meningescesar.x

Embed Size (px)

Citation preview

  • 8/6/2019 meningescesar.x

    1/14

    The differential response of human dendritic cells tolive and killed Neisseria meningitidis

    Hannah E. Jones,1,2 Heli Uronen-Hansson,3

    Robin E. Callard,2 Nigel Klein1 and

    Garth L. J. Dixon1,4*1Infectious Diseases and Microbiology Unit and2Immunobiology Unit, Institute of Child Health, UCL,

    London, UK.3Department of Experimental Medical Science,

    Immunology Lund University, Lund, Sweden.4Department of Microbiology, Camelia Botnar

    Laboratories, Great Ormond Street Hospital, London,

    UK.

    Summary

    There is currently no effective vaccine for Neisseria

    meningitidis (Nm) serogroup B. Generation of

    optimal immune responses to meningococci could

    be achieved by targeting meningococcal antigens to

    human dendritic cells (DCs). Recent studies have

    shown that diverse DC responses and subsequent

    generation of protective immunity can be observed if

    the microbes are viable or killed. This is important

    because the host is likely to be exposed to both live

    and killed bacteria during natural infection. There are

    currently few data on comparative DC responses to

    live and killed meningococci. We show here that

    exposure of human DC to live meningococci does

    not result in a typical maturation response, as deter-

    mined by the failure to upregulate CD40, CD86,

    HLA-DR and HLA-Class I. Despite this, live meningo-

    cocci were potent inducers of IL-12 and IL-10,

    although the ratios of these cytokines differed from

    those to killed organisms. Our data also suggest that

    enhanced phagocytosis of killed organisms com-

    pared with live may be responsible for the differential

    cytokine responses, involving an autocrine IL-10-

    dependent mechanism. The consequences of thesefindings upon the effectiveness of antigen presen-

    tation and T-cell responses are currently under

    investigation.

    Introduction

    Neisseria meningitidis (Nm) remains a major cause of

    bacterial meningitis and septicaemia worldwide. While

    conjugate capsular vaccines have been developed suc-

    cessfully against serogroups A, C, W135 and Y, the devel-

    opment of a safe and effective vaccine against serogroup

    B bacteria has remained elusive (Jodar et al., 2002; Har-

    rison, 2006). Use of serogroup B polysaccharide capsule

    as a vaccine is regarded as being problematic due to its

    poor immunogenicity and concerns over safety due to the

    similarity of group B polysialic acid capsule to human

    neural cell adhesion molecules (Finne et al., 1983). Thecurrent serogroup B vaccines are based on subcapsular

    outer membrane antigens; however, their efficacy has

    been found to be limited especially in infants, who are at

    most risk of the disease (Bjune et al., 1991; Cartwright

    et al., 1999; Tappero et al., 1999). Although a number of

    other promising vaccine candidates (Pizza et al., 2000;

    Giuliani et al., 2006) have been identified, there are

    limited data on how these should be presented to the

    immune system in order to induce immunity to a broad

    range of meningococcal antigens, elicit long-term memory

    and provide protection for all age groups. Understanding

    how meningococci interact with dendritic cells (DCs) islikely to be key to achieving these aims.

    Dendritic cells are the key orchestrators of the immune

    responses to the microbial world (Banchereau and Stein-

    man, 1998). Immature DCs in the periphery and submu-

    cosa sample the external environment and capture

    antigen, including whole bacteria, after which they migrate

    to secondary lymphoid tissue where they present pro-

    cessed antigen to stimulate antigen-specific T-cells

    (Banchereau et al., 2000). The outcome of the interaction

    between DCs and lymphocyte is critically influenced by

    the release of both cytokines and chemokines and by the

    expression and function of co-stimulatory molecules by

    DCs. The nature of the DC response to microbial encoun-

    ter appears to be dictated to a large extent by the

    presence of pathogen-associated molecules, such as

    lipopolysaccharide (LPS) present on the microbe, and the

    signals generated as a result of the interaction of these

    microbial components and host receptors, principally toll-

    like receptors (TLRs) (Pulendran, 2005).

    We have previously shown that both the presence and

    structure of LPS in the outer membrane of Nm are critical

    Received 12 March, 2007; revised 30 May, 2007; accepted 6 June,2007. *For correspondence. E-mail [email protected]; Tel.(+44) 207813 8594; Fax (+44) 207813 8494.

    Cellular Microbiology (2007) 9(12), 28562869 doi:10.1111/j.1462-5822.2007.01001.xFirst published online 10 July 2007

    2007 The AuthorsJournal compilation 2007 Blackwell Publishing Ltd

    mailto:[email protected]:[email protected]
  • 8/6/2019 meningescesar.x

    2/14

    in defining the response of human DCs to the bacteria.

    Wild-type killed meningococci induce both DC maturation

    and production of cytokines, particularly IL-10 and bioac-

    tive IL-12 p70, whereas neither purified LPS nor LPS

    deficient meningococci, even with exogenously added

    LPS, are able to induce high levels of these cytokines

    (Dixon et al., 2001). The reason why wild-type meningo-

    cocci are capable of inducing high levels of IL-10 and

    IL-12 appears to stem from the requirement of LPS within

    the outer membrane for efficient internalization by human

    DCs (Uronen-Hansson et al., 2004a).

    It is clear, however, that both DC maturation and pro-

    duction of certain cytokines (particularly IL-6) are inde-

    pendent of phagocytosis, and moreover can be induced in

    the absence of LPS (Uronen-Hansson et al., 2004a). For

    example, it has been shown that major meningococcal

    outer membrane proteins porA (Al Bader et al., 2004) and

    PorB (Singleton et al., 2005) can induce both DC matu-

    ration and the production of certain cytokines. Therefore,

    in addition to being target antigens these outer membrane

    components also appear to possess adjuvant activitywhich could be advantageous for their use as vaccines.

    Such preparations, however, do not appear to support

    significant production of IL-10 or bioactive IL-12 by DCs,

    both considered to have a critical influence on T-cell

    responses (Al Bader et al., 2004). Targeting DCs by uti-

    lizing whole bacteria through the phagocytic pathway has

    the advantage that it is a highly efficient route for antigen

    delivery. Once within the DC, LPS and other bacterial

    components such as porins in the bacteria then can

    engage with a variety of receptors, including TLRs,

    located within endosomal and phagosomal compartments

    (Underhill et al., 1999; Underhill and Ozinsky, 2002;Uronen-Hansson et al., 2004b). It may be that the signals

    generated by these processes are critical in induction of

    T-cell polarizing cytokines.

    As immune responses to live and killed organisms can

    differ markedly (Kikuchi et al., 2004; Skinner et al., 2004;

    Rey-Ladino et al., 2005), we sought to compare the

    response of DC to live and killed Nm. We postulated that

    live organisms would be at least as effective as killed

    counterparts at inducing DC maturation and cytokine

    production. Surprisingly, we found that live Nm were inca-

    pable of inducing full DC maturation as compared with

    their killed counterparts. Despite the lack of a typical

    maturation response, live Nm could induce both IL-12

    production and IL-10 by DCs, although the ratios of these

    two cytokines were quite different compared with the

    response to killed organisms. The results presented here

    suggest that live meningococci may specifically modulate

    DC function which could have important influences on the

    nature of T-cell and B-cell responses that occur during

    natural infection/carriage and may have implications in

    the design of novel vaccines against this pathogen.

    Results

    A distinct surface phenotype is observed when DCs are

    exposed to live Nm compared with killed bacteria

    We have previously shown that killed Nm increase

    surface expression of co-stimulatory molecules CD86,

    CD40, CD83, HLA-Class I and HLA-DR on human DCs,

    consistent with their maturation and activation (Dixon

    et al., 2001). We first sought to determine whether live Nm

    were able to induce similar DC phenotypic changes. DCs

    were incubated for 1824 h with either live or killed Nm at

    a multiplicity of infection (moi) ranging from 1 to 100 and

    expression of these activation markers determined by

    flow cytometry. Unexpectedly, there was no increase in

    surface expression of CD40, CD86 and MHC Class I

    when DCs were cultured with live Nm (Fig. 1A). This

    observation was independent of the donor from whom the

    DCs were derived and was consistent over a range of

    1100 moi (Fig. 1B). A modest increase in expression of

    HLA-DR and CD83 was observed in response to live Nm

    (Fig. 1A) suggesting that these cells were undergoingsome maturation. The possibility that the induction of

    co-stimulatory molecules expression in response to live

    Nm was delayed compared with killed bacteria was

    explored. The differential pattern of DC cell surface

    expression between live and dead meningococci was the

    same at 48 h as observed at 24 h (data not shown).

    We investigated the possibility that these observed dif-

    ferences could be due to excessive DC necrosis or apo-

    ptosis in co-culture with proliferating bacteria. Despite the

    exposure of DCs to continually proliferating bacteria,

    there was no difference in the proportion of dead DCs

    (ranging from 5% to 15%) for all culture conditions asdetermined by trypan blue staining. To address the pro-

    portion of DCs that were undergoing apoptosis, we mea-

    sured caspase activity by flow cytometry. Figure 2 shows

    the percentage of DCs positive for caspase activity follow-

    ing stimulation with medium, live and killed Nm (100 moi).

    There is a modest increase in apoptotic cells after activa-

    tion with live Nm but still less than 24% compared with

    14% with dead bacteria. This amount of death is not

    sufficient to explain the inability of live bacteria to induce

    CD40 and CD86 expression. We conclude from these

    data that the observed differences between proliferating

    and killed bacteria are unlikely to be due to excessive

    cellular necrosis or apoptosis in live co-cultures.

    Brief exposure to dividing bacteria determines the

    distinct maturation response of DCs to live Nm

    We next investigated if bacterial proliferation and protein

    synthesis was necessary for the distinct DC surface mol-

    ecule expression pattern we had already observed in

    response to livebacteria. To achieve this, live bacteria were

    Dendritic cell response to Neisseria meningitidis 2857

    2007 The AuthorsJournal compilation 2007 Blackwell Publishing Ltd, Cellular Microbiology, 9, 28562869

  • 8/6/2019 meningescesar.x

    3/14

    100 101 102 103 104

    FL1-H

    0

    100

    Events

    100 101 102 103 104

    FL1-H

    0

    100

    Events

    100 101 102 103 104

    FL2-H

    0

    60

    Events

    100 101 102 103 104

    FL2-H

    0

    60

    Events

    CD40 CD86 HLA-Class I HLA-DR CD83

    Killed

    Live

    A

    B

    100 101 102 103 104

    FL2-H

    0

    60

    Events

    100 101 102 103 104

    FL2-H

    0

    64

    Events

    100 101 102 103 104

    FL1-H

    0

    50

    Events

    100 101 102 103 104

    FL1-H

    0

    64

    Events

    100 101 102 103 104

    FL1-H

    0

    70

    Events

    100 101 102 103 104

    FL1-H

    0

    64

    Events

    Medium

    MOI100live

    M

    OI100killed

    50.00

    100.00

    150.00

    200.00

    MFICD86

    p

  • 8/6/2019 meningescesar.x

    4/14

    exposed to bacteriostatic levels (2.510 mg ml-1) of

    chloramphenicol (cm) prior to and during co-culture with

    DCs. Cm inhibits both microbial protein synthesis by

    binding to the 50S subunit of the ribosome and impairing

    peptidyl transferase activity, and is rapidly but rever-

    sibly bacteristatic. In this culture system, the concentra-

    tions of antibiotic used completely inhibit meningococcal

    proliferation, but do not alter viability (see Experimental

    procedures).

    Our results showed that when cm is added prior to

    co-culture with DCs, the pattern of CD40, CD83, CD86,

    HLA-Class 1 and HLA-DR expression was identical to that

    seen with DCs exposed to killed Nm (Fig. 3A). This was

    consistent among all donors tested and was observed at

    all starting concentrations of bacteria (moi of 1100). We

    then investigated over what time period DCs have to be

    exposed to proliferating meningococci to result in the dis-

    tinct DC maturation response. When cm was added after

    6 h exposure to live bacteria, the pattern of cell surface

    molecule expression was similar to that seen with live

    bacteria. This is illustrated in Fig. 3B using CD40 expres-

    sion as an example. Cm had no effect on DC maturation

    100101 102 103 10

    4

    FL1-H

    0

    125

    Events

    10 0 10 1 10 2 10 3 10 4

    FL1-H

    0

    125

    Events

    100 101 102 103 104

    FL1-H

    0

    125

    Events

    CD40 CD86 HLA-Class I HLA-DR CD83

    10 0 10 1 10 2 10 3 10 4

    FL2-H

    0

    90

    Events

    100 101 102 103 104

    FL2-H

    0

    90

    Events

    100 101 102 103 104

    FL2-H

    0

    90

    Events

    10 0 101 10 2 103 10 4

    FL1-H

    0

    100

    Events

    10 0 101 10 2 103 10 4

    FL1-H

    0

    100

    Events

    10 0 10 1 102 10 3 10 4

    FL1-H

    0

    100

    Events

    100 101 102 103 104

    FL1-H

    0

    100

    Events

    100 101 102 103 104

    FL1-H

    0

    100

    Events

    100 101 102 103 104

    FL1-H

    0

    100

    Events

    100 101 102 103 104

    FL2-H

    0

    60

    Events

    100 101 102 103 104

    FL2-H

    0

    60

    Events

    100 101 102 103 104

    FL2-H

    0

    60

    Events

    Killed

    Live

    Live +

    cm at

    0 h

    postinfection

    A

    Isotype control

    Unstimulated

    Killed

    Live

    Live + cm at 0 h post infection

    Live + cm at 6 h post infection

    Isotype control

    Unstimulated

    Stimulated

    B

    100 101 102 103 104

    FL1-H

    0

    100

    Events

    Fig. 3. Bacteristatic doses of chloramphenicol restore capacity of live Nm to induce DC maturation.A. DCs were stimulated with live or killed Nm (moi 100) with and without cm (5 mg ml-1) for 18 h and expression of CD40, CD83, CD86,HLA-Class 1 and HLA-DR determined by flow cytometry.B. A delay in addition of chloramphenicol to live meningococcal/DC co-culture by 6 h results in no induction of CD40 expression.The data shown here are representative of experiments from five separate donors.

    Dendritic cell response to Neisseria meningitidis 2859

    2007 The AuthorsJournal compilation 2007 Blackwell Publishing Ltd, Cellular Microbiology, 9, 28562869

  • 8/6/2019 meningescesar.x

    5/14

    when added alone, nor did it alter the induction of matu-

    ration markers in response to either killed bacteria or LPS

    (data not shown). These data suggest brief exposure of

    DCs to proliferating Nm was required to induce poor

    maturation. It also implies that bacterial proliferation or

    protein synthesis is required for the effect of the livebacteria.

    Distinct DC maturation phenotype response to live

    meningococci is dominant over the stimulus of killed

    bacteria

    In natural infection it is likely there is a combination of

    viable and dead bacteria, so we investigated whether the

    presence of both live and killed Nm affected the distinct

    DC phenotype we observed from live Nm-stimulated DCs.

    To test this, DCs were co-cultured with live and killed Nm

    and the surface phenotype of DCs assessed as before by

    flow cytometry. In the presence of live bacteria, fixed

    bacteria were unable to induce DC maturation to levels

    observed when incubated with fixed bacteria alone, as

    illustrated in Fig. 4 for CD40, CD83, CD86 and HLA-Class

    1 expression. The same pattern of results was observed

    when the ratio of live to killed bacteria was varied (moi 100

    killed and moi 10 live) or when LPS (a potent inducer of

    DC surface maturation) was substituted for the killed

    bacteria. These data show that distinct DC maturation in

    response to live bacteria is dominant over normally potent

    inducers of DC maturation.

    Distinct DC surface phenotype in response to live

    meningococci is dependent on contact between bacteria

    and DC

    We next investigated whether the pattern of surface

    markers obtained on co-culture with live bacteria required

    contact with DCs. Responses of DC to live and killed

    meningococci, either in direct contact or separated by

    0.1 mm transwell (preventing passage of intact meningo-

    cocci but allowing free passage of membrane fragments

    including neisserial blebs and liquid phase molecules),

    were examined. As expected, when live bacteria and DCs

    were not separated by the transwell membrane, little or no

    induction of cell surface markers was observed, as illus-

    trated in Fig. 5A using HLA-Class I expression as an

    example. In contrast, when live bacteria were separated

    by the 0.1 mm membrane, HLA-Class I was upregulated

    (Fig. 5A). These results suggest that bacterial compo-nents released into the media are capable of inducing DC

    maturation. To confirm these findings, DCs were incu-

    bated with supernatants from live bacteria grown in mid-

    log phase. We found that bacterial supernatant from live

    bacteria was a potent stimulator of DC maturation

    (Fig. 5B). Similar patterns were observed for CD83,

    CD86, CD40 and HLA-DR (data not shown). We postulate

    that a likely reason for this is the activity of inflammatory

    moieties such as outer membrane vesicles (OMVs) or

    released neisserial products (DNA and peptidoglycan),

    which would be able to pass through the transwell mem-

    brane or would be present in supernatant following growthof Nm. This is perhaps not surprising as it is known that

    meningococcal OMVs are potent inducers of DC matura-

    tion (Al Bader et al., 2003).

    To further explore the mechanisms that may explain the

    distinct DC phenotype observed in response to intact live

    Nm, DCs were incubated with lysates from live bacteria.

    As Fig. 6 demonstrates, lysates from live bacteria induced

    upregulation of DC maturation markers (using CD40 and

    HLA-DR expression as an example). Similar results were

    also observed for killed bacterial lysates (data not shown).

    In contrast, intact live Nm failed to induce CD40 expres-

    sion and only moderate increase in HLA-DR expression

    (Fig. 6 and as previously demonstrated).

    Taken together, these data strongly suggest that the lack

    of DC maturation induced by Nm is due to a mechanism

    that requires close contact with bacteria and DC. The

    finding that lysates of live bacteria induce similar matura-

    tion to that seen with whole killed bacteria suggests that it

    is not simply due to increase in production of a bacterial

    product but rather a function of intact viable bacteria requir-

    ing bacterial cell division or protein synthesis or both.

    100 101 102 103 104

    FL1-H

    0

    80

    Events

    10 0 10 1 10 2 10 3 10 4

    FL2-H

    0

    80

    Events

    10 0 10 1 10 2 10 3 10 4FL1-H

    0

    80

    Events

    100 10 1 10 2 10 3 104

    FL2-H

    0

    80

    Events

    CD40 HLA-Class I

    CD86 CD83

    Isotype control

    Killed

    Killed/Live

    Cells

    Live

    Fig. 4. Distinct DC phenotypic response to live Nm is dominantover the response to killed bacteria. DCs were co-cultured with live,killed and live/killed Nm at moi 100 for 18 h. Expression of CD40,CD83, CD86 and HLA-Class 1 were then determined by flowcytometry. Flow cytometry profiles are from one donor out of fivestudied, with similar results.

    2860 H. E. Jones et al.

    2007 The AuthorsJournal compilation 2007 Blackwell Publishing Ltd, Cellular Microbiology, 9, 28562869

  • 8/6/2019 meningescesar.x

    6/14

    Live Nm induces different profiles of DC IL-12 p70 and

    IL-10 production compared with killed bacteria

    Our previous work has demonstrated that DC cytokine

    responses to Nm, especially IL-12 p70 and IL-10, are

    dependent upon internalization of bacteria by DC (Uronen-

    Hansson et al., 2004a). Given the lack of induction of

    co-stimulatory molecule expression in response to live

    bacteria shown in this study, we wanted to determine

    whether this would be reflected in poor cytokine responses

    compared with killed Nm. Interestingly, we found that live

    bacteria induced similar levels of IL-6 and TNF-a to those

    we had previously published with killed Nm (Uronen-

    Hansson et al., 2004a). However, when IL-12 p70 and

    IL-10 production was examined, an entirely different pat-

    tern emerged. As Fig. 7 illustrates, although IL-10 and

    IL-12 production can be detected in response to both live

    and killed meningococci, live Nm induced significantly

    greater levels of IL-12 than killed Nm. This was significant,

    despite interindividual variation of cytokine responses

    to these bacteria (moi of 100 and 10, P= 0.007 and

    P= 0.046, respectively, n= 10). In contrast, IL-10 produc-tion in response to killed organisms was significantly

    greater than that seen with live bacteria (moi 100,

    P= 0.001; moi 10, P= 0.005).

    Internalization of live meningococci by human DC is

    reduced compared with killed meningococci

    Our previous work showed that phagocytosis of killed

    Nm is critical for IL-12 production (Uronen-Hansson

    Fig. 5. Distinct DC phenotype response tolive Nm requires contact between bacteriaand DC. DCs cultured in the lower chamberof a transwell were separated from thebacteria (moi 100) added to the upperchamber by a membrane with 0.1 mm pores.This was compared with DC responses in theabsence of transwell, but otherwise identicalculture conditions (A). In parallel with thisexperiment DCs were stimulated with the

    supernatant equivalent to that released frommoi 100 of live and killed Nm (B). After 18 hexpression of HLA-Class I on DCs wasdetermined by flow cytometry. Data arerepresentative of three experiments from

    three different donors yielding similar results.

    100 101 102 103 104

    FL1-H

    0

    50

    Events

    100 101 102 103 104

    FL1-H

    0

    50

    Events

    100 101 102 103 104

    FL1-H

    0

    50

    Events

    100 101 102 103 104

    FL1-H

    0

    50

    Events

    100 101 102 103 104

    FL1-H

    0

    50

    Events

    100 101 102 103 104

    FL1-H

    0

    50

    Events

    HLA-Class I expression

    No transwell Transwell Bacterial

    Supernatant

    A B

    Live

    Killed

    Isotype control

    Unstimulated

    Stimulated

    100 101 102 103 104

    FL1-H

    0

    80

    Events

    100 101 102 103 104

    FL1-H

    0

    80

    Events

    100 101 102 103 104

    FL1-H

    0

    80

    Events

    100 101 102 103 104

    FL1-H

    0

    80

    Events

    100 101 102 103 104

    FL1-H

    0

    80

    Events

    100 101 102 103 104

    FL1-H

    0

    80

    Events

    Isotype controlUnstimulated

    Stimulated

    CD40 HLA-DR

    Live

    Killed

    Live

    lysate

    Fig. 6. Live Nm lysates are potent stimulators of DC maturation.Nm lysates were prepared with 0.05% NP40 detergent(Experimental procedures). DCs were stimulated with live Nmlysate equivalent to an moi of 10. After 18 h expression of CD40and HLA-DR was compared with levels induced by live and killedNm (moi 10) by flow cytometry. NP40 detergent alone had no effecton DC maturation (data not shown).

    Dendritic cell response to Neisseria meningitidis 2861

    2007 The AuthorsJournal compilation 2007 Blackwell Publishing Ltd, Cellular Microbiology, 9, 28562869

  • 8/6/2019 meningescesar.x

    7/14

  • 8/6/2019 meningescesar.x

    8/14

    protein (eGFP). The difference in percentage of DCs

    associated with bacteria were clearly reduced in live DC

    co-culture compared with those exposed to killed bacteria

    (Fig. 8B). These data further confirm that results with the

    FITC-labelled bacteria are valid for the time period (06 h)

    studied.

    We confirmed our flow cytometric analysis of phagocy-

    tosis by using confocal laser scanning microscopy

    (Uronen-Hansson et al., 2004a). Figure 9 shows DC incu-

    bated with killed (Fig. 9A) and live (Fig. 9B) bacteria after

    4 h of incubation. Internalized bacteria were assessed by

    examining optical sections using ALEXA 586-conjugated

    HLA-DR antibody as a counterstain. To distinguish

    between internalized and surface-bound bacteria, a differ-

    ential antibody staining technique with antibody specific for

    porin A serosubtype 1.7 was used to detect if bacteria were

    bound at the surface. In these experiments, surface-bound

    bacteria were FITC positive and stained with ALEXA 568-

    conjugated porin antibody. They appeared as red or yellow

    if overlay was used whereas internalized bacteria appear

    green only, as the cells were not permeabilized and the

    bacteria were inaccessible to antibody binding.

    IL-12 p70 and IL-10 production by DC in response to

    live and killed Nm are dependent upon phagocytosis

    The results presented in the previous section initially ap-

    peared at odds with our previous work (Uronen-Hansson

    Fig. 9. Nm internalization by DCs. DCs wereincubated with either killed (A) or live (B)FITC-labelled Nm for 1, 2, 4 and 6 h (4 h timepoint shown here). Cells were allowed toadhere to adhesion slides and then stainedwith either anti-HLA-DR antibody (i), orTo-Pro3 to stain the nucleus (ii), or acombination of To-Pro3 andanti-meningococcal serosubtype P1.7antibody followed by Alexa Fluor 568 goat

    anti-mouse IgG (H+L) (iii) and then visualizedby scanning laser confocal microscopy.

    Dendritic cell response to Neisseria meningitidis 2863

    2007 The AuthorsJournal compilation 2007 Blackwell Publishing Ltd, Cellular Microbiology, 9, 28562869

  • 8/6/2019 meningescesar.x

    9/14

    et al., 2004a) as we would expect that high IL-12 produc-

    tion would be associated with efficient phagocytosis of

    meningococci. Our previous studies with killed bacteria

    showed that IL-12 production could be abrogated when

    experiments were performed in the presence of cytocha-

    lasin D, a potent inhibitor of phagocytosis. One possibility

    was that in contrast to what occurs with killed bacteria,

    IL-12 production in response to live bacteria may not be

    dependent on internalization. However, as Fig. 10 clearly

    demonstrates, blocking internalization by adding cytocha-

    lasin D prior to co-culture with live bacteria and DCs

    completely abrogated IL-12 p70 production. In addition, we

    also found complete inhibition of IL-10 production in

    response to both live and killed Nm by cytochalasin D.

    Interestingly, cytochalasin D has not effect on IL-6 and

    TNF-a production and DC maturation profiles for menin-

    gococcal stimulated DCs (Uronen-Hansson et al., 2004a).

    These data show that internalization of live bacteria is

    required for IL-12 and IL-10 production and confirm that

    phagocytosis of Nm is required for production of these two

    important cytokines but is not essential for DC maturation.

    Blocking exogenous IL-10 activity restores IL-12 p70

    production by DCs to killed Nm

    The reason behind the disparity between IL-12 and IL-10

    production in DCs in response to live and killed meningo-

    cocci required clarification. Given the dependence of

    phagocytosis for the generation of both of these cytokines

    to both killed and live bacteria, we postulated that IL-12

    production may be reduced by activity of exogenouslyreleased IL-10. To test this possibility, DCs were pre-

    incubated with a blocking antibody to IL-10 prior to

    co-culture with DCs and killed bacteria. Levels of detect-

    able IL-10 in supernatants from stimulated DCs were

    reduced to near baseline levels (Fig. 11). Blocking the

    activity of endogenously released IL-10 was found to

    restore IL-12 production by DCs in response to both killed

    and live bacteria (Fig. 11). No effect was seen when a

    control antibody was added. We conclude that the differ-

    ential production of IL-12 in response to live and killed

    bacteria may be to a large part controlled by the action of

    released IL-10. As IL-10 production in response to killed

    0

    2000

    4000

    6000

    8000

    10000

    12000

    Medium

    Medium+

    CD

    Live

    Live+CD

    Killed

    Killed+CD

    IL-10pgml1

    0

    500

    1000

    1500

    2000

    2500

    3000

    3500

    Medium

    Medium+

    CD

    Live

    Live+CD

    Killed

    Killed+CD

    IL-12pgml1

    Fig. 10. Blocking phagocytosis inhibits production of IL-12 p70 andIL-10 by human DC in response to both live and killed Nm. DCswere incubated with live or killed Nm (moi 100) with or without10 mg ml-1 cytochalasin D (CD). After 18 h supernatants wereharvested and analysed for IL-10 and IL-12 by ELISA. Results areexpressed as the mean and standard error of the mean of fourseparate experiments.

    0

    2000

    4000

    6000

    8000

    10000

    12000

    Cells CellsIC

    CellsAb

    Live LiveIC

    LiveAb

    KilledKilledIC

    KilledAb

    1L-10pgml1

    0Cells Cells

    IC

    Live Live

    IC

    Live

    Ab

    Killed Killed

    IC

    Killed

    Ab

    IL-12pgml1

    8000

    7000

    6000

    5000

    4000

    3000

    2000

    1000

    Cells

    +Ab

    Fig. 11. Cytokine production in DCs following treatment with anIL-10 blocking antibody. DCs were stimulated with killed or live(moi 100) Nm for 18 h with or without mouse anti-human IL-10antibody (Ab) or mouse IgG2b isotype control (IC) at 30 mg ml-1.Supernatants were then collected and analysed for IL-10 and IL-12by ELISA. Results are expressed as the mean and standard errorof the mean of three separate experiments.

    2864 H. E. Jones et al.

    2007 The AuthorsJournal compilation 2007 Blackwell Publishing Ltd, Cellular Microbiology, 9, 28562869

  • 8/6/2019 meningescesar.x

    10/14

    bacteria is higher than in response to live bacteria, the

    effect seen on IL-12 production stimulated by killed Nm is

    greater.

    Discussion

    Dendritic cells provide a critical link between the innate and

    adaptive immune response. What has emerged in recent

    years is the requirement for coordinated antigen presenta-

    tion, together with signals via the action of cytokines and

    chemokines and receptor cross-talk between APC and

    T-cells to achieve optimal adaptive immune responses

    (Banchereau et al., 2000). It is increasingly clear that

    engagement of microbial products such as LPS, with host

    pathogen recognition systems, predominantly TLRs, dic-

    tates these signals (Kapsenberg, 2003; Pulendran and

    Ahmed, 2006). There is evidence that perturbations in

    these signals can have important and lasting conse-

    quences for the nature of adaptive immune responses to

    microbes and vaccine components derived from them. It is

    noteworthy that certain vaccine preparations that havesuboptimal immunogenicity in vivoalso lack the capacity to

    stimulate DCs in vitro (Skowera et al., 2005). In contrast,

    the success of some vaccines appears to be as a result of

    their capacity to engage multiple TLR ligands on DCs

    (Querec et al., 2006). Further understanding on how Nm

    interacts with DCs in terms of induction of co-stimulation

    molecules and cytokines provides a logical strategy for

    improving the efficacy of vaccines against Nm.

    Our previous work has shown that killed Nm are an

    effective way to target DCs because they are readily

    phagocytosed. We have also shown that expression of

    LPS on Nm is essential for uptake and subsequent cytok-ine production by DCs (Uronen-Hansson et al., 2004a).

    This current study adds an extra dimension to the story as

    it shows that, even taking into account the presence of

    adjuvant properties of meningococcal LPS, viable dividing

    bacteria behave differently in their capacity to stimulate

    DCs compared with killed bacteria and even viable bac-

    teria that are not dividing. Despite the fact that live men-

    ingococci are able to induce cytokine production, they

    appear remarkably inefficient at inducing increased

    expression of DC maturation markers. To our knowledge,

    this has not been demonstrated before. These data raise

    the possibility that Nm may have the capacity to interfere

    with or modulate DC function. This is supported by data

    presented in this study that show the phenotypic response

    to live meningococci is dominant over the normally potent

    stimulus of maturation.

    The finding that Nm can modulate DC function is not

    without president in other organisms. For example, a

    number of pathogens appear to be able to target and

    interfere with DC function resulting in ineffective cellular

    and humoral immunity. Microbial pathogens achieve this

    via a number of different mechanisms including direct

    cytotoxicity or specific interference of DC signalling path-

    ways required for effective cytokine responses or matu-

    ration (Urban et al., 1999; Agrawal et al., 2003; Skinner

    et al., 2004; Tobar et al., 2004; Marketon et al., 2005).

    It is not known whether the lack of upregulation of cell

    surface markers in DCs in response to live Nm shown in

    this study is likely to have any effect on subsequent T-cell

    responses. Co-stimulatory molecules are still expressed

    on the DCs exposed to live bacteria, albeit at a lower level

    than seen in response to killed organisms, and this may

    still allow for efficient interactions of DC with T-cell.

    However, there is evidence from other work that it is not

    only the expression of these molecules (e.g. CD40 and

    CD86) but also the density of these receptors that can

    have important effects upon DCT-cell interactions.

    In addition to the lack of co-stimulatory molecule

    induction, there was only modest or no expression of

    HLA-Class I and HLA-DR on DCs stimulated by live bac-

    teria. While the significance of this remains to be eluci-

    dated, this could influence the nature of the DCT-cellsynapse, as antigen density is a determinant of T-cell

    polarization (Langenkamp et al., 2000). Further work in

    our laboratory is investigating whether efficiency of

    antigen presentation is also reduced in DC stimulated

    with live meningococci.

    One important matter raised by these data is: what are

    the mechanisms that may be operating that result in the

    poor maturation response? We have shown that viable but

    non-proliferating (cm treated) Nm induce a DC phenotype

    similar to that induced by killed Nm. As cm rapidly blocks

    bacterial protein synthesis, there is a possibility that men-

    ingococci, upon contact with DC, increase expression offactors that may directly interfere with processes leading to

    increase in expression of maturation markers on DC.

    However, the finding that lysates of live bacteria are

    stimulatory to DCs does not support the notion that

    over-production of bacterial products alone is responsible

    for the lack of maturation seen with live bacteria.

    As the effect of live bacteria requires contact and is

    dominant over normally potent maturation signal such as

    killed bacteria or LPS, we propose that the likely mecha-

    nism is an active process involving a functional surface

    expressed component of meningococci. We cannot rule

    out the activity of molecules locally released by either

    bacteria or DCs in response to live infection. However,

    as neither supernatants from bacteria alone (seen in

    Fig. 5 in this study) nor indeed supernatants from DC

    live bacterial co-cultures (H. Jones, pers. comm.) display

    this inhibitory/dominant effect on DC maturation we feel

    this is unlikely. There are a number of potential menin-

    gococcal candidate genes that could potentially affect

    host cell function. Work is in progress to identify genes

    and factors that might be suppressing phenotypic

    Dendritic cell response to Neisseria meningitidis 2865

    2007 The AuthorsJournal compilation 2007 Blackwell Publishing Ltd, Cellular Microbiology, 9, 28562869

  • 8/6/2019 meningescesar.x

    11/14

    expression of DC co-stimulatory molecules and HLA-

    Class I and HLA-DR. Clearly, the identity of such factors

    would be advantageous as these could potentially be

    deleted or modified in attenuated strains that could be

    used in novel vaccines preparations.

    This study focused on the DC response to one strain of

    wild-type capsulated bacteria (H44/76). However, work

    carried out by two of the authors (G.L.J.D., H.E.J.)

    showed that serogroup B strains B1940 and MC58 have

    identical disparity of DC maturation responses between

    live and killed organisms. Furthermore, we have not found

    any influence of capsulation or modification of LPS struc-

    ture on the distinct maturation phenotype of DCs, as

    mutants lacking these components behave the same way

    as wild-type strains (G.L.J. Dixon, unpubl. data).

    The other clear difference between live and killed bac-

    teria shown in these studies is the difference in rate of

    their internalization by human DC. Other workers have

    shown that live Nm are internalized by DC, and this is also

    affected by capsulation status and LPS oligosaccharide

    structure (Kurzai et al., 2005). Most of these studies wereperformed in serum-low or even serum-free conditions,

    and therefore differ to our studies which do include serum.

    We have previously shown that in the absence of serum

    factors, uptake (presumably by non-opsonic mecha-

    nisms) does occur at a low level but this greatly enhanced

    by both serum and specifically by lipopolysaccharide-

    binding protein (LBP) (Uronen-Hansson et al., 2004a).

    Accordingly, one likely hypothesis is that differences in

    uptake between live and killed organisms are due to inef-

    ficiency of receptor engagement. It is also likely that this is

    dependent on the presence of serum factors such as LBP,

    soluble CD14 and RGD motif containing factors such asvitronectin and fibronectin. At present it is unclear which

    receptors are critical for uptake of Nm by DC, but under

    non-opsonic conditions there is indirect evidence that

    scavenger receptors support internalization of unencap-

    sulated Nm into DC (Kurzai et al., 2005). Recent work we

    have performed has suggested that b2-integrins, involving

    LBP as an opsonin, may facilitate uptake of killed Nm (our

    manuscript in preparation). It would be interesting to see

    whether this pathway operates efficiently with respect to

    live bacteria uptake. The differences between rate and

    efficiency of uptake of killed Nm compared with live and

    the difference in cytokine responses may be linked. We

    show that both IL-10 and IL-12 production is dependent

    upon bacterial uptake by DC. However, we also show that

    that IL-10 release by DC reduces IL-12 production, and

    this may help to explain the unexpected finding that killed

    bacteria, which are phagocytosed more efficiently than

    live, produce less IL-12. While increase in effective dose

    of live bacteria is a potential confounding factor in these

    experiments, this cannot explain the increase in potency

    of live bacteria for inducing IL-12 production in DCs, as

    uptake of live bacteria is consistently far lower than that of

    killed bacteria even in the presence of increased bacteria

    division in live cultures.

    It is not known what are the consequences of the dif-

    ferent profiles of IL-10 and IL-12 p70 induced by live and

    killed meningococci on T-cell responses. It is generally

    accepted that Gram-negative bacteria such as Nm are

    potent inducers of T-helper type 1 responses (de Jong

    et al., 2002) and the likely reason for this is the propensity

    of LPS in these bacteria to induce production of the IL-12

    family of cytokines by DCs (Trinchieri, 1993; Pulendran

    et al., 2001). Production of IL-12 by DCs is responsible for

    multiple effects on lymphocytes, in particular promoting

    IFN-g production by CD4 cells, favouring B-cell differen-

    tiation and isotype switching to Th1-associated immuno-

    globulin IgG2, considered to be most effective isotype

    against polysaccharide antigens (Trinchieri, 1998). In con-

    trast, IL-10 released by DCs exerts other effects, such as

    the class switching of IgD bearing B-cells to produce IgG1

    and IgG3 antibody (Briere et al., 1994). This is pertinent

    as bactericidal antibodies against the subcapsular proteinantigens such as porA, induced by both OMV vaccines

    and after natural infection, are of IgG1 and IgG3 isotypes

    (Sjursen et al., 1990; Naess et al., 1999; Martin et al.,

    2001). Although IL-10 is considered an anti-inflammatory

    cytokine (Igietseme et al., 2000) it may have effects that

    could influence the T-cell priming capacity of meningococ-

    cal pulsed DCs. For example, IL-10 promotes DC survival,

    and this may be important for maintaining contact

    between DC and Ag-specific T-cell.

    In this article, we present data that show live meningo-

    cocci induce different pattern of maturation/activation

    from that seen with dead bacteria. The biological conse-quences of these different DC phenotypes will become

    apparent when tested in a system that assesses the

    effects on meningococcal stimulated DCs on antigen-

    specific T-cell responses and subsequent effects on B-cell

    function. Our findings may be important for meningococ-

    cal vaccine development.

    Experimental procedures

    Bacteria

    The Nm strain used in this study H44/76 (B:15:P1.7,16:) is

    encapsulated and piliated and expresses L3 immunotype (Holten

    et al., 1979). Bacteria were grown on GC agar base (BD Bio-

    sciences, Oxford, UK) supplemented with 1% Vitox (Oxoid) and

    cultured in 5% CO2 in air at 36C. Bacteria were grown overnight

    (18 h) and then suspended in RPMI 1640 medium without phenol

    red (Invitrogen). Optical density was measured at 540 nm and a

    suspension at OD 1.0 was determined to contain 1 109 bacte-

    ria ml-1 by serial dilution and plating using standard microbiologi-

    cal techniques. Bacteria were fixed in 0.5% paraformaldehyde for

    20 min and then washed extensively. For fluorescent labelling,

    live Nm were incubated with 0.5 mg ml-1 FITC (Sigma, Poole,

    2866 H. E. Jones et al.

    2007 The AuthorsJournal compilation 2007 Blackwell Publishing Ltd, Cellular Microbiology, 9, 28562869

  • 8/6/2019 meningescesar.x

    12/14

    UK) for 20 min at 37C and then an aliquot was taken and then

    killed as described above. The level of FITC incorporation was

    confirmed by flow cytometry. In previous optimization experi-

    ments, method of killing bacteria (UV irradiation, heat treatment

    for 56C for 30 min) does not alter the pattern of DC activation or

    cytokine production (H. Uronen-Hansson, pers. comm.). To gen-

    erate bacteria expressing green fluorescent protein, wild-type

    bacteria were transfected with plasmid containing enhanced

    green fluorescence protein under PorA promoter (Christodoul-

    ides et al., 2000). This plasmid was modified by replacement of

    ampicillin resistance cassette with ermC gene conferring resis-

    tance to erythromycin (kind gift from Peter van der Lay, NVI,

    Bilthoven, the Netherlands). Successful transfectants were

    screened for fluorescence and subcultured onto GC agar plates

    containing 50 mg ml-1 erythromycin. Level of fluorescence was

    checked by flow cytometry and immunofluorescence microscopy.

    For studies using cm (clinical grade obtained from Pharmacy,

    Great Ormond Street Hospital, UK), optimization of time kill

    curves was performed in our tissue culture system. At doses of

    2.510 mg ml-1, and at concentrations over 105-107 colony-

    forming units (cfu) ml-1, bacterial concentration remained static

    over incubation period of up to 18 h, as determined by serial

    dilution and plating and quantifying bacteria after further over-

    night incubation on GC agar plates.

    For preparation of bacterial lysates 2 108 cfu ml-1 were sus-

    pended in 0.05% NP40 detergent (Sigma) in PBS and mixed

    rigorously for 15 min. Cellular debris was removed by centrifuga-

    tion and 0.2 mm filtration. Lysis of live bacteria was confirmed by

    plating lysates on GC agar overnight.

    Dendritic cell culture and activation

    Dendritic cells were generated from human peripheral blood

    mononuclear cells (PBMCs) as described previously (Sallusto

    and Lanzavecchia, 1994; Dixon et al., 2001) but with some

    modifications. Briefly, monocytes were prepared from PMBCs

    using positive selection using CD14 immunomagnetic beads

    (Miltenyi Biotec, Surrey, UK). CD14-isolated cells were then cul-

    tured in RPMI supplemented with 10% FCS, 2.4 mM L-glutamine

    (all from Invitrogen), 100 ng ml-1 human recombinant GM-CSF

    and 50 ng ml-1 human recombinant IL-4 (obtained from Professor

    Benjamin Chain). DCs were used after 57 days of culture and

    phenotype was determined by flow cytometry (FACS calibur BD

    Biosciences, Oxon, UK). Immature DCs were CD3-negative,

    CD14-low, CD19-negative, CD83-negative, CD25-negative and

    expressed low levels of HLA-DR, HLA-DQ, HLA-Class 1, CD40,

    CD86 and CD1 as previously described (Dixon et al., 2001).

    For stimulation experiments, DCs (5 105 ml-1) were cultured

    with live or killed Nm at an moi of 100, 10 or 1 for 1824 h. In

    some experiments, 2.510 mg ml-1 cm was added to the DC

    bacteria co-culture either at the start of stimulation or at various

    time points after addition of bacteria, medium or LPS. Prior to

    staining, an aliquot of DC culture was stained with trypan blue

    (Sigma Aldrich, Poole, UK) to assess the proportion of cell death

    in co-cultures using a haemocytometer.

    For transwell experiments, DCs were added to 24-well tissue

    culture plates at a density of 5 105 cells ml-1. The 0.1 mm pore

    size inserts were placed in contact with media and the bacteria

    were added to the top chamber. After overnight culture, DCs were

    removed and cell surface markers analysed as described below.

    Lack of any ingress of live meningococci to the lower chamber

    was proven by culturing an aliquot of the stimulated cells on GC

    agar.

    Surface marker expression and cytokine measurement

    Expression of surface molecules associated with DC maturation

    and activation (CD40, CD83, CD86, HLA-Class 1 and HLA-DR)

    was detected by staining with 2 mg ml-1 of the appropriate FITC-or phycoerythrin (PE)-conjugated monoclonal antibodies (all

    Caltag Medsystems, Silverstone, UK) for 30 min on ice. Follow-

    ing incubation, cells were washed with phosphate-buffered saline

    (PBS) with 0.2% bovine serum albumin (BSA) and then fixed in

    1% formaldehyde. Cells were then analysed by flow cytometry on

    a FACScalibur using CellQuest Pro software (BD Biosciences

    Oxford, UK). DCs form a discrete population when separated by

    side and forward scatter parameters; this population formed the

    collection gate and at least 5000 events within this gate were

    collected for analysis.

    Cytokine (IL-6, IL-1b, TNF-a, IL-12 p70 and IL-10) production

    in cell culture supernatants of co-culture experiments was carried

    out by eBioscience ELISA kits (Insight) following the manufactur-

    ers instructions. Lower limit of detection of IL-12 and IL-10 are4 pg ml-1 and 2 pg ml-1 respectively.

    Phagocytosis assay

    The association of Nm with DCs was determined by flow cytom-

    etry and confocal microscopy. DCs were incubated with FITC-

    labelled Nm and then aliquots of cells taken at various time

    points, as previously described (Uronen-Hansson et al., 2004a).

    Cells were separated into two aliquots. One was washed exten-

    sively with PBS and 0.2% BSA, fixed with 1% formaldehyde then

    analysed by flow cytometry. DC and Nm association was shown

    by the expression FITC within the DC-gated population. To facili-

    tate gating, DCs were counterstained with phycoerythrin-conjugated DC-SIGN antibody, which is expressed highly on DCs

    regardless of maturation status. In some experiments, DC asso-

    ciation with meningococci was judged by per cent positive events

    and mean fluorescence intensity in DC-SIGNbright events. Using

    this method, a clear distinction between DC associated and non-

    associated with bacteria could be achieved. Inhibition of phago-

    cytosis was achieved using the inhibitor of actin polymerization

    cytochalasin D (Sigma) at 10 mg ml-1, which was added to DCs

    just prior to incubation with Nm. In optimization experiments,

    addition of cytochalasin D prior to co-culture with fluorescent

    bacteria reduced bacteria fluorescence to near background

    levels, showing that the majority of FITC/GFP-positive events are

    due to bacteria that have been internalized by DC.

    The remaining aliquot of cells were washed with PBS and then

    allowed to adhere to poly lysine-coated slides for 10 min (Paul

    Marienfeld Gmbh and CO, Germany). Cells were then fixed with

    4% paraformaldehyde. In some experiments, DCs were visual-

    ized by either staining the nuclei with To-Pro3 (Molecular Probes,

    Cambridge Biosciences, Cambridge, UK) or staining the DC

    surface with 5 mg ml-1 anti-HLA-DR antibody (Dako, Glostrup,

    Denmark) followed by Alexa Fluor 568 goat anti-mouse IgG

    (Molecular Probes). To determine if Nm was internalized or

    surface bound, extracellular bacteria were identified by staining

    with 10 mg ml-1 P1.7 antibodies specific for Nm (NIBSC, South

    Dendritic cell response to Neisseria meningitidis 2867

    2007 The AuthorsJournal compilation 2007 Blackwell Publishing Ltd, Cellular Microbiology, 9, 28562869

  • 8/6/2019 meningescesar.x

    13/14

    Mimms, UK) followed by Alexa Fluor 568 goat anti-mouse IgG

    (Invitrogen Molecular Probes, Paisley, UK). Slides were then

    washed and mounted with Vectashield (Vector laboratories,

    Peterborough, UK) and then images were obtained using a Leica

    SP2 confocal laser scanning microscope system (Leica, Milton

    Keynes, UK). To identify internalized bacteria at least 10 optical

    sections (0.20.5 mm) spanning the entire DC were visualized by

    Leica confocal imaging software.

    IL-10 inhibition studies

    Prior to incubation with bacteria, DCs were incubated with

    30 mg ml-1 anti-IL-10 blocking antibody (R&D Systems, Oxon,

    UK) for 10 min.

    Viability

    The percentage of viable cells was assessed by trypan blue

    staining of an aliquot of cells prior to fixation using a

    haemocytometer. In all culture conditions, a proportion of cells

    (ranging from 5% to 15%) were trypan blue positive. However,

    there was no difference observed in the proportion in culturesstimulated with medium, live or killed Nm. The proportion of DCs

    undergoing apoptosis was measured by the detection of active

    caspases using a Vybrant FAM Poly Caspase Assay Kit

    (Molecular probes) according to manufacturers instructions.

    Statistical analysis

    Where shown, statistical analysis was performed using SPSS

    version 12.01 software. Due to variability of biological responses

    (such as individual variation of cytokine responses to microbial

    derived products and the arbitrary unit of fluorescent intensities

    derived from flow cytometric analysis) paired t-tests were per-

    formed on individual donor-derived DC responses to live and

    killed bacteria. The number of donors studied allowed for para-

    metric testing to be performed.

    Acknowledgement

    This work was supported by Meningitis UK.

    References

    Agrawal, A., Lingappa, J., Leppla, S.H., Agrawal, S., Jabbar,

    A., Quinn, C., and Pulendran, B. (2003) Impairment of

    dendritic cells and adaptive immunity by anthrax lethal

    toxin. Nature 424: 329334.

    Al Bader, T., Christodoulides, M., Heckels, J.E., Holloway, J.,

    Semper, A.E., and Friedmann, P.S. (2003) Activation ofhuman dendritic cells is modulated by components of the

    outer membranes of Neisseria meningitidis. Infect Immun

    71: 55905597.

    Al Bader, T., Jolley, K.A., Humphries, H.E., Holloway, J.,

    Heckels, J.E., Semper, A.E., et al. (2004) Activation of

    human dendritic cells by the PorA protein of Neisseria

    meningitidis. Cell Microbiol 6: 651662.

    Banchereau, J., and Steinman, R.M. (1998) Dendritic cells

    and the control of immunity. Nature 392: 245252.

    Banchereau, J., Briere, F., Caux, C., Davoust, J., Lebecque,

    S., Liu, Y.J., et al. (2000) Immunobiology of dendritic cells.

    Annu Rev Immunol 18: 767811.

    Bjune, G., Hoiby, E.A., Gronnesby, J.K., Arnesen, O., Fre-

    driksen, J.H., Halstensen, A., et al. (1991) Effect of outer

    membrane vesicle vaccine against group B meningococcal

    disease in Norway [see comments]. Lancet 338: 1093

    1096.

    Briere, F., Servet-Delprat, C., Bridon, J.M., Saint-Remy, J.M.,

    and Banchereau, J. (1994) Human interleukin 10 induces

    naive surface immunoglobulin D+ (sIgD+) B cells to secrete

    IgG1 and IgG3. J Exp Med 179: 757762.

    Cartwright, K., Morris, R., Rumke, H., Fox, A., Borrow, R.,

    Begg, N., et al. (1999) Immunogenicity and reactogenicity

    in UK infants of a novel meningococcal vesicle vaccine

    containing multiple class 1 (PorA) outer membrane

    proteins. Vaccine 17: 26122619.

    Christodoulides, M., Everson, J.S., Liu, B.L., Lambden, P.R.,

    Watt, P.J., Thomas, E.J., and Heckels, J.E. (2000) Inter-

    action of primary human endometrial cells with Neisseria

    gonorrhoeae expressing green fluorescent protein. Mol

    Microbiol 35: 3243.

    Dixon, G.L., Newton, P.J., Chain, B.M., Katz, D., Andersen,

    S.R., van der Wong, S., et al. (2001) Dendritic cell activa-

    tion and cytokine production induced by group B Neisseria

    meningitidis: interleukin-12 production depends on

    lipopolysaccharide expression in intact bacteria. Infect

    Immun 69: 43514357.

    Finne, J., Leinonen, M., and Makela, P.H. (1983) Antigenic

    similarities between brain components and bacteria

    causing meningitis. Implications for vaccine development

    and pathogenesis. Lancet 2: 355357.

    Giuliani, M.M., Adu-Bobie, J., Comanducci, M., Arico, B.,

    Savino, S., Santini, L., et al. (2006) A universal vaccine for

    serogroup B meningococcus. Proc Natl Acad Sci USA 103:

    1083410839.

    Harrison, L.H. (2006) Prospects for vaccine prevention of

    meningococcal infection. Clin Microbiol Rev 19: 142164.Holten, E. (1979) Serotypes of Neisseria meningitidisisolated

    from patients in Norway during the first six months of 1978.

    J Clin Microbiol 9: 186188.

    Igietseme, J.U., Ananaba, G.A., Bolier, J., Bowers, S.,

    Moore, T., Belay, T., et al. (2000) Suppression of endog-

    enous IL-10 gene expression in dendritic cells enhances

    antigen presentation for specific Th1 induction: potential for

    cellular vaccine development. J Immunol164: 42124219.

    Jodar, L., Feavers, I.M., Salisbury, D., and Granoff, D.M.

    (2002) Development of vaccines against meningococcal

    disease. Lancet 359: 14991508.

    de Jong, E.C., Vieira, P.L., Kalinski, P., Schuitemaker, J.H.,

    Tanaka, Y., Wierenga, E.A., Yazdanbakhsh, M., and

    Kapsenberg, M.L. (2002) Microbiol compounds selectivelyinduce Th1 cell-promoting or Th2 cell-promoting dendritic

    cells in vitro with diverse Th cell-polarizing signals.

    J Immunol 168: 17041709.

    Kapsenberg, M.L. (2003) Dendritic-cell control of pathogen-

    driven T-cell polarization. Nat Rev Immunol 3: 984993.

    Kikuchi, T., Kobayashy, T., Gomi, K., Suzuki, T., Takue, Y.,

    Watanabe, A., and Nukiwa, T. (2004) Dendritic cells pulsed

    with live and dead Legionella pneumophila elicit distinct

    immune responses. J Immunol 172: 17271734.

    2868 H. E. Jones et al.

    2007 The AuthorsJournal compilation 2007 Blackwell Publishing Ltd, Cellular Microbiology, 9, 28562869

  • 8/6/2019 meningescesar.x

    14/14

    Kurzai, O., Schmitt, C., Claus, H., Vogel, U., Frosch, M., and

    Kolb-Maurer, A. (2005) Carbohydrate composition of men-

    ingococcal lipopolysaccharide modulates the interaction of

    Neisseria meningitidis with human dendritic cells. Cell

    Microbiol 7: 13191334.

    Langenkamp, A., Messi, M., Lanzavecchia, A., and Sallusto,

    F. (2000) Kinetics of dendritic cell activation: impact on

    priming of TH1, TH2 and nonpolarized T cells. Nat Immunol

    1: 311316.

    Marketon, M.M., DePaolo, R.W., DeBord, K.L., Jabri, B., and

    Schneewind, O. (2005) Plague bacteria target immune

    cells during infection. Science 309: 17391741.

    Martin, S., Sadler, F., Borrow, R., Dawson, M., Fox, A., and

    Cartwright, K. (2001) IgG antibody subclass responses

    determined by immunoblot in infants sera following vacci-

    nation with a meningococcal recombinant hexavalent PorA

    OMV vaccine. Vaccine 19: 44044408.

    Naess, L.M., Aarvak, T., Aase, A., Oftung, F., Hoiby, E.A.,

    Sandin, R., and Michaelsen, T.E. (1999) Human IgG sub-

    class responses in relation to serum bactericidal and

    opsonic activities after immunization with three doses of

    the Norwegian serogroup B meningococcal outer mem-

    brane vesicle vaccine. Vaccine 17: 754764.

    Pizza, M., Scarlato, V., Masignani, V., Giuliani, M.M., Arico,

    B., Comanducci, M., et al. (2000) Identification of vaccine

    candidates against serogroup B meningococcus by whole-

    genome sequencing. Science 287: 18161820.

    Pulendran, B., Kumar, P., Cutler, C.W., Mohamadzadeh, M.,

    Van Dyke, T., and Banchereau, J. (2001) Lipopolysaccha-

    rides from distinct pathogens induce different classes of

    immune responses in vivo. J Immunol 167: 50675076.

    Pulendran, B. (2005) Variegation of the immune response

    with dendritic cells and pathogen recognition receptors.

    J Immunol 174: 24572465.

    Pulendran, B., and Ahmed, R. (2006) Translating innate

    immunity into immunological memory: implications for

    vaccine development. Cell 124: 849863.

    Querec, T., Bennouna, S., Alkan, S., Laouar, Y., Gorden, K.,Flavell, R., et al. (2006) Yellow fever vaccine YF-17D acti-

    vates multiple dendritic cell subsets via TLR2, 7, 8, and 9 to

    stimulate polyvalent immunity. J Exp Med 203: 413

    424.

    Rey-Ladino, J., Koochesfahani, K.M., Zaharik, M.L., Shen,

    C., and Brunham, R.C. (2005) A live and inactivated

    Chlamydia trachomatis mouse pneumonitis strain induces

    he maturation of dendritc cells that are phenotypically

    and immunologically distinct. Infect Immun 73: 1568

    1577.

    Sallusto, F., and Lanzavecchia, A. (1994) Efficient presen-

    tation of soluble antigen by cultured human dendritic

    cells is maintained by granulocyte/macrophage colony-

    stimulating factor plus interleukin 4 and downregulated bytumour necrosis factor alpha. J Exp Med 179: 11091118.

    Singleton, T.E., Massari, P., and Wetzler, L.M. (2005) Neis-

    serial porin-induced dendritic cell activation is MyD88 and

    TLR2 dependent. J Immunol 174: 35453550.

    Sjursen, H., Wedege, E., Rosenqvist, E., Naess, A.,

    Halstensen, A., Matre, R., and Solberg, C.O. (1990) IgG

    subclass antibodies to serogroup B meningococcal

    outer membrane antigens following infection and vaccin-

    ation. APMIS 98: 10611069.

    Skinner, J.A., Reissinger, A., Shen, H., and Yuk, M.H. (2004)

    Bordetella type III secretion and adenylate cyclase toxin

    synergize to drive dendritic cells into a semimature state.

    J Immunol 173: 19341940.

    Skowera, A., de Jong, E.C., Schuitemaker, J.H., Allen, J.S.,

    Wessely, S.C., Griffiths, G., et al. (2005) Analysis of

    anthrax and plague biowarfare vaccine interactions with

    human monocyte-derived dendritic cells. J Immunol 175:

    72357243.

    Tappero, J.W., Lagos, R., Ballesteros, A.M., Plikaytis, B.,

    Williams, D., Dykes, J., et al. (1999) Immunogenicity of 2

    serogroup B outer-membrane protein meningococcal

    vaccines: a randomized controlled trial in Chile

    [see comments]. JAMA 281: 15201527.

    Tobar, J.A., Gonzalez, P.A., and Kalergis, A.M. (2004) Sal-

    monella escape from antigen presentation can be over-

    come by targeting bacteria to Fc gamma receptors on

    dendritic cells. J Immunol 173: 40584065.

    Trinchieri, G. (1993) Interleukin-12 and its role in the genera-

    tion of TH1 cells. Immunol Today 14: 335338.

    Trinchieri, G. (1998) Interleukin-12: a cytokine at the interface

    of inflammation and immunity. Adv Immunol 70: 83243.

    Underhill, D.M., and Ozinsky, A. (2002) Phagocytosis of

    microbes: complexity in action. Annu Rev Immunol 20:

    825852.

    Underhill, D.M., Ozinsky, A., Hajjar, A.M., Stevens, A.,

    Wilson, C.B., Bassetti, M., and Aderem, A. (1999) The

    Toll-like receptor 2 is recruited to macrophage phago-

    somes and discriminates between pathogens. Nature401:

    811815.

    Urban, B.C., Ferguson, D.J., Pain, A., Willcox, N., Plebanski,M., Austyn, J.M., and Roberts, D.J. (1999) Plasmodium

    falciparum-infected erythrocytes modulate the maturation

    of dendritic cells. Nature 400: 7377.

    Uronen-Hansson, H., Steeghs, L., Allen, J., Dixon, G.L., van

    der Osman, M., Ley, P., et al. (2004a) Human dendritic cell

    activation by Neisseria meningitidis: phagocytosis depends

    on expression of lipooligosaccharide (LOS) by the bacteria

    and is required for optimal cytokine production. Cell Micro-

    biol 6: 625637.

    Uronen-Hansson, H., Allen, J., Osman, M., Squires, G.,

    Klein, N., and Callard, R.E. (2004b) Toll-like receptor 2

    (TLR2) and TLR4 are present inside human dendritic cells,

    associated with microtubules and the Golgi apparatus but

    are not detectable on the cell surface: integrity of microtu-bules is required for interleukin-12 production in response

    to internalized bacteria. Immunology 111: 173178.

    Dendritic cell response to Neisseria meningitidis 2869

    2007 The AuthorsJournal compilation 2007 Blackwell Publishing Ltd, Cellular Microbiology, 9, 28562869