Enhancement of Adaptive Immunity to Neisseria Gonorrhoeae

Embed Size (px)

Citation preview

  • 8/12/2019 Enhancement of Adaptive Immunity to Neisseria Gonorrhoeae

    1/9

    M A J O R A R T I C L E

    Enhancement of Adaptive Immunity to

    Neisseria gonorrhoeaeby Local IntravaginalAdministration of MicroencapsulatedInterleukin 12

    Yingru Liu, Nejat K. Egilmez, and Michael W. Russell

    Department of Microbiology and Immunology, Witebsky Center for Microbial Pathogenesis and Immunology, University at Buffalo, New York

    Gonorrhea remains one of the most frequent infectious diseases, andNeisseria gonorrhoeaeis emerging as re-

    sistant to most available antibiotics, yet it does not induce a state of specic protective immunity against rein-

    fection. Our recent studies have demonstrated that N. gonorrhoeaeproactively suppresses host T-helper (Th) 1/

    Th2-mediated adaptive immune responses, which can be manipulated to generate protective immunity. Herewe show that intravaginally administered interleukin 12 (IL-12) encapsulated in sustained-release polymer mi-

    crospheres signicantly enhanced both Th1 and humoral immune responses in a mouse model of genital gono-

    coccal infection. Treatment of mice with IL-12 microspheres during gonococcal challenge led to faster

    clearance of infection and induced resistance to reinfection, with the generation of gonococcus-specic circulat-

    ing immunoglobulin G and vaginal immunoglobulin A and G antibodies. These results suggest that local ad-

    ministration of microencapsulated IL-12 can serve as a novel therapeutic and prophylactic strategy against

    gonorrhea, with implications for the development of an effective vaccine.

    Keywords. Neisseria gonorrhoeae; immunity; cytokine therapy; IL-12; microencapsulation.

    Infection with gonorrhea does not induce a state ofspecic protective immunity and as a result it can be

    acquired repeatedly [1, 2]. The World Health Organiza-

    tion estimates that >100 million new gonococcal infec-

    tions occur worldwide every year [3]. No vaccine is

    available against gonorrhea, and therapy depends upon

    antibiotics [4]. However, antibiotic resistance ofN. gon-

    orrhoeae continues to emerge and is becoming prob-

    lematic for effective treatment [5].

    The mechanisms underlying the paucity of protec-

    tive immune responses against Neisseria gonorrhoeae

    infection remain elusive. It is generally believed that theextensive and rapid antigenic variability of the gono-

    coccus and its capacity to resist complement-mediated

    bacteriolysis result in immune evasion [1, 68].However,

    our recent studies using experimental murine infections

    have demonstrated thatN. gonorrhoeae proactively sup-

    presses host T-helper (Th) 1and Th2-mediated specic

    immune responses [912]. Concomitantly, gonococcal

    infection elicits Th17 responses, which recruit innate

    defense mechanisms including the characteristic neu-

    trophil inux, and interference with interleukin 17 (IL-

    17) signaling prolongs the infection [13]. Induction of

    transforming growth factor (TGF) and interleukin 10

    (IL-10) is critically involved in the suppression of adap-

    tive immunity by N. gonorrhoeae, and systemic treat-

    ment with antiTGF- or/and antiIL-10 antibodies

    reverses the natural capacity of N. gonorrhoeae to

    inhibit Th1 and Th2 responses, allowing the develop-

    ment of protective immune responses that not only ac-

    celerate clearance of an existing infection but also

    generate resistance to future infection [1012].

    Received 29 January 2013; accepted 21 May 2013; electronically published 18

    September 2013.

    Presented in part: 18th International Pathogenic Neisseria Conference, Wrz-

    burg, Germany, 914 September 2012.

    Correspondence: Yingru Liu, MD, PhD, Department of Microbiology and Immu-

    nology, Farber 138, University at Buffalo, 3435 Main St, Buffalo, NY 14214

    ([email protected]; [email protected]).

    The Journal of Infectious Diseases 2013;208:18219

    The Author 2013. Published by Oxford University Press on behalf of the Infectious

    Diseases Society of America. All rights reserved. For Permissions, please e-mail:

    [email protected].

    DOI: 10.1093/infdis/jit354

    Immunity toN. gonorrhoeae JID 2013:208 (1 December) 1821

    mailto:[email protected]:[email protected]:[email protected]:[email protected]:[email protected]:[email protected]
  • 8/12/2019 Enhancement of Adaptive Immunity to Neisseria Gonorrhoeae

    2/9

    Interleukin 12 (IL-12) is a potent proinammatory cytokine

    that strongly stimulates Th1-associated cellular immunity [14].

    Some studies have shown that IL-12 also activates humoral im-

    munity through both T-celldependent and T-cellindependent

    mechanisms, and enhances immunoglobulin A (IgA) and im-

    munoglobulin G (IgG) antibody responses to foreign proteins

    and pathogens [1517]. It has therefore been exploited as an an-

    titumor therapeutic as well as a vaccine adjuvant for protection

    against bacterial and viral infectious diseases [1821]. However,therapeutic use of soluble IL-12 requires repeated parenteral ad-

    ministration in large doses resulting in systemic toxicity and

    limited efcacy [22,23]. To avoid this problem, we have used

    biodegradable polymeric microparticles for the local and sus-

    tained delivery of cytokines directly to the cancer microenviron-

    ment [24]. Others have demonstrated that local administration

    of similar formulations leads to effective particle uptake and

    release of encapsulated therapeutics in the mucosa [25, 26].

    In the current study, we have assessed the effect of local in-

    travaginal treatment with microencapsulated IL-12 in a murine

    gonococcal infection model and found that IL-12 microspheres

    dramatically accelerated the clearance of infection. The treat-

    ment was associated with signicant enhancement of Th1 cel-

    lular and specic antibody responses, and it induced protective

    immunity against reinfection. We have further found similar

    therapeutic effects when mice were treated with vaginal instilla-

    tion of low doses of antiTGF- or antiIL-10 antibodies en-

    capsulated in sustained-release microspheres.

    MATERIALSANDMETHODS

    Mice

    BALB/c mice were purchased from Jackson Laboratories and

    were maintained under standard conditions in the Laboratory

    Animal Facility at the University at Buffalo. All animal use pro-

    tocols were approved by the Institutional Animal Care and Use

    Committee of the University at Buffalo.

    Bacteria

    Neisseria gonorrhoeae FA1090 was kindly provided by Janne

    Cannon, PhD, (University of North Carolina at Chapel Hill)

    and was cultured on GC agar supplemented with hemoglobin

    and Isovitalex (BD Diagnostic Systems). Growth was checked

    for colony morphology consistent with Opa protein and pilusexpression, and gonococci were harvested from plates and the

    cell density was determined as detailed elsewhere [11]. Opa ex-

    pression, as determined previously [11], was Opa A, B/D/G,

    and E/K.

    Microspheres

    Cytokines and antibodies were encapsulated into polylactic

    acid (PLA) microspheres using the phase inversion nanoencap-

    sulation technology as described elsewhere, except that bovine

    serum albumin was replaced by sucrose (0.1%, wt/wt) [27]. Five

    formulations were produced: (1) control microspheres containing

    no cytokine or antibody, (2) murine IL-12 (0.25-g/mg parti-

    cles), (3) murine IL-17 (0.25-g/mg particles), (4) murine anti-

    mouse TGF- (10-g/mg particles), and (5) rat anti-mouse

    IL-10 (10-g/mg particles).

    Mouse Vaginal Infection Model

    Female mice between 7 and 9 weeks old were infected vaginallyon day 0 with live N. gonorrhoeae FA1090, as described else-

    where [10, 28]. Vaginal mucus was quantitatively cultured daily

    on GC agar supplemented with selective antibiotics to deter-

    mine the bacterial colonization loads. The limit of detection

    was 100 colony-forming units recovered per mouse. Intravagi-

    nal treatments with microsphere preparations were given every

    second day from day 0 to day 8, by instillation of 40-L suspen-

    sions in phosphate-buffered saline of microspheres containing

    IL-12, IL-17, antiTGF-, antiIL-10, or control microspheres.

    Cell Isolation and Flow Cytometry

    Mice were killed, and their iliac lymph nodes (ILNs) and

    genital tracts were excised aseptically. The ILNs were teased in

    Hanksbuffered salt solution to release cells. Vaginal single-cell

    suspensions were prepared by enzymatic digestion, as described

    elsewhere [10, 29]. Isolated cells were washed with staining

    buffer twice, then incubated with the indicated antibodies for

    30 minutes on ice, washed twice, and analyzed on a FACS

    Calibur cytometer. For determination of intracellular cytokine

    expression, cells were restimulated with phorbol myristate

    acetateionomycinGolgiStop (eBioscience) for 5 hours, and

    then xed with Cytox/Cytoperm (eBioscience). Antibodies to

    mouse CD4, CD8, CD19, CD11b, CD11c, NKG2D, Gr-1, inter-feron (IFN), interleukin 4 (IL-4), and IL-17A conjugated with

    uorescein isothiocyanate, phycoerythrin, or allophycocyanin

    were purchased from eBioscience.

    Cytokine Enzyme-Linked Immunosorbent Assay

    Levels of IL-12p70, IFN-, IL-4, interleukin 5, and IL-17A in

    serum or vaginal wash samples were measured in triplicate

    using enzyme-linked immunosorbent assay (ELISA) kits pur-

    chased from eBioscience.

    Real-Time Reverse-Transcription Polymerase Chain ReactionTotal cellular RNA of whole vaginas harvested from the mice

    was isolated with RNeasy Mini Kits (Qiagen) and transcribed

    to complementary DNA (cDNA) using the iScript cDNA syn-

    thesis kit (Bio-Rad). Real-time reverse-transcription polymer-

    ase chain reaction (RT-PCR) was performed on an iCycler iQ

    detection system (Bio-Rad) using Sybergreen (Bio-Rad) for

    real-time monitoring of the PCR. The sequences of IFN-, IL-4,

    IL-17A, and -actin primers used were same as described else-

    where [12]. Relative quantication of target genes was analyzed

    1822 JID 2013:208 (1 December) Liu et al

  • 8/12/2019 Enhancement of Adaptive Immunity to Neisseria Gonorrhoeae

    3/9

    based on the threshold cycle determined by Bio-Rad iQ5

    optical system software.

    Assay of Serum and Mucosal Antibodies

    Saliva, vaginal wash, and serum samples were collected from

    individual mice on day 15 after inoculation. Gonococcus-

    specic IgA, IgG, and immunoglobulin M (IgM) in saliva,

    serum, and vaginal wash samples and total IgA, IgG, and IgM

    concentrations in secretions were assayed by ELISA, as detailedelsewhere [10].

    Statistical Analysis

    Data are expressed as means standard errors of the mean.

    Data on the effects of IL-12, IL-17, antiTGF-, and

    antiIL-10loaded vs blank microsphere treatments on vaginal

    N. gonorrhoeaeinfection were analyzed using repeated-measures

    analysis of variance with Bonferroni corrected post hoc testing of

    pairwise comparisons. Kaplan-Meier analysis with log-rank

    testing was also used to compare infection clearance. Data from

    in vitro experiments were analyzed with unpaired 2-tailedttests

    to compare the mean values between 2 selected groups. Differ-

    ences were considered statistically signicant atP< .05.

    RESULTS

    Intravaginal Administration of IL-12 Microspheres as

    Protection Against Genital TractN. gonorrhoeaeInfection in

    Mice

    To examine the therapeutic effect of IL-12loaded micro-

    spheres, groups of female BALB/c mice were infected with

    N. gonorrhoeae and the bacterial burden was monitored daily

    by vaginal swab culture. Preliminary dose-ranging experimentsshowed that intravaginal instillation of microspheres contain-

    ing 1.0 g of IL-12 every second day was sufcient to accelerate

    clearance of the infection relative to treatment with blank mi-

    crospheres; no further enhancement of clearance was obtained

    with 2.0 g of microencapsulated IL-12, and lower doses were

    progressively less effective (Figure1A).

    Untreated or blank microsphere-treated mice cleared the

    infection in approximately 15 days (Figure1Band1C). Intrava-

    ginal instillation of microencapsulated IL-12 at the optimal 1.0-

    g dose signicantly reduced the recoverable N. gonorrhoeae

    load starting from day 4, and these mice cleared the infectionby day 7, or 8 days earlier than blank microsphere-treated or

    untreated mice (Figure 1B and 1C). The infection did not

    relapse after treatment ceased on day 7. In contrast, intravaginal

    administration of free, soluble IL-12 was completely ineffective

    in enhancing clearance ofN. gonorrhoeae(Figure1Band1C).

    Our previous studies in the same murine model had demon-

    strated that N. gonorrhoeae mainly elicits Th17 responses,

    which are involved in the recruitment of host innate defense

    mechanisms, including the inux of neutrophils to the genital

    tract [13]. We therefore investigated whether local IL-17 treat-

    ment could also promote the clearance of gonococcal infection.

    Intravaginal administration of IL-17loaded microspheres at

    the optimal dose (1.0 g) also accelerated clearance ofN. gonor-

    rhoeae infection but to a lesser extent than IL-12 microspheres

    given on the same schedule (Figure1Band1C).

    Effects of IL-12 Microsphere Treatment on Th1 and Antibody

    Responses to Vaginal Gonococcal InfectionTo elucidate the mechanisms underlying the therapeutic effects

    of IL-12, we characterized the local immune responses to

    genital gonococcal infection in mice treated with IL-12loaded

    or blank microspheres. Single-cell suspensions were prepared

    from ILNs and vaginas of 7 mice per group at 3, 5, 7, and 14

    days after inoculation with N. gonorrhoeae or vehicle only for

    evaluation byow cytometry to detect intracellular IFN-, IL-4,

    and IL-17. Starting on day 3 after inoculation, IL-17+/CD4+

    T cells were observed in the local draining ILNs, with produc-

    tion peaking at day 5 and continuing for the duration of infec-

    tion. At day 5, approximately 22% of CD4+ T cells present in

    the ILNs of control-treated infected mice were IL-17+, whereas

    only approximately 3.5% were IFN-+ and few IL-4+/CD4+

    T cells were detected (Figure 1D). The IL-12microsphere treatment

    markedly enhanced Th1 immune responses toN. gonorrhoeae,

    indicated by signicantly increased numbers of IFN-+/CD4+

    T cells (Figure 1D). In contrast, IL-12 microspheres did not

    change Th2 or Th17 responses, because the numbers of IL-4+/

    CD4+ and IL-17+/CD4+ T cells in ILNs were similar between

    the treated groups (Figure 1D). The RT-PCR analyses showed

    that IFN- but not IL-4 or IL-17 messenger RNA expression

    was elevated in the vaginas of infected mice after IL-12 micro-

    sphere treatment (Figure 1E). Although IL-17 microspheresameliorated gonococcal infection, this treatment was not asso-

    ciated with enhanced Th1 or Th2 responses (Figure 1D and

    1E), but there was increased inux of Gr-1+ neutrophils into

    the genital tract (Figure1F).

    We also used ELISA to measure IL-12p70, IFN-, IL-4, and

    IL-17 concentrations in vaginal wash and serum samples col-

    lected 7 days after inoculation. We detected IL-12 (176.5 48.6

    pg/mL) in vaginal wash samples from infected mice treated

    with IL-12 microspheres. Low levels of IL-12 (41.7 10.7 pg/mL)

    were found in the serum samples from these mice, suggesting

    that the effects of IL-12 microsphere treatment on gonococcalinfection did not result primarily from the passage of the cyto-

    kine into the circulation. Consistent with the ow cytometric

    studies, IFN- was present in the vaginal wash (32.6 9.8 pg/

    mL) and serum (43.3 11.5 pg/mL) samples from infected

    mice treated with IL-12 microspheres, but IL-4 and IL-17 were

    not detected. None of these cytokines was detected in control-

    treated infected mice.

    Interleukin 12 can stimulate humoral immune responses in

    an IFN-dependent manner or directly [16]. We therefore

    Immunity toN. gonorrhoeae JID 2013:208 (1 December) 1823

  • 8/12/2019 Enhancement of Adaptive Immunity to Neisseria Gonorrhoeae

    4/9

    determined whether IL-12 microsphere treatment during N.

    gonorrhoeae infection led to the production of anti-gonococcal

    antibodies in vaginal wash, saliva, and serum samples collected

    15 days after inoculation. The IgM antibodies were at low levels

    with little difference between experimental groups (data not

    shown). No salivary gonococcus-specic antibody was detected in

    any group of mice (data not shown). As reported elsewhere [10],

    N. gonorrhoeae infection of control-treated mice did not

    Figure 1. Effect of intravaginal IL-12 microsphere (ms) treatment on primary gonococcal infection in BALB/c mice.A, interleukin 12 (IL-12) ms dose opti-mization experiment. Microspheres containing the stated doses of IL-12 were given on days 0, 2, 4, 6, and 8 (8 mice per group). The Neisseria gonorrhoeae

    burden was monitored daily by vaginal swab culture. Signicant differences in infection burdens were found between mice treated with 2.0 g (P< .01),

    1.0 g (P< .01), or 0.5 g (P< .05) of microencapsulated IL-12 and controls (analysis of variance). B, Time course of infection in mice treated with IL-12 ms,

    soluble IL-12, interleukin 17 (IL-17) ms, or control ms or in untreated mice (cytokine dose, 1.0 g given on days 1, 1, 3, 5, and 7; 8 mice per group). Signi-

    cant differences in infection burdens were found between mice treated with IL-12 ms (P< .01) or IL-17 ms (P< .02) and controls (analysis of variance). C,

    Data from the experiment shown in B, plotted as percentage of mice remaining infected with the indicated cytokine treatments. Infection was cleared sig-

    nicantly faster in mice treated with IL-12 ms (P< .001) or IL-17 ms (P< .001) than in controls (Kaplan-Meier analysis). D, Cytokine expression in isolated

    iliac lymph node cells from sham-infected or infected mice with IL-12 ms, IL-17 ms, or control ms treatment (7 mice per group). Expression of interferon

    (IFN), interleukin 4 (IL-4), and IL-17 in CD4+ T cells isolated at day 5 after infection was analyzed with ow cytometry.E, Reverse-transcription polymerase

    chain reaction (RT-PCR) analysis of IFN-, IL-4, and IL-17 messenger RNA (mRNA) levels in vaginal tissue harvested at day 3 from sham-infected or infected

    mice with IL-12 ms, IL-17 ms, or control ms treatment (7 mice per group). Cytokine gene expression levels detected with RT-PCR were normalized relative

    to expression of-actin and set at 1.0 for the sham-infected group. F, Phenotypic prole of vaginal cells isolated on day 5 from sham-infected or infected

    mice treated with IL-17 ms or control ms (7 mice per group). G, H, Vaginal (G) and (H) serum anti-gonococcal immunoglobulin A (IgA) and immunoglobulin

    G (IgG) antibody responses in sham-infected or infected mice with IL-12 ms, IL-17 ms, or control ms treatment (7 mice per group). Vaginal wash and serumsamples were collected 15 days after inoculation, and gonococcus-specic and total IgA and IgG were measured with enzyme-linked immunosorbent

    assay. Results from 1 of 3 independent experiments are shown. Data shown as means SEM. #P< .05; *P< .01 (unpairedttest inDH).

    1824 JID 2013:208 (1 December) Liu et al

  • 8/12/2019 Enhancement of Adaptive Immunity to Neisseria Gonorrhoeae

    5/9

    signicantly elevate gonococcus-specic IgA or IgG antibodies

    in either vaginal wash or serum samples. However, IL-12 mi-

    crosphere treatment increased vaginal and serum specic IgG

    antibody (Figure 1G and 1H), as well as vaginal specic IgA

    antibody production (Figure1G).

    IL-12 Microsphere Treatment Induction of Protective

    Anamnestic Immunity Against SecondaryN. gonorrhoeae

    InfectionWe further assessed whether IL-12 microsphere treatment re-

    sulted in the generation of immune memory and protective

    immunity against reinfection. Groups of mice infected with

    N. gonorrhoeaewere treated with IL-12loaded or blank micro-

    spheres, and after the infection had run its course, the mice

    were treated with ceftriaxone (300 g intraperitoneal) on day

    15 to ensure complete elimination of the gonococci. An addi-

    tional group of sham-infected mice treated with IL-12 micro-

    spheres was used to evaluate the possible persistent effect of

    IL-12 in the absence of infection; 56 weeks later, all mice were

    inoculated withN. gonorrhoeae of the same strain without any

    further treatment. As observed previously, primary infection of

    control-treated mice did not protect them against subsequent

    secondary challenge; the duration and bacterial burden of sec-ondary gonococcal infection in previously blank microsphere-

    treated mice were the same as for the primary infection of

    age-matched naive mice (Figure2Aand2B). In contrast, intra-

    vaginal treatment with IL-12loaded microspheres during pri-

    mary infection protected the mice against secondary infection;

    Figure 2. Effect of intravaginal interleukin 12 (IL-12) microsphere (ms) treatment during primary infection on secondaryNeisseria gonorrhoeaeinfection.

    A, Time course of secondary infection in mice treated with IL-12 ms, soluble IL-12, interleukin 17 (IL-17) ms, or control ms during primary infection or in

    previously sham-infected mice with or without IL-12 ms treatment (8 mice per group). Signicant differences in infection burdens were found between

    mice previously treated with IL-12 ms (P< .02) and controls (analysis of variance). B, Data from the experiment shown in A plotted as percentage of mice

    remaining infected after reinfection under the indicated treatments during primary infection. Infection was cleared signicantly faster in mice previouslytreated with IL-12 ms (P< .001) than in controls (Kaplan-Meier analysis). C, Flow cytometric analysis of cytokine expression in iliac lymph node CD4+

    T cells isolated at day 5 from reinfected mice treated with IL-12 ms, IL-17 ms, or control ms during primary infection, or from mice that were sham infected

    in both primary and secondary phases (sham reinfected) (7 mice per group).D, Reverse-transcription polymerase chain reaction (RT-PCR) analysis of in-

    terferon (IFN) , interleukin 4 (IL-4), and IL-17 messenger RNA (mRNA) levels in vaginas harvested at day 3 from sham-reinfected or reinfected mice

    treated with IL-12 ms, IL-17 ms, or blank ms during primary infection (7 mice per group). Cytokine gene expression levels detected by RT-PCR were normal-

    ized relative to expression of -actin and set at 1.0 for sham-reinfected group. E, F, Vaginal (E) and (F) serum anti-gonococcal immunoglobulin A (IgA) and

    immunoglobulin G (IgG) antibody responses to secondary infection in sham-reinfected or reinfected mice treated with IL-12 ms, IL-17 ms, or blank ms

    during primary infection (7 mice per group). Vaginal wash and serum samples were collected 15 days after inoculation, and gonococcus-specic and total

    IgA and IgG were measured with enzyme-linked immunosorbent assay. Results from 1 of 3 independent experiments are shown. Data shown as means

    SEM. #P< .05; *P< .01 (unpairedttest inCF).

    Immunity toN. gonorrhoeae JID 2013:208 (1 December) 1825

  • 8/12/2019 Enhancement of Adaptive Immunity to Neisseria Gonorrhoeae

    6/9

    reinfected mice that had been treated with IL-12 microspheres

    during the primary infection resisted the challenge more effec-

    tively than controls (Figure2Aand2B). However, previous IL-

    12 microsphere treatment of sham-infected mice did not induce

    protection against subsequent infection (Figure2Aand2B). This

    result also excluded the possibility that any persisting micro-

    spheres still affected the secondaryN. gonorrhoeaeinfection.

    Flow cytometric and RT-PCR analyses of ILN cells and

    vaginas performed on day 5 and day 3 of secondary infection,respectively, indicated that the protective effect of previous

    IL-12 microsphere treatment on secondary gonococcal infection

    was also associated with signicantly enhanced Th1 (IFN-)

    responses (Figure2Cand2D). There was also a robust specic

    secondary antibody response in IL-12 microspheretreated

    mice after they were rechallenged with N. gonorrhoeae. Gono-

    coccus-specic IgA and IgG antibodies in vaginal wash

    samples and IgG antibodies in serum samples from reinfected

    mice previously treated with IL-12 microspheres were signi-

    cantly higher than those of control groups (Figure2Eand2F).

    In contrast to the effects of IL-12 microsphere treatment, treat-

    ment with IL-17 microspheres during primary gonococcal in-

    fection did not lead to any protective immunity to secondary

    gonococcal infection, or induce any anamnestic T-cell or anti-

    body responses (Figure2AF).

    Protective Effects of AntiTGF-and AntiIL-10 Microsphere

    Treatments

    We next ascertained whether the local application of microspheres

    could be used to deliver other therapeutic agents for the immuno-

    modulatory treatment of vaginal gonococcal infection. We had

    previously found that treatment with 300 g of antiTGF- or

    antiIL-10 antibody by systemic injection every second dayprotected mice against genital gonococcal infection and induced

    specic protective immunity [10, 12]. We therefore treated

    N. gonorrhoeae-infected mice with PLA microspheres containing

    antiTGF- or antiIL-10 antibody by intravaginal instillation.

    Treatment with 10-fold lower amounts of antiTGF- or anti

    IL-10 antibody (30 g) encapsulated in microspheres achieved a

    similar therapeutic effect as observed previously with parenteral

    injection of these antibodies (Figure 3A and3B). No signicant

    weight loss (10%) or signs of distress (hunched posture, inactivi-

    ty, rufed fur) were observed. In addition, treatment with anti

    TGF-- or antiIL-10loaded microspheres during primary gono-

    coccal infection induced resistance to secondary infection

    (Figure3Dand3E). Local antiTGF-or antiIL-10 microsphere

    treatment was also associated with enhanced Th1 (IFN-) and

    Th2 (IL-4) responses in ILNs (Figure3Cand3F).

    DISCUSSION

    Although it has long been recognized that gonorrhea can be con-

    tracted repeatedly with no apparent development of protective

    immunity or immunological memory, the reasons for this are

    poorly understood. Our recent studies have demonstrated that

    the lack of protective immunity to N. gonorrhoeae is due not

    only to immunoevasion through antigenic variation and other

    strategies, but also to the ability of the pathogen to suppress

    host-specic immune responses in the rst place [9]. Induction

    of immunosuppressive cytokines, TGF-and IL-10, and type 1

    regulatory T cells play an important role in this process [10

    12]. Importantly, our results demonstrate that the natural in-hibitory effect ofN. gonorrhoeaeon Th1/Th2-mediated specic

    immunity can be immunologically reversed.

    The current study showed that intravaginal administration of

    microencapsulated IL-12 elicited the development of adaptive

    immunity against N. gonorrhoeae genital infection in mice.

    Consistent with its well-known type 1stimulatory properties,

    IL-12 treatment signicantly enhanced Th1 immune responses

    to N. gonorrhoeae, indicated by increased production of IFN-

    +/CD4+ T cells in both ILNs and the genital tracts of treated

    mice compared with those from control mice. The treatment

    also signicantly increased the production of gonococcus-

    specic vaginal IgA and IgG and serum IgG antibodies, al-

    though it did not up-regulate Th2 (IL-4) responses. It has been

    reported that IL-12 promotes the production of IgG2a, IgG3

    and mucosal IgA antibodies through both IFN-dependent

    and direct actions [reviewed in 16]. It initially stimulates Th1

    and natural killer cells to secrete large amount of IFN-, which

    induces selective isotype switching in B cells and further stimu-

    lates antibody production [30]. The IL-12 receptor is expressed

    on murine and human B cells through all stages of maturation

    [31], and direct interaction of IL-12 with B cells leads to activa-

    tion of the p50 and c-Rel NF-B family members, B-cell prolif-

    eration and differentiation, and antibody production [3234].All these mechanisms may contribute to the induction of

    humoral immune responses to N. gonorrhoeae infection by

    IL-12 microsphere treatment, in the absence of an elevated Th2

    (IL-4) response.

    Treatment of primary gonococcal infection with IL-12

    loaded microspheres also protected the mice against subse-

    quent N. gonorrhoeae infection. There were both strong Th1

    and specic antibody responses in IL-12 microsphere-treated

    mice after they were reexposed to N. gonorrhoeae without

    further IL-12 treatment. The levels of gonococcus-specic anti-

    bodies in vaginal wash and serum samples from mice that werereinfected after previous IL-12 microsphere treatment were sig-

    nicantly higher than those from the same mice during

    primaryN. gonorrhoeaeinfection, suggesting that the treatment

    had resulted in the generation of immune memory.

    However, intravaginal treatment of mice with the same dose

    and schedule of free, soluble IL-12 did not show any protective

    effect against either primary or secondary gonococcal infection,

    possibly because the cytokine was susceptible to proteolytic

    degradation or had limited vaginal mucosal uptake. The potential

    1826 JID 2013:208 (1 December) Liu et al

  • 8/12/2019 Enhancement of Adaptive Immunity to Neisseria Gonorrhoeae

    7/9

    advantages of local, targeted microencapsulated drug delivery

    include reduction of harmful side effects, decreased dosages

    needed, and continuous maintenance of drug levels in a thera-

    peutically effective range. In our studies the passage of intrava-

    ginally delivered microencapsulated IL-12 into the circulationwas minimal, yet the levels achieved in the genital tract were

    sufcient to induce acquired immunity to gonococcal infection.

    In addition, we determined whether the enhancement of

    natural Th17-governed innate immune responses to N. gonor-

    rhoeaeby local IL-17 microsphere treatment could protect the

    mice against gonococcal infection. Consistent with our previ-

    ous ndings that antiIL-17 antibody treatment, or deciency

    of IL-17RA, leads to prolonged genital tract infection with

    N. gonorrhoeae [13], intravaginal instillation of IL-17 micro-

    spheres ameliorated primary gonococcal infection. However, it

    was less effective than treatment with IL-12 microspheres; fur-

    thermore it did not signicantly protect against subsequent in-

    fection or induce Th1/Th2 responses. Whereas antiTGF-

    treatment interferes with Th17 responses [10, 11], adminis-tration of IL-12 microspheres enhanced Th1-driven adaptive

    responses without affecting IL-17 production and might there-

    fore have the advantage of promoting both adaptive and innate

    defense againstN. gonorrhoeae[35].

    We also explored the use of PLA microspheres for intravagi-

    nal delivery of other potential therapeutic agents in the treat-

    ment of gonococcal infection. We had shown elsewhere that

    systemic administration of TGF-- or IL-10-neutralizing anti-

    bodies could elicit the generation of Th1/Th2 adaptive immunity

    Figure 3. Effect of intravaginal antitransforming growth factor (TGF) antibody or antiinterleukin 10 (IL-10) antibody microsphere (ms) treatment

    during primary infection on primary and secondary genital Neisseria gonorrhoeaeinfection in BALB/c mice. A, Time course of primary infection in mice

    treated with antiTGF-ms, antiIL-10 ms, or control ms (8 mice per group). Signicant differences in infection burdens were found between mice treated

    with antiTGF-ms (P< .02) or antiIL-10 ms (P< .05) and controls (analysis of variance).B, Data from experiment shown in A plotted as percentage of

    mice remaining infected with the indicated treatments. Infection was cleared signicantly faster in mice treated with antiTGF-ms (P< .01) or antiIL-10

    ms (P< .01) than in controls (Kaplan-Meier analysis). C, Flow cytometric analysis of cytokine expression in isolated iliac lymph node CD4+ T cells from

    sham-infected or infected mice with antiTGF-antibody ms, antiIL-10 antibody ms, or control ms treatment (7 mice per group). D, Time course of sec-

    ondary infection in mice treated with antiTGF-antibody ms, antiIL-10 antibody ms, or control ms during primary infection (8 mice per group). Signicant

    differences in infection burdens were found between mice previously treated with antiTGF-ms (P< .02) or antiIL-10 ms (P< .02) and controls (analysis

    of variance).E, Data from experiment shown inDplotted as percentage of mice remaining infected after reinfection under the indicated treatments during

    primary infection. Infection was cleared signicantly faster in mice previously treated with antiTGF-ms (P< .01) or antiIL-10 ms (P< .001) than in con-

    trols (Kaplan-Meier analysis).F, Flow cytometric analysis of cytokine expression in iliac lymph node CD4+ T cells isolated at day 5 from sham-reinfected or

    reinfected mice treated with antiTGF-antibody ms, antiIL-10 antibody ms, or control ms during primary infection (7 mice per group). Results from 1 of

    3 independent experiments are shown. Data shown as means SEM. Abbreviations: IFN, interferon; IL-4, interleukin 4. #P< .05; *P< .01 (unpairedttest in

    CandF).

    Immunity toN. gonorrhoeae JID 2013:208 (1 December) 1827

  • 8/12/2019 Enhancement of Adaptive Immunity to Neisseria Gonorrhoeae

    8/9

    to N. gonorrhoeae [10,12]. However, systemic treatment with

    these antibodies is unlikely to be a viable therapeutic option for

    gonococcal infection owing to the high dosages needed and po-

    tential toxicity. Our current results showed that intravaginally

    administered microencapsulated antiTGF-- or antiIL-10 at

    much lower doses similarly protected the mice against genital

    tract N. gonorrhoeae infection, demonstrating the clinical po-

    tential of this technology. Because it is known that IL-12 can

    antagonize some of the effects of IL-10 and reverse the regula-tory effects of TGF- [3638], these results are consistent, and

    it is possible that treatment with IL-12 microspheres could be

    combined with antiIL-10 or anti TGF- microspheres to

    achieve a greater effect.

    Despite public health measures, gonorrhea remains one of the

    most frequent reportable infectious diseases for which we have

    no effective vaccine.N. gonorrhoeaehas steadily developed resis-

    tance to each class of antibiotics deployed against it, including

    uoroquinolones and, most recently, even cephalosporins [5]; as

    a result, there are serious concerns that gonorrhea might become

    untreatable [3]. The results from the current study indicate that

    local manipulation of cytokines that control Th1, Th2, and regu-

    latory immune responses can reverse the inhibitory effect ofN.

    gonorrhoeaeon adaptive immunity and may serve as novel, safe

    approaches to the treatment and prevention of this very

    common sexually transmitted infection.

    In addition, our ndings may be useful in formulating novel

    approaches to gonococcal vaccine development. Indeed it can

    be argued that intravaginal administration of IL-12 micro-

    spheres serves as an adjuvant that converts gonococcal infec-

    tion into a live vaccine. This has the added advantage of

    targeting the vaccine toward a population that is at risk of

    repeat infection with gonorrhea.

    Notes

    Acknowledgments. We are grateful to the assistance of the Confocal

    Microscope and Flow Cytometry Facility in the School of Medicine and Bi-

    omedical Sciences, University at Buffalo. We thank Ann E. Jerse, PhD, for

    helpful advice and valuable discussions during the course of this work,

    Terry Mashtare, MS, for statistical analysis and advice, and Thomas Russo,

    MD, for critical reading of the manuscript.

    Financial support. This work was supported by the National Institute

    of Allergy and Infectious Diseases at the National Institutes of Health

    (grant AI074791 to M. W. R., and grant AI104067 to Y. L.), and by a grant

    from The John R. Oishei Foundation, Buffalo (grant to M. W. R.).

    Potential conicts of interest. N. K. E. has ownership interest in Thera-pyX, which is developing sustained-release particulate formulations of

    IL-12 for cancer therapy. All other authors report no potential conicts.

    All authors have submitted the ICMJE Form for Disclosure of Potential

    Conicts of Interest. Conicts that the editors consider relevant to the

    content of the manuscript have been disclosed.

    References

    1. Russell MW, Hook EW III. Gonorrhea. In: Barrett ADT, Stanberry LR,

    eds. Vaccines for biodefense and emerging and neglected diseases.

    London, UK: Academic Press,2009:96381.

    2. Zhu W, Chen CJ, Thomas CE, et al. Vaccines for gonorrhea: can we

    rise to the challenge? Front Microbiol 2011; 2:124.

    3. World Health Organization. Global action plan to control the spread

    and impact of antimicrobial resistance inNeisseria gonorrhoeae,2012.

    4. Hook EW, Handseld HH. Gonococcal infections in the adult. In:

    Holmes KK, Sparling PF, Mrdh PA, et al., eds. Sexually transmitted dis-

    eases. New York, NY: McGraw-Hill Health Professions Division,1999.

    5. Bolan GA, Sparling PF, Wasserheit JN. The emerging threat of untreat-

    able gonococcal infection. N Engl J Med2012; 366:4857.

    6. Ram S, Cullinane M, Blom AM, et al. Binding of C4b-binding protein

    to porin: a molecular mechanism of serum resistance ofNeisseria gon-

    orrhoeae. J Exp Med2001; 193:28195.

    7. Smith H, Parsons NJ, Cole JA. Sialylation of neisserial lipopolysaccharide:

    a major inuence on pathogenicity. Microb Pathog1995; 19:36577.

    8. Lewis LA, Burrowes E, Rice PA, Ram S. Interactions ofNeisseriawith

    complement. In: Genco CA, Wetzler L, eds. Neisseria: molecular mech-

    anisms of pathogenesis. Norfolk, UK: Caister Academic Press,2010.

    9. Liu Y, Feinen B, Russell MW. New concepts in immunity toNeisseria

    gonorrhoeae: innate responses and suppression of adaptive immunity

    favor the pathogen, not the host. Front Microbiol2011; 2:52.

    10. Liu Y, Russell MW. Diversion of the immune response toNeisseria gon-

    orrhoeaefrom Th17 to Th1/Th2 by treatment with anti-transforming

    growth factorantibody generates immunological memory and protec-

    tive immunity. MBio2011; 2:e0009511.

    11. Liu Y, Islam EA, Jarvis GA, et al.Neisseria gonorrhoeaeselectively sup-

    presses the development of Th1 and Th2 cells, and enhances Th17 cellresponses, through TGF--dependent mechanisms. Mucosal Immunol

    2012; 5:320331.

    12. Liu Y, Liu W, Russell MW. Suppression of host adaptive immune re-

    sponses byNeisseria gonorrhoeae: role of interleukin 10 and type 1 reg-

    ulatory T cells [published online ahead of print June 12, 2013].

    Mucosal Immunol.2013. doi:10.1038/mi.2013.36.

    13. Feinen B, Jerse AE, Gaffen SL, Russell MW. Critical role of Th17 re-

    sponses in a murine model ofNeisseria gonorrhoeae genital infection.

    Mucosal Immunol2010; 3:312321.

    14. Egilmez NK. Interleukin-12: effector mechanisms and homeostatic

    counter-regulation. In: Manjili MH, ed. Cytokines: mechanisms, func-

    tions and abnormalities. 1st ed. Hauppauge, NY: Nova Science Publish-

    ers,2011:322.

    15. Metzger DW. IL-12 as an adjuvant for the enhancement of protective

    humoral immunity. Expert Rev Vaccines 2009; 8:515

    8.16. Metzger DW. Interleukin-12 as an adjuvant for induction of protective

    antibody responses. Cytokine2010; 52:1027.

    17. Boyaka PN, Marinaro M, Jackson RJ, et al. IL-12 is an effective adjuvant

    for induction of mucosal immunity. J Immunol 1999; 162:1228.

    18. Arulanandam BP, OToole M, Metzger DW. Intranasal interleukin-12

    is a powerful adjuvant for protective mucosal immunity. J Infect Dis

    1999; 180:9409.

    19. Lynch JM, Briles DE, Metzger DW. Increased protection against pneu-

    mococcal disease by mucosal administration of conjugate vaccine plus

    interleukin-12. Infect Immun2003; 71:47808.

    20. Baron SD, Singh R, Metzger DW. InactivatedFrancisella tularensislive

    vaccine strain protects against respiratory tularemia by intranasal vacci-

    nation in an immunoglobulin A-dependent fashion. Infect Immun

    2007; 75:215262.

    21. Kumar D, Kirimanjeswara G, Metzger DW. Intranasal administrationof an inactivated Yersinia pestis vaccine with interleukin-12 generates

    protective immunity against pneumonic plague. Clin Vaccine Immunol

    2011; 18:192535.

    22. Cohen J. IL-12 deaths: explanation and a puzzle. Science1995; 270:908.

    23. Hedlund J, Langer B, Konradsen HB, Ortqvist A. Negligible adjuvant

    effect for antibody responses and frequent adverse events associated

    with IL-12 treatment in humans vaccinated with pneumococcal poly-

    saccharide. Vaccine2001; 20:1649.

    24. Egilmez NK, Kilinc MO, Gu T, Conway TF. Controlled-release particu-

    late cytokine adjuvants for cancer therapy. Endocr Metab Immune

    Disord Drug Targets2007; 7:26670.

    1828 JID 2013:208 (1 December) Liu et al

  • 8/12/2019 Enhancement of Adaptive Immunity to Neisseria Gonorrhoeae

    9/9

    25. Mathiowitz E, Jacob JS, Jong YS, et al. Biologically erodable microspheres

    as potential oral drug delivery systems. Nature 1997; 386:4104.

    26. Langer R. Drug delivery and targeting. Nature1998; 392:510.

    27. Egilmez NK, Jong YS, Mathiowitz E, Bankert RB. Tumor vaccination with

    cytokine-encapsulated microspheres. Methods Mol Med2003; 75:68796.

    28. Jerse AE. Experimental gonococcal genital tract infection and opacity

    protein expression in estradiol-treated mice. Infect Immun 1999;

    67:5699708.

    29. Weigmann B, Tubbe I, Seidel D, et al. Isolation and subsequent analysis

    of murine lamina propria mononuclear cells from colonic tissue. Nat

    Protoc2007; 2:230711.

    30. Snapper CM, Paul WE. Interferon-gamma and B cell stimulatory

    factor-1 reciprocally regulate Ig isotype production. Science 1987;

    236:9447.

    31. Airoldi I, Gri G, Marshall JD, et al. Expression and function of IL-12

    and IL-18 receptors on human tonsillar B cells. J Immunol 2000;

    165:68808.

    32. Jelinek DF, Braaten JK. Role of IL-12 in human B lymphocyte prolifera-

    tion and differentiation. J Immunol 1995; 154:160613.

    33. Thibodeaux DK, Hunter SE, Waldburger KE, et al. Autocrine regula-

    tion of IL-12 receptor expression is independent of secondary IFN-se-

    cretion and not restricted to T and NK cells. J Immunol 1999; 163:

    525764.

    34. Skok J, Poudrier J, Gray D. Dendritic cell-derived IL-12 promotes B cell

    induction of Th2 differentiation: a feedback regulation of Th1 develop-

    ment. J Immunol1999; 163:428491.

    35. Nady S, Ignatz-Hoover J, Shata MT. Interleukin-12 is the optimum cy-

    tokine to expand human Th17 cells in vitro. Clin Vacc Immunol 2009;

    16:798805.

    36. Watkins SK, Egilmez NK, Suttles J, Stout RD. IL-12 rapidly alters the

    functional prole of tumor-associated and tumor-inltrating macro-

    phages in vitro and in vivo. J Immunol2007; 178:135762.

    37. Yu J, Wei M, Becknell B, et al. Pro- and antiinammatory cytokine sig-

    naling: reciprocal antagonism regulates interferon-gamma production

    by human natural killer cells. Immunity2006; 24:57590.

    38. Thomas DA, Massague J. TGF-beta directly targets cytotoxic T cell

    functions during tumor evasion of immune surveillance. Cancer Cell

    2005; 8:36980.

    Immunity toN. gonorrhoeae JID 2013:208 (1 December) 1829