Basic Science Concepts in Otitis Media Pathophysiology and Immunity: Role of Mucins and Inflammation




(1)
Sheikh Zayed Institute, The Otologic Laboratory, Children’s National Health System, Center for Genetic Medicine Research, 111 Michigan Avenue NW, 20010 Washington, DC, USA

 



Keywords
Otitis mediaEffusionsMiddle ear epitheliumInflammationMucusMucinInnate immunityMolecular mechanismToll-like receptor



Part I: The Innate Immunity in Otitis Media


The first line of defense against pathogens entering in the middle ear is innate immunity. It plays very diverse and important roles:



  • Creating a physical and chemical barrier to pathogens: cellular barriers that are the epithelial surfaces and mucus layers on the top of epithelia


  • Identifying pathogens with nonspecific receptors or sensing molecules


  • Producing factors to activate inflammation, called pro-inflammatory mediators as cytokines and chemokines, to attract inflammatory cells


  • Activate the process of adaptive immunity response by recruiting cells and presenting them antigens


  • Kill pathogens and clean them from the tissue.

Several pathogens were identified in the middle ear of patients suffering from OM: diverse bacteria and viruses . Sometimes both at the same time have been found in middle ear effusions (MEEs) and seem to help each other [1]. Against these invaders, cellular and molecular barriers, recognition molecules and receptors, inflammatory mediators, and inducible effectors of the epithelium constitute the innate immune mechanisms that protect the middle ear.


The First Line of Defense of the Innate Immunity: Cellular and Humoral Barriers


The very first lines of defenses of the innate immunity are physical and functional barriers. They are the epithelium of the middle ear and eventually the layer of mucoid gel on the top of it to protect the cells of the epithelium from the invasion by pathogens. The middle ear cavity of healthy patients does not contain liquid. Contrary to the airways, this line of defense has to be activated in the middle ear.


The Middle Ear Epithelium


The epithelium of the middle ear is mostly a single layer of cubical squamous cells. Some patches of the middle ear epithelium, and especially close to the Eustachian tube, gradually change in a pseudostratified columnar epithelium similar to the mucociliary epithelium of respiratory epithelia [2]. This epithelium is constituted of basal cells, goblet cells producing mucins, and other cells that can be ciliated or not. On top of these patches, mucus is present and protects these regions of the epithelium from the infection. The ciliated cells ensure the movement of the mucus in direction to the Eustachian tube orifice where it is evacuated from the middle ear to the oral cavity.

Mucus glands, a normal feature of the Eustachian tube, can also be present in the middle ear of patients with OM. They constitute invaginations of the epithelium in the lamina propria, regions very rich in goblet cells that are able to produce large quantities of mucins. In healthy subjects that did not have prior disease of the middle ear, very few mucus glands are usually observed, whereas in subjects that had a history of OM events, these glands are more numerous [3]. This suggests that in the middle ear, mucus glands probably appear after several episodes of OM and then remain even after the disease is resolved. In patients with chronic suppurative otitis media (CSOM), the density of mucus glands is very high [4] and appears as a sequelae of CSOM [5]. Studies of the structure of mucus glands in the middle ear showed that the epithelium first invaginates at the location of high-density goblet cells and then different ramifications develop to lead to different structure types and sizes. Nevertheless, it was noticed that mucus glands of the middle ear can degenerate and lose their ability to produce mucins [3] likely because when OM resolves, the ear contains less factors sustaining inflammation and mucin production.

As explained before, the healthy middle ear epithelium contains few goblet cells that are concentrated in some patches of mucociliary epithelium mainly close to the Eustachian tube. But in the case of OM, the simple layer epithelium remodels into a pseudostratified epithelium. Several studies have demonstrated by histology techniques (Hematoxilyn and eosin staining on cuts of paraffin-embedded tissues) that the middle ear epithelium exhibits more secretory cells as well as ciliated cells in numerous parts of the middle ear epithelium [68]. Secretory cells are positive to periodic acid Schiff (PAS) staining detecting the presence of glycoconjugates, which are mainly in mucin proteins. Smirnova et al. [9] demonstrated that these mucins are secreted in the MEEs as they were PAS positive using a slot blot.


The Mucus in the Middle Ear: An Important Role for Mucins


OM is characterized by the presence of fluid in the middle ear cavity, called effusions that can be serous or mucous. Serous effusions do not contain mucins and are not viscous. On the contrary, mucous effusions are highly viscous and contain a high content of mucins [10]. An in vitro test of transportability of a bead under magnetic attraction also indicated that mucous effusions are less transportable than serous ones, suggesting that mucous effusions are difficult to clear in the middle ear [11]. Serous effusions contain proteins similar to the blood, so it is suggested that serous fluids are the result of a passive transudate of blood components in the middle ear due to a negative pressure in the middle ear likely because of the Eustachian blockade during inflammatory OM [12, 13]. Even if there are some conflicting findings from different research groups, serous effusions are believed to show better outcomes of the disease, whereas mucoid effusions are suggested to predict chronic otitis media with effusions (COME) [11, 14]. The study of Matkovic et al. [15] also contributed to this hypothesis as among 108 effusions collected, only 6 % were mucoid for patients having OM diagnosed for less than 3 months, whereas 95 % were mucoid for patients having the disease for more than 3 months. The large differences in medical outcomes and effusion and middle ear mucosa (MEM) characteristics prove that various cellular and molecular pathways act in the evolution of the disease. The production of large amounts of mucins necessitates the differentiation of goblet cells in the epithelium and the development of mucin glands. Indeed, the reabsorption of water in serous fluids and the concentration of their proteins are believed to participate to turning serous effusions into mucous ones [16]. Contrary to serous effusions, mucous effusion production necessitates the active process of producing mucins (exudates). But as a large number of proteins from the blood are also present in the mucous effusions (as albumin the predominant one), a passive diffusion of proteins and liquid is also probably implicated in the accumulation of mucous effusions.

Ion transport and water channels are also believed to play an important role in bringing water in the middle ear cavity and participate to serous and mucous effusion production. The healthy middle ear has to be kept without fluid contrary to the inner ear for a good transmission of the sound vibrations. Herman et al. [17] suggested the importance of water channels and ion transports. Experiments conducted in Mongolian gerbil’s middle ear cells showed that the absorption of fluid in the middle ear was dependent on an osmotic gradient created by sodium and potassium adenylpyrophosphatase (ATPase)-dependent channels. The impairment of this ion flux has been shown in the lungs of rabbits in response to hydrogen peroxide that is produced during oxidative processes as well as hypoxia, a condition likely to appear in OM [18]. The aquaporins (AQP)1, 4, and 5, channels regulating the water homeostasis in cells, were detected in the Eustachian tube and MEM of rats as well as the epithelial sodium channels (ENaCs) [19]. In experimental OM in rats induced by Eustachian tube obstruction, ENaC and AQP were deregulated from 1 to 8 weeks after Eustachian tube obstruction, suggesting their implication in the water imbalance leading to fluid presence in the middle ear [20].

Effusions are composed of mucins but contain other proteins (antibacterial proteins, cytokines, etc.), lipids, deoxyribonucleic acid (DNA), and bacterial components [10, 14, 21], some of these substances being remains of dead bacteria and epithelial cells. Mucins , the major macromolecular component of epithelial mucus, are very high-molecular-weight proteins constituted of a backbone where numerous sugar side chains are added as a posttranslational modification (glycosylation with glycotransferases enzymes). These glycoconjugates are linked to the mucins in the Golgi and are then stored in secretory granules, waiting to have a signal to merge with the membrane and be released in the extracellular compartment [22]. Mucins are widely studied as their regulation is often a key determinant of diseases as cancer, lung diseases, and gastrointestinal diseases.

Mucins are classified by their protein backbone that is encoded by different mucin genes called MUC. MUC transcripts are big (until 15 kilo bases), so is the protein backbone, accounting for 15–50 % of mucin mass, and can contain 400 to more than 11,000 amino acids [22].The major posttranslational modification of mucins is O-glycosylations, consisting in O-glycans attached to tandem repeats rich in serine and threonine all along the backbone. N-glycosylation is also observed but in a lesser extent. More than 20 human MUC genes have been identified—about the same number in mice. Considering the size of their gene transcript and protein backbone, but also their many glycoconjugates, the analysis of the MUC proteins is difficult. In the respiratory tract, 12 MUC genes have been identified and less in the ear, probably because mucins are more studied in the airways. Studies of mucins in the MEM and MEEs seem to show that MUC5B is the predominant mucin in the ear of patients having OM, whereas healthy subjects show very low levels of mucin [68, 23, 24]. But other mucins have been detected, apparently in lower amounts, which are MUC5AC, MUC2, and MUC4 [7, 13, 24, 25]. MUC5B has been detected by transcript analysis and protein assay: Preciado et al. [23] detected MUC5B protein by mass spectrometry in MEEs from COME patients and Lin et al. [7] by immunohistochemistry on the mucosa of patients with mucoid OM, whereas non-inflamed mucosa did not react with either of the antibodies anti-MUC5B and anti-MUC4. It has been noticed that the Eustachian tube of mucoid OM patients had MUC5B, MUC4, and also MUC5AC and MUC1 glycoproteins [8]. From the same study, electron microscopy of secretions from COME patients showed the presence of chain-like polymeric mucin. Some studies have detected the presence of messenger ribonucleic acid (mRNA) transcripts of MUC1, MUC2, MUC3, MUC4, MUC5AC, MUC5B, MUC7, MUC8, MUC9, MUC11, MUC13, MUC15, MUC16, MUC18, MUC19, and MUC20 [2426], but gene expression does not always reflect the protein production and secretion. Indeed, Thornton et al. [27] showed that mucin gene expression in the airways was not always correlated to the presence of the protein. Mucins can be secreted or attached to the cell membrane. Among the mucins detected in the middle ear, we can notice that MUC5B and MUC5AC are secreted mucins, whereas MUC1 and MUC4 and are membrane-tethered mucins [22].

The overproduction of mucins leads to mucoid effusions that are hard to clear by the middle ear. Efforts have been made to try to prevent mucin overproduction, but a recent article pointed to the necessary presence of MUC5B in the innate immune response of airways and the ear. A study directed by Dr. Christopher Evans showed that the knockout of Muc5b in mice had a fast and dramatic effect on the mortality and morbidity due to infection of the airways leading to systemic infection [28]. Histology of the lungs showed an overproduction of Muc5ac probably to compensate the lack of Muc5b, but failed to protect the airways from infection. The ears were also infected by different bacteria and contained liquid as well as signs of inflammation. Thus, Muc5b glycoprotein is needed in the airways and the ear to protect mice against bacterial invasion and shows its central role in the innate immunity.


Antimicrobial Molecules in Effusions


MEEs contain other molecules that participate to the defense against pathogens. Antibacterial proteins efficiently kill bacteria and are very important in the innate immunity mechanisms. Defensins are broad-spectrum antimicrobial peptides, small (30–45 amino acids), rich in cationic amino acids, and stabilized by disulfide bounds that protect them from proteases [29]. Defensins have antimicrobial properties towards bacteria and viruses, are able to inhibit some bacteria toxins [30], and have pro-inflammatory activities stimulating cytokine and chemokine production [31]. Surprisingly, defensins have not been studied in patient samples, but in vivo and in vitro studies of experimental OM showed their induction in response to bacterial infection of the middle ear [32, 33]. Human β-defensin 2 (HBD2) was studied in vitro in order to determine the molecular pathways implicated in its induction. In human middle ear epithelial cells (HMEEsCs), HBD2 is under the control of the pro-inflammatory cytokine interleukin (IL)1-β that activates Raf-MEK1/2 (mitogen-activated protein kinase kinase), the mitogen-activated protein kinase (MAPK) pathway [34]. Non-typable Haemophilus influenzae (NTHi) is also able to induce the expression of HBD2 first activating the toll-like receptor (TLR) 2 and then inducing protein 38 (p38) MAPK pathway [32]. HBD1 and HBD2 have also shown their ability to reduce Streptococcus pneumonia (SP), Haemophilus influenzae (Hi), and Moraxella catarrhalis (MC) growth in a liquid broth assay [35]. In Chinchilla, the orthologue of human β-defensin 3, chinchilla β-defensin 1, CBD1, had potent antimicrobial activity against SP, Hi, and MC [36]. Furthermore, chinchillas pretreated with recombinant CBD1 resulted in lower colonization of NTHi in the nasapharynx [37]. But bacteria are able to resist to defensins when they are growing in biofilms: Jones et al. [38] demonstrated that HBD3 binds to extracellular DNA constituting the matrix of NTHi biofilms, leading to the sequestration of HBD3 and thus diminishing the biological activity of an important defense of innate immunity.

Other antibacterial molecules are also part of the innate immune defense. Among them, the lysozyme is a cathelicidin (a cationic peptide) that has various effects, primarily damaging the membrane of bacteria. Lysozyme is present in MEEs of pediatric patients having OM, especially in mucous effusions compared to serous ones [12, 13, 39]. Giebink et al. [40] showed that the concentrations of lysozyme are more important in the MEEs of patients with COME positive for bacteria culture and suggested that this antibacterial agent was not only produced by polymorphonuclear leukocytes but also by the middle ear epithelium that accounted for 50–80 % of the lysozyme in the middle ear. Experimental OM in animals also demonstrated higher lysozyme detection in the middle ear: in response to MC in the Guiney pig [41] and in response to SP in chinchilla, this study also showed that more lysozyme were observed even when heat-killed bacteria were injected in the middle ear, suggesting that the production of lysozyme might be activated in response to membrane components of bacteria. Furthermore, mouse depleted of lysozyme also showed a higher susceptibility to OM development after SP infection [42], underlining the importance of lysozyme in the innate immune defense against bacteria.

Finally, some other antibacterial molecules poorly studied seem to play a role in the middle ear defense to pathogens: surfactant proteins as short palate, lung, and nasal epithelium clone (SPLUNC)-1, small cationic peptides, halocidin, and xylitol [29, 43].


Recognition of Pathogens



The System of the Complement


The complement system is a biochemical cascade composed of several peptides normally present as inactive forms. This system can be activated by different sequential cascades of enzymatic reactions in which proteins are sequentially cleaved and activated. The resulting effector molecules are C3a and C5a, also called anaphylatoxins. They are the most potent activation products of the complement that are able to induce a large diversity of effects as bacterial cytotoxicity, induction of pro-inflammatory cytokines production, and inflammatory cell activation [44]. The activation of the complement system depends on three pathways. The classical pathway consists in the recognition of immunoglobulin IgG and IgM complexes formed around pathogens that activate the C1 complex, activating C4 molecules to induce the activation of C3 and C5. The alternative pathway is triggered by carbohydrates, lipids, and proteins found on pathogens: C3 mediates the activation of the cascade. And the lectin pathway recognizes sugars at the microbial surface and leads to the activation of C4, C3, and finally C5.

Mediators of the complement activation have been found in MEEs and the MEM. Recently, He et al. [45] analyzed molecules of the component in effusions of children with recurrent OM by the enzyme-linked immunosorbent assay (ELISA). High amounts of C3a, C5a, and sC5-b9 were detected in the MEEs of patients having OM for more than 6 weeks. The concentration of C5a was also strongly correlated to the concentration of IL-6 and IL-8 pro-inflammatory cytokines, suggesting a link between complement activation and the inflammatory effect they induce. Complement transcript induction was also observed in HMEEsCs in vitro in response to SP and influenza A virus (IAV). Another study was conducted on effusions of patients with COME to assay the complement activation (C3a and C3 cleavage fragments) by ELISA and western blot analysis [46]. High concentrations of complement molecules were found and C3 activation was evaluated at 40 % of the total amount of C3 protein. They also noticed that C3a concentration was higher when effusions stayed longer in the ear and when children had multiple tube insertions, pointing C3a levels as a marker of the chronicity of OM. The complement activation leads to lysis of pathogens; this has been verified by Niarko-Markela and Meri [47] with erythrocytes of Guiney pigs exposed to MEEs from patients with otitis media with effusion (OME). Thirteen of the 38 MEEs tested had direct endogenous hemolytic activity, and 27 enhanced serum-initiated lysis. They also detected high levels of terminal complement complexes demonstrating the strong activation of the complement.


Receptors of the Innate Immunity


Multiple cell types and especially epithelial cells that are in contact with the external environment express innate immune receptors as Toll Like Receptors (TLRs). In the mucosal environment, mast cells and dendritic cells also express TLRs. The TLRs are pattern recognition receptors that recognize pathogen-associated molecular patterns (PAMPs). The activation of TLRs leads to the production of molecules also implicated in the innate immune response, as chemokines, cytokines, interferons (IFNs), and antimicrobial molecules described before. TLRs are type I transmembrane receptors with an extracellular N-terminal region with leucin-rich repeats and an intracellular toll-IL-1 receptor (TIR) domain. TLRs can form homodimers or heterodimers. The homodimers of TLR4 , TLR5, TLR11, and the heterodimers of TLR2-TLR1 or TLR2-TLR6 bind to their respective ligands at the cell surface, whereas TLR3, TLR7-TLR8, TLR9, and TLR13 localize to the endosomes, where they sense microbial and host-derived nucleic acids. TLR4 localizes at both the plasma membrane and the endosomes. They each recognize specific types of PAMPs, for example, TLR1-TLR2 and TLR1-TLR6 recognize acylated peptides, TLR4 recognizes lipopolysaccharide (LPS), TLR3 targets double-stranded ribonucleic acid (RNA), and TLR9 recognizes bacterial DNA. TLR signaling is induced by their dimerization, dependent on ligand binding. All TLRs except TLR3 have a signaling pathway dependent on myeloid differentiation factor 88 (MyD88), activating the transcription factor NF-ĸB and then the expression of pro-inflammatory cytokines [48].

Several TLRs have been identified in the MEM and MEEs. TLR2, TLR4, TLR5, and TLR9 were found at the level of RNA and proteins in the MEM of both OM and non-OM patients [49]. For TLR2, TLR4, and TLR5, no difference of expression was found between non-OM and OM MEM, but their concentration was lower in the mucosa of patients with CSOM. In consequence, it was suggested that the clinical recovery of OM depends on TLR expression in the middle ear. There are conflicting evidences considering TLRs in MEEs and the correlation with the presence of bacteria. Lee et al. [50] observed effusions of patients with OME having lower TLR2, TLR6, and TLR9 mRNA when they are prone to have persistent OM and the level of TLRs is higher in culture-positive MEEs. Another study demonstrated the inverse for TLR9: less TLR9 mRNA is detected in culture-positive MEEs [51], whereas Lee et al. [52] failed to see any difference in TLR2, TLR4, TLR5, and TLR9 mRNA. Studying TLRs in MEEs might be accurate for assaying their presence in immune cells but does not take into account the epithelial cells playing an important role in the immune defense through TLRs. This might explain the differences described before. Nevertheless, animal studies showed that defects in TLR2 and TLR4 lead to the persistence of inflammation and mucosal metaplasia during OM [53].


Role of the Inflammation in OM


Inflammation is a central innate immune response activated by pro-inflammatory mediators (chemokines, cytokines) in order to attract and activate immune cells, stimulate the various innate immune defenses, and initiate the adaptive immune response to pathogens. This is a very efficient process involving different mediators and cells, but also deleterious if it does not resolve when the pathogens are no longer present. Inflammation is suspected to participate to the absence of resolution of OM especially in the case of COME and CSOM.


Pro-inflammatory Cytokines and Chemokines in OM

Pro-inflammatory cytokines and chemokines are characteristic of the inflammatory process: They are induced at early stages of the innate immune response until advanced stages to sustain the inflammation and to stimulate the adaptive immune response. Cytokines are usually associated to different types of immune response and can be produced by different cell types: epithelial cells, macrophages, neutrophils, dendritic cells, etc. Thus, cytokines that are known to play an important role in the innate immunity are tumor necrosis factor α (TNF-α), interleukins IL-1, IL-10, IL-12, IFNs, and chemokines like IL-8. The adaptive immunity is usually characterized by the cytokines IL-2, IL-4, IL-5, transforming growth factor β (TGF-β) , IL-10, and IFN-γ production, TGF-β and IL-10 being able to repress inflammation. In addition, granulocyte-macrophage-colony-stimulating factor (GM-CSF) and granulocyte-colony-stimulating factor (G-CSF) are cytokines known to stimulate the differentiation of hematopoetic cells. Pro-inflammatory cytokines and chemokines have been detected many times in MEEs; the Table 7.1 summarizes some of the more recent studies assaying the content of cytokine protein in MEEs or MEM by ELISA or their transcripts by polymerase chain reaction (PCR) [15, 49, 50, 5461]. Among the 11 studies listed, 12 different cytokines were detected in samples collected from children or adults with acute otitis media (AOM), OME, COME, and CSOM. Despite the fact that it is complicated to compare the quantity of cytokines in each study, it seems that IL-8, IL-6, IL-12, and IL-2 are detected in almost all the effusions. IL-1β, IL-4, IL-5, TNF-α, IFN-δ, TGF-β, and IL-10 were detected in less MEEs (40–80 % for the studies detailing this parameter), and TNF-β showed conflicting evidences considering its presence. IL-8, IL-6, IL-12, IL-1β, TNF-α, IFN-δ, and IL-10 seem to be in higher concentrations in culture-positive samples and CSOM that are usually characterized by the presence of a strong bacterial infection. IL-8 and IL-10 were detected in higher concentrations in mucoid effusions compared to serous ones.


Table 7.1
Pro-inflammatory mediator detection in middle ear effusions (MEEs) or middle ear mucosa (MEM) of patients with otitis media (OM)








































































































































































Cytokine

Detected in

References

IL-8

36 MEEs, COME patients, 92 %

[54]


108 MEEs, OM +/− 3 months, +

[15]


96 MEEs, OME, + (RNA)

[50]


46 MEEs, OME, +

[55]

IL-6

20 MEEs, AOM, +

[56]


96 MEEs, OME, + (RNA)

[50]


72 ears, MEM, COM/CSOM, + (RNA)

[49]


75 MEEs, OME persistent and/or recurrent, 83 %

[57]

IL-12

96 MEEs, OME, + (RNA)

[50]


80 MEEs, OME adults, 100 %

[58]

IL-1β

36 MEEs, COME patients, 67 %

[54]


108 MEEs, OM +/− 3 months, +

[15]


30 MEEs children, 38 MEEs adults OM, +

[59]


72 ears, MEM, COM/CSOM, + (RNA)

[49]


75 MEEs, OME persistent and/or recurrent, 58 %

[57]

IL-2

108 MEEs, OM +/− 3 months, +

[15]


80 MEEs, OME adults, 75 %

[58]

IL-4

80 MEEs, OME adults, 41 %

[58]


26 MEEs, OME, +

[60]

IL-5

80 MEEs, OME adults, 52 %

[58]


26 MEEs, OME, +

[60]

TNF-α

36 MEEs, COME patients, 77 %

[54]


108 MEEs, OM +/− 3 months, +

[15]


30 MEEs children, 38 MEEs adults OM, +

[59]


96 MEEs, OME, + (RNA)

[50]


72 ears, MEM, COM/CSOM, + (RNA)

[49]


75 MEEs, OME persistent and/or recurrent, 38 %

[57]

IFN-δ

108 MEEs, OM +/− 3 months, +

[15]


96 MEEs, OME, + (RNA)

[50]


72 ears, MEM, COM/CSOM, + (RNA)

[49]


80 MEEs, OME adults, 83 %

[58]


75 MEEs, OME persistent and/or recurrent, 51 %

[57]

TGF-β

45 MEEs, adults, OME, +

[61]

IL-10

108 MEEs, OM +/− 3 months, +

[15]


96 MEEs, OME, + (RNA)

[50]


80 MEEs, OME adults, 18 %

[58]


45 MEEs, adults, OME, +

[61]

TNF-β

36 MEEs, COME patients, 0 %

[54]


108 MEEs, OM +/− 3 months, +

[15]


In the column “Detected in,” the following information is given: number of MEEs or samples of MEM; type of OM detected; adult is specified—if nothing written, the samples come from children; % of samples positive for the analysis (+ means not specified in the study, assuming 100 %)

OME otitis media with effusion, AOM acute otitis media, COM chronic otitis media, COME chronic otitis media with effusion, CSOM chronic suppurative otitis media, RNA ribonucleic acid

MEEs contain a variety of cytokines acting both in promoting inflammation and regulating the adaptive immune response. Some of them are produced in very high content especially when the bacterial infection persists. The chronic stages of OM also show a diversity of cytokines in high concentration in the middle ear, suggesting a persistence of inflammation in absence of pathogens. The Eustachian tube obstruction due to inflammation and the low transportability of mucoid effusions might limit the efficiency of the clearance of killed pathogens, letting PAMPs in the middle ear that still stimulate the immune responses, so do the cytokines in mucoid fluids that might accumulate without the possibility of being cleared from the middle ear. Some defects in cytokine production, dependent on genetic and environmental influence, might also explain why children tend to be prone to recurrent and persistent OM. Cytokines exhibit strong effects that, if not balanced, can lead to a disproportionate immune response. These cytokines are produced by epithelial cells but also immune cells. They are granulocytes as neutrophils, basophils, and eosinophils and phagocytic cells as macrophages and dendritic cells, all detected in MEEs of patients with OME [60, 6264].


Innate Immunity to Adaptive Immunity in OM: Activation of Lymphocytes

As described before, several immunoregulator cytokines are present in MEEs of patients, underlining the importance of the role of the adaptive system in OM. They can be divided in two groups: TH1 and TH2 (meaning lymphocyte T helper). They represent the ability of lymphocytes T to differentiate in TH1 type, inducing cell-mediated immunity and inflammation, or TH2 that mediates the humoral immunity through the production of antibodies by differentiated lymphocytes B. The different cytokines detected in the MEEs show the activation of both pathways. CD4+ T cells were detected in MEEs several times [9, 61], T cells that are naïve or differentiated. The lymphocyte subpopulation in MEEs was analyzed by flow cytometry in the study of Skotnicka et al. [65]. CD3+ T cells were dominating the population of lymphocytes, and the T helpers CD4+ were the majority. The ratio of CD4+/CD8+ cells was significantly higher in MEEs, but the proportion of CD8+ cells was lower in MEEs than in blood. These immune cells are suspected to come from adenoids in patients presenting this abnormality, as the population of lymphocytes of adenoids is important and similar to the middle ear [66]. Lymphocytes B have been identified in the middle ear as well. A study assayed the presence of lymphocytes in relation to the presence of antibodies against specific bacteria in 238 MEEs of patients with AOM [67]. The percentage of lymphocytes was higher in the ears with bacteria-specific antibodies than in the ears without, which correlated with a faster resolution of OM. The activation of the TH2-specific pathway inducing the differentiation of lymphocytes B to produce specific antibodies seem to also play an important role in the resolution of OM.


Part II: Molecular and Cellular Mechanisms Implicated in OM Pathogenesis


OM is a very common disease in children that sometimes evolves into chronic OM for reasons not yet understood. In order to prevent the evolution of the disease in a chronic stage that is difficult to treat, we need to understand the mechanisms implicated in OM development in response to bacteria, and how their interaction evolves into chronic OM . The innate immune system plays a central role in OM, so researchers investigated the different mechanisms implicated in its activation during the infection of the middle ear. In vivo and in vitro models were developed to better understand how the middle ear epithelium responds to bacteria, hoping to find new strategies to treat patients with OM.


In Vivo and In Vitro Models to Study OM Pathogenesis



Animal Models


Animals are useful models to investigate the cellular and molecular mechanisms implicated in OM. They permit to control the type of infection, the different stages of a disease, as well as the genetic background of the biological material. Comparing to in vitro studies, in vivo models allow taking into account the entire immune system and the interaction between different cell types being important in the resolution of infections. But we have to keep in mind that animal models have their limitations as their responses to pathogens might not be the same as the human ones, the differences being dependent on the species chosen. For the study of OM, rodents are widely used: chinchilla, mouse, rat, Guiney pig, and gerbil. According to the literature, mice and chinchillas are the main animals used in laboratories to study OM. Mouse is the first animal models used now as they are small, with a very controllable genetic background, easy to use in laboratories because a high diversity of reagents are compatible with this species. Nevertheless, mice have a very small middle ear, which is less convenient to induce OM by surgery as well as collecting MEEs. Chinchilla offers the possibility to have a bigger middle ear: the review of Ryan et al. [68] compared the middle ear volume observed in different studies. The average middle ear volume of the chinchilla is about 1.5 ml3, whereas the one of a mouse is about 0.05 ml3, so the middle ear volume of the chinchilla is 30 times bigger than the one of the mouse. It is consequently easier to manipulate the middle ear and recover MEEs that are sufficient in quantity to do several biological assays. The anatomy of the chinchilla ear has also been shown to be very close to the human one [69]; they do not often develop spontaneous OM [70], and they show similar responses to virus and bacteria in the course of OM compared to humans even if the pathogens colonizing humans are not usually the same as those of chinchillas (see [71]). Rats are also used in several studies and have the advantage of having a bigger middle ear and more availability of reagents than chinchillas. Several interesting studies used rats to do a time course analysis of OM development.

OM is often induced by experimental obstruction of the Eustachian tube, leading to a negative pressure in the middle ear [72, 73]. Infection by bacteria can be coupled to this procedure to mimic better human OM. Bacterial injection can be made through the tympanic membrane but damaging this membrane lets other contaminants the possibility to enter in the middle ear. Injection via the ventral bulla is preferred as it does not damage the tympanic membrane and avoids contaminations. But it necessitates skills in microsurgery to avoid damaging the vessels and airways around the bulla. Infections post surgery can occur and may modify the immune response in the middle ear. Considering these limitations, Stol et al. [74] developed a noninvasive murine model adapted from a previous rat model. They used a pressure cabin at 40 kPa which induced pneumococci translocation from the nasopharyngeal cavity to the middle ear; the maximum bacteria load appearing 96 h post infection with the bacteria. Inflammation was confirmed with the secretion of IL-1β and TNF-α in the middle ear. This model has the advantage to avoid the limitations due to the surgery but probably does not permit to have homogenous OM development between animals. Another disadvantage should be considered: other parts of the body might be affected by the difference of pressure, especially for medium- or long-term experiments. Finally, pressure cabins might not be easy to use, expensive to buy, and might not permit to expose enough animals at the same time.

Human pathogens are studied in animal models as relevant clinical strains. But they are evolutionary adapted to humans and usually not animals, which can bring bias in these experimental studies. Nevertheless, the effects observed in experimental OM induced in animals and especially mice are very close to the observations made in humans: OM induced in mice having different genetic backgrounds with different strains of SP, Hi, and MC have shown similar inflammatory and mucosal effects even if the duration of the disease, the intensity of the responses, and the ability of resolution of OM where variable [68].

OM was also evaluated in mutant mouse strains; a strategy often used to investigate the implication of a specific gene in the apparition or the course of a disease. Mutations are natural or induced in laboratories. For the study of OM, we have to be careful choosing the type of mutations. Mutations in genes acting in the development of the middle ear might create some morphologic defects, influencing the responses of the middle ear. Mutating central genes in the immunity might also compromise the response to pathogens. And the deletion of a gene sharing similar functions with other genes sometimes leads to compensation mechanisms that may compensate the loss of functionality. Otherwise, this type of biological material offers great possibilities in studying spontaneous OM or pathogen-induced OM.


In Vitro Models


Transformed middle ear epithelial cells are now widely used to investigate the mechanisms implicated in bacteria effects, especially focusing on inflammatory and mucoid effects. This type of biological material permits to assay the effect of live bacteria, bacteria lysates, purified bacteria proteins, inflammatory mediators, etc., in a homogenous cell type which is useful but lacks the interaction with other cell types, especially the immune cells that produce mediators regulating epithelial cells. Knowing the limitations of cells in vitro, it is a very useful tool that gives us opportunities we cannot have with animals: the analysis of mechanisms implicated in a biological effect is more easy.

The human middle ear epithelial cell line HMEEC-1 was created by Dr. David Lim in 2002 using a retrovirus containing E6/E7 genes of human papillomavirus type 16 to transform primary middle ear epithelial cells from adults [75]. This type of transformation is known to regulate the cell cycle acting on the retinoblastoma (RB) tumor suppressor gene limiting the repression of the cell cycle and mediating the degradation of p53 protein also implicated in cell cycle repression [76, 77].

The mouse middle ear epithelial cell (mMEEsC) line was made by Dr. Jizhen Lin laboratory in 2005 [78]. Middle ear epithelial cells were isolated from mice and transformed by the large T-antigen of the simian virus 40 (SV40) A-gene. These cells have the property to be temperature sensitive: At 33 °C, the SV40 antigen is active and stimulates the cell cycle. But at 37/39 °C, SV40 is inactivated and cells differentiate, expressing markers of epithelial cells such as keratins and collagens. In our laboratory, we have noticed that these cells can be cultured several weeks at air liquid interface and form a single layer epithelium.

Other cell types were used: the middle ear cell line from chinchillas immortalized by SV40 [79] and the primary chinchilla middle ear epithelial cells (CMEEsCs) [80] or primary middle ear epithelial cells from adults successfully differentiated at air liquid interface in a ciliary and secretory epithelium [81]. Our laboratory tried to culture middle ear epithelial cells from children middle ear but because of the low amount of cells available during these procedures, we were unable to successfully grow them.


Interactions Between Pathogens and Ear Epithelial Cells


After having passed the eventual innate immune barriers in the middle ear, bacteria reach the middle ear epithelium. There, they adhere to the cells using adherence molecules varying depending on the bacteria. This part is focused on NTHi adhesion and invasion in airway and middle ear cells as NTHi is the main pathogen implicated in OM and as its interactions with epithelial cells has been widely studied.

NTHi is a gram-negative nonencapsulated bacterium that adheres and invades the middle ear. Several factors are necessary to its ability to invade epithelial cells. NTHi is able to secrete IgA proteases that increase its ability to adhere and invade the bronchial epithelial cells NCI-H292 [82]. Several factors produced by NTHi bind to host proteins: The protein F, a homolog of SP lamin-binding proteins, is an adhesion factor that binds to the lamin of host cells [83]. Protein E has also been implicated in epithelial cell adhesion and the interaction with extracellular matrix proteins [84, 85]. Protein D, an outer membrane lipoprotein highly conserved, is important for NTHi adherence and is now used in pneumococcal polysaccharide conjugate vaccines that include monoacetylated protein D carriers, vaccines that showed their efficiency preventing OM development [86]. Finally, the phosphocholine (PCho) groups associated to the lipooligosaccharide of NTHi showed several times its implication in NTHi adherence as well as its ability to form biofilms [8688]. PCho also present in SP was found to interact with the platelet-activating factor receptor (PAF receptor) as PAF present also PCho motifs recognized by this receptor [88]. In addition, the study of Van Schilfgaarde et al. [89] demonstrated that different clinical strains of NTHi elicited different patterns of adhesion, implying that some factors produced by specific strains might play a critical role in NTHi adhesion. They suggested that high molecular weight proteins are implicated in the virulence of the different clinical strains of NTHi.

NTHi has been detected on cells (adherence) and in cells (invasion). The presence of NTHi at the surface of epithelial cells was demonstrated by bacteria culture after infection [90], fluorescent microscopy techniques [89], and scanning electron microscopy, bacteria being mainly located on the top of non-ciliated cells [91]. Different molecular pathways in epithelial cells were found to play an important role in NTHi adhesion and invasion. The cytoskeleton with microtubules and actin were rearranged and necessary for NTHi virulence [9092]. Macropinocytosis was demonstrated to be an important internalization mechanism of NTHi [91], and other studies found the implication of lipid rafts [92]. NTHi is also able to produce outer membrane vesicles that contain factors that will help the bacteria to invade hosts. These vesicles have a diameter of 20–200 nm and contain DNA, adhesins, and other enzymes [93]. These vesicles are internalized by caveolin-dependent mechanisms and elicit the production of immune proteins as IL-8 and the antibacterial protein LL-37, surprisingly enhancing NTHi invasion in epithelial cells. Thus, different mechanisms are implicated in NTHi internalization in epithelial cells and might be dependent on cell culture conditions and the NTHi strain used.

In the middle ear, bacteria are found planktonic or organized in biofilms [94, 95]. The growth of bacteria in biofilms gives them the ability to hide from the immune system of the host and resist to antibiotics due to the extracellular matrix the bacteria create around them [95]. Biofilms of main pathogens in OM were detected in the middle ear of patients: SP, Hi, and MC but also Staphylococcus aureus and Staphylococcus epidermidis. But even if these bacteria are known to resist to high antibiotic quantities, we do not know yet how they can invade human cells. It is possible that biofilms are a defense mechanism to protect bacteria from a hostile inflammatory environment, offering them a niche to wait that immune responses decrease in order to better infect the host.


Regulation of Mucin Production and Mucous Cell Metaplasia in OM: Role of Pro-inflammatory Mediators


Inflammation and effusion production are characteristic of OM. As explained before, clinical inflammation seems to appear at the early stages of OM development when the middle ear tries to fight the infection by bacteria and/or viruses until more chronic stages even if the bacteria count seems lower. But this is not the case for mucin production that is mainly observed at later stages of OM course. Serous effusions are suspected to mainly come from the transudation of liquid and proteins from the blood, whereas mucous effusions need the active process of mucin production. Mucins are produced by goblet cells of the middle ear epithelium and mucus glands, which are easily detected in the MEM of patients with OME and COME, but at very low levels in healthy middle ears. These observations suggest that several factors play a role in the remodeling of the epithelium of the middle ear. This part of the chapter will try to review the different factors implicated in mucin production and mucous cell metaplasia, focusing on the inflammatory mediators that seem to play a crucial role in this process.


Infection of the Middle Ear by Bacteria Results in Pro-inflammatory Mediators Expression and Secretion In In Vivo And In Vitro Models


MEEs from patients with OM contain high concentrations of a panel of pro-inflammatory cytokines (see Sect. 3.3.1). In vivo studies were conducted to try to replicate the conditions of infection occurring in OM in order to analyze the expression and secretion of pro-inflammatory cytokines in the MEM and in MEEs mainly in mice, chinchillas, and rats (Table 7.1 and 7.2).


Table 7.2
Pro-inflammatory mediators detected in mouse models of otitis media (OM)


































































Reference

Bacterial species/component

OM induction

Duration of infection

Cytokines deregulated (time point)

Technique

[98]

Heat killed Hi

TTI one ear, other ear not injected as control

6 h

TNF-α, IL-1α, IL-10, IL1β, IL-6, MIP-2, KC

Gene chip (mRNA) on MEM

[101]

LPS

TTI, saline as control

3, 6, 12, or 24 h

IL-1β, TNF-α, MIP-2, KC (24h, other ND); GM-CSF (6h)

ELISA of ME wash

[74]

SP

Pressure cabin, nasopharyngeal infection

48, 96, 144 h after nasopharyngeal infection

IL-1β, TNF-α (48hrs and 96hrs)

ELISA of MEH

[73]

NTHi purified endotoxins

ETO and then TTI, saline as control

3 days, 2 weeks, 2 months

TNF-α (3 time points), IL-1β (3 days), IFN-δ not consistent

ELISA of MEEs, in situ hybridization MEM

[99]

SP then Influenza virus

Intranasal exposure, saline as control

15 days after SP infection

IL-1α, pro-IL-1β, TNF-α, MIP-2

PCR (mRNA) of MEM

[97]

NTHi lysates

TTI, saline as control

1 day and 7 days

MIP-2 most induced gene

Microarray (mRNA)

[102]

endotoxins from Salmonella typhimurium

TTI, saline as control

6 h, 12 h, 1 day, 3 days, 7 days, and 14 days

IL-1α (up to 3 days), TNF-α (day 1 and 3)

Only gold members can continue reading. Log In or Register to continue

Stay updated, free articles. Join our Telegram channel

Oct 17, 2016 | Posted by in PSYCHIATRY | Comments Off on Basic Science Concepts in Otitis Media Pathophysiology and Immunity: Role of Mucins and Inflammation

Full access? Get Clinical Tree

Get Clinical Tree app for offline access