ArticlePDF Available

Regulatory T Cells Are Locally Induced during Intravaginal Infection of Mice with Neisseria gonorrhoeae

American Society for Microbiology
Infection and Immunity
Authors:

Abstract and Figures

Neisseria gonorrhoeae is a gram-negative diplococcus that in human beings produces gonorrhea. Much clinical evidence has led to the conclusion that gonococcus has important mechanisms to evade host immune functions; however, these mechanisms are only now beginning to be elucidated. In this study, we determined that the BALB/c mouse is a good animal model to study gonococcus infection and examined the immune response against the bacteria. We determined that after intravaginal inoculation of mice with Neisseria gonorrhoeae, the bacteria reached and invaded the upper female reproductive tissues and elicited a T-cell-specific immune response associated with a very weak humoral response, altogether resembling gonococcus infection and disease in women. Remarkably, in the draining lymph nodes of the genital tracts of infected mice, we found an increase of regulatory T lymphocytes, namely, transforming growth factor β1-positive CD4+ T cells and CD4+ CD25+ Foxp3+ T cells. Altogether, results indicate that N. gonorrhoeae induces regulatory T cells, which might be related to the local survival of the pathogen and the establishment of a chronic asymptomatic infection.
This content is subject to copyright. Terms and conditions apply.
INFECTION AND IMMUNITY, Dec. 2008, p. 5456–5465 Vol. 76, No. 12
0019-9567/08/$08.000 doi:10.1128/IAI.00552-08
Copyright © 2008, American Society for Microbiology. All Rights Reserved.
Regulatory T Cells Are Locally Induced during Intravaginal Infection
of Mice with Neisseria gonorrhoeae
Mo´nica Imarai,
1
* Enzo Candia,
1
Carolina Rodriguez-Tirado,
1
Javier Tognarelli,
1
Mirka Pardo,
1
Tomas Pe´rez,
1
Daniel Valde´s,
1
Sebastia´n Reyes-Cerpa,
1
Pablo Nelson,
1
Claudio Acuna-Castillo,
1
and Kevin Maisey
1,2
Laboratorio de Inmunología, Departamento de Biología, Facultad de Química y Biología, Universidad de Santiago de
Chile, Alameda 3363, Correo 40, Casilla 33, Santiago, Chile,
1
and Unidad de Investigacio´n y Desarrollo,
Macrocap S.A., Santiago, Chile
2
Received 6 May 2008/Returned for modification 18 June 2008/Accepted 19 September 2008
Neisseria gonorrhoeae is a gram-negative diplococcus that in human beings produces gonorrhea. Much
clinical evidence has led to the conclusion that gonococcus has important mechanisms to evade host immune
functions; however, these mechanisms are only now beginning to be elucidated. In this study, we determined
that the BALB/c mouse is a good animal model to study gonococcus infection and examined the immune
response against the bacteria. We determined that after intravaginal inoculation of mice with Neisseria
gonorrhoeae, the bacteria reached and invaded the upper female reproductive tissues and elicited a T-cell-
specific immune response associated with a very weak humoral response, altogether resembling gonococcus
infection and disease in women. Remarkably, in the draining lymph nodes of the genital tracts of infected mice,
we found an increase of regulatory T lymphocytes, namely, transforming growth factor 1-positive CD4
T
cells and CD4
CD25
Foxp3
T cells. Altogether, results indicate that N. gonorrhoeae induces regulatory T
cells, which might be related to the local survival of the pathogen and the establishment of a chronic
asymptomatic infection.
Gonorrhea is a sexually transmitted disease produced by the
gram-negative diplococcus Neisseria gonorrhoeae. In women,
infection affects the cervix and may spread to the uterus and
oviduct, inducing endometritis and pelvic inflammatory dis-
ease. Strikingly, about 50% of the cases proceed without symp-
toms, inducing damage mainly in the fallopian tube, while in
men infection occurs with distinctive clinical symptoms (11,
21). The hallmark of humoral immune response against N.
gonorrhoeae is the extremely low levels of antigonococcal an-
tibodies found in serum and secretions of the human (male and
female) during infection (19, 20). Antibodies are directed
against several major membrane molecules, such as the pilus-
and opacity-associated outer membrane proteins (Pil and Opa,
respectively), the porin protein (Por), and the lipooligosaccha-
ride (5, 19, 20, 38, 39, 55). Although some of these have
bactericidal activity, they are not protective and seem to be
blocked by outer membrane protein 3 (RmP)-specific immu-
noglobulin Gs (IgGs) (38).
The highly asymptomatic infection in women and the poor
immune response related to gonococcus prime and multiple
challenges might actually be related to mechanisms of immune
evasion acquired by the bacteria to constrain the immune
response. Moreover, the presence of a significant, although
weak, humoral immune response during gonococcus infection
(19) suggests that the bacteria might stimulate a regulatory
type of immune response. One of these mechanisms might be
the induction of noninflammatory responses dominated by
Th2-type cytokines and the activation of regulatory T cells
(Tregs), which in turn would contribute to the suppression of
most of the mechanisms of protection against intracellular
pathogens (2, 40). Tregs are CD4
T lymphocytes involved in
the induction of suppressor responses; it has been determined
experimentally that they show several phenotypes. One of the
major characteristics is related to the presence of transforming
growth factor 1 (TGF-1) (52), and subgroups can be distin-
guished because of the expression of CD25 and the Foxp3
transcription factor. CD4
CD25
T cells correspond to a
subgroup expressing Foxp3, which originate in the thymus and
are called natural Tregs, while the CD4
CD25
T cells are
induced at the periphery after antigenic stimulation in the
presence of a distinct cytokine environment (28, 31, 50, 52).
TGF-1 blocks T-cell proliferation, inhibits Th1, Th2, and
CTL differentiation, and moreover induces Foxp3 expression
in Tregs (27).
Overall, primary cell and organ culture systems have been
successfully developed to examine the initial phase of gono-
coccal pathogenesis (9, 10, 13, 29, 48) where several gonococ-
cus membrane components, such as Pil and Opa, are highly
relevant (30). However, the mechanisms explaining why female
infection occurs in a high number of cases without inflamma-
tory signs, with low levels of antibody induction, and with no
disease resolution responses are not understood. Understand-
ing pathogenesis at this level has been greatly hindered due to
the ethical considerations associated with human research
work and the lack of an animal model of experimental infec-
tion (9). Only recently, a murine model introduced by Ann
Jerse has allowed studies of protection against gonococcus
infection (23, 37).
* Corresponding author. Mailing address: Laboratorio de Inmu-
nología, Departamento de Biología, Facultad de Química y Biología,
Universidad de Santiago de Chile, Alameda 3363, Correo 40, Casilla
33, Santiago, Chile. Phone: 56-2-718 1164. Fax: 56-2-681 2108. E-mail:
monica.imarai@usach.cl.
Published ahead of print on 29 September 2008.
5456
In this study, we have further investigated the mouse exper-
imental model of N. gonorrhoeae infection and demonstrated
that the bacteria reach and invade the upper female reproduc-
tive tissues (uterus and oviduct), resembling gonococcus infec-
tion and disease in women. The murine model allowed us to
determine that infection elicits a T-cell-specific immune re-
sponse associated with a weak humoral response. In addition,
the local response includes the induction of regulatory TGF-
1
T cells which, acting to suppress the activation of the
immune system, would support the occurrence of infection.
Altogether, results indicate that N. gonorrhoeae induces regu-
latory mechanisms of immunity which, in turn, might explain
the local survival of the pathogen and the establishment of a
chronic asymptomatic infection in women.
MATERIALS AND METHODS
Bacteria and culture conditions. The following three different variants of N.
gonorrhoeae strain P9, kindly provided by Myron Christodoulides (University of
Southampton, United Kingdom), were used in this study: P9-13 (Pil
Opa
a
),
P9-16 (Pil
Opa
b
), and P9-17 (Pil
Opa
b
) (8). Bacteria were routinely grown
on GC agar (Becton Dickinson, Maryland) supplemented with 1% IsoVitaleX
(Becton Dickinson) for 18 h at 37°C in 5% CO
2
. Gonococcal variants containing
a red shift mutant green fluorescent protein (GFP) plasmid were grown in GC
agar containing ampicillin (5 g/ml). Analysis of colony morphology under a
stereomicroscope and Western blotting for the detection of Pil and Opa were
routinely performed to discard phenotypic variability. Monoclonal antibodies
against Pil and Opa were kindly provided by Mumtaz Virji (University of Bristol,
United Kingdom) and Mark Achtman (Max Planck Institute, Berlin, Germany),
respectively.
Animals. BALB/c mice were obtained from the Institute of Public Health
(Santiago, Chile), housed under regulated light and temperature conditions, and
sacrificed by cervical dislocation. Research was conducted in accordance with
institutional guidelines and with the International Guiding Principles for
Biomedical Research Involving Animals of the Society for the Study of
Reproduction.
Mouse uterine cell cultures. Cultures were prepared from mouse uteri as
previously described (17). Briefly, uterine horns were washed in RPMI 1640
medium (GIBCO Invitrogen Co., Carlsbad, CA) containing 100 U/ml penicillin
and 100 g/ml streptomycin (GIBCO Invitrogen Co.). Organs were cut into
small pieces and then transferred to a plate containing 0.25% trypsin and 2.5%
pancreatin (Life Technologies, Grand Island, NY) and incubated for 60 min at
4°C followed by 60 min at 20°C. Organs were transferred to a sterile tube
containing 15 ml of cold Hanks’ balanced salt solution (GIBCO Invitrogen Co.).
Digested uteri were then vortexed and the released epithelial cells were recov-
ered and transferred to a clean tube. The procedure was repeated three times
and cells in suspension were collected, centrifuged for 8 min at 180 g, and
resuspended in RPMI 1640 supplemented with 10% fetal bovine serum (Hy-
Clone Laboratories, Inc., Logan, UT), 1 mM L-glutamine, 100 g/ml streptomy-
cin, and 100 U/ml penicillin. Uterine cells were cultured at 37°C in 5% CO
2
until
they reached 90% confluence. Positive immunostaining with a mouse anticyto-
keratin monoclonal antibody (Chemicon International, Temecula, CA) and neg-
ative staining with antivimentin antibody (Chemicon International) confirmed
the epithelial origin of these cells. Typically, cultures had more than 95% epi-
thelial cells.
Infection of primary uterine cell cultures. Gonococcal isolates were taken
from frozen stocks and cultured on GC agar plates at 37°C in a 5% CO
2
-air
atmosphere. Bacteria were then scraped from confluent culture plates and re-
suspended in 1 ml of Dulbecco’s modified Eagle’s medium without phenol red
(GIBCO Invitrogen Co.). The concentration was estimated by comparison as 1
optical density unit at 600 nm, corresponding to 3.2 10
9
CFU/ml. Cells were
infected with N. gonorrhoeae fluorescence variants at a multiplicity of infection of
10 and cultured at 37°C in 5% CO
2
until they reached 90% confluence. To
determine the uptake of bacteria, epithelial cells were treated with 100 g/ml of
gentamicin for 60 min at 37°C to kill extracellular bacteria. Cells were then
diluted in phosphate-buffered saline (PBS) containing 1% saponin and incubated
for 15 min at 37°C. A tenfold dilution series was prepared and 100-l portions
were spread on GC plates. The internalization of bacteria in epithelial cells was
evident by the recovery of colonies composed of gram-negative, oxidase-positive
diplococci after 18 h of growth at 37°C.
Mouse experimental infection. Experimental infection of BALB/c mice was
performed as previously described (23) with some modifications. Briefly, 7 days
before bacterium inoculation, four groups of five female BALB/c mice (6 to 8
weeks old) were daily injected with 3 g of cetrorelix acetate (subcutaneous
administration). On day 4, half of the groups were additionally inoculated daily
with 300 ng of estradiol [estra-1,3,5(10)-triene-3,17-beta-diol; Sigma] until the
end of the experiment. On day 7, one estradiol-treated group and one control
group were intravaginally inoculated with 10
8
CFU N. gonorrhoeae fluorescence
variants suspended in Dulbecco’s modified Eagle’s medium without phenol red
(GIBCO Invitrogen Co.). The other two groups were treated with medium. The
procedure was performed using a sterile syringe connected to flexible tubing
which allowed instillation. Tubing was inserted into the vagina, first dorsally and
then cranially, until the cervix was reached. Fifty-microliter portions of bacterial
suspension were inoculated in the mice. At 1, 3, and 5 days postinoculation, the
genital tracts of infected and control mice were removed. Uteri were weighed to
assess the effectiveness of estradiol treatment.
Microscopy. For microscopic analysis, cells were cultured on coverslips and
infected as described above. After the indicated times, cells were fixed with 1%
paraformaldehyde in PBS. In the cases of vaginas, uteri, and oviducts, tissues
were fixed in 4% paraformaldehyde in PBS for 1 h before sequential transfer to
10% sucrose in PBS for 1 h, 20% sucrose in PBS for 1 h, and 30% sucrose in PBS
overnight. Organs were mounted in embedding compound (Cryo-M-Bed; Bright
Instrument Co. Ltd., Huntingdon, United Kingdom) and frozen at 20°C. Slices
of5to10m were cut using a Bright Starlet cryostat at 20°C. Fixed cultured
cells and tissue sections were counterstained using a solution of 1 g/ml pro-
pidium iodide in PBS and mounted in a solution of PBS containing 10% (vol/vol)
1,4-diazobicyclo[2.2.2] octane (Dabco; Sigma) and 90% (vol/vol) glycerol. Anal-
ysis of interactions of N. gonorrhoeae with uterine cells was performed by con-
focal microscopy (Zeiss LSM510). Briefly, confocal z-plane slices were obtained
and orthogonal views and three-dimensional images were generated from iso-
lated cells and tissues. For transmission electron microscopy analysis, cells were
fixed in 1% glutaraldehyde in PBS (pH 7.4) and dehydrated for embedding in
epoxy resin. Ultrathin sections were stained with uranyl acetate and lead citrate.
Control and infected samples were viewed with a Philips EM-200 transmission
electron microscope.
ELISA. An enzyme-linked immunosorbent assay (ELISA) was performed for
serum titration. Briefly, whole-protein extracts were prepared by freeze-thawing
N. gonorrhoeae strain P9-17 in water, and protein levels were determined by the
Bradford assay. Extracts prepared from Escherichia coli JM109 were used as a
negative control. Microtiter plates (Falcon; Becton Dickinson Co., Santiago,
Chile) were activated with 10 g of protein per well and incubated overnight at
4°C for 24 h. After the washing, sera collected from infected (n4) and control
(n4) mice were added to plates (at 1:20, 1:50, 1:100, 1:200, and 1:400
dilutions). Specific antibodies to gonococcus were detected with goat anti-mouse
IgG-alkaline phosphatase conjugate (Sigma) and p-nitrophenyl phosphate
(Sigma). Each mouse serum sample was assayed in duplicate.
T-cell antigen proliferation assays. To determine specific T-cell responses,
mice were intravaginally infected as described above and then boosted intraperi-
toneally on day 15. Seven days after the boost (day 22), mice were sacrificed and
the spleens and lymph nodes (renal, iliac, and caudal) were removed (34). CD4
T cells were isolated by negative selection using antibody-coated magnetic beads
(Dynal; Invitrogen). Isolated T cells (1 10
5
cells/well) from treated and control
mice were cultured with mitomycin-treated autologous splenocytes (2 10
4
cells/well) previously pulsed with 1 or 10 g of whole-protein extract of N.
gonorrhoeae. Experiments were carried out in 96-well plates in triplicate. As a
positive control, T cells were stimulated with phytohemagglutinin (PHA) (10
l/ml; GIBCO Invitrogen Co.). Plates were incubated at 37°C and 5% CO
2
for
72 h and then pulsed with 1 Ci methyl-[
3
H]thymidine (Amersham Biosciences,
Buckinghamshire, United Kingdom) for the final 18 h before harvesting. The
amount of incorporated radioactivity was measured using a liquid scintillation
counter (Tri-Carb 2100 TR; Packard). Data were expressed as experimental
minus control counts per minute.
Leukocyte isolation. A previously described protocol was followed (24).
Briefly, uteri dissected from mice (four animals/experiment) were placed in
sterile ice-cold Hanks’ balanced salt solution (GIBCO, Grand Island, NY),
weighed, and then transferred to a mixture of pancreatin (GIBCO), trypsin
(Sigma, St. Louis, MO), and DNase (Worthington, Lakewood, NJ). Under sterile
conditions, uterine tissues in the enzyme mixture were cut into small pieces,
transferred to six-well culture plates, and incubated first for1hat4°Candthen
for an additional hour at room temperature with gentle shaking. Cells were
recovered from the supernatant and were pooled for each experiment.
Immunofluorescence staining and analysis. For TGF-1 detection, leukocytes
(5 10
5
cells/ml) were incubated with anti-TGF-1 antibody (1:100; Santa Cruz
VOL. 76, 2008 T-CELL IMMUNE RESPONSE AGAINST GONOCOCCUS 5457
Biotechnology) for1hat4°C. After being washed with PBS, cells were incubated
with a fluorescein isothiocyanate-conjugated polyclonal anti-rabbit IgG (1:200;
Santa Cruz Biotechnology) for1hat4°C. For CD11b detection, cells were
incubated for 30 min on ice with 2 ml of RPMI-10% fetal bovine serum with 10%
heat-inactivated normal mouse serum to reduce the nonspecific binding of an-
tibodies. After incubation, 10
6
cells/ml were labeled with phycoerythrin-conju-
gated anti-mouse CD11b (Santa Cruz Biotechnology, Inc.) for1hat4°C. For
analysis of CD4
CD25
Foxp3
T lymphocytes, 1 10
6
cells were labeled
using PE-Cy5.5-conjugated anti-CD4 antibody (clone GK1.5; eBioscience, San
Diego, CA) and fluorescein isothiocyanate-conjugated anti-CD25 antibody
(clone PC61.5; eBioscience). After being washed, cells were resuspended in 200
l of Fix/Perm buffer (eBioscience) and left at 4°C for 30 min. After being
washed, cells were incubated with phycoerythrin-conjugated anti-mouse Foxp3
(clone FJK-16S; eBioscience, San Diego, CA) for 30 min. Isotypic controls were
routinely included in all experiments. Labeled cells were analyzed on a FACScan
flow cytometer (Becton Dickinson) with CellQuest software.
Mixed lymphocyte reaction (MLR). Fresh T cells isolated from the lymph
nodes and spleens of control and infected BALB/c mice (H-2
d
) were first labeled
with 5 M carboxyfluorescein diacetate succinimidyl ester (CFSE). T-cell re-
sponders (10
5
) were incubated with spleen cells (5 10
5
) isolated from C57BL/6
(H-2
b
) mice in 200 l of RPMI at 37°C. After 5 days of culture in 96-well
round-bottom plates, cells were washed and fixed in 1% paraformaldehyde, and
CFSE dilution was assessed by flow cytometry.
RESULTS
N. gonorrhoeae infects isolated murine uterine cells. Previ-
ously, it had been extensively described that N. gonorrhoeae is
a strictly human pathogen; nevertheless, recent reports indi-
cated that the bacteria are also able to colonize the mouse
genital female tract. Before initiating studies of infection in
vivo, our first aim was to demonstrate that the bacteria attach,
bind, and invade murine genital tract cells. Isolated mouse
uterine epithelial cells were chosen as targets for in vitro stud-
ies. Cells were infected with N. gonorrhoeae fluorescent (GFP)
strain P9, variant P9-17 (Pil
Opa
b
), at a multiplicity of
infection of 10 bacteria per cell. A simultaneous detection of
GFP-expressing gonococci and the red-stained nucleus on 0.4-
m-thickness cross-sectional images by confocal microscopy
showed that, after 24 h of incubation, gonococci were found
associated with the epithelial cells, with a significant number
inside the cells surrounding the nucleus (Fig. 1A). Similar
results were obtained for variants P9-16 and P9-13. The intra-
cellular localization of the bacteria was confirmed by transmis-
FIG. 1. (A) Confocal photomicrograph of serial sections (0.4 m) shows the presence of N. gonorrhoeae (P9-17) around the nuclei of uterine
epithelial cells. Bacteria appear green, and the cell nuclei are red. (B) Uterine cells; control cultures stained with propidium iodide. (C and D)
Transmission electron microscopy images of isolated uterine epithelial cells infected with N. gonorrhoeae (variant P9-16) show multiple cytoplasmic
bacteria enclosed in membrane-bound vesicles (arrowheads). Bars, 20 m (B); 1 m (C); 0.1 m (D).
5458 IMARAI ET AL. INFECT.IMMUN.
sion electron microscopy, as micrographs exhibited N. gonor-
rhoeae P9-16 enclosed in membrane-bound vesicles of the
mouse uterine cells (Fig. 1C and D). Similar structures were
not found in control cells (not shown). In addition, an average
of 3.5 10
4
intracellular gonococci per well (24-well plate) was
recovered from infected uterine epithelial cells after selective
antibiotic killing of extracellular bacteria. Colonies had the
expected morphology, i.e., variants exhibited small opaque col-
onies with sharp edges, which correspond to Pil
Opa
gono-
cocci. Much higher numbers of bacteria were observed under
fluorescent microscopy, indicating that the major portion of
gonococci appear attached to the uterine cell, exactly as pre-
viously described for the infection of human endometrial and
oviductal cells (8). Results demonstrated that N. gonorrhoeae is
able to attach and invade the uterine epithelial, thus unequiv-
ocally identifying murine cells as a target of gonococcus infec-
tion.
N. gonorrhoeae invades the upper female reproductive tract
of the mouse. Once it was determined that N. gonorrhoeae is
able to colonize isolated uterine epithelial cells, the next step
was to corroborate that the bacteria colonize the upper repro-
ductive tract in mice, as seen for women. Because estradiol
seems to be crucial for gonococcus infection in mice (23), we
inoculated bacterial strain P9-17 expressing GFP (10
8
CFU)
FIG. 2. Confocal images of uterine sections of mice intravaginally infected with GFP-expressing N. gonorrhoeae (variant P9-17). The cell nuclei
are stained with propidium iodide. (A) Three days after inoculation. Bar, 50 m. (B) Five days after inoculation. Bar, 20 m. (C) Orthogonal views
of a midplane zsection; height, 1.3 m. (D) Negative control. Arrows denote fluorescent bacteria.
VOL. 76, 2008 T-CELL IMMUNE RESPONSE AGAINST GONOCOCCUS 5459
into the vaginas of estradiol-treated BALB/c mice. Uteri were
removed after several days postinoculation, processed as de-
scribed in Materials and Methods, and analyzed by confocal
microscopy. Fluorescent bacteria were found in the uteri of all
inoculated mice (n25), particularly in the epithelial and
subepithelial tissues of the organs. On day 1 after inoculation,
gonococci were mostly observed in the epithelium and occa-
sionally in the stroma, while on day 3, most of the bacteria were
present in the stroma (Fig. 2A). On day 5, the bacteria were
distributed throughout the stroma but also in the most external
tissues of the uterus (Fig. 2B). An orthogonal view of the
infected uterus (midplane zsection; height, 1.3 m,) confirmed
the bacterial localization within the depths of the tissue (Fig.
2C). As the bacteria were also found in the epithelial and
subepithelial tissues of the vaginas and the oviducts, overall
these results indicate that intravaginal inoculation of N. gonor-
rhoeae allows the bacteria to reach, attach to, and invade the
mucosal tissues of the lower and upper organs of the mouse
genital tract. Further experiments were performed to evaluate
the contribution of estradiol in allowing infection. The exper-
iments were repeated several times with different mice at ran-
dom stages of the reproductive cycle, and surprisingly, infec-
tion was also observed for all mice tested. Moreover, infection
seemed to be persistent, because the bacteria were detected as
late as 22 days postinoculation; however, as is the case for
women, no clinical signs of the disease were apparent in
the mice.
Immune response. After corroborating that gonococcus in-
fects the mouse genital tract, as it does in humans, we wanted
to establish whether the bacteria also induce an antigen-spe-
cific response in mice. To evaluate the humoral immune re-
sponse, sera from infected and control animals obtained at day
22 after treatment were tested using ELISA against a whole-
protein extract of the P9-17 bacterial variant. Very low anti-
body titers in response to N. gonorrhoeae were detected for two
out of the four infected mice (Fig. 3), while as expected, no
detectable antibody was observed in serum samples of control
mice. To examine the T-cell response, CD4
T lymphocytes
isolated from uterus-draining lymph nodes from infected and
control mice were examined for the ability to proliferate in
response to a whole-protein extract of the bacteria (variant
P9-17). The results show that T cells from infected BALB/c
mice exhibited a dose-dependent T-cell-proliferative response
to gonococcus protein extract (Fig. 4). These findings indicate
that N. gonorrhoeae induces a local antigen-specific T-cell re-
sponse associated with a low- or no-antibody response during
experimental infection in the mouse.
Tregs increase in infected animals. As mentioned previ-
ously, specific stimulation of Tregs might explain the weak
antigonococcal humoral immune response and the absence of
protective immunity. Thus, we evaluated whether the BALB/c
T-cell response to gonococcus also involves the stimulation of
Tregs. As the major phenotype of Tregs is the synthesis of
TGF-1, we first quantified the percentage of TGF-1-produc-
ing T cells locally induced 22 days after infection. In six inde-
pendent experiments, the percentages of CD4
TGF-1
T
cells isolated from the regional lymph nodes of infected mice
(Fig. 5C, top right, and F) were twofold higher than those of
cells of the control group of mice (Fig. 5B, top right, and F).
No changes in the CD4
TGF-1
T lymphocytes were ob-
served for T cells isolated from spleens of the same animals
(Fig. 5D and E, top right, and F). Interestingly, a strong shift
in CD4
TGF-1
splenocytes from infected (Fig. 5E) versus
noninfected (Fig. 5D) mice occurred. This effect was not ob-
served in the regional lymph nodes, and the cells might corre-
spond to a different type of regulatory cells. In addition, we
examined the stimulated T cells in four independent experi-
ments to determine the presence of CD25 and Foxp3, addi-
tional molecular markers of Tregs. Results revealed that the
percentage of CD4
CD25
Foxp3
T cells detected in the
local lymph nodes of the group of infected animals was higher
FIG. 4. Proliferative response of isolated CD4
T cells (LT) from
infected mice to a protein extract of N. gonorrhoeae. T cells were
stimulated with PHA or added to BALB/c splenocytes (antigen-pre-
senting cells [APC]) pretreated with 1 or 10 g of whole-protein
extract of N. gonorrhoeae P9-17 (Ngo). Proliferation was measured by
[
3
H]thymidine incorporation. Each bar represents the mean stan-
dard error of the mean (SEM) from triplicates. Data were expressed as
experimental minus control counts per minute. Two independent ex-
periments were done with similar results. Asterisks and double aster-
isks indicate Pvalues of 0.05 and 0.0001, respectively, versus values
for LT plus APC only by analysis of variance.
FIG. 3. Analysis of serum response to N. gonorrhoeae by ELISA.
Mouse sera were assayed on a whole-protein extract of the P9-17
bacterial variant in a reciprocal dilution series starting at 1:50, and
binding was detected with an alkaline phosphatase anti-mouse conju-
gate. Black symbols represent infected mice (n4) and open symbols
control mice (n4).
5460 IMARAI ET AL. INFECT.IMMUN.
than that of the control group (P0.005) (Fig. 6A and B, top
right, and E). Once again, no statistically significant differences
were found when cells in the spleens of the animals were
quantified (Fig. 6C and D, top right, and E). Then, we tested
the suppressive function of Tregs in an allogeneic MLR. Only
cells from the lymph nodes of infected BALB/c mice, where
Tregs have been expanded by gonococcus infection, showed
suppression in the MLR stimulated by C57BL/6 spleen cells,
FIG. 5. Flow cytometric analysis of TGF-1-producing CD4
T cells. (A to E) Representative experiment. (A) Control of autofluorescence. (B) Cells
isolated from the regional lymph nodes of control mice. (C) Cells isolated from the regional lymph nodes of infected mice. (D) Splenocytes of control mice.
(E) Splenocytes of infected mice. The percentage of TGF-1
CD4
T cells is shown in the top right panel of each figure. (F) Percentages of TGF-1
CD4
T cells in lymph nodes and spleens of infected (white bars) and control (black bars) BALB/c mice. Bars represent means SEM from six independent
experiments. Lymph nodes of infected mice showed a significantly higher percentage of TGF-1-producing CD4
T cells than did the lymph nodes of the control
group. No differences were found in spleens. *,Pvalue of 0.02 by the Mann-Whitney U test.
VOL. 76, 2008 T-CELL IMMUNE RESPONSE AGAINST GONOCOCCUS 5461
while cells from the lymph nodes of control mice and from the
spleens of infected mice, where no T-regulatory expansion was
observed, showed a regular response to allogeneic C57BL/6
stimulation (Fig. 7). Overall, these results indicate that gono-
coccus infection of the murine genital tract induces a signifi-
cant stimulation of Tregs as part of the local immune response.
TGF-1
cells infiltrate the uterine tissues. We further
investigated whether a local immune response was also accom-
panied by a uterine infiltration of TGF-1
cells. Because N.
gonorrhoeae in vitro induces the expression of TGF-1 in mac-
rophages (unpublished data), cells isolated from the uteri of
infected and control animals were labeled with anti-TGF-1
and anti-CD11b (macrophage marker) antibodies. Data from a
flow cytometric analysis show that infected animals have a
moderate but significant increase of CD11b
TGF-1
cells in
the uteri (Fig. 8C and D, top right), which indicates that gono-
coccus induces a mucosal infiltration of macrophages whose
phenotype would favor the differentiation of Tregs. Interest-
FIG. 6. Flow cytometric detection of regulatory CD4
T cells. (A to D) Representative experiment; results are expressed as the percentage of
CD25
or CD25
T cells expressing FoxP3 in the gated CD4
T-cell population of local lymph nodes of control BALB/c mice (A), local lymph
nodes of infected BALB/c mice (B), spleens of control mice (C), and spleens of infected mice (D). (E) Percentages of CD4
CD25
Foxp3
T
cells in lymph nodes and spleens of infected (white bars) and control (black bars) mice. Bars represent means SEM of four independent
experiments. Lymph nodes of infected mice showed a significantly higher percentage of CD4
CD25
Foxp3
T cells than did the lymph nodes
of the control group. No differences were found in spleens. *,Pvalue of 0.05 by the Mann-Whitney U test.
5462 IMARAI ET AL. INFECT.IMMUN.
ingly, in addition to being associated with the infiltration of
macrophages, infection is also associated with the infiltration
of a great number of CD11b
TGF-1
cells (Fig. 8E), which
might correspond to Tregs.
DISCUSSION
In this study, we established that N. gonorrhoeae invades the
murine mucosa of the upper genital tract, results that confirm
and extend the extensive mouse work developed by Ann Jerse
and collaborators (23, 37, 43). Moreover, we demonstrated
that infection induces a weak humoral immune response asso-
ciated with a local increase of TGF-1
CD4
Tregs.
Until now, it has been widely accepted that N. gonorrhoeae is
a strictly human pathogen, which calls the validity of the use of
mice as an animal model of experimental infection into ques-
tion. This is reinforced by the fact that most of human recep-
tors for attachment and invasion do not seem to be expressed
in the mouse (30). However, we demonstrated here that N.
gonorrhoeae is able to infect murine epithelial cells isolated
from the uterus; the uptake of gonococcus by murine uterine
epithelial cells was demonstrated by means of three indepen-
dent assays, a gentamicin resistance test, confocal multiple
scan analysis, and transmission electron microscopy. More-
over, in vivo, we showed that GFP-expressing gonococci are
able to reach the upper genital organs and to invade uterine
tissues in all infected animals. Because in the experiments
there were no sources of green fluorescence other than that of
the bacteria, the uterus is normally sterile, and colonies of
gonococcus were recovered from the uterine cells of infected
mice, we are confident that the 1- to 2-m fluorescent spots
seen in tissues by confocal microscopy correspond to bacteria.
Therefore, although mice are not natural hosts for N. gonor-
rhoeae, results confirmed that gonococcus infects the lower
genital tract of BALB/c mice (23) and demonstrated for the
first time that the bacteria not only reach the upper genital
tract but also invade the upper murine mucosae, as occurs in
humans.
Analyses performed to characterize the immune response of
BALB/c mice showed that once gonococcus colonizes the fe-
male genital tract, bacterial antigens are detected by the im-
mune system, as revealed by the ability of CD4
T lymphocytes
to respond to the bacterial extract and by the presence of
specific antibodies in serum. However, as has been observed
for humans, antibodies that recognize N. gonorrhoeae were
FIG. 7. Geometric mean fluorescence of CFSE-labeled T cells. Re-
sponder cells were isolated from the lymph nodes and spleens of
control (white bars) and infected (black bars) BALB/c mice. For MLR,
stimulators were obtained from C57BL/6 spleens. CSFE-labeled re-
sponder cells were incubated with stimulator cells (ratio of 1:5) for 5
days and CFSE dilution was assessed by flow cytometry at the end of
the experiment. Bars represent geometric means (G mean) SEM
from nine independent experiments. Only cells from the lymph nodes,
where Tregs have been expanded by gonococcus infection, showed
suppression to the MLR. ,Pvalue of 0.0001 by the Kruskal-Wallis
test.
FIG. 8. Flow cytometry analysis of CD11b
TGF-1
cells infil-
trating genital tract tissues following infection with N. gonorrhoeae.
(A) Autofluorescence control. (B) Secondary antibody control. (C and
D) CD11b
TGF-1
cells from control (C) and infected (D) tissues.
(E) White bars, TGF-1
cells from control groups; black bars, TGF-
1
cells from infected mice.
VOL. 76, 2008 T-CELL IMMUNE RESPONSE AGAINST GONOCOCCUS 5463
detected at low levels in the infected mice. Gonococcal acti-
vation of CD4
T cells has also been described for humans, as
gonococcal pilus interaction with CD4
T cells induces the
activation and proliferation of lymphocytes and stimulates the
secretion of interleukin-10 (IL-10) (36). In contrast, it has also
been shown that N. gonorrhoeae Opa proteins mediate binding
to CEACAM-1 expressed by CD4
T cells and suppress the
activation and proliferation of naive lymphocytes (4, 25). Al-
though not fully comparable, this does not seem to occur in
mice, since PHA-stimulated naive T cells from control mice
showed proliferation (not shown).
The analysis of the nature of the CD4
T cells stimulated by
gonococci during the experimental infection leads us to ob-
serve that infection induces TGF-1
CD4
T-cell responders
in the mucosal lymph nodes, including a subset of CD25
Foxp3
Tregs. In particular, these Tregs showed a small but
significant increase in number which is similar to that observed
after infection with parasites (15, 47, 53). Interestingly, the
induction of this type of immunity did not occur at the systemic
level. Even more, the regulatory activity was confirmed in vitro
in an allogeneic MLR, indicating that N. gonorrhoeae might
induce this type of response to avoid the host mechanisms of
protection. The results support the idea that suppression is
induced at least in part by TGF-1, which either in a cell-
surface-bound or a secreted form inhibits the immune re-
sponse at a variety of levels, i.e., inhibits IL-2 production,
IL-12-dependent cell activation, and Th1 development, among
other responses (27, 46). The source of TGF-1 was not dem-
onstrated in this study; nevertheless, epithelial and stromal
cells of the reproductive organs of the mouse and human,
which are targets of gonococcus infection, express high levels
of TGF-1 and other molecules involved in conditioning im-
mune privilege sites (6, 17, 22, 51). Because TGF-1 also has
a role in the induction of Tregs (54) and, moreover, because
CD4
CD25
peripheral T cells can be converted to Foxp3
Tregs by stimulation via the T-cell receptor in the presence of
TGF-1 (7, 12), we believe that the cytokine milieu found in
the reproductive tract subsidizes the induction of Tregs by N.
gonorrhoeae. Moreover, antigen-presenting cells, such as mac-
rophages and dendritic cells, regularly present in the reproduc-
tive tissues (16, 44) might also contribute to Treg differentia-
tion, as they may produce TGF-1 after infection. Actually, we
detected an increase of CD11b
macrophages infiltrating the
reproductive mucosae of infected mice, which is consistent
with such a role. Moreover, a high number of CD11b
TGF-
1
cells, which seem to be T cells by morphology criteria, also
infiltrated the mucosal reproductive tissues during infection,
suggesting that Tregs also play an important role at the mu-
cosal level.
If extrapolated to humans, the results would indicate that
gonococcus infection in women might also be related to a
similar Treg induction, explaining in part the lack of protective
immune response. In fact, there are various examples of eva-
sion through pathogen-induced modulation of the immune
response; one of these has been reported for the human filarial
infection, where the parasite also induces an imbalance toward
the Th2 response, which is at the same time accompanied by a
diminished production of inflammatory factors and an increase
of anti-inflammatory components, including Tregs (1). In hu-
man beings, N. gonorrhoeae can induce other highly efficient
ways to overcome immune defense mechanisms, resulting in
disease or chronic infection (14). Previous studies have focused
attention on antigenic variation (49), epitope mimicry (18, 33),
and phagosome subversion (3) as molecular mechanisms of
immune evasion. Moreover, recent studies have explored a
putative role of T cells as a determinant of successful gono-
coccus infection. As mentioned above, Opa proteins from
gonococci have the ability to inhibit CD4
T-cell proliferation,
which will prevent adaptive immune response. What is more
interesting is the gonococcus-dependent induction of IL-10
(36), a cytokine that it is also involved in the differentiation of
Tregs (Tr1 type) (26, 41). A study of infected patients will shed
light on the mechanism of pathogenesis and the presumed role
of Tregs.
Altogether, the results showed that the infection of mice
with N. gonorrhoeae induces a tolerant type of response which
may correspond to a form of immune evasion that has not been
previously studied for gonorrhea. Most studies describing
these evasion mechanisms have been reported for parasite
infection and chronic diseases caused by viruses (14, 32, 35, 42,
45). In those cases, immune regulation seems to favor the
persistence of infection, which becomes evident when Tregs
are depleted, and the disease is soon controlled by the immune
system and the pathogen cleared. We are currently investigat-
ing the effect that the depletion of Tregs may have on the
development of gonorrhea; moreover, studies of human sub-
jects are currently in progress to investigate the role of these
regulatory cells during infection in women.
ACKNOWLEDGMENTS
This work was supported by grants 1020354 from the Fondo Nacio-
nal de Desarrollo Científico y Tecnolo´gico (FONDECYT), 020743IB
and 020540AC from the Direccio´n de Investigaciones Científicas y
Tecnolo´gicas (DICYT), and IPA06 from the Programa Bicentenario
en Ciencia y Tecnología (PBCT).
REFERENCES
1. Babu, S., C. P. Blauvelt, V. Kumaraswami, and T. B. Nutman. 2006. Regu-
latory networks induced by live parasites impair both Th1 and Th2 pathways
in patent lymphatic filariasis: implications for parasite persistence. J. Immu-
nol. 176:3248–3256.
2. Belkaid, Y., R. B. Blank, and I. Suffia. 2006. Natural regulatory T cells and
parasites: a common quest for host homeostasis. Immunol. Rev. 212:287–
300.
3. Binker, M. G., L. I. Cosen-Binker, M. R. Terebiznik, G. V. Mallo, S. E.
McCaw, E. L. Eskelinen, M. Willenborg, J. H. Brumell, P. Saftig, S. Grin-
stein, and S. D. Gray-Owen. 2007. Arrested maturation of Neisseria-con-
taining phagosomes in the absence of the lysosome-associated membrane
proteins, LAMP-1 and LAMP-2. Cell. Microbiol. 9:2153–2166.
4. Boulton, I. C., and S. D. Gray-Owen. 2002. Neisserial binding to CEACAM1
arrests the activation and proliferation of CD4T lymphocytes. Nat. Im-
munol. 3:229–236.
5. Brooks, G. F., and C. J. Lammel. 1989. Humoral immune response to
gonococcal infections. Clin. Microbiol. Rev. 2(Suppl.):S5–S10.
6. Chegini, N., Y. Zhao, R. S. Williams, and K. C. Flanders. 1994. Human
uterine tissue throughout the menstrual cycle expresses transforming growth
factor-beta 1 (TGF beta 1), TGF beta 2, TGF beta 3, and TGF beta type II
receptor messenger ribonucleic acid and protein and contains [125I]TGF
beta 1-binding sites. Endocrinology 135:439–449.
7. Chen, W., W. Jin, N. Hardegen, K. J. Lei, L. Li, N. Marinos, G. McGrady,
and S. M. Wahl. 2003. Conversion of peripheral CD4CD25naive T cells
to CD4CD25regulatory T cells by TGF-beta induction of transcription
factor Foxp3. J. Exp. Med. 198:1875–1886.
8. Christodoulides, M., J. S. Everson, B. L. Liu, P. R. Lambden, P. J. Watt, E. J.
Thomas, and J. E. Heckels. 2000. Interaction of primary human endometrial
cells with Neisseria gonorrhoeae expressing green fluorescent protein. Mol.
Microbiol. 35:32–43.
9. Dehio, C., S. D. Gray-Owen, and T. F. Meyer. 2000. Host cell invasion by
pathogenic neisseriae. Subcell. Biochem. 33:61–96.
5464 IMARAI ET AL. INFECT.IMMUN.
10. Draper, D. L., E. A. Donegan, J. F. James, R. L. Sweet, and G. F. Brooks.
1980. Scanning electron microscopy of attachment of Neisseria gonorrhoeae
colony phenotypes to surfaces of human genital epithelia. Am. J. Obstet.
Gynecol. 138:818–826.
11. Edwards, J. L., and M. A. Apicella. 2004. The molecular mechanisms used by
Neisseria gonorrhoeae to initiate infection differ between men and women.
Clin. Microbiol. Rev. 17:965–981.
12. Fantini, M. C., C. Becker, G. Monteleone, F. Pallone, P. R. Galle, and M. F.
Neurath. 2004. Cutting edge: TGF-beta induces a regulatory phenotype in
CD4CD25T cells through Foxp3 induction and down-regulation of
Smad7. J. Immunol. 172:5149–5153.
13. Fernandez, R., P. Nelson, J. Delgado, J. Aguilera, R. Massai, L. Velasquez,
M. Imarai, H. B. Croxatto, and H. Cardenas. 2001. Increased adhesiveness
and internalization of Neisseria gonorrhoeae and changes in the expression
of epithelial gonococcal receptors in the Fallopian tube of copper T and
Norplant users. Hum. Reprod. 16:463–468.
14. Finlay, B. B., and G. McFadden. 2006. Anti-immunology: evasion of the host
immune system by bacterial and viral pathogens. Cell 124:767–782.
15. Finney, C. A., M. D. Taylor, M. S. Wilson, and R. M. Maizels. 2007. Expan-
sion and activation of CD4()CD25() regulatory T cells in Heligmo-
somoides polygyrus infection. Eur. J. Immunol. 37:1874–1886.
16. Givan, A. L., H. D. White, J. E. Stern, E. Colby, E. J. Gosselin, P. M. Guyre,
and C. R. Wira. 1997. Flow cytometric analysis of leukocytes in the human
female reproductive tract: comparison of fallopian tube, uterus, cervix, and
vagina. Am. J. Reprod. Immunol. 38:350–359.
17. Grant, K. S., and C. R. Wira. 2003. Effect of mouse uterine stromal cells on
epithelial cell transepithelial resistance (TER) and TNFalpha and TGFbeta
release in culture. Biol. Reprod. 69:1091–1098.
18. Harvey, H. A., W. E. Swords, and M. A. Apicella. 2001. The mimicry of
human glycolipids and glycosphingolipids by the lipooligosaccharides of
pathogenic Neisseria and Haemophilus. J. Autoimmun. 16:257–262.
19. Hedges, S. R., M. S. Mayo, J. Mestecky, E. W. Hook III, and M. W. Russell.
1999. Limited local and systemic antibody responses to Neisseria gonorrhoeae
during uncomplicated genital infections. Infect. Immun. 67:3937–3946.
20. Hedges, S. R., D. A. Sibley, M. S. Mayo, E. W. Hook III, and M. W. Russell.
1998. Cytokine and antibody responses in women infected with Neisseria
gonorrhoeae: effects of concomitant infections. J. Infect. Dis. 178:742–751.
21. Hook, E. I., and H. H. Handsfield. 1999. Gonococcal infections in the adult,
p. 451–466. In K. K. Holmes, P. F. Sparling, P. A. Mårdh, S. M. Lemon,
W. A. Stamm, P. Piot, and J. N. Wasserheit (ed.), Sexually transmitted
diseases, 3rd ed. McGraw-Hill, New York, NY.
22. Imarai, M., L. Varela-Nallar, C. Figueroa-Gaete, P. Gonzalez, D. Valdes,
L. Velasquez, H. Cardenas, and K. Maisey. 2005. Fas ligand in the uterus
of the non-pregnant mouse induces apoptosis of CD4T cells. J. Reprod.
Immunol. 66:13–32.
23. Jerse, A. E. 1999. Experimental gonococcal genital tract infection and opac-
ity protein expression in estradiol-treated mice. Infect. Immun. 67:5699–
5708.
24. Kaushic, C., K. Grant, M. Crane, and C. R. Wira. 2000. Infection of
polarized primary epithelial cells from rat uterus with Chlamydia tracho-
matis: cell-cell interaction and cytokine secretion. Am. J. Reprod. Immu-
nol. 44:73–79.
25. Lee, H. S., M. A. Ostrowski, and S. D. Gray-Owen. 2008. CEACAM1 dy-
namics during Neisseria gonorrhoeae suppression of CD4T lymphocyte
activation. J. Immunol. 180:6827–6835.
26. Levings, M. K., S. Gregori, E. Tresoldi, S. Cazzaniga, C. Bonini, and M. G.
Roncarolo. 2005. Differentiation of Tr1 cells by immature dendritic cells
requires IL-10 but not CD25CD4Tr cells. Blood 105:1162–1169.
27. Li, M. O., Y. Y. Wan, S. Sanjabi, A. K. Robertson, and R. A. Flavell. 2006.
Transforming growth factor-beta regulation of immune responses. Annu.
Rev. Immunol. 24:99–146.
28. Lohr, J., B. Knoechel, and A. K. Abbas. 2006. Regulatory T cells in the
periphery. Immunol. Rev. 212:149–162.
29. Maisey, K., G. Nardocci, M. Imarai, H. Cardenas, M. Rios, H. B. Croxatto,
J. E. Heckels, M. Christodoulides, and L. A. Velasquez. 2003. Expression of
proinflammatory cytokines and receptors by human fallopian tubes in organ
culture following challenge with Neisseria gonorrhoeae. Infect. Immun. 71:
527–532.
30. Merz, A. J., and M. So. 2000. Interactions of pathogenic neisseriae with
epithelial cell membranes. Annu. Rev. Cell Dev. Biol. 16:423–457.
31. Miyara, M., and S. Sakaguchi. 2007. Natural regulatory T cells: mechanisms
of suppression. Trends Mol. Med. 13:108–116.
32. Montagnoli, C., S. Bozza, R. Gaziano, T. Zelante, P. Bonifazi, S. Moretti, S.
Bellocchio, L. Pitzurra, and L. Romani. 2006. Immunity and tolerance to
Aspergillus fumigatus. Novartis Found. Symp. 279:66–79, 216-219.
33. Moran, A. P., M. M. Prendergast, and B. J. Appelmelk. 1996. Molecular
mimicry of host structures by bacterial lipopolysaccharides and its contribu-
tion to disease. FEMS Immunol. Med. Microbiol. 16:105–115.
34. Parr, M. B., and E. L. Parr. 1990. Antigen recognition in the female repro-
ductive tract. I. Uptake of intraluminal protein tracers in the mouse vagina.
J. Reprod. Immunol. 17:101–114.
35. Peters, N., and D. Sacks. 2006. Immune privilege in sites of chronic infection:
Leishmania and regulatory T cells. Immunol. Rev. 213:159–179.
36. Plant, L. J., and A. B. Jonsson. 2006. Type IV pili of Neisseria gonorrhoeae
influence the activation of human CD4
T cells. Infect. Immun. 74:442–448.
37. Plante, M., A. Jerse, J. Hamel, F. Couture, C. R. Rioux, B. R. Brodeur, and
D. Martin. 2000. Intranasal immunization with gonococcal outer membrane
preparations reduces the duration of vaginal colonization of mice by Neis-
seria gonorrhoeae. J. Infect. Dis. 182:848–855.
38. Plummer, F. A., H. Chubb, J. N. Simonsen, M. Bosire, L. Slaney, I. Maclean,
J. O. Ndinya-Achola, P. Waiyaki, and R. C. Brunham. 1993. Antibody to
Rmp (outer membrane protein 3) increases susceptibility to gonococcal
infection. J. Clin. Investig. 91:339–343.
39. Plummer, F. A., H. Chubb, J. N. Simonsen, M. Bosire, L. Slaney, N. J.
Nagelkerke, I. Maclean, J. O. Ndinya-Achola, P. Waiyaki, and R. C. Brun-
ham. 1994. Antibodies to opacity proteins (Opa) correlate with a reduced
risk of gonococcal salpingitis. J. Clin. Investig. 93:1748–1755.
40. Pulendran, B. 2004. Modulating TH1/TH2 responses with microbes, den-
dritic cells, and pathogen recognition receptors. Immunol. Res. 29:187–196.
41. Roncarolo, M. G., S. Gregori, M. Battaglia, R. Bacchetta, K. Fleischhauer,
and M. K. Levings. 2006. Interleukin-10-secreting type 1 regulatory T cells in
rodents and humans. Immunol. Rev. 212:28–50.
42. Rouse, B. T., P. P. Sarangi, and S. Suvas. 2006. Regulatory T cells in virus
infections. Immunol. Rev. 212:272–286.
43. Spencer, S. E., I. E. Valentin-Bon, K. Whaley, and A. E. Jerse. 2004. Inhi-
bition of Neisseria gonorrhoeae genital tract infection by leading-candidate
topical microbicides in a mouse model. J. Infect. Dis. 189:410–419.
44. Stagg, A. J., M. Tuffrey, C. Woods, E. Wunderink, and S. C. Knight. 1998.
Protection against ascending infection of the genital tract by Chlamydia
trachomatis is associated with recruitment of major histocompatibility com-
plex class II antigen-presenting cells into uterine tissue. Infect. Immun.
66:3535–3544.
45. Suvas, S., and B. T. Rouse. 2006. Treg control of antimicrobial T cell
responses. Curr. Opin. Immunol. 18:344–348.
46. Taylor, A., J. Verhagen, K. Blaser, M. Akdis, and C. A. Akdis. 2006. Mech-
anisms of immune suppression by interleukin-10 and transforming growth
factor-beta: the role of T regulatory cells. Immunology 117:433–442.
47. Taylor, M. D., A. Harris, S. A. Babayan, O. Bain, A. Culshaw, J. E. Allen, and
R. M. Maizels. 2007. CTLA-4 and CD4CD25regulatory T cells inhibit
protective immunity to filarial parasites in vivo. J. Immunol. 179:4626–4634.
48. Tjia, K. F., J. P. van Putten, E. Pels, and H. C. Zanen. 1988. The interaction
between Neisseria gonorrhoeae and the human cornea in organ culture. An
electron microscopic study. Graefes Arch. Clin. Exp. Ophthalmol. 226:341–
345.
49. van der Woude, M. W., and A. J. Baumler. 2004. Phase and antigenic
variation in bacteria. Clin. Microbiol. Rev. 17:581–611.
50. von Boehmer, H. 2005. Mechanisms of suppression by suppressor T cells.
Nat. Immunol. 6:338–344.
51. Wada, K., S. Nomura, E. Morii, Y. Kitamura, Y. Nishizawa, A. Miyake, and
N. Terada. 1996. Changes in levels of mRNAs of transforming growth factor
(TGF)-beta1, -beta2, -beta3, TGF-beta type II receptor and sulfated glyco-
protein-2 during apoptosis of mouse uterine epithelium. J. Steroid Biochem.
Mol. Biol. 59:367–375.
52. Wan, Y. Y., and R. A. Flavell. 2006. The roles for cytokines in the generation
and maintenance of regulatory T cells. Immunol. Rev. 212:114–130.
53. Wilson, M. S., M. D. Taylor, A. Balic, C. A. Finney, J. R. Lamb, and R. M.
Maizels. 2005. Suppression of allergic airway inflammation by helminth-
induced regulatory T cells. J. Exp. Med. 202:1199–1212.
54. Yamagiwa, S., J. D. Gray, S. Hashimoto, and D. A. Horwitz. 2001. A role for
TGF-beta in the generation and expansion of CD4CD25regulatory T
cells from human peripheral blood. J. Immunol. 166:7282–7289.
55. Zheng, H. 1997. Analysis of the antigen-antibody specificity in the semen of
patients with Neisseria gonorrhoeae. Chin. Med. Sci. J. 12:47–49.
Editor: B. A. McCormick
VOL. 76, 2008 T-CELL IMMUNE RESPONSE AGAINST GONOCOCCUS 5465
... HIV uninfected women were enrolled from the Sex Workers Outreach Program (SWOP) clinic in Nairobi (n=64, Table 1). The median age was 28 years (IQR: [25][26][27][28][29][30], and median time in sex work was 3 years (IQR: 2-6). Most (86%) reported sex work as their primary mode of income, and nearly three-quarters (72%) had completed secondary or tertiary education. ...
... Similar to previous work, we find Treg are proportionally increased in the endocervix compared to the blood, by approximately two-fold (27). Interestingly, with respect to pathogens, a balance is required between Tregs that protect against excess inflammation, facilitating protective immunity [as is the case with HSV-2 (28,29)] versus those that can be co-opted to limit host immunity as a persistence strategy, as has been suggested for gonorrhea (30). In the HIV acquisition context, the finding that women whose CD4+ T cell populations contained proportionally higher Treg had lower concentrations of inflammatory cytokines and HIV target cells suggests that Tregs may play a protective role by limiting HIV's ability to cross the mucosal barrier and establish local infection, both thought to be critical events in transmission success. ...
Article
Full-text available
Regulatory T cells (Tregs) play important roles in tissue homeostasis, but few studies have investigated tissue Tregs in the context of genital inflammation, HIV target cell density, and vaginal microbiota in humans. In women from Nairobi (n=64), the proportion of CD4+ CD25+ CD127low Tregs in the endocervix correlated with those in blood (r=0.31, p=0.01), with a higher Treg frequency observed in the endocervix (median 3.8 vs 2.0%, p<0.0001). Most Tregs expressed FOXP3 in both compartments, and CTLA-4 expression was higher on endocervical Tregs compared to blood (median 50.8 vs 6.0%, p<0.0001). More than half (34/62, 55%) of participants displayed a non-Lactobacillus dominant vaginal microbiota, which was not associated with endocervical Tregs or CD4+ T cell abundance. In a multivariable linear regression, endocervical Treg proportions were inversely associated with the number of elevated pro-inflammatory cytokines (p=0.03). Inverse Treg associations were also observed for specific cytokines including IL-1β, G-CSF, Eotaxin, IL-1RA, IL-8, and MIP-1 β. Higher endocervical Treg proportions were associated with lower abundance of endocervical CD4+ T cells (0.30 log10 CD4+ T cells per log10 Treg, p=0.00028), with a similar trend for Th17 cells (p=0.09). Selectively increasing endocervical Tregs may represent a pathway to reduce genital tract inflammation in women.
... HIV uninfected women were enrolled from the Sex Workers Outreach Program (SWOP) clinic in Nairobi (n=64, Table 1). The median age was 28 years (IQR: [25][26][27][28][29][30], and median time in sex work was 3 years (IQR: 2-6). Most (86%) reported sex work as their primary mode of income, and nearly threequarters (72%) had completed secondary or tertiary education. ...
... Similar to previous work, we find Treg are proportionally increased in the endocervix compared to the blood, by approximately two-fold 27 . Interestingly, with respect to pathogens, a balance is required between Tregs that protect against excess inflammation, facilitating protective immunity (as is the case with HSV-2, 28,29 ) versus those that can be co-opted to limit host immunity as a persistence strategy, as has been suggested for gonorrhea 30 . In the HIV acquisition context, the finding that women whose CD4+ T cell populations contained proportionally higher Treg had lower concentrations of inflammatory cytokines and HIV target cells suggests that Tregs may play a protective role by limiting HIV's ability to cross the mucosal barrier and establish local infection, . ...
Preprint
Full-text available
Regulatory T cells (Tregs) play important roles in tissue homeostasis, but few studies have investigated tissue Tregs in the context of genital inflammation, HIV target cell density, and vaginal microbiota in humans. In women from Nairobi (n=64), the proportion of CD4+ CD25+ CD127low Tregs in the endocervix correlated with those in blood (r=0.31, p=0.01), with a higher Treg frequency observed in the endocervix (median 3.8 vs 2.0%, p<0.0001). Most Tregs expressed FoxP3 in both compartments, and CTLA-4 expression was higher on endocervical Tregs compared to blood (median 50.8 vs 6.0%, p<0.0001). More than half (34/62, 55%) of participants displayed a non-Lactobacillus dominant vaginal microbiota, which was not associated with endocervical Tregs or CD4+ T cell abundance. In a multivariable linear regression, endocervical Treg proportions were inversely associated with the number of elevated pro-inflammatory cytokines (p=0.03). Inverse Treg associations were also observed for specific cytokines including IL-1β, G-CSF, Eotaxin, IL-1RA, IL-8, and MIP-1 β. Higher endocervical Treg proportions were associated with lower abundance of endocervical CD4+ T cells (0.30 log10 CD4+ T cells per log10 Treg, p=0.00028), with a similar trend for Th17 cells (p=0.09). Selectively increasing endocervical Tregs may represent a pathway to reduce genital tract inflammation in women.
... However, T regs can also have an unfavorable role if they dampen the immune response against sexually transmitted infections such as human immunodeficiency virus (HIV), human papilloma virus (HPV) or chlamydia. In a mouse model of intravaginal N. gonorrhea infection TGF-b + T regs were induced in cervix-draining lymph nodes, thus evading the immune response and enabling pathogen survival (324). The occurrence of T regs in the mucosal tissue is described for several pathogens and conditions, while the mechanisms of their induction still need to be elucidated. ...
Article
Full-text available
Priming of T cells by antigen presenting cells (APCs) is essential for T cell fate decisions, enabling T cells to migrate to specific tissues to exert their effector functions. Previously, these interactions were mainly explored using blood-derived cells or animal models. With great advances in single cell RNA-sequencing techniques enabling analysis of tissue-derived cells, it has become clear that subsets of APCs are responsible for priming and modulating heterogeneous T cell effector responses in different tissues. This composition of APCs and T cells in tissues is essential for maintaining homeostasis and is known to be skewed in infection and inflammation, leading to pathological T cell responses. This review highlights the commonalities and differences of T cell priming and subsequent effector function in multiple barrier tissues such as the skin, intestine and female reproductive tract. Further, we provide an overview of how this process is altered during tissue-specific infections which are known to cause chronic inflammation and how this knowledge could be harnessed to modify T cell responses in barrier tissue.
... Important work using human cervical tissue explants demonstrated that N.g colonizes and invades the ectocervical, transformation zone and endocervical region of the female cervix [22], which contain high concentrations of macrophages [23]. Also, macrophages are recruited to the genital tissues [24] and infiltrate the uterine mucosa [25] in murine models of gonorrhea. The mechanism of macrophage colonization by gonococci as well as the identity and subcellular localization of the gonococci-occupied niche remain important open questions. ...
Article
Full-text available
Dynamic reorganization of the actin cytoskeleton dictates plasma membrane morphogenesis and is frequently subverted by bacterial pathogens for entry and colonization of host cells. The human-adapted bacterial pathogen Neisseria gonorrhoeae can colonize and replicate when cultured with human macrophages, however the basic understanding of how this process occurs is incomplete. N . gonorrhoeae is the etiological agent of the sexually transmitted disease gonorrhea and tissue resident macrophages are present in the urogenital mucosa, which is colonized by the bacteria. We uncovered that when gonococci colonize macrophages, they can establish an intracellular or a cell surface-associated niche that support bacterial replication independently. Unlike other intracellular bacterial pathogens, which enter host cells as single bacterium, establish an intracellular niche and then replicate, gonococci invade human macrophages as a colony. Individual diplococci are rapidly phagocytosed by macrophages and transported to lysosomes for degradation. However, we found that surface-associated gonococcal colonies of various sizes can invade macrophages by triggering actin skeleton rearrangement resulting in plasma membrane invaginations that slowly engulf the colony. The resulting intracellular membrane-bound organelle supports robust bacterial replication. The gonococci-occupied vacuoles evaded fusion with the endosomal compartment and were enveloped by a network of actin filaments. We demonstrate that gonococcal colonies invade macrophages via a process mechanistically distinct from phagocytosis that is regulated by the actin nucleating factor FMNL3 and is independent of the Arp2/3 complex. Our work provides insights into the gonococci life-cycle in association with human macrophages and defines key host determinants for macrophage colonization.
... The utility of this lower genital tract infection model to study ascending infection is unclear because the original study had only 2 mice with recoverable bacteria in the UGT [34]. Another study detected uterine bacteria in all infected mice, although this protocol described a large volume injected against the cervix, which may reflect a direct uterine inoculation [35]. However, no other model has been described to model bacterial ascent, and it remains to be seen whether transgenic mice expressing human factors that facilitate N. gonorrhoeae infection show increased ascent of N. gonorrhoeae into the UGT after their introduction into the vagina. ...
Article
While infection by Neisseria gonorrhoeae is often asymptomatic in women, undetected infections can ascend into the upper genital tract to elicit an inflammatory response that manifests as pelvic inflammatory disease, with the outcomes depending on the intensity and duration of inflammation and whether it is localized to the endometrial, fallopian tube, ovarian, and/or other tissues. This review examines the contribution of N. gonorrhoeae versus other potential causes of pelvic inflammatory disease by considering new insights gained through molecular, immunological, and microbiome-based analyses, and the current epidemiological burden of infection, with an aim to highlighting key areas for future study.
Article
Full-text available
Neisseria gonorrhoeae (Ng) is a uniquely adapted human pathogen and the etiological agent of gonorrhea, a sexually transmitted disease. Ng has developed numerous mechanisms to avoid and actively suppress innate and adaptive immune responses. Ng successfully colonizes and establishes topologically distinct colonies in human macrophages and avoids phagocytic killing. During colonization, Ng manipulates the actin cytoskeleton to invade and create an intracellular niche supportive of bacterial replication. The cellular reservoir(s) supporting bacterial replication and persistence in gonorrhea infections are poorly defined. The manner in which gonococci colonize macrophages points to this innate immune phagocyte as a strong candidate for a cellular niche during natural infection. Here we investigate whether nutrients availability and immunological polarization alter macrophage colonization by Ng. Differentiation of macrophages in pro-inflammatory (M1-like) and tolerogenic (M2-like) phenotypes prior to infection reveals that Ng can invade macrophages in all activation states, albeit with lower efficiency in M1-like macrophages. These results suggest that during natural infection, bacteria could invade and grow within macrophages regardless of the nutrients availability and the macrophage immune activation status.
Preprint
Full-text available
Neisseria gonorrhoeae (Ng) is a uniquely adapted human pathogen and the etiological agent of gonorrhea, a sexually transmitted disease. Ng has developed numerous mechanisms to avoid and actively suppress innate and adaptive immune responses. Ng successfully colonizes and establishes topologically distinct colonies in human macrophages and avoids phagocytic killing. During colonization, Ng manipulates the actin cytoskeleton to invade and create an intracellular niche supportive of bacterial replication. The cellular reservoir(s) supporting bacterial replication and persistence in gonorrhea infections are poorly defined. The manner in which gonococci colonize macrophages points to this innate immune phagocyte as a strong candidate for a cellular niche during natural infection. Here we investigate whether nutrients availability and immunological polarization alter macrophage colonization by Ng . Differentiation of macrophages in pro-inflammatory (M1-like) and tolerogenic (M2-like) phenotypes prior to infection reveals that Ng can invade macrophages in all activation states, albeit with lower efficiency in M1-like macrophages. These results suggest that during natural infection, bacteria could invade and grow within macrophages regardless of the nutrients availability and the macrophage immune activation status.
Article
BACKGROUND The endometrium is a highly dynamic tissue that undergoes dramatic proliferation and differentiation monthly in order to prepare the uterus for implantation and pregnancy. Intrauterine infection and inflammation are being increasingly recognized as potential causes of implantation failure and miscarriage, as well as obstetric complications later in gestation. However, the mechanisms by which the cells of the endometrium respond to infection remain understudied and recent progress is slowed in part owing to similar overlapping studies being performed in different species. OBJECTIVE AND RATIONALE The aim of this scoping review is to systematically summarize all published studies in humans and laboratory animals that have investigated the innate immune sensing and response of the endometrium to bacteria and viruses, and the signaling mechanisms involved. This will enable gaps in our knowledge to be identified to inform future studies. SEARCH METHODS The Cochrane Library, Ovid Embase/Medline, PubMed, Scopus, Google Scholar, and Web of Science databases were searched using a combination of controlled and free text terms for uterus/endometrium, infections, and fertility to March 2022. All primary research papers that have reported on endometrial responses to bacterial and viral infections in the context of reproduction were included. To focus the scope of the current review, studies in domesticated animals, included bovine, porcine, caprine, feline, and canine species were excluded. OUTCOMES This search identified 42 728 studies for screening and 766 full-text studies were assessed for eligibility. Data was extracted from 76 studies. The majority of studies focused on endometrial responses to Escherichia coli and Chlamydia trachomatis, with some studies of Neisseria gonorrhea, Staphylococcus aureus, and the Streptococcus family. Endometrial responses have only been studied in response to three groups of viruses thus far: HIV, Zika virus, and the herpesvirus family. For most infections, both cellular and animal models have been utilized in vitro and in vivo, focusing on endometrial production of cytokines, chemokines, and antiviral/antimicrobial factors, and the expression of innate immune signaling pathway mediators after infection. This review has identified gaps for future research in the field as well as highlighted some recent developments in organoid systems and immune cell co-cultures that offer new avenues for studying endometrial responses to infection in more physiologically relevant models that could accelerate future findings in this area. WIDER IMPLICATIONS This scoping review provides an overarching summary and benchmark of the current state of research on endometrial innate immune responses to bacterial and viral infection. This review also highlights some exciting recent developments that enable future studies to be designed to deepen our understanding of the mechanisms utilized by the endometrium to respond to infection and their downstream effects on uterine function.
Article
Full-text available
Neisseria gonorrhoeae (also known as gonococcus) has been causing gonorrhoea in humans since ancient Egyptian times. Today, global gonorrhoea infections are rising at an alarming rate, in concert with an increasing number of antimicrobial-resistant strains. The gonococcus has concurrently evolved several intricate mechanisms that promote pathogenesis by evading both host immunity and defeating common therapeutic interventions. Central to these adaptations is the ability of the gonococcus to manipulate various host microenvironments upon infection. For example, the gonococcus can survive within neutrophils through direct regulation of both the oxidative burst response and maturation of the phagosome; a concerning trait given the important role neutrophils have in defending against invading pathogens. Hence, a detailed understanding of how N. gonorrhoeae exploits the human host to establish and maintain infection is crucial for combating this pathogen. This review summarizes the mechanisms behind host manipulation, with a central focus on the exploitation of host epithelial cell signaling to promote colonization and invasion of the epithelial lining, the modulation of the host immune response to evade both innate and adaptive defenses, and the manipulation of host cell death pathways to both assist colonization and combat antimicrobial activities of innate immune cells. Collectively, these pathways act in concert to enable N. gonorrhoeae to colonize and invade a wide array of host tissues, both establishing and disseminating gonococcal infection.
Article
Microbial infections are a threat to women's reproductive health. Although reproductive cycles and pregnancy are controlled by sex hormones, the impact of hormones on host-pathogen interactions and immune function in the female reproductive tract are understudied. Furthermore, the changing endocrine environment throughout pregnancy may influence how and when women are susceptible to ascending infection. Because most intrauterine microbial infections originate in the lower reproductive tract, it is vital that future studies determine how different hormonal conditions influence the lower reproductive tract's susceptibility to infection to understand temporal components of infection susceptibilities across pregnancy. These studies should also extend to nonpregnant women, as it is critical to establish how hormonal fluctuations across the menstrual cycle and hormonal contraceptives may influence disease susceptibility. This review summarizes current knowledge of how estrogen and progesterone impact vaginal and cervical mucosal immunity, barrier function, and interactions with microbial communities.
Article
Explants of human corneas in organ culture were used to study the interaction betweenNeisseria gonorrhoeae and human corneal epithelium at an ultrastructural level. Scanning electron microscopy revealed that infection of the corneal explants withN. gonorrhoeae resulted in a rapid adherence of the bacteria to the cell surface. This attachment was probably mediated by pili since only piliated strains were able to adhere to the cells. Upon attachment the bacteria appeared to become engulfed by the epithelial cells. Transmission electron microscopy revealed gonococci apparently lying within vacuoles inside the cells within 1 h after inoculation of the bacteria. At prolonged infection (8–24 h), the thickness of the epithelium was found to be considerably reduced. This thinning of the cornea was probably caused by a continuous desquamation of infected cells. Taken together, the present data demonstrate thatNeisseria gonorrhoeae is able to adhere and penetrate into intact corneal epithelium and furthermore indicate that human cornea explants in organ culture are a useful model in studies of bacterial-epithelial cell interaction.
Article
The objective of this study was to examine the susceptibility of rat uterine epithelial cells (UEC) to infection with Chlamydia trachomatis and to study the epithelial-stromal interactions following infection. UEC were isolated from adult rats and grown in culture. Polarized, confluent monolayers of UEC were infected with 10(6) IFU/well C. trachomatis (MoPn). In order to confirm infection, MoPn was labeled with a fluorescent tracking dye, PKH-26, and then used in epithelial cell infections. Transepithelial resistances were measured prior to and following infection to test the effect of Chlamydia on the integrity of the epithelial monolayers. In other experiments, polarized epithelial cultures were infected in the presence and absence of stromal cells. Media was collected from the apical and basolateral compartments of the cultures before and after infection and analyzed for cytokines IL-1alpha and TNF-alpha. Epithelial cell cultures infected with PKH-26 labeled MoPn were examined 4-5 days later. Bacterial inclusions were detected inside epithelial cells indicating infection had occurred. Co-localization of PKH-26 labeled bacteria with FITC-labelled anti-Chlamydia antibody on the epithelial cells confirmed infection. No changes were found in resistance across the monolayers of epithelial cells in the presence or absence of infection. ELISA results indicate that UEC secrete IL-1alpha constitutively in citro. Stromal cells secrete very little IL-1alpha. When stromal cells were co-incubated with epithelial cells there was a decrease in the amount of IL-1alpha secreted by epithelial cells 48 hr post-infection. On the other hand, maximum TNF-alpha was found in stromal cells. both with and without infection. Epithelial cells, in these studies made very little TNF-alpha. These results show that primary rat epithelial cells can be infected with Chlamydia in vitro. Epithelial and stromal cells from uteri of adult rats make IL-1alpha and TNF-alpha in vitro both prior to and following infection with Chlamydia. This system can be used to analyze the role played by epithelial-stromal interactions in providing protection on this mucosal surface.
Article
Abstract The core oligosaccharides of low-molecular-weight lipopolysaccharide (LPS), also termed lipooligosaccharide (LOS), of pathogenic Neisseria spp. mimic the carbohydrate moieties of glycosphingolipids present on human cells. Such mimicry may serve to camouflage the bacterial surface from the host. The LOS component is antigenically and/or chemically identical to lactoneoseries glycosphingolipids and can become sialylated in Neisseria gonorrhoeae when the bacterium is grown in the presence of cytidine 5′-monophospho-N-acetylneuraminic acid, the nucleotide sugar of sialic acid. Strains of Neisseria meningitidis and Haemophilus influenzae also express similarly sialylated LPS. Sialylation of the LOS influences susceptibility to bactericidal antibody, may decrease or prevent phagocytosis, cause down-regulation of complement activation, and decrease adherence to neutrophils and the subsequent oxidative burst response. The core oligosaccharides of LPS of Campylobacter jejuni serotypes which are associated with the development of the neurological disorder, Guillain-Barré syndrome (GBS), exhibit mimicry of gangliosides. Cross-reactive antibodies between C. jejuni LPS and gangliosides are considered to play an important role in GBS pathogenesis. In contrast, the O-chain of a number of Helicobacter pylori strains exhibit mimicry of Lewisx and Lewisy blood group antigens. The role of this mimicry remains to be investigated, but may play a role in bacterial camouflage, the induction of autoimmunity and immune suppression in H. pylori-associated disease.
Article
Local immunization in the vagina of several species elicits immune responses, but little is known about the uptake, processing and recognition of antigens at this site. We investigated the uptake of intravaginally administered tracers using FITC-bovine albumin, FITC-horse ferritin and FITC-horseradish peroxidase in non-pregnant and pregnant mice. Tracers were detected in cells in the vaginal epithelium and stroma at diestrus, proestrus and metestrus, but not at estrus. During pregnancy, racers were present in vaginal cells on Day 6 but not on Day 13. The distribution of tracers in the vagina was the same in all mice. They were present in vaginal epithelium in cells similar to Langerhans' cells and in the stroma in cells that resembled dendritic cells, fibroblasts or macrophages. In some non-pregnant mice, tracers were present in cells adjacent to lymphatic nodules located in the adventitia between the vagina and urethra. Tracers were seen in phagocytic cells lining the marginal and medullary sinuses of the draining lymph nodes (iliac nodes) in some non-pregnant mice at 4 h after intravaginal administration, or in small, dendritic cells in the paracortex at 17 h. To test the possibility that transfer of proteins into the vagina was due to toxic effects of the tracers, FITC-conjugated proteins were also administered into the lumen of uterine horns, and their distribution in horns, cervix and vagina was studied. In uterine horns, tracers were either absent or were located only in apical vesicles in the luminal epithelium. Tracers were present in the cervix and vagina as described above for intravaginal tracers. This result suggests that uptake of tracers in the vagina was not due to toxic effects, and that the vagina and cervix are major sites of protein uptake into the reproductive tract.
Article
Gonorrhea may be the most extensively studied infection of the past 20 years. The gonorrhea epidemic in the United States began in the early 1960s and peaked in 1975. Ironically, since 1976 the declining overall incidence has been offset by the advent of plasmid-mediated beta-lactamase production by Neisseria gonorrhoeae and by a growing problem with outbreaks due to strains with chromosomally mediated penicillin and tetracycline resistance. This new antimicrobial resistance, coupled with the frequency of concurrent chlamydial infection in developed countries and concurrent syphilis in some developing countries, has created a need for new approaches to gonorrhea therapy. With the introduction of certain new antimicrobial agents, highly effective forms of therapy are again available. New approaches to rapid diagnosis are also becoming available, but require critical appraisal. Unfortunately, in most of the world's population, gonorrhea remains epidemic, diagnosis of gonorrhea in women is extremely difficult, and highly effective antimicrobial agents are no longer affordable. Thus, vaccine development remains an extremely important goal. Although no candidate gonococcal vaccine currently holds high promise, the increasing understanding of the biology of the gonococcus and the pathogenesis of gonorrhea will serve to focus future research on vaccine development.
Article
Attachment of different gonococcal colony phenotypes to explants of human genital tract epithelium was studied by means of scanning electron microscopy and radioisotope-labeled gonococci. Heavily piliated organisms attached in greater numbers than nonpiliated organisms. Gonococci from transparent colony phenotypes attached in higher numbers than gonococci from opaque phenotypes to all tissues studied. Transitional cells from cervical tissues showing a gradual squamocolumnar transition demonstrated more gonococci attached per surface area than either endocervical or fallopian tube epithelium. Squamous epithelium showed the fewest number of attached gonococci. In all tissues, the attachment of the gonococcus was to the tips and surfaces of the microvilli. Gonococcal colony phenotypes as well as the length and location of the cervical transition zone may influence the progression of cervical gonococcal infection to invasive disease.
Article
The use of isoform-specific transforming growth factor-beta (TGF beta) primers, 35S-labeled 40-mer oligonucleotide probes and polyclonal antibodies, reverse transcription-polymerase chain reaction, in situ hybridization, and immunohistochemical observations has revealed that human uterine tissue at various reproductive stages expresses TGF beta s and TGF beta type II receptor messenger RNAs (mRNAs) and proteins. The reverse transcription-polymerase chain reaction revealed the predicted 443-, 310-, 524-, and 431-basepair fragments for TGF beta 1, TGF beta 2, TGF beta 3, and TGF beta type II receptor, respectively, in both endometrial and myometrial tissues, which were further verified by restriction enzyme analysis. In situ hybridization and immunohistochemical observations indicated that all uterine cell types express TGF beta s mRNAs and proteins. In the functionalis region, endometrial luminal and glandular epithelial cells are the primary cell types expressing TGF beta s mRNAs and proteins, with lesser expression in stromal cells, whereas in the basalis region, they are equally expressed in both cell types. In myometrium, TGF beta mRNA and protein expression in smooth muscle cells occurs at a substantially lower level than in endometrial tissue. In endometrial tissue, the highest level of TGF beta mRNA and protein expression appeared in the late proliferative and early to midsecretory phases of the menstrual cycle, with a considerable reduction during the late secretory and postmenopausal periods. The pattern and cellular distribution of TGF beta type II receptor protein were similar to those seen with TGF beta isoforms in both endometrial and myometrial tissues. Quantitative autoradiography (net grain density per 100 microns 2) of specific binding of [125I]TGF beta 1 for different uterine cell types indicated that the stromal cells contain a higher grain density than other uterine cell types (P < 0.05), without a significantly different density in the proliferative, compared with the secretory, phase of the menstrual cycle. These data suggest that TGF beta s acting through their specific receptors may play an important role in a variety of uterine functions in an autocrine/paracrine manner, and ovarian steroids may also regulate their expression in endometrial tissue.