A. Ost. 1984. Cytomegalovirus infection of human blood
cells. J. Infect. Dis. 149:207214.
10. Fish, K. N., A. S. Depto, A. V. Moses, W. Britt, and J. A. Nelson. 1995.
Growth kinetics of human cytomegalovirus are altered in monocyte-derived
macrophages. J. Virol. 69:37373743.
11. Gerna, G., D. Zipeto, E. Percivalle, M. Parea, M. G. Revello, R. Maccario, G.
Peri, and G. Milanesi. 1992. Human cytomegalovirus infection of the major
leukocyte subpopulations and evidence for initial viral replication in poly-
morphonuclear leukocytes from viremic patients. J. Infect. Dis. 166:1236
1244.
12. Gnann, J. W., Jr., J. Ahlmen, C. Svalander, L. Olding, M. B. Oldstone, and
J. A. Nelson. 1988. Inammatory cells in transplanted kidneys are infected by
human cytomegalovirus. Am. J. Pathol. 132:239248.
13. Gonczol, E., P. W. Andrews, and S. A. Plotkin. 1984. Cytomegalovirus rep-
licates in differentiated but not in undifferentiated human embryonal carci-
noma cells. Science 224:159161.
14. Goodrum, F. D., C. T. Jordan, K. High, and T. Shenk. 2002. Human cyto-
megalovirus gene expression during infection of primary hematopoietic pro-
genitor cells: a model for latency. Proc. Natl. Acad. Sci. USA 99:16255
16260.
15. Grattan, M. T., C. E. Moreno-Cabral, V. A. Starnes, P. E. Oyer, E. B.
Stinson, and N. E. Shumway. 1989. Cytomegalovirus infection is associated
with cardiac allograft rejection and atherosclerosis. JAMA 261:35613566.
16. Gredmark, S., and C. So¨derberg-Naucle´r. 2003. Human cytomegalovirus
inhibits differentiation of monocytes into dendritic cells with the conse-
quence of depressed immunological functions. J. Virol. 77:1094310956.
17. Hahn, G., R. Jores, and E. S. Mocarski. 1998. Cytomegalovirus remains
latent in a common precursor of dendritic and myeloid cells. Proc. Natl.
Acad. Sci. USA 95:39373942.
18. Ibanez, C. E., R. Schrier, P. Ghazal, C. Wiley, and J. A. Nelson. 1991. Human
cytomegalovirus productively infects primary differentiated macrophages.
J. Virol. 65:65816588.
19. Kapasi, K., and G. P. Rice. 1988. Cytomegalovirus infection of peripheral
blood mononuclear cells: effects on interleukin-1 and -2 production and
responsiveness. J. Virol. 62:36033607.
20. Kaplan, G., and G. Gaudernack. 1982. In vitro differentiation of human
monocytes. Differences in monocyte phenotypes induced by cultivation on
glass or on collagen. J. Exp. Med. 156:11011114.
21. Kondo, K., H. Kaneshima, and E. S. Mocarski. 1994. Human cytomegalo-
virus latent infection of granulocyte-macrophage progenitors. Proc. Natl.
Acad. Sci. USA 91:1187911883.
22. Kondo, K., and E. S. Mocarski. 1995. Cytomegalovirus latency and latency-
specic transcription in hematopoietic progenitors. Scand. J. Infect. Dis.
Suppl. 99:6367.
23. Kondo, K., J. Xu, and E. S. Mocarski. 1996. Human cytomegalovirus latent
gene expression in granulocyte-macrophage progenitors in culture and in
seropositive individuals. Proc. Natl. Acad. Sci. USA 93:1113711142.
24. Kotenko, S. V., S. Saccani, L. S. Izotova, O. V. Mirochnitchenko, and S.
Pestka. 2000. Human cytomegalovirus harbors its own unique IL-10 ho-
molog (cmvIL-10). Proc. Natl. Acad. Sci. USA 97:16951700.
25. Kutza, A. S., E. Muhl, H. Hackstein, H. Kirchner, and G. Bein. 1998. High
incidence of active cytomegalovirus infection among septic patients. Clin.
Infect. Dis. 26:10761082.
26. Loenen, W. A., C. A. Bruggeman, and E. J. Wiertz. 2001. Immune evasion by
human cytomegalovirus: lessons in immunology and cell biology. Semin.
Immunol. 13:4149.
27. Loetscher, P., M. Seitz, M. Baggiolini, and B. Moser. 1996. Interleukin-2
regulates CC chemokine receptor expression and chemotactic responsive-
ness in T lymphocytes. J. Exp. Med. 184:569577.
28. Lonnqvist, B., O. Ringden, B. Wahren, G. Gahrton, and G. Lundgren. 1984.
Cytomegalovirus infection associated with and preceding chronic graft-ver-
sus-host disease. Transplantation 38:465468.
29. Mendelson, M., S. Monard, P. Sissons, and J. Sinclair. 1996. Detection of
endogenous human cytomegalovirus in CD34bone marrow progenitors.
J. Gen. Virol. 77:30993102.
30. Minton, E. J., C. Tysoe, J. H. Sinclair, and J. G. Sissons. 1994. Human
cytomegalovirus infection of the monocyte/macrophage lineage in bone mar-
row. J. Virol. 68:40174021.
31. Movassagh, M., J. Gozlan, B. Senechal, C. Baillou, J. C. Petit, and F. M.
Lemoine. 1996. Direct infection of CD34progenitor cells by human cyto-
10388 GREDMARK ET AL. J. VIROL.
on June 1, 2013 by guesthttp://jvi.asm.org/Downloaded from
megalovirus: evidence for inhibition of hematopoiesis and viral replication.
Blood 88:12771283.
32. Mutimer, D., D. Mirza, J. Shaw, K. ODonnell, and E. Elias. 1997. Enhanced
(cytomegalovirus) viral replication associated with septic bacterial complica-
tions in liver transplant recipients. Transplantation 63:14111415.
33. Naniche, D., and M. B. Oldstone. 2000. Generalized immunosuppression:
how viruses undermine the immune response. Cell. Mol. Life Sci. 57:1399
1407.
34. Noraz, N., J. L. Lathey, and S. A. Spector. 1997. Human cytomegalovirus-
associated immunosuppression is mediated through interferon-alpha. Blood
89:24432452.
35. Passlick, B., D. Flieger, and H. W. Ziegler-Heitbrock. 1989. Identication
and characterization of a novel monocyte subpopulation in human periph-
eral blood. Blood 74:25272534.
36. Paya, C. V., R. H. Wiesner, P. E. Hermans, J. J. Larson-Keller, D. M. Ilstrup,
R. A. Krom, S. Rettke, and T. F. Smith. 1993. Risk factors for cytomegalo-
virus and severe bacterial infections following liver transplantation: a pro-
spective multivariate time-dependent analysis. J. Hepatol. 18:185195.
37. Pro¨sch, S., A.-K. Heine, H.-D. Volk, and D. H. Kru¨ger. 2001. CCAAT/
enhancer-binding proteins and negatively inuence the capacity of tumor
necrosis factor-to up-regulate the human cytomegalovirus IE1/2 enhancer/
promoter by nuclear factor B during monocyte differentiation. J. Biol.
Chem. 276:4071240720.
38. Prosch, S., K. Staak, J. Stein, C. Liebenthal, T. Stamminger, H. D. Volk, and
D. H. Kru¨ger. 1995. Stimulation of the human cytomegalovirus IE enhancer/
promoter in HL-60 cells by TNFis mediated via induction of NF-B.
Virology 208:197206.
39. Reiser, H., J. Kuhn, H. W. Doerr, H. Kirchner, K. Munk, and R. Braun.
1986. Human cytomegalovirus replicates in primary human bone marrow
cells. J. Gen. Virol. 67:25952604.
40. Ritter, T., C. Brandt, S. Prosch, A. Vergopoulos, K. Vogt, J. Kolls, and H. D.
Volk. 2000. Stimulatory and inhibitory action of cytokines on the regulation
of hCMV-IE promoter activity in human endothelial cells. Cytokine 12:
11631170.
41. Ross, J. A., and M. J. Auger. 2002. The biology of the macrophage, p. 172.
In B. Burke and C. E. Lewis (ed.), The macrophage, 2nd ed. Oxford Uni-
versity Press, Oxford, United Kingdom.
42. Serrander, L., M. Fallman, and O. Stendahl. 1996. Activation of phospho-
lipase D is an early event in integrin-mediated signalling leading to phago-
cytosis in human neutrophils. Inammation 20:439450.
43. Simmons, P., K. Kaushansky, and B. Torok-Storb. 1990. Mechanisms of
cytomegalovirus-mediated myelosuppression: perturbation of stromal cell
function versus direct infection of myeloid cells. Proc. Natl. Acad. Sci. USA
87:13861390.
44. Sinclair, J. H., J. Baillie, L. A. Bryant, J. A. Taylor-Wiedeman, and J. G.
Sissons. 1992. Repression of human cytomegalovirus major immediate early
gene expression in a monocytic cell line. J. Gen. Virol. 73:433435.
45. Sindre, H., G. E. Tjoonnfjord, H. Rollag, T. Ranneberg-Nilsen, O. P. Veiby,
S. Beck, M. Degre, and K. Hestdal. 1996. Human cytomegalovirus suppres-
sion of and latency in early hematopoietic progenitor cells. Blood 88:4526
4533.
46. Slobedman, B., and E. S. Mocarski. 1999. Quantitative analysis of latent
human cytomegalovirus. J. Virol. 73:48064812.
47. So¨derberg, C., S. Larsson, S. Bergstedt-Lindqvist, and E. Mo¨ller. 1993.
Denition of a subset of human peripheral blood mononuclear cells that are
permissive to human cytomegalovirus infection. J. Virol. 67:31663175.
48. So¨derberg-Naucle´r, C., K. N. Fish, and J. A. Nelson. 1997. Interferon-and
tumor necrosis factor-specically induce formation of cytomegalovirus-
permissive monocyte-derived macrophages that are refractory to the antivi-
ral activity of these cytokines. J. Clin. Investig. 100:31543163.
49. So¨derberg-Naucle´r, C., K. N. Fish, and J. A. Nelson. 1997. Reactivation of
latent human cytomegalovirus by allogeneic stimulation of blood cells from
healthy donors. Cell 91:119126.
50. So¨derberg-Naucle´r, C., D. N. Streblow, K. N. Fish, J. Allan-Yorke, P. P.
Smith, and J. A. Nelson. 2001. Reactivation of latent human cytomegalovirus
in CD14
monocytes is differentiation dependent. J. Virol. 75:75437554.
51. Speir, E., R. Modali, E. S. Huang, M. B. Leon, F. Shawl, T. Finkel, and S. E.
Epstein. 1994. Potential role of human cytomegalovirus and p53 interaction
in coronary restenosis. Science 265:391394.
52. Spencer, J. V., K. M. Lockridge, P. A. Barry, G. Lin, M. Tsang, M. E.
Penfold, and T. J. Schall. 2002. Potent immunosuppressive activities of
cytomegalovirus-encoded interleukin-10. J. Virol. 76:12851292.
53. Stein, J., H. D. Volk, C. Liebenthal, D. H. Kruger, and S. Prosch. 1993.
Tumour necrosis factor alpha stimulates the activity of the human cytomeg-
alovirus major immediate early enhancer/promoter in immature monocytic
cells. J. Gen. Virol. 74:23332338.
54. Taylor-Wiedeman, J., G. P. Hayhurst, J. G. Sissons, and J. H. Sinclair. 1993.
Polymorphonuclear cells are not sites of persistence of human cytomegalo-
virus in healthy individuals. J. Gen. Virol. 74:265268.
55. Taylor-Wiedeman, J., J. G. Sissons, L. K. Borysiewicz, and J. H. Sinclair.
1991. Monocytes are a major site of persistence of human cytomegalovirus in
peripheral blood mononuclear cells. J. Gen. Virol. 72:20592064.
56. Taylor-Wiedeman, J., P. Sissons, and J. Sinclair. 1994. Induction of endog-
enous human cytomegalovirus gene expression after differentiation of mono-
cytes from healthy carriers. J. Virol. 68:15971604.
57. Topilko, A., and S. Michelson. 1994. Morphological and cytochemical anal-
ysis of human cytomegalovirus inoculum: correlation of free particles in
inoculum with counterparts in infected cells. Res. Virol. 145:6573.
58. Torok-Storb, B., P. Simmons, D. Khaira, D. Stachel, and D. Myerson. 1992.
Cytomegalovirus and marrow function. Ann. Hematol. 64(Suppl.):A128
A131.
59. van den Berg, A. P., I. J. Klompmaker, E. B. Haagsma, P. M. Peeters, L.
Meerman, R. Verwer, T. H. The, and M. J. Slooff. 1996. Evidence for an
increased rate of bacterial infections in liver transplant patients with cyto-
megalovirus infection. Clin. Transplant. 10:224231.
60. Wentworth, B. B., and L. French. 1970. Plaque assay of cytomegalovirus
strains of human origin. Proc. Soc. Exp. Biol. Med. 135:253258.
61. White, K. L., B. Slobedman, and E. S. Mocarski. 2000. Human cytomega-
lovirus latency-associated protein pORF94 is dispensable for productive and
latent infection. J. Virol. 74:93339337.
62. von Laer, D., U. Meyer-Koenig, A. Serr, J. Finke, L. Kanz, A. A. Fauser, D.
Neumann-Haefelin, W. Brugger, and F. T. Hufert. 1995. Detection of cyto-
megalovirus DNA in CD34cells from blood and bone marrow. Blood
86:40864090.
63. Yanagi, Y., B. A. Cubitt, and M. B. Oldstone. 1992. Measles virus inhibits
mitogen-induced T cell proliferation but does not directly perturb the T cell
activation process inside the cell. Virology 187:280289.
64. Ziegler-Heitbrock, H. W., and R. J. Ulevitch. 1993. CD14: cell surface re-
ceptor and differentiation marker. Immunol. Today 14:121125.
VOL. 78, 2004 HCMV INHIBITS MACROPHAGE DIFFERENTIATION 10389
on June 1, 2013 by guesthttp://jvi.asm.org/Downloaded from
... For an optimal viral replication, the virus must be able to inhibit the macrophage differentiation. This could be a factor of getting the low rate of viral growth in macrophages cultures 298,299 . ...
... After the HCMV binding to monocytes, an intracellular increase in Ca2+ levels is induced and increased Ca2+ levels result in the blockage of monocytes ability to differentiate into macrophages. Observations suggest that HCMV has an efficient strategy that allows its interference with the cellular differentiation pathways, and this can also explain the development of generalized immunosuppression in patients infected with HCMV298,327 . HCMV infection of CD14+ monocytes results in the generation of latency-specific transcripts, the conservation of the viral genomes, and the ability of the virus to enter the lytic cycle again 328 .3-HCMV, ...
Thesis
Since several years, the role played by human cytomegalovirus (HCMV) in the development of inflammatory diseases and cancer has been extensively studied and addressed by different research groups. Various tumor tissues originating from colon, liver, prostate, brain (glioblastoma, medulloblastoma) and breast cancer have shown to harbor either the antigen or the DNA of HCMV. These growing evidences about the implication of HCMV infection in malignant entities had led to the emergence of the concept of “Oncomodulation”. This is explained by the ability of the virus to contribute to the oncogenic processes, however without any direct transformatory potential. HCMV-DB (KT959235) is a clinical isolate obtained from a cervical swab specimen of a 30-year-old pregnant woman previously isolated in our laboratory. This viral strain had shown its ability to infect the primary macrophages and to replicate productively in the human mammary epithelial cells (HMECs). In fact, HMECs infected by HCMV-DB resulted in the establishment of a pro-oncogenic cellular environment characterized by retinoblastoma (Rb) hyperphosphorylation and a decreased p53 functional activity, enhanced telomerase activity, upregulation of c-Myc, activation of Akt and STAT3, and upregulation of cyclin D1 causing an enhanced cellular proliferation. Furthermore, HCMV-DB had shown its potential to transform the primary HMECs by colony formation on soft agar, a well know assay to perceive transformation. Interestingly, HCMV-DB infected HMECs in culture showed the emergence of clusters of spheroid cells that were named CTH cells (CMV Transformed HMECs). In our thesis we characterized the genomic profile of HCMV-DB strain and compared it to either clinical or laboratory strains. HCMV-DB was shown to be close to the genomes of Toledo and JP strains where the JP strain is a clinical strain that was isolated from a glandular tissue, the prostate. We also compared the genes that are involved in virus entry using phylogenetic analyses and we observed that HCMV-DB is close to the prototypic HCMV strain, Merlin. By studying the transcriptomic profile of HMECs infected with HCMV-DB, we found that it displays a triple negative basal-like phenotype, ER−/PR−/HER2−, and presents oncogenic characteristics with upregulated expression of several oncogenes, pro-survival genes (with a down-regulation of caspase 8), proliferation markers, stemcellness and epithelial mesenchymal transition (EMT). The transcriptomic profile of HMECs infected with HCMV-DB also displays variant modifications in cell signaling, angiogenesis and proteolysis. At the chromatin level, HMECs infected with HCMV-DB reveals a global hypomethylation state. By screening for the presence of HCMV-DB genome in the formed CTH cells, we detected a signature of the HCMV-DB genome, namely a lncRNA4.9. Taken together, our data showed that the transcriptome of HMECs infected with HCMV-DB clearly reveals a pro-oncogenic traits and the detection of part of the HCMV-DB genome suggests that this part of the viral genome might be responsible for the obtained cellular transformation.
... Recently, it has been reported that several blocks occur in the viral cycle following HCMV infection of cancer cells [33]. The virus ability to inhibit macrophage differentiation is required for optimal viral replication, and might be a factor of the low rate of viral growth in macrophage cultures [34,35]. In addition, studies showed that the presence of an intact ULb' sequence in the genome of clinical HCMV isolates had lead to their efficient growth in macrophages, epithelial cells and endothelial cells [36 -39]. ...
... One of the results of the Ca 2+ rise is the block of the monocytes ability to differentiate into macrophages. Observations suggest that the virus has an efficient strategy permitting it to interfere with cellular differentiation pathways, and this may also elucidate the generalized immunosuppression that is often observed in HCMV-infected patients [34,69]. HCMV infection of CD14+ monocytes gives rise to the generation of latency-specific transcripts, conservation of viral genomes, and the capacity of the virus to reenter the lytic cycle. ...
Article
Full-text available
The human cytomegalovirus (HCMV) is a betaherpesvirus that is highly host specific, infects among others epithelial cells and macrophages, and has been recently mentioned as having oncomodulatory properties. HCMV is detected in the breast tumor tissue where macrophages, especially tumor associated macrophages, are associated with a poor prognosis. In this review, we will discuss the potential implication of HCMV in breast cancer with emphasis on the role played by macrophages.
... Infected macrophages serve as "mobile vectors" for virus spreading and dissemination to different organs mainly by transendothelial migration (17). HCMV-infected macrophages show classical macrophage markers but less phagocytic and immunogenic capacity (89). Furthermore, HCMV + IBC cancer tissues are characterized by significantly high infiltration of CD163 + and MAC387 + TAMs in comparison with HCMV -IBC cancer tissues. ...
Article
Full-text available
Inflammatory breast cancer (IBC) is a highly aggressive phenotype of breast cancer that is characterized by a high incidence early metastasis. We previously reported a significant association of human cytomegalovirus (HCMV) DNA in the carcinoma tissues of IBC patients but not in the adjacent normal tissues. HCMV-infected macrophages serve as “mobile vectors” for spreading and disseminating virus to different organs, and IBC cancer tissues are highly infiltrated by tumor-associated macrophages (TAMs) that enhance IBC progression and promote breast cancer stem cell (BCSC)-like properties. Therefore, there is a need to understand the role of HCMV-infected TAMs in IBC progression. The present study aimed to test the effect of the secretome (cytokines and secreted factors) of TAMs derived from HCMV⁺ monocytes isolated from IBC specimens on the proliferation, invasion, and BCSC abundance when tested on the IBC cell line SUM149. HCMV⁺ monocytes were isolated from IBC patients during modified radical mastectomy surgery and tested in vitro for polarization into TAMs using the secretome of SUM149 cells. MTT, clonogenic, invasion, real-time PCR arrays, PathScan Intracellular Signaling array, and cytokine arrays were used to characterize the secretome of HCMV⁺ TAMs for their effect on the progression of SUM149 cells. The results showed that the secretome of HCMV⁺ TAMs expressed high levels of IL-6, IL-8, and MCP-1 cytokines compared to HCMV⁻ TAMs. In addition, the secretome of HCMV⁺ TAMs induced the proliferation, invasion, colony formation, and expression of BCSC-related genes in SUM149 cells compared to mock untreated cells. In addition, the secretome of HCMV⁺ TAMs activated the phosphorylation of intracellular signaling molecules p-STAT3, p-AMPKα, p-PRAS40, and p-SAPK/JNK in SUM149 cells. In conclusion, this study shows that the secretome of HCMV⁺ TAMs enhances the proliferation, invasion, colony formation, and BCSC properties by activating the phosphorylation of p-STAT3, p-AMPKα, p-PRAS40, and p-SAPK/JNK intracellular signaling molecules in IBC cells.
... Besides being different systems, the data provides biological clues to disease. Perhaps the block in differentiation due to cytokines produced during alloreactivity explain HCMV disease in allogenic stem cell transplant patients (Gredmark et al., 2004b). Collectively, the data suggest HCMV regulates the polarization and differentiation of monocytes towards a unique M1/M2 macrophage that favors viral persistence under normal infection conditions. ...
Article
Full-text available
Viral dissemination is a key mechanism responsible for persistence and disease following human cytomegalovirus (HCMV) infection. Monocytes play a pivotal role in viral dissemination to organ tissue during primary infection and following reactivation from latency. For example, during primary infection, infected monocytes migrate into tissues and differentiate into macrophages, which then become a source of viral replication. In addition, because differentiated macrophages can survive for months to years, they provide a potential persistent infection source in various organ systems. We broadly note that there are three phases to infection and differentiation of HCMV-infected monocytes: (1) Virus enters and traffics to the nucleus through a virus receptor ligand engagement event that activates a unique signalsome that initiates the monocyte-to-macrophage differentiation process. (2) Following initial infection, HCMV undergoes a “quiescence-like state” in monocytes lasting for several weeks and promotes monocyte differentiation into macrophages. While, the initial event is triggered by the receptor-ligand engagement, the long-term cellular activation is maintained by chronic viral-mediated signaling events. (3) Once HCMV infected monocytes differentiate into macrophages, the expression of immediate early viral (IE) genes is detectable, followed by viral replication and long term infectious viral particles release. Herein, we review the detailed mechanisms of each phase during infection and differentiation into macrophages and discuss the biological significance of the differentiation of monocytes in the pathogenesis of HCMV.
... cytosis.[38][39][40][41][42][43][44][45] CMV has also been shown to modulate TLR-mediated pathways, although generally these data have been derived from in vitro studies using cell lines. For example, CMV microRNAs such pression of several cytokines after stimulation with a TLR2 ligand.48 ...
Article
Full-text available
CMV is an immunomodulatory virus that indirectly increases the risk for bacterial, fungal and viral infections. However, the pathogenesis of this phenomenon is poorly understood. We determined whether inflammatory responses to different Toll‐like receptor (TLR) ligands are blunted during CMV infection in solid organ transplant (SOT) patients. Peripheral blood mononuclear cells from 38 SOT patients with and without CMV were incubated in the presence of various viral, fungal and bacterial TLR ligands. Cytokines were measured in the supernatant by multiplex ELISA. Patients had blunted cytokine responses to bacterial, fungal and viral ligands during CMV infection when compared to the absence of CMV infection. This was independent of viral load, clinical presentation of CMV infection or immunosuppression, supporting the clinical observation in SOT recipients that CMV infection increases susceptibility to bacterial, fungal, and other viral infections. Moreover, in the absence of CMV infection, patients with subsequent CMV infection had lower cytokines in response to TLR ligands compared to those without subsequent CMV infection, suggesting that inherent differences in patients not directly related to CMV also contribute to this increased susceptibility. In summary, these data provide novel ex vivo evidence to support indirect effects of CMV. This article is protected by copyright. All rights reserved.
... Upon co-culture with HCMV-infected cells pDC, but not monocyte-derived cells, mount abundant IFN-α responses As reported previously by us and others [15,[29][30][31][32][33][34], direct HCMV stimulation of pDC as well as monocyte-derived DC, GM-CSF MΦ, and M-CSF MΦ induced abundant IFN-α expression (Figure 1(a)) and secretion of IFN-α 24 hours post infection (hpi) (Figure 1(b)). To study whether also contact with HCMV-infected cells induced antiviral IFN-I responses, retinal pigment epithelial cells (RPE cells), which are permissive for HCMV replication [35,36], were used. ...
Article
Full-text available
Infection of healthy individuals with human cytomegalovirus (HCMV) is usually unnoticed and results in life-long latency, whereas HCMV reactivation as well as infection of newborns or immunocompromised patients can cause life-threatening disease. To better understand HCMV pathogenesis we studied mechanisms that restrict HCMV spread. We discovered that HCMV-infected cells can directly trigger plasmacytoid dendritic cells (pDC) to mount antiviral type I interferon (IFN-I) responses, even in the absence of cell-free virus. In contrast, monocyte-derived cells only expressed IFN-I when stimulated by cell-free HCMV, or upon encounter of HCMV-infected cells that already produced cell-free virus. Nevertheless, also in the absence of cell-free virus, i.e., upon co-culture of infected epithelial/endothelial cells and monocyte-derived macrophages (moMΦ) or dendritic cells (moDC), antiviral responses were induced that limited HCMV spread. The induction of this antiviral effect was dependent on cell-cell contact, whereas cell-free supernatants from co-culture experiments also inhibited virus spread, implying that soluble factors were critically needed. Interestingly, the antiviral effect was independent of IFN-γ, TNF-α, and IFN-I as indicated by cytokine inhibition experiments using neutralizing antibodies or the vaccinia virus-derived soluble IFN-I binding protein B18R, which traps human IFN-α and IFN-β. In conclusion, our results indicate that human macrophages and dendritic cells can limit HCMV spread by IFN-I dependent as well as independent mechanisms, whereas the latter ones might be particularly relevant for the restriction of HCMV transmission via cell-to-cell spread.
... Although many questions are unanswered, it has been shown that pathogens can trigger the innate immunity by binding TLRs and could activate the adaptive immune responses through several mechanisms including the molecular mimicry, direct T-cell activation and autoimmune reactions; moreover, the infectious agents can affect the vascular biology through the direct infection of tissue. Based on this, it has been proposed that chlamydia pneumoniae and cytomegalovirus in atherogenesis can play a role in the atherosclerosis pathology (14)(15)(16)(17)(18)(19)(20)(21)(22)(23)(24)(25). ...
... In particular, IFN-α is produced in infected monocytes [148,183] and IFN-β in infected M2 macrophages [165]. Further studies on the effect of CMV on T reg and on other immune system cells in a GBM tumor are required. ...
Article
Full-text available
Recent years have seen considerable progress in understanding the biochemistry of cancer. For example, more significance is now assigned to the tumor microenvironment, especially with regard to intercellular signaling in the tumor niche which depends on many factors secreted by tumor cells. In addition, great progress has been made in understanding the influence of factors such as neurotensin, growth differentiation factor-15 (GDF-15), sphingosine-1-phosphate (S1P), and infection with cytomegalovirus (CMV) on the ‘hallmarks of cancer’ in glioblastoma multiforme. Therefore, in the present work we describe the influence of these factors on the proliferation and apoptosis of neoplastic cells, cancer stem cells, angiogenesis, migration and invasion, and cancer immune evasion in a glioblastoma multiforme tumor. In particular, we discuss the effect of neurotensin, GDF-15, S1P (including the drug FTY720), and infection with CMV on tumor-associated macrophages (TAM), microglial cells, neutrophil and regulatory T cells (Treg), on the tumor microenvironment. In order to better understand the role of the aforementioned factors in tumoral processes, we outline the latest models of intratumoral heterogeneity in glioblastoma multiforme. Based on the most recent reports, we discuss the problems of multi-drug therapy in treating glioblastoma multiforme.
... Since the viral genome is maintained (42) even in the absence of a productive infection (17), monocytes are considered to be reservoirs during latency and vehicles for viral dissemination (40,46). It is known that monocytes exposed to HCMV undergo a number of physiological changes which include changes in Ca 2ϩ homeostasis and phospholipid turnover, induction of second messengers, upregulation of transcription factors (55), and alteration of their capacity to differentiate to dendritic cells and macrophages (15,16,43). Since chemokine receptors play crucial roles in monocyte activation and trafficking, we investigated the effects of HCMV infection on their expression and function. ...
Article
Background Intracranial large artery stenosis (ILAS) is an important contributor to ischemic stroke in the United States and worldwide. There is evidence to suggest that chronic exposure to certain infectious agents may also be associated with ILAS. We aimed to study this association further in an ethnically diverse, prospective, population-based sample of Northern Manhattan. Methods We enrolled a random sample of stroke-free participants from an urban, racially, and ethnically diverse community in 1993. Participants have been followed prospectively and a subset underwent brain magnetic resonance angiograms from 2003 to 2008. Intracranial stenoses of the circle of Willis and vertebrobasilar arteries were scored as 0=no stenosis, 1≤50% (or luminal irregularities), 2=50% to 69%, 3≥70% stenosis, and 4=flow gap. We summed the individual score of each artery to produce a global ILAS score (possible range, 0–44). Past infectious exposure to Chlamydia pneumoniae , Helicobacter pylori , cytomegalovirus, and herpes simplex virus 1 and 2 was determined using serum antibody titers. Results Among 572 NOMAS (Northern Manhattan Study) participants (mean age 71.0±8.0 years, 60% women, 68% Hispanic) with available magnetic resonance angiogram and serological data, herpes simplex virus 2 (beta=0.051, P <0.001) and cytomegalovirus (beta=0.071, P <0.05) were associated with ILAS score after adjusting for demographics and vascular risk factors. Stratifying by anterior and posterior circulations, herpes simplex virus 2 remained associated with the anterior circulation (beta=0.055 P <0.01) but not with posterior circulation ILAS score. Conclusions Chronic infectious exposures, specifically herpes simplex virus 2 and cytomegalovirus were associated with asymptomatic ILAS as seen on magnetic resonance angiogram imaging. This may represent an additional target of intervention in the ongoing effort to stem the substantial global burden of strokes related to ILAS.
Article
Full-text available
Both tumour necrosis factor ~ (TNF-~) and phorbol 12- myristate 13-acetate (PMA) stimulated human cyto- megalovirus (HCMV) major immediate early (IE) enhancer/promoter activity in the HL-60 granulocyte/ monocyte progenitor cell line when added to transfected cells. In U-937 monocytic cells, by contrast, TNF-~ had no stimulatory effect and the addition of PMA produced only marginal stimulation. In the mature THP-1 mono- cytic cell line and in differentiated HL-60 cells, addition of TNF-~ caused inhibition of the IE enhancer/promoter activity. The stimulating effect of PMA, as observed in
Article
The majority of the human population harbors latent cytomegalovirus. Although CD14+ peripheral blood mononuclear cells have been implicated as sites of latency, the conformation of the latent viral genome in these cells is unknown. In this study, the conformation of viral genomic DNA was assessed in CD14+ cells from healthy virus seropositive carriers using an electrophoretic separation on native agarose gels in combination with polymerase chain reaction detection. Here we show that the viral genome migrates as a circular plasmid with a mobility equivalent to a circular 230-kb Shigella flexneri megaplasmid marker. Neither linear nor complex or integrated forms of the viral genome were detected. This report provides further evidence that the CD14+ cell population is an important site of viral latency in the naturally infected human host. Detection of the viral genome as a circular plasmid during latency suggests that this virus maintains its genome in a manner analogous to other herpesviruses where latent viral genome conformation has been studied.
Article
With the aid of two-color immunofluorescence and flow cytometry, a new subset of cells coexpressing CD14 and CD16 antigens can be identified in human peripheral blood. Using the monoclonal antibody My4, these CD14+/CD16+ cells account for 2.2% of the mononuclear cells and form about 13% of all cells identified by the monocyte-specific CD14 monoclonal antibody. The CD14+/CD16+ cells can be assigned to the monocyte lineage based on typical morphology, on expression of additional monocyte-associated molecules, on the ability to form reactive oxygen intermediates and on the expression of monocyte- specific NaF-sensitive esterase. Light scatter analysis revealed lower forward angle and right angle light scatter for the CD14+/CD16+ cells compared with the regular monocytes, and the average cell size was determined to be 13.8 and 18.4 microns, respectively. Expression of class II antigens on these “small monocytes” was twofold higher compared with the regular monocytes. By contrast, the capacity to perform adherence to plastic surfaces, as well as the ability to phagocytize antibody-coated erythrocytes was clearly reduced in the CD14+/CD16+ monocyte subset as compared with the regular monocytes. Hence the CD14+/CD16+ cells appear to represent a new monocyte subset with a distinct functional repertoire. A survey of various tissues revealed that a large proportion of the alveolar macrophages, but not of the peritoneal macrophages, express the CD14+/CD16+ phenotype.
Article
In the past, CD14 has been viewed simply as a useful marker molecule for monocytes and macrophages. Now, new findings on its role in binding of LPS-LBP complexes and in signal transduction have engendered renewed interest in the properties of CD14. Here, CD14 function, its expression in different cell types and the regulation of expression, including the generation of soluble CD14, are described, and the diagnostic value of CD14 in various diseases is discussed.
Article
We studied the effects of cytomegalovirus (CMV) infection on 301 cardiac transplant recipients who were treated during the cyclosporine era of immunosuppression (1980 to the present). These patients received varying combinations of cyclosporine, azathioprine, prednisone, rabbit antithymocyte globulin, and OKT3 as their immunosuppressive therapy. Two hundred ten patients were free of CMV infection (non-CMV group). During the same period CMV infection developed in 91 patients, as manifested by a fourfold IgG serologic titer rise, demonstration of CMV inclusion bodies in tissue, or positive cultures for the virus (CMV group). The rate of graft rejection was significantly higher in the CMV group. Graft atherosclerosis was significantly more severe in the CMV group as judged by angiographic criteria or by pathologic study. Patient survival rates were significantly lower in the CMV group. Death caused by graft atherosclerosis was significantly more common among patients in the CMV group. Finally, the graft loss rate (from either death or retransplantation for atherosclerosis) was significantly greater in the CMV group. These data demonstrate that CMV infection in cardiac transplant recipients is associated with more frequent rejection, graft atherosclerosis, and death. (JAMA. 1989;261:3561-3566)
Article
In the attempt to evaluate the role of Autologous and Allogeneic Bone Marrow Transplantation, we have retrospectively analyzed 159 patients with Acute Myeloid Leukemia in first complete remission treated in our Unit, most of whom were referred from other Institutions. High-dose therapy was uniform and consisted of cyclophosphamide 60 mg/kg/d on two consecutive days and TBI in a single dose (10 Gy) for ABMT patients and in fractionated doses (3.3 Gy 3 days) for BMT patients. Eight years actuarial survival was similar in two groups (52% for BMT and 49% for ABMT). The actuarial risk of relapse for BMT and ABMT was 29% and 43%, respectively. Considering that none of ABMT patients was purged with in vitro technique, this review seems to confirm the importance of in vivo purging with postremission intensification, immediately before the harvesting. Of course, more patients and a longer follow-up are needed to drow final conclusions.
Article
Viral promoters are commonly used as regulatory elements in gene therapy vectors due to their strong activity in various cell lines in vitro. However, transgene expression under the control of viral promoters in vivo has been shown to be limited to a short period of time. Several mechanisms for the transient expression of the delivered transgene may be important including deletion of transduced cells or promoter downregulation. Recently we reported that cytokines may either decrease or increase the activity of the human cytomegalovirus (hCMV) promoter in monocytes depending on the differentiation status of the transduced cells. For many applications, the gene of interest has to be delivered into an inflammatory milieu (tumour, ischaemia/reperfusion, vector-induced inflammation etc.). In this report we investigated the influence of various inflammatory cytokines on the hCMV-IE promoter activity in transduced human primary endothelial cells (Huvec) in vitro, which may be the first target cells after gene transfer into different organs. Cultured cells were infected with an E1-deleted adenoviral vector encoding for E. coliβ-galactosidase (Adβ-gal) driven by the hCMV-IE promoter and incubated either with or without various cytokines. Our results indicate that interferon-γ (IFN-γ) and interleukin-10 (IL-10) downregulate promoter activity in endothelial cells whereas, in contrast, tumour necrosis factor (TNF-α), interleukin 1β (IL-1β) and interleukin 4 (IL-4) increased the promoter activity. These results suggest that inflammatory processes influence the in vivo expression of transferred viral promoter controlled genes of interest.
Article
Hematopoietic cells and their progenitors play important roles in human cytomegalovirus latency and reactivation. Latent infection has been evaluated in defined populations of myeloid-lineage-committed progenitor cells coexpressing CD33 and CD15 or CD33 and CD14 along with the dendritic cell markers CD1a and CD10. These CD33+ cell populations were found to support latency and expression of viral latency-associated transcripts and to undergo reactivation of productive viral replication when differentiated in the presence of human fibroblasts. Reactivation was also observed when myeloid cells were carried in the presence of fibroblast-conditioned medium or medium supplemented with certain cytokines (interferon gamma, tumor necrosis factor alpha, interleukin 4, or granulocyte-macrophage colony-simulating factor), suggesting that cell differentiation pathways act as determinants of reactivation. More primitive CD34+ hematopoietic cells were also found to be susceptible to viral infection and latency was maintained as these cells differentiated into CD33+-lineage-committed populations. Between 0.01% and 0.001% of CD33+ CD14+ or CD33+ CD15+ bone marrow mononuclear cells isolated from naturally infected individuals were found to express latent transcripts. Thus, cytomegalovirus is carried within a small percentage of myeloid and dendritic cell progenitors in the healthy seropositive host. Virus reactivation may be triggered by factors associated with the inflammatory response.
Article
We have previously shown that a major site of persistence of human cytomegalovirus (HCMV) in healthy carriers is in peripheral blood monocytes. However, monocytes are difficult to infect in vitro with HCMV, and HCMV gene expression cannot be reproducibly detected in peripheral blood cells of healthy carriers. Here we show that the monocytic cell line THP1 is non-permissive for HCMV infection due to a block in expression of the HCMV major immediate early (IE) promoter. This repression is correlated with the presence of a differentiation-specific cellular factor which binds to the imperfect dyad symmetry and the 21 bp enhancer repeats of the major IE promoter regulatory region and which has characteristics of MBF1, a factor which we have previously defined in HCMV non-permissive, undifferentiated teratocarcinoma cells. Both differentiation of THP1 cells into macrophages, which results in a decrease in this factor, or deletion of the factor's binding sites from the IE promoter/enhancer lifts this repression and permits expression from the major IE promoter.