You are on page 1of 10

Journal of Cystic Fibrosis 17 (2018) 143 – 152

www.elsevier.com/locate/jcf

Review
Mechanisms of humoral immune response against Pseudomonas aeruginosa
biofilm infection in cystic fibrosis
Renan Marrichi Mauch a , Peter Østrup Jensen b , Claus Moser c ,
Carlos Emilio Levy a,d , Niels Høiby b,c, *
a
Department of Clinical Pathology, School of Medical Sciences, University of Campinas, Brazil
b
Department of International Health, Immunology and Microbiology, Faculty of Health and Medical Sciences, Panum Institute,
University of Copenhagen, Denmark
c
Department of Clinical Microbiology, Rigshospitalet (Copenhagen University Hospital), Denmark
d
Laboratory of Microbiology, Division of Clinical Pathology, Hospital de Clínicas (Campinas University Hospital), Brazil

Received 26 July 2017; revised 21 August 2017; accepted 22 August 2017


Available online 21 October 2017

Abstract

P. aeruginosa chronic lung infection is the major cause of morbidity and mortality in patients with cystic fibrosis (CF), and is characterized by a
biofilm mode of growth, increased levels of specific IgG antibodies and immune complex formation. However, despite being designed to combat
this infection, such elevated humoral response is not associated with clinical improvement, pointing to a lack of anti-pseudomonas effectiveness.
The mode of action of specific antibodies, as well as their structural features, and even the background involving B-cell production, stimulation and
differentiation into antibody-producing cells in the CF airways are poorly understood. Thus, the aim of this review is to discuss studies that have
addressed the intrinsic features of the humoral immune response and provide new insights regarding its insufficiency in the CF context.
© 2017 European Cystic Fibrosis Society. Published by Elsevier B.V. All rights reserved.

Keywords: Cystic fibrosis; Pseudomonas aeruginosa; Lung infection; Humoral immune response; Antibodies

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 144
2. B-cells, antibodies and the humoral immune response in CF . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 145
2.1. B-cells in CF . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 145
2.2. Functionality of antibody response in CF . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 147
2.2.1. Immune complex formation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 147
2.2.2. Opsonization capacity and phagocytic killing . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 147
2.2.3. Defective interaction among pathogen, antibodies and phagocytes . . . . . . . . . . . . . . . . . . . . . . . 148
2.2.4. Alginate: cause or consequence of the ineffective humoral response? . . . . . . . . . . . . . . . . . . . . . . 149
3. Concluding remarks . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 149
Conflict of interests . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 149
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 150

⁎ Corresponding author at: Rigshospitalet (Session 9301), Juliane Maries Vej, 2100 Copenhagen, Capital Region, Denmark.
E-mail address: hoiby@hoibyniels.dk (N. Høiby).

https://doi.org/10.1016/j.jcf.2017.08.012
1569-1993/© 2017 European Cystic Fibrosis Society. Published by Elsevier B.V. All rights reserved.
144 R.M. Mauch et al. / Journal of Cystic Fibrosis 17 (2018) 143–152

1. Introduction caused by the CFTR defects leads to changing chemical


characteristics of the ASL, where an acidified environment is
Pulmonary disease is the preponderant manifestation in formed, reducing or inactivating defensins, lysozymes and
cystic fibrosis (CF), where a dehydration in the airway surface lactoferrins, natural defense molecules of the primary airway
liquid (ASL) leads to mucus accumulation in the lower barrier, making bacterial killing even more difficult [4,5].
airways, impairing the mucociliary clearance (MCC) and Still, a number of studies in the last two decades have shown
facilitating colonization and infection of the lower airway that not only the primary immune barriers are impaired in the CF
environment by bacterial pathogens, especially Pseudomonas airways, but also the cellular and signaling artifacts of the immune
aeruginosa, the major morbidity and mortality cause in the CF system as a whole is involved [6]. An interesting fact is that such
population [1]. P. aeruginosa infection starts with a non- impairment does not necessarily mean deficiency, but mostly
mucoid planktonic variant, which is frequently succeeded by overproduction of several immune components. CF airway
colonization-treatment-recolonization cycles, marked by inter- epithelial cells (AECs), when challenged with P. aeruginosa,
mittent bacterial isolations in microbiological respiratory tract show increased activation of the Nuclear Transcription Factor –
cultures. This can further develop into a chronic stage of Kappa B (NF-κB), which leads to overproduction of pro-
infection, marked by a mucoid biofilm-growing P. aeruginosa. inflammatory cytokines interleukin (IL-) 6 and 8, and Tumor
Biofilm formation provides protection against the immune Necrosis Factor Alpha (TNF-α) disproportionately to the bacterial
system (Figs. 1 and 2) and antibiotics, as well as provoking an load in the airways, when compared with normal AECs. NF-κB
intense inoperative tissue-damaging inflammatory response, overproduction also reduces the synthesis of glutathione, an
rendering P. aeruginosa practically impossible to eradicate in antioxidant peptide that neutralizes reactive oxygen species (ROS)
this phase of infection [2,3]. Moreover, the ionic unbalance and increases the production of inflammatory mediators like

Fig. 1. Mucus with a P. aeruginosa biofilm surrounded by polymorphonuclear neutrophils (PMN) and IgG antibodies specific to P. aeruginosa antigens, such as
alginate, LPS and proteins. Immune complexes are formed between P. aeruginosa antigens and IgG, which activate complement and attract PMNs. Activated PMNs
consume oxygen and liberate ROS, proteases and DNA, starting the inflammatory reaction. Such inflammatory reaction consumes all oxygen in mucus, which
becomes anaerobic, impairing the PMNs' ability to phagocytose and kill P. aeruginosa in sputum due to the oxygen starvation needed for PMN activity, and leading
to a frustrated phagocytosis. The pronounced IgG antibody response occurs especially due to the high NF-κB production by CF airway epithelial cells (AECs), and to
the Th2-skewing of the adaptive immune reponse, resulting in hyperproduction of proinflammatory cytokines, like IL-5 and IL-8, and low production of
anti-inflammatory cytokines, like IL-10 and INF-γ. Since the PMNs' ability is impaired, this hyperinflammatory response leads to tissue damage rather than killing P.
aeruginosa biofilms. IgA is produced in the Bronchi-Associated Lymphoyd Tissue (BALT) submucosa (1), where it combines with the secretory component,
yielding sIgA, which is exported through the epithelal cells to the airway surface liquid (ASL) (2), preventing P. aeruginosa to attach to the epithelial cells, in a
macrophage-mediated non-phlogistic response, thereby not contributing to airway inflammation.
R.M. Mauch et al. / Journal of Cystic Fibrosis 17 (2018) 143–152 145

The Th17 response has also been recently addressed in CF as


having the ability to stimulate the neutrophil mobiliser G-CSF and
the neutrophil chemoattractant IL-8, thus contributing to the
pulmonary pathology during chronic P. aeruginosa lung infec-
tion. The rate of naive T-cells that differentiate into Th17 cells is
almost twice as high as in healthy subjects. This amount correlates
with the severity of the lung disease, especially caused by P.
aeruginosa, suggesting that IL-17 and associated cytokines can
serve both as biomarkers for early detection of the disease and
potential therapeutic targets [18–20]. Furthermore, a Th2-Th17
axis in CF may predispose for development P. aeruginosa lung
infection [17,18,20,21].

2. B-cells, antibodies and the humoral immune response in CF

Fig. 2. Fluorescence in situ hybridization staining of P. aeruginosa in a biofilm from Although the elucidation of CF immunological disorders has
an explanted chronically infected CF lung. Bacteria are stained red and the
been facilitating the understanding of the lung disease natural
surrounding PMNs are stained blue (the nuclei). Extracted from Bjarnsholt et al. [61].
history, the efficacy of the humoral immune response remains a
poorly explored field concerning the pathogen-immune system
Prostaglandin E2 and Cycloxygenase-2 (COX-2) [7]. CF AECs interaction. In P. aeruginosa infection, particularly, the high
also produce a decreased amount of interferon-gamma (IFN-γ), antibody level followed by immune complex (IC) formation is a
causing upregulation of IL-33, which, like IL-6 and IL-8, are hallmark (Fig. 1). Pioneer studies had already shown that the level
neutrophil chemoattractants [8]. Consequently, neutrophils of specific P. aeruginosa precipitating antibodies were increased
extravasate to the airways and are activated, releasing substantial in the presence of the mucoid phenotype and associated with poor
levels of proteases and cationic peptides, deteriorating the airway prognosis [22]. From then on, the antibody response with
surface and permeability [9]. Under normal conditions, neutro- diagnostic purposes was tested against a broad variety of P.
phils undergo apoptosis and are swept from the airways by cough aeruginosa antigens, showing frequently enhanced antibody levels
clearance or phagocytosis by alveolar macrophages (AM). In the (mainly IgG), even in patients without P. aeruginosa chronic lung
CF context, notwithstanding the impaired MCC, macrophages' infection [23]. The response against particular antigens appears to
ability to phagocyte apoptotic neutrophils is also decreased; thus, depend on the infection stage, with some antigens provoking a
neutrophils undergo necrosis rather than apoptosis, releasing their more intense response in the acute phase, while others are more
intracellular content in the airways, including chemoattractants targeted during the chronic phase [24]. However, the relation
(attracting other neutrophils to the inflammation site, thus between response intensity and pulmonary damage, as well as the
perpetuating the immune response), DNA content (that serves lack of the association between humoral response and clinical
as a biofilm matrix), proteases and ROS (damaging the airway improvement are well reported [25,26]. Besides, antibiotic
structure) [10,11]. treatment may decrease anti-Pseudomonas antibody production,
In the adaptive immune response, more specifically in the but even after treatment, there is an increased risk of recurrent
T-cell response, a predilection to mount a Th2-skewed response infection if patients display high antibody levels [27].
is observed, with upregulation of IL-5 and IL-13, and higher Despite the evidences of the inefficiency of humoral response
B-cell sensitivity to IL-4. Such response is well known to fight to mediate the clearance of infection from the airways, the mode
parasites, not being efficient against bacterial pathogens like P. of action of specific IgG anti-Pseudomonas antibodies, as well as
aeruginosa [6]. There are evidences that CF Th cells also their subclass distribution and the scenario involving B-cell
produce low levels of IFN-γ (a Th1-associated cytokine) and production, stimulation and differentiation into plasma cells in
elevated levels of IL-10, a Th2-associated cytokine with ability the airways remains poorly studied in CF.
to downregulate IFN-γ and to decrease co-stimulatory mole-
cules in macrophages, thereby hindering antigen presentation 2.1. B-cells in CF
and preventing a proper immune response to P. aeruginosa,
Aspergillus fumigatus and other CF pathogens [12–14]. It has B-cells primarily rise from hematopoietic stem cells and their
been suggested that the Th1 response can be protective against development is marked by a continuum of stages that starts in the
P. aeruginosa infection and confer resistance to reinfection primary lymphoid tissues – mainly the bone marrow (BM). B-cell
[15]. IFN-γ treatment in a chronic lung infection rat model maturation progresses in sequential steps in the BM and, after this,
using alginate embedded P. aeruginosa changed the pulmonary the B-cells migrate to and complete the maturation in the secondary
response from an inflammation dominated by neutrophils to lymphoid tissues (SLT) - e.g. lymph node and spleen [28].
mononuclear leucocytes [16]. Moreover, IFN-γ induction of Antigen-induced activation and differentiation in the SLTs are
alveolar macrophages may mediate removal of apoptotic mediated by dynamic changes in gene expression that originate in
neutrophils, preventing further increased inflammation due to the germinal center (GC) reaction. GCs are transient structures
progression into necrosis [17]. formed inside the SLT and dedicated to the production of high
146 R.M. Mauch et al. / Journal of Cystic Fibrosis 17 (2018) 143–152

affinity antibodies, where the B-cells expressing such membrane appears to be insufficient to remove the bacteria, prevent chronic
bound antibodies mature, undergo stimulation and develop into infection or acquisition of new strains [6]. In another report, CF
antibody-producing cells (plasma cells) or memory B-cells. patients showed a significantly higher spontaneous formation of
Developing B-cells progresses through rearrangement of gene plaque-forming cells (which reflect the B-cell differentiation into
segments of the light and heavy immunoglobulin (Ig) chains (V, D plasma cells in vivo), when compared with normal individuals
and J) of pre- and pro-B-cells, thus culminating in the expression of after challenge with mitogen or staphyloccoci, as well as a
the mature B-cell receptor (BCR) (IgD or IgM) in the cell surface, significant impairment in the B-cell differentiation in response to
which can bind antigens (Fig. 3). The maturation steps depend on the polyclonal activation in vitro, which could not be explained
close interactions between developing B-cells and BM stromal by the increased numbers of adherent suppressor cells or by the
cells, which provide critical adhesive integrins, growth factors, presence of suppression enhanced by T-cells [30]. Neill et al. [31]
chemokines and cytokines. Immature B-cells that leave the BM recently showed, for the first time, the kinetic of B-cell responses
towards the SLTs have the maturation process guided by BCR to chemoattractant factors and the kinetic of B-cell differentia-
signals, B-cell Activating Factor (BAFF), and expression of tion, both following P. aeruginosa infection and in relation to the
transcription factors, like NOTCH2 and BTK [29]. B-cell recruitment, in a CF murine model. P. aeruginosa
High numbers of B-cells have been documented in CF infection showed association with elevated levels of BAFF and
patients infected with P. aeruginosa, and low levels in patients another B-cell chemoattractants, like CXCL13, CCL19 and
under treatment for pulmonary exacerbation. Patients with CCL21. An elevated BAFF level was also found in bronchoal-
persistent infection with P. aeruginosa showed high levels of veolar lavage (BAL) of CF pediatric patients, irrespective of
immunoglobulins in ASL, mainly IgA; however, this response absence or presence of P. aeruginosa infection, implying that the

Fig. 3. After synthesis in the bone marrow, naïve B cells are activated in the secondary lymphoid organs (e.g. CF BALT) during antigen presentation by an
antigen-presenting cell (e.g. dendritic cell), and interaction with the antigen. B cells then proliferate and generate plasma cells, which produce antibodies (e.g. IgG and
IgA). Antigen-specific naïve B cells can differentiate within secondary lymphoid tissue into short-lived low-affinity plasma cells or undergo a rapid proliferative
phase known as the germinal center (GC) reaction, where clonal expansion and affinity/avidity maturation take place, yielding either memory B cells or high-affinity
long-lived plasma cells. When memory B cells are re-activated by a specific antigen, they quickly respond to the cognate antigen, proliferate and differentiate into the
high-affinity long-lived antibody producing plasma cells. CF antibodies undergo deficient affinity/avidity maturation [89]. This may be due to the continous
pronounced antigen stimulation, which takes place over decades in well-treated patients, where both high-affinity and low affinity clones are activated. This is
completely different from the controlled situation during vaccination, where small amounts of antigens are repeatedly administered to obtain protective immunity and
where only high affinity clones are activated. IgA is mainly transported to the ASL to keep bacteria and their products at a distance from the epithelial surface in order
to prevent damage to the epithelial surface (see Fig. 1). IgG, on the other hand, is then transported to the interstial fluid and to the blood, reaching the respiratory zone
(alveoli) (1), where it binds the bacteria present in this zone, contributing to the inflammatory response, attracting macrophages and neutrophils, leading to secretion
of proinflammatory cytokines. These cytokines attract more neutrophils through transmigration from the capillary endothelia to the alveolar space (2). Neutrophils
then produce proteases and ROS, provoking a gradual and extensive lung injury.
R.M. Mauch et al. / Journal of Cystic Fibrosis 17 (2018) 143–152 147

expression is not specific for this pathogen and can be a feature of 2.2.2. Opsonization capacity and phagocytic killing
the CF airways, and suggesting that not only the B-cells recruited Derived from the greek word opson (condiment), opsonization
to the airway, but the local environment as well, is able to support is the process by which a foreign particle, particularly a microbe, is
the B-cell survival, differentiation and antibody production; coated with plasma proteins (opsonins) aiming to facilitate the
however, persistence of P. aeruginosa infection suggests that this microbe attachment and internalization by a professional phago-
response is ineffective. Still, there are no studies confirming cytic cell. Overall, the process refers to the microbe coating with Ig
whether there is any failure in B-cell production, regulation and molecules (antibodies) that are specific for the antigenic determi-
proliferation in CF. nants of that organism, or with complement proteins (particularly
C3b) that are deposited upon the surface of the microorganism both
through the classic or alternative activation pathway. The presence
2.2. Functionality of antibody response in CF of these proteins in the microbial surface facilitates its sequential
interaction with the Ig receptors (Fc receptors) or complement
2.2.1. Immune complex formation receptors in the phagocyte surface. These interactions result in
IC formation during P. aeruginosa lung infection in sputum encirclement of the particle by the cytoplasmic membrane of the
(Fig. 1) and serum from CF patients is long-established [32,33]. A phagocytic cell, until the particle is contained in a membrane-
significantly higher IgG2 concentration was reported in sera of CF bound vacuole (phagosome) within the cell [47].
patients chronically infected with P. aeruginosa, as well as higher Early reports found inability of bactericidal activity in sera
mean levels of circulating immune complexes (CIC). CIC removal from CF patients with advanced lung disease, particularly against
was accompanied by significant decrease in IgG2 concentration autologous P. aeruginosa isolates, suggesting the presence of
without changes in other subclasses, and by a significant increase “bactericidal block” antibodies [48,49], and attributing this defect
of both macrophage and neutrophil-mediated phagocytosis, to IgG [50]. Such findings headed the investigation of the opsonic
therefore suggesting IgG2 to significantly contribute to CIC capacity of CF sera, revealing a deficiency of sera from patients
formation and to have antiphagocytic activity through direct infected with P. aeruginosa to promote phagocytosis (in relation
inhibition in CF [34]. ICs in sputum of CF patients chronically to normal individuals and patients without P. aeruginosa
infected with P. aeruginosa were found to be basically composed infection) particularly by alveolar macrophages [51–54], but not
of lipopolysaccharide (LPS), IgG1–4, IgA and IgM. Still, high by PMNs [51,55]. Complement activation is reported as being
CIC concentration was shown to be positively correlated with the normal [49,51,52,55–57] and opsonic capacity appeared to
TNF-α amount. In vitro analyses showed that the ICs containing include a non-complement cofactor for optimal activity [52].
LPS stimulated a higher TNF-α release than LPS alone, and also Immunoglobulin activity, on the other hand, was suggested to
induced oxidative stress in neutrophils [35]. One study found that be deficient and even inhibitory, confirming previous reports. A
C4 activation was highly associated with CIC [36], but two other series of studies by Moss et al. [36,52,56,58] found that CF patients
did not find any relation between CIC and complement activation with P. aeruginosa infection displayed a broadly distributed
[37,38]. hyperimmunoglobulinemia G, and even not infected patients were
Conflicting results were obtained in attempts to correlate IC subject to have elevated IgG3 and IgG4 levels. The major antigenic
formation and level with clinical state. CF patients with acute lung determinant of the elevated IgG serum response was the P.
disease had higher CIC formation than stable patients [38] and aeruginosa LPS. Such elevation showed distribution among all
higher CIC levels were found in patients with worst clinical states IgG subclasses, with a shift towards IgG3. Notwithstanding, when
in three studies, being associated with worst clinical and X-ray serum opsonic capacity was compared with level of autologous
scores, lower lung function results, lower weight, and higher antibodies, increased levels of IgG4, but not IgG1–3 were found in
anti-P. aeruginosa IgG levels [39–41]. A multifactorial analysis patients with poor serum opsonic activity. Moreover, an inverse
with 10 years of follow-up [42] reported correlation between CIC correlation was found between IgG4 levels and opsonic activity,
and only four of 38 clinical parameters – low National Institute of suggesting a blocking capacity of AM-mediated P. aeruginosa
Health (NIH) score, higher age, low rate of respiratory peak flow phagocytosis by antibodies of this particular isotype. In addition,
and high total IgG level. There was no correlation between CIC patients who were not infected by P. aeruginosa had decreased
and severity of individual pulmonary exacerbation; however, all IgG2 levels and increased IgG4 levels to LPS when compared with
patients who died during the follow-up had higher CIC levels, normal controls. IgG4 indeed has a poor opsonizing capacity and
comparing with those who survived. Two studies observed an efficient response of this subclass is not expected, since IgG2 is
association between CIC evidence and/or formation with the subclass that better responds to polysaccharide antigens like
mortality [36,43], while other studies did not find association LPS [56]. The low IgG2 levels raises the possibility of restricted
between ICs and colonization or response to P. aeruginosa, immunodeficiency to polysaccharide antigens, with an attempt of
Staphylococcus aureus, Haemophilus influenzae and streptococ- compensatory shift towards IgG3 and IgG4 doomed to failure.
cal species, nor correlation of ICs with severity or progression of Indeed, almost one decade later, it was documented that patients
lung disease, pulmonary exacerbations, or non-specified humoral with high IgG3 levels to LPS, alginate and a mixed P. aeruginosa
and cellular functions [37,44,45]. Proteolytic cleavage of ICs was antigen (sonicate) are able to induce a high complement activation
suggested to contribute to the failure to detect ICs in CF and level that, however, is associated with a more aggressive lung
therefore to the lack of correlation with disease severity in some damage, probably secondary to local IC formation and reflecting
studies [46]. the inefficiency of this response [57].
148 R.M. Mauch et al. / Journal of Cystic Fibrosis 17 (2018) 143–152

Although PMN-mediated phagocytosis induction has been lead to a prolonged circulation of ICs, causing tissue deposition
reported as being normal, after P. aeruginosa opsonization with CF and damage [66].
sera, chemiluminescence analyses found that, while opsonization A study addressing the immunization of CF patients and
of P. aeruginosa with P. aeruginosa-infected sera resulted in healthy adults with a LPS-based vaccine demonstrated that this
increased levels of PMN chemiluminescence, when corrected for vaccine elicited a significant increase in the total anti-LPS IgG
equimolar P. aeruginosa LPS-specific IgG content, P. aeruginosa- levels and in the affinity of these antibodies. On the other hand, CF
infected sera displayed markedly lower opsonizing ability patients chronically colonized by P. aeruginosa (not immunized)
compared to P. aeruginosa-uninfected sera, normal sera, or normal showed affinity values at least 100 times lower than antibodies
serum-derived P. aeruginosa hyperimmune globulin [58]. Recent induced by the vaccine, despite of the elevated levels of naturally
reports [59,60] showed that P. aeruginosa opsonized with egg yolk acquired anti-LPS IgG. These high affinity antibodies seemed to
antibodies (IgY), the analogue avian antibody for human IgG, mediate the opsonophagocytic killing of P. aeruginosa by
enhanced the PMN-mediated respiratory burst and subsequent macrophages [67]. Other analysis [68] found that P. aeruginosa
bacterial killing in vitro. Such effects were observed against isolates recovered from CF patients were reactive mainly with a
different P. aeruginosa strains, and possibly reflect a decreased monoclonal antibody directed to the outer core of the P.
opsonizing capacity of CF IgG to head PMN-mediated aeruginosa LPS, suggesting reactivity to LPS as having potential
phagocytosis. value in P. aeruginosa eradication in these patients. Similar
P. aeruginosa biofilm aggregates in the lungs and sputum of conclusions were proposed by Lang et al. [69], who found
CF patients were shown to be surrounded by PMNs, but have association between P. aeruginosa infection and absence of high
never been observed inside PMNs (Figs. 1 and 2). In contrary, affinity anti-LPS antibodies. The frequency of infection was
planktonic P. aeruginosa cells were frequently seen inside the significantly higher among patients who were not vaccinated with
PMNs, engulfed, but presumably not destroyed. Since CF LPS and vaccinated patients who failed to mount a high affinity
sputum becomes anaerobic, due to the PMN activity, these cells response, than among immunized patients. Most P. aeruginosa-
are deprived form oxygen and may not be able to produce a positive respiratory cultures were obtained from specimens from
metabolic burst to kill the phagocytosed bacteria (Fig. 1) [61] immunized patients only after the loss of the high affinity
and thus functionally may be somewhat like PMNs from circulating anti-LPS antibodies, which occurred between the
chronic granulomatous disease patients [62], who are able to second and third years (out of four years) of observation. No
phagocytose bacteria but not to kill catalase positive bacteria such correlation was found when total anti-P. aeruginosa LPS
like e.g. S. aureus since they are defective in NADP-oxidase antibody levels versus infection rate were analyzed. Mucoid P.
and, therefore, cannot produce O2−. This is therefore a possible aeruginosa was cultured from a single vaccinated patient (out of
additional explanation for the functional inability of the four patients) who failed to mount a high affinity response, while
immune system of CF patients to clear lung infections. As a six (out of 10) non-vaccinated patients become infected with the
matter of fact, our group has previously shown that hyperbaric mucoid variants. Therefore, it was suggested that the presence of
oxygen treatment of O2-depleted P. aeruginosa biofilms could high affinity anti-LPS antibodies can act to keep the bacterial load
significantly enhance the bactericidal activity of ciprofloxacin below a critical level needed for the phenotypic switch.
on the bacteria, due to the increase of the diffusive supply for IgG avidity against P. aeruginosa exotoxin A (ExoA) was
aerobic respiration during ciprofloxacin treatment of these shown to be decreased in CF patients with P. aeruginosa chronic
biofilms, thereby showing the potential of combined treatments lung infection, despite being significantly different from patients
with hyperbaric oxygen [63]. without infection and healthy subjects. Also, no increase in
anti-ExoA IgG avidity was seen in 6/8 chronically infected
2.2.3. Defective interaction among pathogen, antibodies and patients after a period of six months of follow-up [70]. A possible
phagocytes explanation is the long term exposure to the antigen via CF lungs,
Evidences of defective antibody functional capacity reported which can cause a specific suppression of the IgG response [71].
to date suggest that CF antibodies may have a low avidity In a cohort of 11 CF patients with poor lung function and 9
(overall strength of an antigen-antibody complex) or affinity patients with good lung function, all patients developed increasing
(strength of the interaction between the antigen binding site in levels of antibodies against P. aeruginosa beta-lactamase and
the antibody and the antigen epitope) (Fig. 3) [64,65]. against the 60–65 kDa heat shock protein (anti-GroEL), but no
P. aeruginosa phagocytosis, by means of its AM-mediated avidity maturation of these antibodies was observed. In patients
internalization and opsonic killing, was shown to be reduced with good lung function, the avidity of anti-beta-lactamase
when the bacteria were opsonized with CF sera with high antibodies was higher than in patients with poor lung function,
specific IgG levels, even when compared with non-opsonized suggesting a more efficient inhibition of the beta-lactamase by
bacteria. Besides, radioimmunoassay studies observing the these antibodies, resulting in better lung function [72].
AM-bacteria interaction found only a few bacteria attached to In recurrent Staphylococcus aureus infections, high levels of
the AM membrane, thereby suggesting that a defect in the Fcγ antibodies to a variety of antigens are seen in CF, including
portion of the Pseudomonas-specific IgG in CF interferes with against the polysaccharide capsule, suggesting the presence of
the phagocytic function by preventing a proper receptor virulence factors that can inactivate specific antibodies, like those
attachment and subsequent triggering of internalization. Such directed to the A-protein, a T-cell-independent (TI) mitogen,
defect, with consequent decrease in the bacterial clearance, can which was suggested to induce the production of monoclonal
R.M. Mauch et al. / Journal of Cystic Fibrosis 17 (2018) 143–152 149

antibodies and to non-specifically bind immunoglobulin and, [79,80,87]. IgA was suggested to block alginate opsonization by
subsequently, also complement, thus possibly compromising S. IgG and, due to its poor opsonic capacity, to contribute to poor
aureus phagocytosis [73,74]. phagocytosis [88]. Furthermore, we have shown that IgG avidity
against alginate does not significantly increase with the progress
2.2.4. Alginate: cause or consequence of the ineffective of chronic infection, possibly playing a role in the difficulty to
humoral response? mediate efficient phagocytosis against this antigen to clear P.
Despite the reports by Lang et al. suggesting that induction of aeruginosa infection [89]. Previous studies [90,91] reported that
high affinity anti-LPS antibodies may help to prevent the P. alginate-based vaccines elicited opsonic antibodies in human,
aeruginosa phenotypic switch, no additional study has corrobo- mice and rabbit even in the presence of preexisting non-opsonic
rated this suggestion, as far as it is known. On the other hand, P. antibodies [91]. Antibodies elicited by two vaccines enhanced
aeruginosa alginate is well known to be a major component of the deposition of complement onto mucoid P. aeruginosa cells and
P. aeruginosa biofilm, helping in its maturation, architecture and mediated opsonic killing of heterologous P. aeruginosa strains
stability, as well as helping to protect the bacteria against expressing different alginates [90]. However, no studies have so
phagocytosis and clearance from the lungs, by acting like an far addressed avidity changes against alginates during the course
oxygen-radical scavenger, and inducing damage in the surrounding of infection.
tissues [75,76]. The fact of many PMNs are found surrounding the
P. aeruginosa biofilms, but few or no PMNs are seen inside the
biofilm (Figs. 1 and 2) is in accordance with the protective activity 3. Concluding remarks
of alginate, as well as with the lytic action of P. aeruginosa
rhamnolipids on PMNs [61,77,78]. The findings of immune dysregulation have strongly contrib-
A pronounced antibody response against alginate is uted in understanding CF inflammation mechanisms and directed
produced by CF patients (Fig. 1) [79–81], but opsonic quality researches of new therapeutic strategies for prevention and
has shown to be significantly declined in longitudinal analyses. treatment. Inflammatory responses are designed for acute
Patients who passed age 12 free of P. aeruginosa colonization infection, aiming a fast clearance of the corresponding pathogen.
developed chronic P. aeruginosa lung infection later, suggest- In a chronic infection scenario, like CF, with repeated exposure to
ing that naturally occurring antibodies do not protect CF the pathogen, several and highly detrimental collateral damages
patients from mucoid P. aeruginosa infection, and opsonic are worrisome. To date, treatment regimens have focused on
quality of serum antibodies deteriorates as infection becomes controlling the pro-inflammatory response in order to reduce the
established [82]. NF-kB signaling and consequently the neutrophilic influx in the
Alginate is a polysaccharide antigen, and similar carbohydrate airways. A therapeutic alternative would be to upregulate the
antigens are known to have poor immunogenicity due to the anti-inflammatory mechanisms, thus directing the immune
T-cell-independent (TI) nature [83]. TI antigens induce an response to a protective phenotype. Therapies targeting the
immunological memory characterized by high avidity antibodies, molecular and submolecular mechanisms and of biofilm
however they lack antibody production after new challenge with formation will likely have a key role in clearing infection,
the antigen [84]. It has been shown that alginate is not able to fix helping to halt the perpetual neutrophilic response.
complement and, therefore, cannot cause neutrophil chemotaxis by The role of adaptive immune response also needs to be better
itself, but has the ability to enhance neutrophil-mediated oxidative exploited, especially regarding to the humoral response. Despite
burst [79]. One report [85] suggested that the opsonic activity of B-cells being directly activated by invading organisms, and CF
specific anti-alginate antibodies against mucoid P. aeruginosa in chronic lung infections recruiting an elevated and apparently not
biofilms is related to their ability to deposit complement C3 effective antibody response, very few studies addressing antibody
fragments next to the bacterial surface, but sufficiently exposed so function in CF were published in the last 20 years, and B-cell
they can bind to complement receptors on the phagocytes, since features in CF has been reported only once, as far as it is known.
alginate-specific opsonins were shown to deposit significantly Therefore, this field remains mostly unknown. Such antibody
more C3 fragments on alginate than non-alginate-specific response is not inefficient by chance, and the underlying
opsonins. The authors still suggest that the alginate biofilm layer mechanisms, such as avidity/affinity maturation, class switch,
prevent complement receptors on the phagocytes to bind C3 memory formation and cytokine synthesis must be better
fragments deposited by non-alginate-specific antibodies on other elucidated.
antigens, since biofilms treated with alginate lyase improved the The discovery of new disease mechanisms and development
opsonic killing mediated by non-alginate-specific antibodies in 12 of new therapies can create windows of opportunity to reduce
from 16 P. aeruginosa chronically infected patients. the impact of inflammation, consequently reducing the decline
Other reason for the difficulty to efficiently opsonize in lung function in CF.
alginate-producing P. aeruginosa may be the predominance of
the IgG2 response, since this IgG subclass responds to
polysaccharide antigens and has poor opsonic quality [86]. IgG Conflict of interests
competition with IgA by the alginate epitope may also play a role,
since both serum and secretory IgA were shown to respond On behalf all authors, the corresponding author states that
specifically when directed against purified P. aeruginosa alginate there are no competing interests.
150 R.M. Mauch et al. / Journal of Cystic Fibrosis 17 (2018) 143–152

References [20] Bayes HK, Bicknell S, MacGregor G, Evans TJ. T helper cell subsets
specific for Pseudomonas aeruginosa in healthy individuals and patients
with cystic fibrosis. PLoS One 2014;9. https://doi.org/10.1371/journal.
[1] Stoltz D, Meyerholz DK, Welsh MJ. Origins of cystic fibrosis lung disease. pone.0090263.
NEJM 2015;372:351–62. https://doi.org/10.1056/NEJMra1300109. [21] Decraene A, Willems-Widyastuti A, Kasran A, De Boeck K, Bullens DM,
[2] Folkesson A, Jelsbak L, Yang L, Johansen HK, Ciofu O, Høiby N, et al. Dupont LJ. Elevated expression of both mRNA and protein levels of IL-
Adaptation of Pseudomonas aeruginosa to the cystic fibrosis airway: an 17A in sputum of stable cystic fibrosis patients. Respir Res 2010;11:
evolutionary perspective. Nat Rev Microbiol 2012;10:841–51. https://doi. 177–84. https://doi.org/10.1186/1465-9921-11-177.
org/10.1038/nrmicro2907. [22] Høiby N, Flensborg EW, Beck B, Friis B, Jacobsen SV, Jacobsen L.
[3] Moradali MF, Ghods S, Rehm BHA. Pseudomonas aeruginosa lifestyle: a Pseudomonas aeruginosa infection in cystic fibrosis. Diagnostic and
paradigm for adaptation, survival, and persistence. Front Cell Infect prognostic significance of Pseudomonas aeruginosa precipitins deter-
Microbiol 2017;7:1–29. https://doi.org/10.3389/fcimb.2017.00039. mined by means of crossed immunoelectrophoresis. Scand J Respir Dis
[4] Abou Alaiwa MH, Reznikov LR, Gansemer ND, Sheets KA, Horswill AR, 1977;58:65–79.
Stoltz DA, et al. pH modulates the activity and synergism of the airway [23] Mauch RM, Rossi CL, Ribeiro JD, Ribeiro AF, Nolasco da Silva MT,
surface liquid antimicrobials β-defensin-3 and LL-37. Proc Natl Acad Sci Levy CE. Assessment of IgG antibodies to Pseudomonas aeruginosa in
2014;111:18703–8. https://doi.org/10.1073/pnas.1422091112. patients with cystic fibrosis by an enzyme-linked immunosorbent assay
[5] Berkebile AR, McCray PB. Effects of airway surface liquid pH on host (ELISA). Diagn Pathol 2014;9:158–65. https://doi.org/10.1186/s13000-
defense in cystic fibrosis. Int J Biochem Cell Biol 2014;52:124–9. https:// 014-0158-z.
doi.org/10.1016/j.biocel.2014.02.009.Effects. [24] Mauch RM, Levy CE. Serum antibodies to Pseudomonas aeruginosa in
[6] Yonker LM, Cigana C, Hurley BP, Bragonzi A. Host-pathogen interplay cystic fibrosis as a diagnostic tool: a systematic review. J Cyst Fibros
in the respiratory environment of cystic fibrosis. J Cyst Fibros 2015;14: 2014;13:499–507. https://doi.org/10.1016/j.jcf.2014.01.005.
1–9. https://doi.org/10.1016/j.jcf.2015.02.008. [25] Brett MM, Ghoneim AT, Littlewood JM. Prediction and diagnosis of early
[7] Ziady AG, Hansen J. Redox balance in cystic fibrosis. Int J Biochem Cell Pseudomonas aeruginosa infection in cystic fibrosis: a follow-up study
Biol 2014;52:113–23. https://doi.org/10.1016/j.biocel.2014.03.006. [published erratum appears in J Clin Microbiol 1989 Jan 27 (1):230]. J
[8] Parker D, Cohen TS, Alhede M, Harfenist BS, Martin FJ, Prince A. Clin Microbiol 1988;26:1565–70.
Induction of type I interferon signaling by Pseudomonas aeruginosa is [26] West SEH, Zeng L, Lee BL, Kosorok MR, Laxova A, Rock MJ, et al.
diminished in cystic fibrosis epithelial cells. Am J Respir Cell Mol Biol Respiratory infections with Pseudomonas aeruginosa in children with
2012;46:6–13. https://doi.org/10.1165/rcmb.2011-0080OC. cystic fibrosis: early detection by serology and assessment of risk factors.
[9] Twigg MS, Brockbank S, Lowry P, FitzGerald SP, Taggart C, Weldon S. JAMA 2002;287:2958–67.
The role of serine proteases and antiproteases in the cystic fibrosis lung. [27] Anstead M, Heltshe SL, Khan U, Barbieri JT, Langkamp M, Döring G,
Mediat Inflamm 2015;2015:1–10. https://doi.org/10.1155/2015/293053. et al. Pseudomonas aeruginosa serology and risk for re-isolation in the
[10] McCaslin CA, Petrusca DN, Poirier C, Serban KA, Anderson GG, EPIC trial. J Cyst Fibros 2013;12:147–53. https://doi.org/10.1016/j.jcf.
Petrache I. Impact of alginate-producing Pseudomonas aeruginosa on 2012.08.001.
alveolar macrophage apoptotic cell clearance. J Cyst Fibros 2015;14:70–7. [28] Lebien TW, Tedder TF. B lymphocytes : how they develop and function.
https://doi.org/10.1016/j.jcf.2014.06.009. Am Soc Hematol 2008;112:1570–80. https://doi.org/10.1182/blood-2008-
[11] Bruscia EM, Bonfield TL. Cystic fibrosis lung immunity: the role of the 02-078071.
macrophage. J Innate Immun 2016;8:550–63. https://doi.org/10.1159/ [29] Hoffman W, Lakkis FG, Chalasani G. B cells, antibodies, and more. Clin J Am
000446825. Soc Nephrol 2016;11:137–54. https://doi.org/10.2215/CJN.09430915.
[12] Moss RB, Hsu YP, Olds L. Cytokine dysregulation in activated cystic [30] Sorensen RU, Ruuskanen O, Miller K, Stern RC. B-lymphocyte function
fibrosis (CF) peripheral lymphocytes. Clin Exp Immunol 2000;120: in cystic fibrosis. Eur J Respir Dis 1983;64:524–33.
518–25. https://doi.org/10.1046/j.1365-2249.2000.01232.x. [31] Neill DR, Saint GL, Bricio-Moreno L, Fothergill JL, Southern KW,
[13] Casaulta C, Schöni MH, Weichel M, Crameri R, Jutel M, Daigle I, et al. Winstanley C, et al. The B lymphocyte differentiation factor (BAFF) is
IL-10 controls aspergillus fumigatus- and Pseudomonas aeruginosa- expressed in the airways of children with CF and in lungs of mice infected
specific T-cell response in cystic fibrosis. Pediatr Res 2003;53:313–9. with Pseudomonas aeruginosa. PLoS One 2014;9:3–9. https://doi.org/10.
https://doi.org/10.1203/01.PDR.0000047528.79014.CF. 1371/journal.pone.0095892.
[14] Hartl D, Griese M, Kappler M, Zissel G, Reinhardt D, Rebhan C, et al. [32] McFarlane H, Holzel A, Brenchley P, Allan JD, Wallwork JC, Singer BE,
Pulmonary TH2 response in Pseudomonas aeruginosa-infected patients et al. Immune complexes in cystic fibrosis. Br Med J 1975;1:423–8.
with cystic fibrosis. J Allergy Clin Immunol 2006;117:204–11. https://doi. [33] Schiøtz PO, Nietsen H, Høiby N, Glikmann G, Svehag SE. Immune
org/10.1016/j.jaci.2005.09.023. complexes in the sputum of patients with cystic fibrosis suffering from
[15] Moser C, Jensen PO, Kobayashi O, Hougen HP, Song Z, Rygaard J, et al. chronic Pseudomonas aeruginosa lung infection. APMIS 1978;86C:
Improved outcome of chronic Pseudomonas aeruginosa lung infection is 37–40. https://doi.org/10.1111/j.1699-0463.1978.tb02555.x.
associated with induction of a Th1-dominated cytokine response. Clin Exp [34] Hornick DB, Fick Jr RB. The immunoglobulin G subclass composition of
Immunol 2002;127:206–13. https://doi.org/10.1046/j.1365-2249.2002. immune complexes in cystic fibrosis. J Clin Invest 1990;86:1285–92.
01731.x. [35] Kronborg G, Fomsgaard A, Galanos C, Freudenberg M, Høiby N.
[16] Johansen HK, Hougen HP, Rygaard J, Høiby N. Interferon-gamma (IFN- Antibody responses to lipid A, core, and O sugars of the Pseudomonas
gamma) treatment decreases the inflammatory response in chronic aeruginosa lipopolysaccharide in chronically infected cystic fibrosis
Pseudomonas aeruginosa pneumonia in rats. Clin Exp Immunol 1996;103: patients. J Clin Microbiol 1992;30:1848–55.
212–8. [36] Moss RB, Hsu YP, Lewiston NJ, Curd JG, Milgrom H, Hart S, et al.
[17] Moser C, Pedersen HT, Lerche CJ, Kolpen M, Line L, Thomsen K, et al. Association of systemic immune complexes, complement activation,
Biofilms and host response: helpful or harmful? APMIS 2017;125: and antibodies to Pseudomonas aeruginosa lipopolysaccharide and
320–38. https://doi.org/10.1111/apm.12674. exotoxin A with mortality in cystic fibrosis. Am Rev Respir Dis 1986;
[18] Tiringer K, Treis A, Fucik P, Gona M, Gruber S, Renner S, et al. A Th17- 133:648–52.
and Th2-skewed cytokine profile in cystic fibrosis lungs represents a [37] Moss RB, Lewiston NJ. Immune complexes and humoral response to
potential risk factor for Pseudomonas aeruginosa infection. Am J Respir Pseudomonas aeruginosa in cystic fibrosis. Am Rev Respir Dis 1980;121:
Crit Care Med 2013. 23–9.
[19] Kushwah R, Gagnon S, Sweezey NB. Intrinsic predisposition of naïve [38] Church JA, Jordan SC, Keens TG, Wang CI. Circulating immune
cystic fibrosis T cells to differentiate towards a Th17 phenotype. Respir complexes in patients with cystic fibrosis. Chest 1981;80:405–11. https://
Res 2013;14:138. https://doi.org/10.1186/1465-9921-14-138. doi.org/10.1378/chest.80.4.405.
R.M. Mauch et al. / Journal of Cystic Fibrosis 17 (2018) 143–152 151

[39] Hodson ME, Beldon I, Batten JC. Circulating immune complexes in [59] Thomsen K, Christophersen L, Bjarnsholt T, Jensen P, Moser C, Høiby N.
patients with cystic fibrosis. Clin Allergy 1985;15:363–70. https://doi.org/ Anti-Pseudomonas aeruginosa IgY antibodies promote specific bacterial
10.1111/j.1365-2222.1985.tb03004.x. aggregation and internalization in polymorphonuclear neutrophils. Infect
[40] Van Bever HP, Gigase PL, De Clerck LS, Bridts CH, Franckx H, Stevens Immun 2013;83:2686–93. https://doi.org/10.1128/IAI.02970-14.
WJ. Immune complexes and Pseudomonas aeruginosa antibodies in [60] Thomsen K, Christophersen L, Jensen PØ, Bjarnsholt T, Moser C, Høiby
cystic fibrosis. Arch Dis Child 1988;63:1222–8. https://doi.org/10.1136/ N. Anti-Pseudomonas aeruginosa IgY antibodies promote bacterial
adc.63.10.1222. opsonization and augment the phagocytic activity of polymorphonuclear
[41] Lagacé J, Mercier J, Frechette M, Fournier D, Dubreuil M, Lamarre A, et al. neutrophils. Hum Vaccines Immunother 2016;12:1690–9. https://doi.org/
Circulating immune complexes, antibodies to Pseudomonas aeruginosa, and 10.1080/21645515.2016.1145848.
pulmonary status in cystic fibrosis. J Clin Lab Immunol 1989;30:7–11. [61] Bjarnsholt T, Jensen PØ, Fiandaca MJ, Pedersen J, Hansen CR, Andersen
[42] Disis ML, McDonald TL, Colombo JL, Kobayashi RH, Angle CR, CB, et al. Pseudomonas aeruginosa biofilms in the respiratory tract of
Murray S. Circulating immune complexes in cystic fibrosis and their cystic fibrosis patients. Pediatr Pulmonol 2009;44:547–58. https://doi.org/
correlation to clinical parameters. Pediatr Res 1986;20:385–90. https://doi. 10.1002/ppul.21011.
org/10.1203/00006450-198605000-00002. [62] Holland SM. Chronic granulomatous disease. Clin Rev Allergy Immunol
[43] Wisnieski JJ, Todd EW, Fuller RK, Jones PK, Dearborn DG, Boat TF, 2010;38:3–10. https://doi.org/10.1007/s12016-009-8136-z.
et al. Immune complexes and complement abnormalities in patients with [63] Kolpen M, Mousavi N, Sams T, Bjarnsholt T, Ciofu O, Moser C, et al.
cystic fibrosis. Increased mortality associated with circulating immune Reinforcement of the bactericidal effect of ciprofloxacin on Pseudomonas
complexes and decreased function of the alternative complement pathway. aeruginosa biofilm by hyperbaric oxygen treatment. Int J Antimicrob Agents
Am Rev Respir Dis 1985;132:770–6. 2016;47:163–7. https://doi.org/10.1016/j.ijantimicag.2015.12.005.
[44] Berdischewsky M, Pollack M, Young LS, Chia D, Osher AB, Barnett EV. [64] Gutiérrez J, Maroto C. Are IgG antibody avidity assays useful in the
Circulating immune complexes in cystic. Pediatr Res 1980;14:830–3. diagnosis of infectious diseases? A review. Microbios 1996;87:113–21.
[45] Pitcher-Wilmott RW, Levinsky RJ, Matthew DJ. Circulating soluble [65] Dimitrov JD, Planchais C, Roumenina LT, Vassilev TL, Kaveri SV, Lacroix-
immune complexes containing pseudomonas antigens in cystic fibrosis. Desmazes S. Antibody polyreactivity in health and disease: statu variabilis. J
Arch Dis Child 1982;57:577–81. https://doi.org/10.1136/adc.57.8.577. Immunol 2013;191:993–9. https://doi.org/10.4049/jimmunol.1300880.
[46] Döring G, Albus A, Høiby N. Immunologic aspects of cystic fibrosis. [66] Fick RB, Naegel GP, Matthay RA, Reynolds HY. Cystic fibrosis
Chest 1988;94:109S–15S. Pseudomonas opsonins. Inhibitory nature in an in vitro phagocytic assay. J
[47] Czuprynski CJ. Opsonization and phagocytosis. Encycl. ref. immunotoxicol. Clin Invest 1981;68:899–914.
Berlin/Heidelberg: Springer-Verlag; 2005. p. 488–9. https://doi.org/10.1007/ [67] Bruderer U, Cryz SJ, Schaad UB, Deusinger M, Que JU, Lang AB. Affinity
3–540-27806-0_1113. constants of naturally acquired and vaccine-induced anti-Pseudomonas
[48] Høiby N, Schiøtz PO. Immune complex mediated tissue damage in the aeruginosa antibodies in healthy adults and cystic fibrosis patients. J Infect
lungs of cystic fibrosis patients with chronic Pseudomonas aeruginosa Dis 1992;166:344–9.
infection. Acta Paediatr 1982:63–73. [68] Torensma R, Fluit AC, Verhoef J. Reactivity of monoclonal antibodies to
[49] Thomassen MJ, Demko CA. Serum bactericidal effect on Pseudomonas Pseudomonas aeruginosa isolates from hospitalized adults and patients
aeruginosa isolates from cystic fibrosis patients. Infect Immun 1981;33:512–8. with cystic fibrosis. Clin Infect Dis 1994;19:11–4.
[50] Guttman RM, Waisbren BA. Bacterial blocking activity of specific IgG in [69] Lang AB, Schaad UB, Rüdeberg A, Wedgwood J, Que JU, Fürer E, et al.
chronic Pseudomonas aeruginosa infection. Clin Exp Immunol 1975;19: Effect of high-affinity anti-Pseudomonas aeruginosa lipopolysaccharide
121–30. antibodies induced by immunization on the rate of Pseudomonas aeruginosa
[51] LeBlanc CMA, Bortolussi R, Issekutz AC, Gillespie T. Opsonization of infection in patients with cystic fibrosis. J Pediatr 1995;127:711–7.
mucoid and non-mucoid Pseudomonas aeruginosa by serum from patients [70] Polanec J, Patzer J, Grzybowski J, Strukelj M, Pavelic ZP. Amount and
with cystic fibrosis assessed by a chemiluminescence assay. Clin Investig avidity of IgG antibodies to Pseudomonas aeruginosa exotoxin a antigen
Med 1982;5:125–8. in cystic fibrosis patients. Pathol Oncol Res 1997;3:26–9.
[52] Moss RB, Hsu YP, Sullivan MM, Lewiston NJ. Altered antibody isotype [71] Brauner A, Cryz SJ, Granström M, Hanson HS, Löfstrand L, Strandvik B,
in cystic fibrosis: possible role in opsonic deficiency. Pediatr Res 1986;20: et al. Immunoglobulin G antibodies to Pseudomonas aeruginosa
453–9. lipopolysaccharides and exotoxin A in patients with cystic fibrosis or
[53] Thomassen MJ, Boxerbaum B, Demko CA, Kuchenbrod PJ, Dearborn bacteremia. Eur J Clin Microbiol Infect Dis 1993;12:430–6.
DG, Wood RE. Inhibitory effect of cystic fibrosis serum on Pseudomonas [72] Ciofu O, Petersen TD, Jensen PØ, Høiby N. Avidity of anti-P. aeruginosa
phagocytosis by rabbit and human alveolar macrophages. Pediatr Res antibodies during chronic infection in patients with cystic fibrosis. Thorax
1979;13:1085–8. 1999;54:141–4. https://doi.org/10.1136/thx.54.2.141.
[54] Thomassen MJ, Demko CA, Wood RE, Tandler B, Dearborn DG, [73] Albus A, Fournier JM, Wolz C, Boutonnier A, Ranke M, Høiby N, et al.
Boxerbaum B, et al. Ultrastructure and function of alveolar macrophages Staphylococcus aureus capsular types and antibody response to lung
from cystic fibrosis patients. Pediatr Res 1980;14:715–21. https://doi.org/ infection in patients with cystic fibrosis. J Clin Microbiol 1988;26:
10.1203/00006450-198005000-00003. 2505–9.
[55] Pier GB, Saunders MJ, Ames P, Edwards MS, Auerbach H, Goldfarb J, [74] Kobayashi SD, DeLeo FR. Staphylococcus aureus protein A promotes
et al. Opsonophagocytic killing antibody to Pseudomonas aeruginosa immune suppression. MBio 2013;4:e00764-3. https://doi.org/10.1128/
mucoid exopolysaccharide in older noncolonized patients with cystic mBio.00764-13.
fibrosis. N Engl J Med 1987;317:793–8. [75] Song Z, Wu H, Ciofu O, Kong KF, Høiby N, Rygaard J, et al.
[56] Moss RB. The role of IgG subclass antibodies in chronic infection: the Pseudomonas aeruginosa alginate is refractory to Th1 immune response
case of cystic fibrosis. N Engl Reg Allergy Proc 1988;9:57–61. and impedes host immune clearance in a mouse model of acute lung
[57] Pressler T, Jensen ET, Espersen F, Pedersen SS, Høiby N. High levels of infection. J Med Microbiol 2003;52:731–40. https://doi.org/10.1099/jmm.
complement-activation capacity in sera from patients with cystic fibrosis 0.05122-0.
correlate with high levels of IgG3 antibodies to Pseudomonas aeruginosa [76] Ciofu O, Tolker-Nielsen T, Jensen PØ, Wang H, Høiby N. Antimicrobial
antigens and poor lung function. Pediatr Pulmonol 1995;20:71–7. https:// resistance, respiratory tract infections and role of biofilms in lung
doi.org/10.1002/ppul.1950200204. infections in cystic fibrosis patients. Adv Drug Deliv Rev 2015;85:7–23.
[58] Eichler I, Joris L, Hsu YP, Van Wye J, Bram R, Moss R. Nonopsonic https://doi.org/10.1016/j.addr.2014.11.017.
antibodies in cystic fibrosis. Pseudomonas aeruginosa lipopolysaccharide- [77] Høiby N, Ciofu O, Bjarnsholt T. Pseudomonas aeruginosa biofilms in
specific immunoglobulin G antibodies from infected patient sera inhibit cystic fibrosis. Future Microbiol 2010;5:1663–74.
neutrophil oxidative responses. J Clin Invest 1989;84:1794–804. https://doi. [78] Jensen PØ, Bjarnsholt T, Phipps R, Rasmussen TB, Calum H, Christoffersen
org/10.1172/JCI114364. L, et al. Rapid necrotic killing of polymorphonuclear leukocytes is caused by
152 R.M. Mauch et al. / Journal of Cystic Fibrosis 17 (2018) 143–152

quorum-sensing-controlled production of rhamnolipid by Pseudomonas during chronic lung infection in cystic fibrosis patients. J Immunol 1995;
aeruginosa. Microbiology 2007;153:1329–38. https://doi.org/10.1099/mic. 155:2029–38. https://doi.org/10.4049/jimmunol.173.5.3320.
0.2006/003863-0. [86] Sigal LH. Basic science for the clinician 58: IgG subclasses. J Clin Rheumatol
[79] Pedersen SS, Kharazmi A, Espersen F, Høiby N. Pseudomonas 2012;18:316–8. https://doi.org/10.1097/RHU.0b013e318269446b.
aeruginosa alginate in cystic fibrosis sputum and the inflammatory [87] Aanaes K, Johansen HK, Poulsen SS, Pressler T, Buchwald C, Høiby N.
response. Infect Immun 1990;58:3363–8. Secretory IgA as a diagnostic tool for Pseudomonas aeruginosa
[80] Pedersen SS, Møller H, Espersen F, Sørensen CH, Jensen T, Høiby N. respiratory colonization. J Cyst Fibros 2013;12:81–7. https://doi.org/10.
Mucosal immunity to Pseudomonas aeruginosa alginate in cystic fibrosis. 1016/j.jcf.2012.07.001.
APMIS 1992;100:326–34. [88] Pedersen SS, Espersen F, Høiby N, Jensen T. Immunoglobulin A and
[81] Craig A, Mai J, Cai S, Jeyaseelan S. Neutrophil recruitment to the lungs immunoglobulin G antibody responses to alginates from Pseudomonas
during bacterial pneumonia. Infect Immun 2009;77:568–75. https://doi. aeruginosa in patients with cystic fibrosis. J Clin Microbiol 1990;28:
org/10.1128/IAI.00832-08. 747–55.
[82] Tosi MF, Zakem-Cloud H, Demko CA, Schreiber JR, Stern RC, Konstan [89] Mauch RM, Nørregard LL, Jensen PØ, Levy CE, Høiby N. 76 avidity of IgG
MW, et al. Cross-sectional and longitudinal studies of naturally occurring antibodies and opsonic quality of serum samples against Pseudomonas
antibodies to Pseudomonas aeruginosa in cystic fibrosis. J Infect Dis aeruginosa during chronic lung infection in cystic fibrosis. J Cyst Fibros
1995;172:453–61. 2017;16:S83–4. https://doi.org/10.1016/S1569-1993(17)30440-X.
[83] Weintraub A. Immunology of bacterial polysaccharide antigens. Carbohydr [90] Pier GB, DesJardin D, Grout M, Garner C, Bennett SE, Pekoe G, et al. Human
Res 2003;338:2539–47. https://doi.org/10.1016/j.carres.2003.07.008. immune response to Pseudomonas aeruginosa mucoid exopolysaccharide
[84] Mazmanian SK, Kasper DL. The love–hate relationship between bacterial (alginate) vaccine. Infect Immun 1994;62:3972–9.
polysaccharides and the host immune system. Nat Rev Immunol 2006;6: [91] Theilacker C, Coleman FT, Mueschenborn S, Llosa N, Grout M, Pier GB.
849–58. https://doi.org/10.1038/nri1956. Construction and characterization of a Pseudomonas Aeruginosa mucoid
[85] Meluleni GJ, Grout M, Evans DJ, Pier GB. Mucoid Pseudomonas exopolysaccharide-alginate conjugate vaccine. Infect Immun 2003;71:
aeruginosa growing in a biofilm in vitro are killed by opsonic antibodies 3875–84. https://doi.org/10.1128/IAI.71.7.3875.
to the mucoid exopolysaccharide capsule but not by antibodies produced

You might also like