Skip to content
Publicly Available Published by De Gruyter September 29, 2018

The role of the innate immune system on pulmonary infections

  • Michelle Galeas-Pena , Nathaniel McLaughlin and Derek Pociask EMAIL logo
From the journal Biological Chemistry

Abstract

Inhalation is required for respiration and life in all vertebrates. This process is not without risk, as it potentially exposes the host to environmental pathogens with every breath. This makes the upper respiratory tract one of the most common routes of infection and one of the leading causes of morbidity and mortality in the world. To combat this, the lung relies on the innate immune defenses. In contrast to the adaptive immune system, the innate immune system does not require sensitization, previous exposure or priming to attack foreign particles. In the lung, the innate immune response starts with the epithelial barrier and mucus production and is reinforced by phagocytic cells and T cells. These cells are vital for the production of cytokines, chemokines and anti-microbial peptides that are critical for clearance of infectious agents. In this review, we discuss all aspects of the innate immune response, with a special emphasis on ways to target aspects of the immune response to combat antibiotic resistant bacteria.

Introduction

The lung is a warzone. The primary function – gas exchange, puts this internal organ in direct contact with the outside world. As such, the pulmonary epithelium is exposed to a daily onslaught of infectious agents, hazardous pollutants and toxic particles. While the lung has evolved many defensive mechanisms to maintain the epithelial borders, pneumonia remains the most common cause of hospitalizations in adults and it is the fifth cause of death in children 5–9 years old (Greenbaum et al., 2014). It is therefore imperative to understand the immune responses in this complex organ in order to help reduce infection and promote pulmonary well-being.

Figure 1: 
Abbreviated examples of innate immune system response in the respiratory epithelium interface to different types of pathogens.
(A) Influenza infection: viral particles in the airway lumen are detected by TLR3 and TLR7 in the epithelial cells, triggering an initial response by NF-κβ, activating Type I and II interferons, producing CXCL2 and other chemokines that attract effector cells like macrophages and T cells as well as cytokines, such as IL-1β, IL-17 and IL-18. (B) Klebsiella pneumoniae infection: this Gram-negative bacteria is sensed by TLR2 and TLR3, unfolding a signaling cascade by MyD88, TRIF and NF-κβ, this cascade results in chemokines such as CXCL1, CXCL2 and CXCL5, promoting the recruitment of immune cells and releasing cytokines, most notably, IL-17, IL-6 and TNF-α. (C) Aspergillus fumigatus infection: the airway epithelium detects connidia or hyphae by TLR-2, TLR4 and Dectin-1. Decting-1 activates PCK- , recruiting neutrophils and other immune cells that exert antifungal activity by NADPH and reactive oxygen species (ROS). TLR2 and TLR4 activate PI3K, MAPK and ERK1/2 which results in production of cytokines and chemokines such as IL-8, IL-1α, IL-1β, IL-17, TNF-α, CCL3, CCL4, CxCL1 from the epithelium and immune cells. This response not only contributes to the acute defense of the lung, but also further activate the adaptive immune response.
Figure 1:

Abbreviated examples of innate immune system response in the respiratory epithelium interface to different types of pathogens.

(A) Influenza infection: viral particles in the airway lumen are detected by TLR3 and TLR7 in the epithelial cells, triggering an initial response by NF-κβ, activating Type I and II interferons, producing CXCL2 and other chemokines that attract effector cells like macrophages and T cells as well as cytokines, such as IL-1β, IL-17 and IL-18. (B) Klebsiella pneumoniae infection: this Gram-negative bacteria is sensed by TLR2 and TLR3, unfolding a signaling cascade by MyD88, TRIF and NF-κβ, this cascade results in chemokines such as CXCL1, CXCL2 and CXCL5, promoting the recruitment of immune cells and releasing cytokines, most notably, IL-17, IL-6 and TNF-α. (C) Aspergillus fumigatus infection: the airway epithelium detects connidia or hyphae by TLR-2, TLR4 and Dectin-1. Decting-1 activates PCK- , recruiting neutrophils and other immune cells that exert antifungal activity by NADPH and reactive oxygen species (ROS). TLR2 and TLR4 activate PI3K, MAPK and ERK1/2 which results in production of cytokines and chemokines such as IL-8, IL-1α, IL-1β, IL-17, TNF-α, CCL3, CCL4, CxCL1 from the epithelium and immune cells. This response not only contributes to the acute defense of the lung, but also further activate the adaptive immune response.

The scope of this article is to review the role of the innate immune system in combating respiratory borne pathogens and provide an update with the latest research in the field.

Epithelial cells and mucosal immunity

Barrier and mucosal immunity activities of the lung epithelium

The epithelium is the front line of defense against infection. The initial responsibility of the pulmonary epithelium is to act as a physical barrier between the lumen and vasculature. It does this through the formation of tight junctions involving claudins, occludins and adherens such as E-cadherin and β-catenin, among others (for an excellent review see Wittekindt, 2017). The integrity of the epithelial layer in the lungs and alveolar spaces is central to preventing infectious agents from colonizing and disseminating from the lung as well as preventing fluid build-up (edema) in the lung. Tight junction formation can be altered by viral infections that target the epithelium. The two most common are influenza, which impairs claudin-4 (Short et al., 2016) and respiratory syncytial virus (RSV) which downregulates the expression of occludins and claudin-1 (Kast et al., 2017). Bacterial infections such as Pseudomonas aeruginosa can also result in edema due to loss of zonula occludens-1 proteins (Higgins et al., 2016).

Pattern recognition receptors (PRRs)

Loss of epithelial integrity can have dire consequences, allowing dissemination of infectious agents into the blood stream while at the same time, letting fluid build-up in the lung and reducing gas exchange. It is therefore critical that the pulmonary epithelium monitor the apical surface for signs of infection. It does this through PRRs, which are integral to the innate immune response.

Human lung epithelium and innate immune cells express TLR1, TLR2, TLR3, TLR4, TLR5, TLR6, TLR7, TLR8, TLR8, TLR9 and TLR10, each recognizing different antigens (Table 1). In gene expression studies, it was found that dsRNA (such as in influenza infection), recognized by TLR3, was the most effective stimulus of airway epithelial cells, inducing a strong cytokine and chemokine response (Doyle et al., 2003; Sha et al., 2004). In contrast, the TLR2 and TLR5 response from alveolar macrophages elicits a more effective response against bacterial agents and allergens, promoting an enhanced response from the airway epithelium (Hess et al., 2010). TLR4 recognizes Gram-negative bacteria and can induce up-regulation of TLR3 in alveolar macrophages (Ding et al., 2017). Clinically, this can result in elevated viral susceptibility due to synergistic amplification, triggering an exacerbated inflammatory response during subsequent viral infections. This is important because patients surviving severe bacterial pneumonia can have extensive tissue damage or even death in secondary viral infections (Ding et al., 2017).

Table 1:

Human TLRs and their ligands.

TLR Ligand
TLR1 Triacyl lipoproteins
TLR2 Peptidoglycans and heat shock proteins, lipoprotein and high mobility group 1 (HMGB1) amphoterin, lypotechoic acid, zymosan.
TLR3 dsRNA (self and viral)
TLR4 Heat shock proteins, LPS, RSV fusion protein, fibrinogen, fibronectin, heparan sulfate, hyaluronic acid, mouse mammary tumor virus envelope, HMGB1.
TLR5 Flagellin
TLR6 Lypotechoic acid, zymosan, triacyl lipoproteins
TLR7 ssRNA (self and viral)
TLR8 ssRNA (self and viral)
TLR9 DNA (self, viral and bacterial)
TLR10 Unknown

The function of TLRs is to induce inflammation that can lead to clearance of the infectious agent. However, TLRs can also be immunosuppressant, depending on context. For example, LPS along with certain growth factors, such as vascular endothelial growth factor (VEGF), granulocyte-macrophage colony-stimulating factor (GM-CSF) and granulocyte colony stimulating factor (G-CSF) and cytokines like interleukin (IL)-1β, IL-6, can induce myeloid-derived suppressor cells in the lungs, that exert an anti-inflammatory response and limit inflammation-derived tissue damage (Ray et al., 2013).

The importance of TLR signaling in response to infection make them attractive therapeutic targets. According to the National Institutes of Health (NIH), there are 48 clinical trials exploring TLR activation as targets for cancer, metabolic disorders and infectious diseases (https://clinicaltrials.gov). For example, the synthetic molecule PUL-042 targets TLR2 and TLR9, enhancing their effects on innate immune response. In a model of influenza infection in mice, oseltamivir, a common drug used to treat influenza A, was given in combination with PUL-042, improving the rate of survival after receiving lethal doses of influenza A (Leiva-Juarez et al., 2018). TLRs are also being used as potential adjuvants to increase vaccine efficacy. The combinatorial stimulation of TLR4 and NOD2 can enhance the cellular and humoral response when used in vaccines (Tukhvatulin et al., 2016). The concomitant activation of TLR1/2, TLR3, TLR4 and NOD1 can augment the inflammatory response to asthma and infections after stimulation with their agonists such as Pam3CSK5, LPS, poly (I:C), R-837 and iE-DAP, promoting the release of IL-6, IL-8, GM-CSF, ICAM1, RANTES and HLA-DR (Månsson Kvarnhammar et al., 2013). In Table 2, we summarize the clinical trials pertaining to infectious diseases in the lung.

Table 2:

Clinical trials using TLR to address lung infections.

Title Conditions Interventions Locations URL
Enhancing influenza vaccination in seniors with TLR (Toll like receptor) agonists Influenza Vaccination in Seniors Drug: resiquimod | Drug: placebo gel University of British Columbia, Vancouver General Hospital, Vancouver, British Columbia, Canada https://ClinicalTrials.gov/show/NCT01737580
Toll-like receptor 2 gene polymorphism, serum cytokines and susceptibility to disease severity or treatment response of pulmonary tuberculosis Pulmonary tuberculosis| genetic variants of host|immune response of host Kaohsiung Chang Gung Memorial Hospital, Kaohsiung Hsien, Taiwan|Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan https://ClinicalTrials.gov/show/NCT00772408
Possible relation of Toll-like receptors and Nitric oxide to chronic lung disease lung diseases Procedure: tracheal aspirate fluid Children’s Mercy Hospital & Clinic, Kansas City, MO, USA|Children’s Hospital and Regional Medical Center (Seattle), Seattle, WA, USA https://ClinicalTrials.gov/show/NCT00245167
Ventilator-associated pneumonia (VAP) in intensive care unit (ICU) Ventilator-associated pneumonia Zakynthinos, Larisa, Mezourlo, Greece https://ClinicalTrials.gov/show/NCT00935285
Innate immunity and respiratory syncytial virus (RSV) infection in children Respiratory syncytial virus infection University of Wisconsin-Madison, Madison, WI, USA https://ClinicalTrials.gov/show/NCT00593918
Immune response to Toll-like receptor 9-agonist adjuvanted pneumococcal vaccination in HIV infected adults HIV Infections Biological: Pneumococcal vaccines + CPG 7909|Biological: pneumococcal vaccines Department of Infectious Diseases, Aarhus University Hospital, Aarhus, Denmark https://ClinicalTrials.gov/show/NCT00562939
Intradermal influenza vaccine in the young Influenza viral Infections Drug: Imiquimod ointment|Drug: aqueous cream|biological: intradermal influenza vaccine|biological: Intramuscular influenza vaccine The University of Hong Kong, Queen Mary Hospital, Hong Kong, China https://ClinicalTrials.gov/show/NCT02103023
Safety and immunogenicity of VAX125 influenza vaccine in community-living adults >=65 years of age Influenza Biological: VAX125 JCCT, Lanexa, KS, USA|University of Rochester, Rochester, NY, USA|Coastal Carolina Research Center, Charleston, SC, USA https://ClinicalTrials.gov/show/NCT00966238
Multi-drug resistant tuberculosis in Korea Tuberculosis National Masan Tuberculosis Hospital, Clinical Research Center, Masan, Republic of Korea|National Medical Center, Seoul, Republic of Korea https://ClinicalTrials.gov/show/NCT00341601

Another type of PRRs are the NOD-like receptors (NLR). The main NLRs are NLR3 and NLR4. NLRs form inflammasomes, which are multimeric proteins regulating caspase-1 activity and triggering inflammation via IL-1β, and IL-18 (Guo et al., 2015). During infection, inflammation is a helpful mechanism for microbial clearance. However, inflammasome activation and the subsequent release of IL-1β also have deleterious effects on the local tissue through the recruitment and activation of neutrophils (Ceballos-Olvera et al., 2011; Rathinam et al., 2012). NLRs are important in limiting infections. NLR4 is a potent inducer of pyropoptosis, limits bacterial intracellular growth (Ceballos-Olvera et al., 2011) and is important in the clearance of multiple bacterial infections including Salmonella typhimurium, Shigella flexneri, P. aeruginosa and Burkholderia pseudomallei (Chiang et al., 2018). NLRs are also the link between intestinal commensal flora and early innate response to infections in the lung, as demonstrated in a study where depletion of gut microbiota, but not upper respiratory tract commensals impaired early innate clearance of Klebsiella pneumoniae infection via NLRs (Clarke, 2014).

Mucins as defense mechanisms

Mucins are extracellular proteins produced primarily by goblet cells and club cells (Fahy and Dickey, 2010), as well as pseudostratified columnar ciliated cells throughout the airway epithelium. The most common mucins in the lung are MUC5AC and MUC5B which play important roles in defensive barrier function as a component of the viscous mucus that covers the ciliated structures in the airway (Thornton et al., 2008; Kim, 2012).

During the acute inflammation phase of viral and bacterial infections, tumor necrosis factor (TNF)-α, up-regulates MUC1 production promoting resolution of inflammation by inducing IL-10 production in dendritic cells (Li et al., 2010; Choi et al., 2011). Despite its antimicrobial affects, certain bacteria have developed mechanisms to overcome mucin. Pseudomonas aeruginosa can bind to MUC1 (Kim, 2012) and modulate inflammatory responses by reducing the phagocytic activity of classically activated (M1) macrophages (Kato et al., 2017a,b), contributing to microbial resistance by hijacking part of the host’s innate immune response.

Antimicrobial peptides (AMPs) and other immunoregulatory peptides

AMPs were initially discovered and named for their direct antimicrobial properties (Zasloff, 2002). They are abundantly produced in the lung, where they act as a first line of defense against infection. Defensins (alpha and beta), LL-37, RegIIIγ and surfactant proteins are examples of predominant AMPs in the lung.

Defensins alpha and beta, are produced primarily by neutrophils and epithelial cells in humans. In mice, alpha-defensins are found in Paneth cells, but not in neutrophils (Eisenhauer and Lehrer, 1992). Early studies determined that alpha defensins have a potent anti-viral action by directly binding to viral particles, impairing their ability to infect cells (Daher et al., 1986; Wilson et al., 2013). Beta-defensins also possess chemokine activity, indirectly enhancing the immune response against infections by inducing the production of MCP-1, Gro-α and monocyte-derived chemokine (MDC) from monocytes (Petrov et al., 2013). Beta-defensins have been a focus of intense study due to their roles not only as antimicrobial agents but also as immunomodulators. In this context beta-defensins have a pro-inflammatory effect, acting as chemoattractants to T cells, immature dendritic cells and macrophages (Semple and Dorin, 2012). Beta-defensin can be induced directly by P. aeruginosa (Scharf et al., 2012), whereas during Brucella abortus infection, it is indirectly induced by infected monocytes production of IL-1β (Hielpos et al., 2015).

LL-37 is a member of the cathelicidin family and is produced by epithelial and myeloid cells (Dürr et al., 2006). In humans, LL-37 can inactivate LPS and possesses direct antimicrobial effects by disrupting the bacterial membrane (Choi et al., 2017). It also stimulates epithelial cell release of IL-8 and transactivation of epithelial growth factor receptor (EGFR) (Sandra Tjabringa et al., 2005). CRAMP (cathelicidin-related AMP), the murine orthologue, plays an immunomodulatory role during bacterial sepsis through the reduction of TNF-α and nitric oxide production, as well as serving as a chemoattractant (Cirioni et al., 2006). Because of these properties, LL-37 has been actively studied as a potential therapeutic agent in sepsis (Bowdish et al., 2005; Cirioni et al., 2006).

Another important antimicrobial peptide is RegIIIγ, a C-Type lectin AMP that specifically targets Gram-positive bacteria. In the adult lung, RegIIIγ is induced in a STAT3 dependent manner and is critical for the clearance of methicillin-resistant Staphylococcus aureus (MRSA) pneumonia (Choi et al., 2013). Interestingly, neonates have reduced expression of RegIIIγ, contributing to the susceptibility of infection and impairment of MRSA clearance (Fitzpatrick et al., 2017).

In the airway, surfactant proteins A (SP-A) and D (SP-D) are part of the collectin AMP family (Coya et al., 2015). Pathogens interact with the leptin domain of collectins, resulting in opsonization and subsequent phagocytosis (Wright, 2005). Unlike SP-A, SP-D has a strong antiviral response to influenza and has been suggested as a possible therapy to enhance mucosal immunity during influenza outbreaks (Hillaire et al., 2014), or as a biomarker due to its elevated serum levels during certain acute and chronic infections (Gaunsbaek et al., 2013; Güzel et al., 2014). Surfactant protein C (SP-C) is less characterized, but it has been shown to have immunomodulatory effects via JAK/STAT activation in lung repair after ARDS (Jin et al., 2018).

Lipocalin-2 is a siderophore binding AMP required for the clearance of K. pneumoniae (Chan et al., 2009) and Streptococcus pneumoniae (Warszawska et al., 2013). However, lipocalin can suppress macrophage activation and impairing bacterial clearance. Furthermore, the higher the levels of lipocalin-2 correlate with worse outcomes in pneumococcal infection (Warszawska et al., 2013). Similarly, in a sepsis model, high levels of lipocalin-2 expression in lung tissue were observed in C57BL/6 J mice, but not in A/J mice, suggesting that differential genetic background in IL-10 production by macrophages, which regulates lipocalin-2 expression might play a role in the severity of sepsis and could be used as a prognosis marker (Vazquez et al., 2015).

Complement

The complement system are plasma proteins that function as key effectors in the immune response against pathogens by enhancing phagocytosis, production of active mediators of inflammation like C5a and C3a and by attacking the pathogen’s cell membrane. Most of the complement proteins are made by hepatocytes, but can also be produced by circulating monocytes, tissue macrophages and epithelial cells in the lung. Complement circulates as inactive proteins, but can be activated by three different pathways, classical, alternate and leptin.

Hyper-activation of complement can result in tissue damage. For instance, it has been reported that H5N1 avian influenza causes excessive activation of complement causing acute lung injury (Sun et al., 2013) leading to acute respiratory distress syndrome (ARDS). Inhibition of complement activation in this model helps alleviate lung injury (Sun et al., 2013). In a model of Mycobacterium tuberculosis component factor C7 contributes to lung injury and bacterial persistence by promoting the formation and maintenance of granulomas (Welsh et al., 2012). The modulation of the complement system is an active field of research in inflammation-related tissue damage. Interestingly, alpha-defensins can modulate complement by inhibiting its production (Hiemstra, 2015).

Cellular immunity, components and effectors

Cellular immunity is central to the innate immune response in the lung where resident immune cells monitor and maintain homeostasis as well as cells that emigrate to the tissue when activated by infection or tissue damage. The main components of cellular immunity are macrophages, monocytes, dendritic cells, neutrophils and innate immune cells. For examples and summary of the innate response during infection by three different pathogens, see Figure 1.

Monocytes and macrophages

Macrophages are the central innate cellular response to infection. They kill directly by phagocytizing infecting agents, while recruiting other immune cells through the release of multiple cytokines and chemokines. Following insult, circulating monocytes differentiate into macrophages or dendritic cells (DCs) to exert their innate immune response in the lung tissue. In contrast, tissue resident macrophages are already in the lung. These first responders play important roles in immunomodulation, tissue repair and homeostasis in the lung after microbial clearance. Lung macrophages are commonly divided in two groups: alveolar macrophages (AM) and interstitial macrophages (IM). For a detailed description of how these cells are characterized, see Kopf et al. (2014). While research suggests macrophages can exist in a transient state from one type to another, both AMs and IMs play important roles in homeostasis and host protection (Sugimoto et al., 2015).

Alveolar macrophages patrol the alveolar surface and are generally the predominant immune cell in a healthy lung. These cells develop during the first week after birth from fetal monocytes that populate the lung during development (Guilliams et al., 2013; Gomez Perdiguero et al., 2015), and are able to self-replenish with very little contribution from circulating monocytes (Yona et al., 2013). Most AM are polarized towards an anti-inflammatory phenotype called M2 and collaborate in lung development in newborns (Jones et al., 2013).

AMs have varying roles in response to viral infection, depending on the agent. During RSV and influenza infection, AMs promote viral clearance (Wang et al., 2012; Eichinger et al., 2015). While influenza clearance requires the production of high amounts of type I and type III interferons (Wang et al., 2012), AMs suppress RSV replication independent of interferon production (Makris et al., 2016).

Interestingly, AMs are not always beneficial. During human metapneumovirus (hMPV) infection, AMs contribute to lung pathology by facilitating early entry and replication in the lung, as well as production of inflammatory soluble factors (Kolli et al., 2014). Further, AMs are potential HIV-1 reservoirs in otherwise healthy patients with undetectable plasma viral loads, which impairs AMs phagocytic function making these patients more susceptible to respiratory infections (Cribbs et al., 2015). In tuberculosis, low genetic expression of macrophage migration inhibitory factor (MIF) is a risk factor for certain populations due to a decreased ability to manage early infection and dissemination of mycobacteria (Das et al., 2013). This phenomenon highlights the seemingly opposing roles for the innate immune system.

IMs are also believed to have homeostatic functions and defend against pathogens (Sugimoto et al., 2015). In contrast to AMs, proliferation of IMs is high when stimulated with LPS or IFN-γ, but are shorter lived (Sugimoto et al., 2015), suggesting that IMs have an important role in innate immunity in the lungs.

In general, it has been proposed that AMs are central in pathogen clearance, whereas IMs play a greater role in immunomodulation of the adaptive response due to their higher expression of MHCII (Steinmüller et al., 2000). However, in some infections, IMs play a key role in clearance. IMs are important in Chlamydia muridarum infection, where the pathogen targets IMs for infection, thus activating them. Upon IM activation, an IFN-γ induced cytokine response promotes pathogen clearance, suggesting that imbalances in the different macrophage populations can lead to C. muridarum associated pathology (Gracey et al., 2015).

Similar to macrophages, circulating Monocytes also have antimicrobial functions. Monocytes expressing CCR2 and Ly6C as well as monocyte-derived DCs have a pivotal role in conidial clearance during A. fumigatus infection indirectly by recruiting neutrophils that will attack conidia and directly by ingesting conidia during the process of DC differentiation, resulting in direct conicidial activity (Espinosa et al., 2014).

Dendritic cells

Lung DCs serve as sentinels against infections, representing the interface between innate and adaptive immune response (Belz et al., 2004; Condon et al., 2011). Plasmacytoid DCs play a central role in viral infections such as RSV or H1N1, eliciting a type I interferon protective response, activating the adaptive immune machinery (Garg et al., 2012; Cormier et al., 2014). Although, in certain highly pathogenic influenza strains, it is suggested that the increased amount of DC recruitment may contribute to the pathogenicity of the strain (Soloff et al., 2014). Recently, researchers are also focusing on DCs participation in bacterial infections. For instance, there is active research on the role of plasmacytoid DCs against early stages of Chlamydia pneumoniae, where they promote an innate pro-inflammatory state in the lung but have a suppressive role in the lymph node (Crother et al., 2012).

Neutrophils

Neutrophils are part of the early innate immune response. In humans, they are the most common white blood cell in the circulation. During infection, neutrophils respond to chemokines (IL-8, MIP-1) and travel to the site of inflammation. Lung migrating leukocytes do not use the usual rolling and tethering that is required in other peripheral organs for extravasation, allowing higher neutrophil concentration in the lungs during inflammation (Burns et al., 2003). Confocal microscopy studies have demonstrated that pericytes, contractile cells found around the endothelium of capillaries, assist in the process of neutrophil transmigration (Proebstl et al., 2012).

Neutrophils exert direct and indirect antimicrobial effects during the innate immune response to infection. They directly kill the infecting agent by phagocytosis, by creating neutrophil extracellular traps (NET) or through activation of other innate immune cells, like macrophages. For example, it has been reported that neutrophils are required for macrophage activation against helminth Nippostrongylus brasiliensis infection, accelerating clearance (Chen et al., 2014). Neutrophils also participate in the clearing of Aspergillus fumigatus hyphae through myeloperoxidase and NADPH oxidase and conidia through lactoferrin-mediated iron sequestration (Garth and Steele, 2017). Recent reports have postulated that Dectin-1 and Mac-1 induced activation of PKC-γ is key to fungal clearance because it is required for reactive oxygen species production upon cellular uptake (Li et al., 2016). Because of their relevance in infectious and autoinflammatory diseases, neutrophils have become an interesting target for therapy. It has been found that CCR5 and FPR1 are required for neutrophil recruitment and blocking these molecules could be advantageous in limiting acute lung injury during bacterial pneumonia (Grommes et al., 2014). Neutrophil recruitment in bacterial clearance requires the action of chemokines CXCL5 and CXCL1, which are activated via IL-17/IL-17R axis (Chen et al., 2016). Furthermore, it has been suggested that neutrophil recruitment and damage in the lung is a dysregulation of the CXCL1/CXCR2 at the target tissue (Sawant et al., 2015).

Innate lymphoid cells (ILCs)

ILCs are recently described tissue resident T-cells. In 2013, Spits et al. proposed a nomenclature for ILCs, categorizing them in three groups based on the cytokines and transcription factors they produce (Spits et al., 2013). NK cells and ILC1 make up Group I. Group I cells produce IFN-γ and TNF. ILC2 are the only member of Group 2. Upon activation by IL-33 and IL-25, ILC2 are the main producers of IL-4, IL-5, IL-9, IL-13 and AREG (Ebbo et al., 2017; Richards, 2017). NCR+ILC3, NCR-ILC3 and LTi cells are all part of Group 3, which produce IL-17, IL-22 and IFN-γ (Spits et al., 2013). Although there are similarities with conventional T-cells, they play different roles in immunity, tissue remodeling and development (Ebbo et al., 2017). ILCs are unique in that they lack RAG genes but share TCR signaling molecules such as ICOS, LAT and LCK (Paclik et al., 2015). This allows them to be non-specifically activated, as they lack antigen-specific receptors (Spits et al., 2013; Mjösberg and Spits, 2016). However, ILCs are capable of presenting antigens to T-cells during bacterial and parasitic infections (Hepworth et al., 2013; Oliphant et al., 2014). Despite the proposed classification, ILCs have plasticity, which makes them change their phenotype upon challenges in the microenvironment (Silver et al., 2016).

An important subgroup of ILC1, NK cells, are an important player during control of cancer development and certain infectious diseases due to their cytotoxic activity. During Toxoplasma gondii infection (Dunay and Diefenbach, 2018), NK cells are the major source of the anti-inflammatory cytokine IL-10 (Wagage et al., 2014) or in response to other pathogens, IFN-γ (Richards, 2017). In response to K. pneumoniae pneumonia, NK cells orchestrate the innate immune response by producing type I interferons, which in turn activate alveolar macrophages to exert their protective role in the infected lung (Ivin et al., 2017). NK cells have also been demonstrated to be essential for the control of murine cytomegalovirus acute stage of infection, underlining its well-known role in viral infections (Andrews et al., 2003). Despite the classification proposed by Spits, under certain circumstances, ILCs can produce cytokines that do not fit within his typical nomenclature. For example, during influenza pneumonia NK cells produce IL-22, which is an important cytokine primarily involved in repair and tissue regeneration (Pociask et al., 2013; McAleer and Kolls, 2014). ILC2s have been associated with lung protective effects during viral infection, particularly during influenza by producing amphiregulin, which helps with tissue regeneration (Monticelli et al., 2011; Califano et al., 2018).

Since the characterization of ILCs, it was thought that ILC1 and ILC2 are the only groups found in the lung. However, several studies have found that ILC3 is also present in the mouse lung (De Grove et al., 2016). Many of these studies are in the context of cancer, where ILC3s are linked to the production of IL-22, TNF-α, IL-8 and IL-2, and contribute to the formation of tertiary lymphoid structures, eliciting a protective role in cancers (Carrega et al., 2015; Ferlazzo et al., 2016). Some studies have also found ILC3s in the context of allergy and helminthic infections in the lower respiratory tract (Taube et al., 2011; Tait Wojno and Artis, 2016). A recent study by Gray et al., found that in the lungs of newborn mice there is a large population of ILC3 cells. These cells are proposed to migrate during early stages of development from the small intestine to the lung, where they are crucial in the resistance to pneumonia in the newborn mice (Gray et al., 2017), connecting the gut microbiota and mucosal immunity with lung innate immunity.

T cells

Some immune cells, like the T helper type 17 (Th17) cells produce cytokines that not only are relevant in the immune innate response to infections in the lungs, but also promote the integrity of the epithelium by promoting proliferation of epithelial cells. The cytokines IL-17A and IL-22 are crucial in the homeostasis and tissue repair. As part of the mucosal immunity, IL-17A and IL-22, produced by CD4 T cells, are associated with innate defense in K. pneumoniae pneumonia (Aujla et al., 2008).

γδ T-cells are a subset of T cells that have innate immune cell properties and thought to act as a bridge between the innate and adaptive responses. They migrate to mucosal surfaces early in development and are considered a tissue resident T cell. γδ T-cells are critical in clearing many infections. In Pneumocysti carinii infection, γδ T-cells modulate host’s susceptibility due to their interaction with CD8+ T cells and local production of IFN-γ (Steele et al., 2002). In M. tuberculosis infection they produce IFN-γ that triggers macrophages to produce nitric oxide as a defense mechanism (Pahari et al., 2018). Enhancing the γδ T-cell population and promoting their activation is under study as a viable approach for the development of tuberculosis vaccines (Xia et al., 2016). In other bacterial infections, such as K. pneumoniae, γδ T-cells are required for TNF-α and IFN-γ production and bacterial clearance and survival (Moore et al., 2000), making these cells a critical component of the innate immune system.

Inducible structures

Following an infection, inflammatory cells may persist in the lung and can form tertiary lymphoid structures. These structures are categorized by the cellular make up. The two most common inducible pulmonary lymphoid structures are granulomas and inducible brochus-associated lymphoid tissue (iBALT). These structures form to contain pathological agents or to help promote localized immunity to other infections.

Granulomas are commonly formed through infection by fungi and mycobacteria such as M. tuberculosis (TB). The TB granuloma is formed by macrophages and T cells that surround the mycobacteria, creating a pathogen-host interaction. These granulomas vary in general composition but must maintain a balance between pro-inflammatory and anti-inflammatory to protect from the bacteria getting out (Gideon et al., 2015). However, the pro-inflammatory and antimicrobial effects in the granuloma are not just based on cytokines, but also in antimicrobial peptides (AMPs) and reactive oxygen species (ROS). A recent proteomic study of the spatial distribution in granulomas revealed that the necrotic center contains a highly pro-inflammatory environment with high levels of AMPs and ROS, as well as pro-inflammatory cytokines like IFN-γ, whereas the surroundings tend to be anti-inflammatory, hypothesizing that such distribution is relevant to contain the pathogen while limiting tissue destruction (Marakalala et al., 2016).

Another inducible structure is the induced iBALT. This structure is composed of B and T cell zones, dendritic cells as well as high endothelial venules and lymphatics (Moyron-Quiroz et al., 2004). These structures contain antigen presenting cells that generate memory T and B cell responses in response to subsequent infection promoting a rapid, focal response to secondary infection (Moyron-Quiroz et al., 2004, 2006; Halle et al., 2009). iBALT formation occurs in response to a number of bacterial and viral infections (Hwang et al., 2016) and is dependent upon the production of IL-17 (Rangel-Moreno et al., 2011) and requires a number of chemokines such as CXCL13, CCL19 and CXCL21 (Rangel-Moreno et al., 2007).

Summary and outlook: innate immunity and antibiotic resistance

Antibiotic resistance is a major public health problem although many new drugs are being discovered. Unfortunately, resistance to those drugs quickly develops, raising concerns about new treatments and management. Multi-target mechanisms, targeting virulence factors as well as several steps in the same pathway, could enhance the innate immune response (Oldfield and Feng, 2014). Many have proposed using AMPs as alternatives to conventional agents to mitigate antibiotic resistance. While bacteria can develop resistance to AMPs, they retain the ability to enhance the innate immune response (Andersson et al., 2016). While direct administration of AMPs is an attractive therapy, concerns of toxicity (Falagas et al., 2006) and efficacy have limited this approach to mostly topical applications (Andersson et al., 2016). Therefore, alternative approaches that locally induce AMPs at the site of infection may be a safer therapy.

Cytokine administration may offer a more practical therapeutic approach. A number of cytokines can induce AMPs in localized areas of infection and injury. IL-22 is once such cytokine. It is not only critical for protection of pulmonary epithelium during infection (Kumar et al., 2013; Pociask et al., 2013) it induces production of lipocalin (Aujla et al., 2008) and RegIIIγ (Barthelemy et al., 2018) which are critical for clearance of Gram-negative bacteria (Aujla et al., 2008) and protection from secondary bacterial infection (Barthelemy et al., 2018). It also induces complement (C3) which aids in the clearance of pneumococcal pneumonia (Trevejo-Nunez et al., 2016).

Certain drugs can also have indirect or secondary antimicrobial properties through increased AMPs, thus increasing innate immunity. For example, statins promote the formation of extracellular traps, although they also have a negative impact in oxidative stress formation and phagocytosis (Chow et al., 2010). It has been demonstrated that by targeting a step in in the sterol pathway has the effect of boosting the production of neutrophil and macrophage extracellular traps (NETs and METs) and AMP LL-37 (Liu et al., 2008; Chow et al., 2010; Lin et al., 2012), showing a protective effect of statins to S. aureus pneumonia.

In a different study, it was found that the administration of picostin, a molecule similar to the isoprenoid metabolite (E)-4-hydroxy-3-methyl-but-2-enyl pyrophosphate (HMBPP) in combination with IL-2, elicited an expansion of Vγ2Vδ2 T cells. This expansion conferred protection against tuberculosis-related lesions following mycobacterial infection, helping to reduce bacterial burden (Chen et al., 2013). Interestingly, a bioengineered vaccine providing HMBPP as a booster expanded γδ T cells subsets which effectively increased early innate response against S. typhimurium (Workalemahu et al., 2014).

While many studies aim to affect the T-cell compartment, increasing macrophage activation and killing have also been studied. Delivery of lipids in apoptotic body-like liposomes can promote phagosome activation and maturation, increasing intracellular killing of BCG through acidification and reactive oxygen species generation. This approach has also been used against P. aeruginosa conferring a higher capacity to innate system to handle drug resistant infections (Poerio et al., 2017).

Other methods have been developed that do not rely on AMPs. An example of this is the use chimeric proteins derived from domains 6 and 7 of human complement factor H fused with the IgG Fc domain. In this approach, the alternative pathway of the complement system is uninhibited, resulting in bacterial clearance by opsonization and subsequent phagocytosis (Blom et al., 2017). This was tested in models of infection with Haemophilus influenzae, Neisseria meningitidis and Neisseria gonorrhoeae with similar bactericidal effects (Shaughnessy et al., 2016; Wong et al., 2016). Another area of research is the use of peptidoglycan recognition proteins (PGRP). These proteins can contribute to limit infections by three potent bacteriostatic effects: oxidative, thiol and metal stress that combined become bactericidal, resulting in death of bacteria exposed to it. Also, it has been established that PGRPs provide enhanced innate immune responses as well as complement-dependent responses, such as phagocytosis and AMP production (Dziarski and Gupta, 2018).

Award Identifier / Grant number: 1RO1HL122760-01

Funding statement: Division of Intramural Research, National Institute of Allergy and Infectious Diseases, Funder Id: 10.13039/100006492, Grant Number: 1R21AI117569-01A1. National Heart, Lung, and Blood Institute, Funder Id: 10.13039/100000050, Grant Number: 1RO1HL122760-01.

References

Andersson, D.I., Hughes, D., and Kubicek-Sutherland, J.Z. (2016). Mechanisms and consequences of bacterial resistance to antimicrobial peptides. Drug Resist. Updat. 26, 43–57.10.1016/j.drup.2016.04.002Search in Google Scholar PubMed

Andrews, D.M., Scalzo, A.A., Yokoyama, W.M., Smyth, M.J., and Degli-Esposti, M.A. (2003). Functional interactions between dendritic cells and NK cells during viral infection. Nat. Immunol. 4, 175–181.10.1038/ni880Search in Google Scholar PubMed

Aujla, S.J., Chan, Y.R., Zheng, M., Fei, M., Askew, D.J., Pociask, D.A., Reinhart, T.A., McAllister, F., Edeal, J., Gaus, K., et al. (2008). IL-22 mediates mucosal host defense against Gram-negative bacterial pneumonia. Nat. Med. 14, 275–281.10.1038/nm1710Search in Google Scholar PubMed PubMed Central

Barthelemy, A., Sencio, V., Soulard, D., Deruyter, L., Faveeuw, C., Le Goffic, R., and Trottein, F. (2018). Interleukin-22 immunotherapy during severe influenza enhances lung tissue integrity and reduces secondary bacterial systemic invasion. Infect. Immun. 86, e00706–e00717.10.1128/IAI.00706-17Search in Google Scholar PubMed PubMed Central

Belz, G.T., Smith, C.M., Kleinert, L., Reading, P., Brooks, A., Shortman, K., Carbone, F.R., and Heath, W.R. (2004). Distinct migrating and nonmigrating dendritic cell populations are involved in MHC class I-restricted antigen presentation after lung infection with virus. Proc. Natl. Acad. Sci. USA 101, 8670–8675.10.1073/pnas.0402644101Search in Google Scholar PubMed PubMed Central

Blom, A.M., Magda, M., Kohl, L., Shaughnessy, J., Lambris, J.D., Ram, S., and Ermert, D. (2017). Factor H-IgG chimeric proteins as a therapeutic approach against the Gram-positive bacterial pathogen Streptococcus pyogenes. J. Immunol. 199, 3828–3839.10.4049/jimmunol.1700426Search in Google Scholar PubMed PubMed Central

Bowdish, D.M.E., Davidson, D.J., Scott, M.G., and Hancock, R.E.W. (2005). Immunomodulatory activities of small host defense peptides. Antimicrob. Agents Chemother. 49, 1727–1732.10.1128/AAC.49.5.1727-1732.2005Search in Google Scholar PubMed PubMed Central

Burns, A.R., Smith, C.W., and Walker, D.C. (2003). Unique structural features that influence neutrophil emigration into the lung. Physiol. Rev. 83, 309–336.10.1152/physrev.00023.2002Search in Google Scholar PubMed

Califano, D., Furuya, Y., Roberts, S., Avram, D., McKenzie, A.N.J., and Metzger, D.W. (2018). IFN-γ increases susceptibility to influenza A infection through suppression of group II innate lymphoid cells. Mucosal Immunol. 11, 209–219.10.1038/mi.2017.41Search in Google Scholar PubMed PubMed Central

Carrega, P., Loiacono, F., Di Carlo, E., Scaramuccia, A., Mora, M., Conte, R., Benelli, R., Spaggiari, G.M., Cantoni, C., Campana S., et al. (2015). NCR+ILC3 concentrate in human lung cancer and associate with intratumoral lymphoid structures. Nat. Commun. 6, 8280.10.1038/ncomms9280Search in Google Scholar PubMed

Ceballos-Olvera, I., Sahoo, M., Miller, M.A., Del Barrio, L., and Re, F. (2011). Inflammasome-dependent pyroptosis and IL-18 protect against Burkholderia pseudomallei lung infection while IL-1β is deleterious. PLoS Pathog. 7, e1002452.10.1371/journal.ppat.1002452Search in Google Scholar PubMed PubMed Central

Chan, Y.R., Liu, J.S., Pociask, D.A., Zheng, M., Mietzner, T.A., Berger, T., Mak, T.W., Clifton, M.C., Strong, R.K., Ray, P., et al. (2009). Lipocalin 2 is required for pulmonary host defense against Klebsiella infection. J. Immunol. 182, 4947–4956.10.4049/jimmunol.0803282Search in Google Scholar PubMed PubMed Central

Chen, C.Y., Yao, S., Huang, D., Wei, H., Sicard, H., Zeng, G., Jomaa, H., Larsen, M.H., Jacobs Jr., W.R., Wang, R., et al. (2013). Phosphoantigen/IL2 expansion and differentiation of Vγ2Vδ2 T cells increase resistance to tuberculosis in nonhuman primates. PLoS Pathog. 9, e1003501.10.1371/journal.ppat.1003501Search in Google Scholar PubMed PubMed Central

Chen, F., Wu, W., Millman, A., Craft, J.F., Chen, E., Patel, N., Boucher, J.L., Urban, J.F., Kim, C.C., and Gause, W.C. (2014). Neutrophils prime a long-lived effector macrophage phenotype that mediates accelerated helminth expulsion. Nat. Immunol. 15, 938–946.10.1038/ni.2984Search in Google Scholar PubMed PubMed Central

Chen, K., Eddens, T., Trevejo-Nunez, G., Way, E.E., Elsegeiny, W., Ricks, D.M., Garg, A. V, Erb, C.J., Bo, M., Wang, T., et al. (2016). IL-17 Receptor signaling in the lung epithelium is required for mucosal chemokine gradients and pulmonary host defense against K. pneumoniae. Cell Host Microbe 20, 596–605.10.1016/j.chom.2016.10.003Search in Google Scholar PubMed PubMed Central

Chiang, C.-Y., Uzoma, I., Moore, R.T., Gilbert, M., Duplantier, A.J., and Panchal, R.G. (2018). Mitigating the impact of antibacterial drug resistance through host-directed therapies: current progress, outlook, and challenges. MBio. 9, e01932-17.10.1128/mBio.01932-17Search in Google Scholar PubMed PubMed Central

Choi, S., Park, Y.S., Koga, T., Treloar, A., and Kim, K.C. (2011). TNF-α is a key regulator of MUC1, an anti-inflammatory molecule, during airway Pseudomonas aeruginosa infection. Am. J. Respir. Cell Mol. Biol. 44, 255–260.10.1165/rcmb.2009-0323OCSearch in Google Scholar PubMed PubMed Central

Choi, S.-M., McAleer, J.P., Zheng, M., Pociask, D.A., Kaplan, M.H., Qin, S., Reinhart, T.A., and Kolls, J.K. (2013). Innate Stat3-mediated induction of the antimicrobial protein Reg3γ is required for host defense against MRSA pneumonia. J. Exp. Med. 210, 551–561.10.1084/jem.20120260Search in Google Scholar PubMed PubMed Central

Choi, H., Yang, Z., and Weisshaar, J.C. (2017). Oxidative stress induced in E. coli by the human antimicrobial peptide LL-37. PLoS Pathog. 13, e1006481.10.1371/journal.ppat.1006481Search in Google Scholar PubMed PubMed Central

Chow, O.A., von Köckritz-Blickwede, M., Bright, A.T., Hensler, M.E., Zinkernagel, A.S., Cogen, A.L., Gallo, R.L., Monestier, M., Wang, Y., Glass, C.K., et al. (2010). Statins enhance formation of phagocyte extracellular traps. Cell Host Microbe. 8, 445–454.10.1016/j.chom.2010.10.005Search in Google Scholar PubMed PubMed Central

Cirioni, O., Giacometti, A., Ghiselli, R., Bergnach, C., Orlando, F., Silvestri, C., Mocchegiani, F., Licci, A., Skerlavaj, B., Rocchi, M., et al. (2006). LL-37 protects rats against lethal sepsis caused by Gram-negative bacteria. Antimicrob. Agents Chemother. 50, 1672–1679.10.1128/AAC.50.5.1672-1679.2006Search in Google Scholar PubMed PubMed Central

Clarke, T.B. (2014). Early innate immunity to bacterial infection in the lung is regulated systemically by the commensal microbiota via nod-like receptor ligands. Infect. Immun. 82, 4596–4606.10.1128/IAI.02212-14Search in Google Scholar PubMed PubMed Central

Condon, T.V., Sawyer, R.T., Fenton, M.J., and Riches, D.W.H. (2011). Lung dendritic cells at the innate-adaptive immune interface. J. Leukoc. Biol. 90, 883–895.10.1189/jlb.0311134Search in Google Scholar PubMed PubMed Central

Cormier, S.A., Shrestha, B., Saravia, J., Lee, G.I., Shen, L., DeVincenzo, J.P., Kim, Y.-I., and You, D. (2014). Limited type I interferons and plasmacytoid dendritic cells during neonatal respiratory syncytial virus infection permit immunopathogenesis upon reinfection. J. Virol. 88, 9350–9360.10.1128/JVI.00818-14Search in Google Scholar PubMed PubMed Central

Coya, J.M., Akinbi, H.T., Sáenz, A., Yang, L., Weaver, T.E., and Casals, C. (2015). Natural anti-infective pulmonary proteins: in vivo cooperative action of surfactant protein SP-A and the lung antimicrobial peptide SP-BN. J. Immunol. 195, 1628–1636.10.4049/jimmunol.1500778Search in Google Scholar PubMed PubMed Central

Cribbs, S.K., Lennox, J., Caliendo, A.M., Brown, L.A., and Guidot, D.M. (2015). Healthy HIV-1-infected individuals on highly active antiretroviral therapy harbor HIV-1 in their alveolar macrophages. AIDS Res. Hum. Retroviruses 31, 64–70.10.1089/aid.2014.0133Search in Google Scholar PubMed PubMed Central

Crother, T.R., Ma, J., Jupelli, M., Chiba, N., Chen, S., Slepenkin, A., Alsabeh, R., Peterson, E., Shimada, K., and Arditi, M. (2012). Plasmacytoid dendritic cells play a role for effective innate immune responses during Chlamydia pneumoniae infection in mice. PLoS One 7, e48655.10.1371/journal.pone.0048655Search in Google Scholar PubMed PubMed Central

Daher, K.A., Selsted, M.E., and Lehrer, R.I. (1986). Direct inactivation of viruses by human granulocyte defensins. J. Virol. 60, 1068–1074.10.1128/jvi.60.3.1068-1074.1986Search in Google Scholar PubMed PubMed Central

Das, R., Koo, M.-S., Kim, B.H., Jacob, S.T., Subbian, S., Yao, J., Leng, L., Levy, R., Murchison, C., Burman, W.J., et al. (2013). Macrophage migration inhibitory factor (MIF) is a critical mediator of the innate immune response to Mycobacterium tuberculosis. Proc. Natl. Acad. Sci. USA 110, E2997–E3006.10.1073/pnas.1301128110Search in Google Scholar PubMed PubMed Central

Ding, X., Jin, S., Tong, Y., Jiang, X., Chen, Z., Mei, S., Zhang, L., Billiar, T.R., and Li, Q. (2017). TLR4 signaling induces TLR3 up-regulation in alveolar macrophages during acute lung injury. Sci. Rep. 7, 34278.10.1038/srep34278Search in Google Scholar PubMed PubMed Central

Doyle, S.E., O’Connell, R., Vaidya, S.A., Chow, E.K., Yee, K., and Cheng, G. (2003). Toll-like receptor 3 mediates a more potent antiviral response than Toll-like receptor 4. J. Immunol. 170, 3565–3571.10.4049/jimmunol.170.7.3565Search in Google Scholar PubMed

Dunay, I.R. and Diefenbach, A. (2018). Group 1 innate lymphoid cells in Toxoplasma gondii infection. Parasite Immunol. 40, e12516.10.1111/pim.12516Search in Google Scholar PubMed

Dürr, U.H.N., Sudheendra, U.S., and Ramamoorthy, A. (2006). LL-37, the only human member of the cathelicidin family of antimicrobial peptides. Biochim. Biophys. Acta Biomembr. 1758, 1408–1425.10.1016/j.bbamem.2006.03.030Search in Google Scholar PubMed

Dziarski, R. and Gupta, D. (2018). How innate immunity proteins kill bacteria and why they are not prone to resistance. Curr. Genet. 64, 125–129.10.1007/s00294-017-0737-0Search in Google Scholar PubMed PubMed Central

Ebbo, M., Crinier, A., Vély, F., and Vivier, E. (2017). Innate lymphoid cells: major players in inflammatory diseases. Nat. Rev. Immunol. 17, 665–678.10.1038/nri.2017.86Search in Google Scholar PubMed

Eichinger, K.M., Egaña, L., Orend, J.G., Resetar, E., Anderson, K.B., Patel, R., and Empey, K.M. (2015). Alveolar macrophages support interferon gamma-mediated viral clearance in RSV-infected neonatal mice. Respir. Res. 16, 122.10.1186/s12931-015-0282-7Search in Google Scholar PubMed PubMed Central

Eisenhauer, P.B. and Lehrer, R.I. (1992). Mouse neutrophils lack defensins. Infect. Immun. 60, 3446–3447.10.1128/iai.60.8.3446-3447.1992Search in Google Scholar PubMed PubMed Central

Espinosa, V., Jhingran, A., Dutta, O., Kasahara, S., Donnelly, R., Du, P., Rosenfeld, J., Leiner, I., Chen, C.C., Ron, Y., et al. (2014). Inflammatory monocytes orchestrate innate antifungal immunity in the lung. PLoS Pathog. 10, e1003940.10.1371/journal.ppat.1003940Search in Google Scholar PubMed PubMed Central

Fahy, J.V. and Dickey, B.F. (2010). Airway mucus function and dysfunction. N. Engl. J. Med. 363, 2233–2247.10.1056/NEJMra0910061Search in Google Scholar PubMed PubMed Central

Falagas, M.E., Rafailidis, P.I., Kasiakou, S.K., Hatzopoulou, P., and Michalopoulos, A. (2006). Effectiveness and nephrotoxicity of colistin monotherapy vs. colistin–meropenem combination therapy for multidrug-resistant Gram-negative bacterial infections. Clin. Microbiol. Infect. 12, 1227–1230.10.1111/j.1469-0691.2006.01559.xSearch in Google Scholar PubMed

Ferlazzo, G., Mingari, M., and Carrega, P. (2016). Group 3 NCR+ innate lymphoid cells accumulate in human lung cancer. J. Immunol. 196 (Suppl 1), 142.18.10.4049/jimmunol.196.Supp.142.18Search in Google Scholar

Fitzpatrick, E.A., You, D., Shrestha, B., Siefker, D., Patel, V.S., Yadav, N., Jaligama, S., and Cormier, S.A. (2017). A neonatal murine model of MRSA pneumonia. PLoS One 12, e0169273.10.1371/journal.pone.0169273Search in Google Scholar PubMed PubMed Central

Garg, R., Shrivastava, P., and van Drunen Littel-van den Hurk, S. (2012). The role of dendritic cells in innate and adaptive immunity to respiratory syncytial virus, and implications for vaccine development. Expert Rev. Vaccines 11, 1441–1457.10.1586/erv.12.117Search in Google Scholar PubMed

Garth, J.M. and Steele, C. (2017). Innate lung defense during invasive aspergillosis: new mechanisms. J. Innate Immun. 9, 271–280.10.1159/000455125Search in Google Scholar PubMed PubMed Central

Gaunsbaek, M.Q., Rasmussen, K.J., Beers, M.F., Atochina-Vasserman, E.N., and Hansen, S. (2013). Lung surfactant protein D (SP-D) response and regulation during acute and chronic lung injury. Lung. 191, 295–303.10.1007/s00408-013-9452-xSearch in Google Scholar PubMed PubMed Central

Gideon, H.P., Phuah, J., Myers, A.J., Bryson, B.D., Rodgers, M.A., Coleman, M.T., Maiello, P., Rutledge, T., Marino, S., Fortune, S.M., et al. (2015). Variability in tuberculosis granuloma T cell responses exists, but a balance of pro- and anti-inflammatory cytokines is associated with sterilization. PLoS Pathog. 11, e1004603.10.1371/journal.ppat.1004603Search in Google Scholar PubMed PubMed Central

Gomez Perdiguero, E., Klapproth, K., Schulz, C., Busch, K., Azzoni, E., Crozet, L., Garner, H., Trouillet, C., de Bruijn, M.F., Geissmann, F., et al. (2015). Tissue-resident macrophages originate from yolk-sac-derived erythro-myeloid progenitors. Nature 518, 547–551.10.1016/j.exphem.2015.06.130Search in Google Scholar

Gracey, E., Baglaenko, Y., Prayitno, N., Van Rooijen, N., Akram, A., Lin, A., Chiu, B., and Inman, R.D. (2015). Pulmonary Chlamydia muridarum challenge activates lung interstitial macrophages which correlate with IFN-γ production and infection control in mice. Eur. J. Immunol. 45, 3417–3430.10.1002/eji.201545874Search in Google Scholar PubMed

Gray, J., Oehrle, K., Worthen, G., Alenghat, T., Whitsett, J., and Deshmukh, H. (2017). Intestinal commensal bacteria mediate lung mucosal immunity and promote resistance of newborn mice to infection. Sci. Transl. Med. 9, eaaf9412.10.1126/scitranslmed.aaf9412Search in Google Scholar PubMed PubMed Central

Greenbaum, A.H., Chen, J., Reed, C., Beavers, S., Callahan, D., Christensen, D., Finelli, L., and Fry, A.M. (2014). Hospitalizations for severe lower respiratory tract infections. Pediatrics 134, 546–554.10.1542/peds.2014-0244Search in Google Scholar PubMed

Grommes, J., Drechsler, M., and Soehnlein, O. (2014). CCR5 and FPR1 mediate neutrophil recruitment in endotoxin-induced lung injury. J. Innate Immun. 6, 111–116.10.1159/000353229Search in Google Scholar PubMed PubMed Central

De Grove, K.C., Provoost, S., Verhamme, F.M., Bracke, K.R., Joos, G.F., Maes, T., and Brusselle, G.G. (2016). Characterization and quantification of innate lymphoid cell subsets in human lung. PLoS One 11, e0145961.10.1371/journal.pone.0145961Search in Google Scholar PubMed PubMed Central

Guilliams, M., De Kleer, I., Henri, S., Post, S., Vanhoutte, L., De Prijck, S., Deswarte, K., Malissen, B., Hammad, H., and Lambrecht, B.N. (2013). Alveolar macrophages develop from fetal monocytes that differentiate into long-lived cells in the first week of life via GM-CSF. J. Exp. Med. 210, 1977–1992.10.1084/jem.20131199Search in Google Scholar PubMed PubMed Central

Guo, H., Callaway, J.B., and Ting, J.P.-Y. (2015). Inflammasomes: mechanism of action, role in disease and therapeutics. Nat. Med. 21, 677–687.10.1038/nm.3893Search in Google Scholar PubMed PubMed Central

Güzel, A., Karadağ, A., Okuyucu, A., Alaçam, H., and Küçük, Y. (2014). The evaluation of serum surfactant protein D (SP-D) levels as a biomarker of lung injury in tuberculosis and different lung diseases. Clin. Lab. 60, 1091–1098.10.7754/Clin.Lab.2013.130323Search in Google Scholar

Halle, S., Dujardin, H.C., Bakocevic, N., Fleige, H., Danzer, H., Willenzon, S., Suezer, Y., Hämmerling, G., Garbi, N., Sutter, G., et al. (2009). Induced bronchus-associated lymphoid tissue serves as a general priming site for T cells and is maintained by dendritic cells. J. Exp. Med. 206, 2593–2601.10.1084/jem.20091472Search in Google Scholar PubMed PubMed Central

Hepworth, M.R., Monticelli, L.A., Fung, T.C., Ziegler, C.G.K., Grunberg, S., Sinha, R., Mantegazza, A.R., Ma, H.-L., Crawford, A., Angelosanto, J.M., et al. (2013). Innate lymphoid cells regulate CD4+ T-cell responses to intestinal commensal bacteria. Nature 498, 113–117.10.1038/nature12240Search in Google Scholar PubMed PubMed Central

Hess, C., Herr, C., Beisswenger, C., Zakharkina, T., Schmid, R.M., and Bals, R. (2010). Myeloid RelA regulates pulmonary host defense networks. Eur. Respir. J. 35, 343–352.10.1183/09031936.00196408Search in Google Scholar PubMed

Hielpos, M.S., Ferrero, M.C., Fernández, A.G., Bonetto, J., Giambartolomei, G.H., Fossati, C.A. and Baldi, P.C. (2015). CCL20 and b-defensin 2 production by human lung epithelial cells and macrophages in response to Brucella abortus infection. PLoS One 10, e0140408.10.1371/journal.pone.0140408Search in Google Scholar PubMed PubMed Central

Hiemstra, P.S. (2015). Parallel activities and interactions between antimicrobial peptides and complement in host defense at the airway epithelial surface. Mol. Immunol. 68, 28–30.10.1016/j.molimm.2015.07.030Search in Google Scholar PubMed

Higgins, G., Fustero Torre, C., Tyrrell, J., McNally, P., Harvey, B.J., and Urbach, V. (2016). Lipoxin A 4 prevents tight junction disruption and delays the colonization of cystic fibrosis bronchial epithelial cells by Pseudomonas aeruginosa. Am. J. Physiol. Cell. Mol. Physiol. 310, L1053–L1061.10.1152/ajplung.00368.2015Search in Google Scholar PubMed

Hillaire, M.L.B., van Eijk, M., Vogelzang-van Trierum, S.E., Fouchier, R.A.M., Osterhaus, A.D.M.E., Haagsman, H.P., and Rimmelzwaan, G.F. (2014). Recombinant porcine surfactant protein D inhibits influenza A virus replication ex vivo. Virus Res. 181, 22–26.10.1016/j.virusres.2013.12.032Search in Google Scholar PubMed

Hwang, J.Y., Randall, T.D., and Silva-Sanchez, A. (2016). Inducible bronchus-associated lymphoid tissue: taming inflammation in the lung. Front. Immunol. 7, 258.10.3389/fimmu.2016.00258Search in Google Scholar PubMed PubMed Central

Ivin, M., Dumigan, A., de Vasconcelos, F.N., Ebner, F., Borroni, M., Kavirayani, A., Przybyszewska, K.N., Ingram, R.J., Lienenklaus, S., Kalinke, U., et al. (2017). Natural killer cell-intrinsic type I IFN signaling controls Klebsiella pneumoniae growth during lung infection. PLoS Pathog. 13, e1006696.10.1371/journal.ppat.1006696Search in Google Scholar PubMed PubMed Central

Jin, H., Ciechanowicz, A.K., Kaplan, A.R., Wang, L., Zhang, P.X., Lu, Y., Tobin, R.E., Tobin, B.A., Cohn, L., Zeiss, C.J., et al. (2018). Surfactant protein C dampens inflammation by decreasing JAK/STAT activation during lung repair. Am. J. Physiol. Cell. Mol. Physiol. L882–L892.10.1152/ajplung.00418.2017Search in Google Scholar PubMed PubMed Central

Jones, C. V, Williams, T.M., Walker, K.A., Dickinson, H., Sakkal, S., Rumballe, B.A., Little, M.H., Jenkin, G., and Ricardo, S.D. (2013). M2 macrophage polarisation is associated with alveolar formation during postnatal lung development. Respir. Res. 14, 41.10.1186/1465-9921-14-41Search in Google Scholar PubMed PubMed Central

Kast, J.I., McFarlane, A.J., Głobińska, A., Sokolowska, M., Wawrzyniak, P., Sanak, M., Schwarze, J., Akdis, C.A., and Wanke, K. (2017). Respiratory syncytial virus infection influences tight junction integrity. Clin. Exp. Immunol. 190, 351–359.10.1111/cei.13042Search in Google Scholar PubMed PubMed Central

Kato, K., Hanss, A.D., Zemskova, M.A., Morgan, N.E., Kim, M., Knox, K.S., Lin, Y., Lillehoj, E.P., and Kim, K.C. (2017a). Pseudomonas aeruginosa increases MUC1 expression in macrophages through the TLR4-p38 pathway. Biochem. Biophys. Res. Commun. 492, 231–235.10.1016/j.bbrc.2017.08.056Search in Google Scholar PubMed PubMed Central

Kato, K., Lillehoj, E.P., Lu, W., and Kim, K.C. (2017b). MUC1: the first respiratory mucin with an anti-inflammatory function. J. Clin. Med. 6, 110.10.3390/jcm6120110Search in Google Scholar PubMed PubMed Central

Kim, K.C. (2012). Role of epithelial mucins during airway infection. Pulm. Pharmacol. Ther. 25, 415–419.10.1016/j.pupt.2011.12.003Search in Google Scholar PubMed PubMed Central

Kolli, D., Gupta, M.R., Sbrana, E., Velayutham, T.S., Chao, H., Casola, A., and Garofalo, R.P. (2014). Alveolar macrophages contribute to the pathogenesis of human metapneumovirus infection while protecting against respiratory syncytial virus infection. Am. J. Respir. Cell Mol. Biol. 51, 502–515.10.1165/rcmb.2013-0414OCSearch in Google Scholar PubMed PubMed Central

Kopf, M., Schneider, C., and Nobs, S.P. (2014). The development and function of lung-resident macrophages and dendritic cells. Nat. Immunol. 16, 36–44.10.1038/ni.3052Search in Google Scholar PubMed

Kumar, P., Thakar, M.S., Ouyang, W., and Malarkannan, S. (2013). IL-22 from conventional NK cells is epithelial regenerative and inflammation protective during influenza infection. Mucosal Immunol. 6, 69–82.10.1038/mi.2012.49Search in Google Scholar PubMed PubMed Central

Leiva-Juarez, M.M., Kirkpatrick, C.T., Gilbert, B.E., Scott, B., Tuvim, M.J., Dickey, B.F., Evans, S.E., and Markesich, D. (2018). Combined aerosolized Toll-like receptor ligands are an effective therapeutic agent against influenza pneumonia when co-administered with oseltamivir. Eur. J. Pharmacol. 818, 191–197.10.1016/j.ejphar.2017.10.035Search in Google Scholar PubMed PubMed Central

Li, Y., Dinwiddie, D.L., Harrod, K.S., Jiang, Y., and Kim, K.C. (2010). Anti-inflammatory effect of MUC1 during respiratory syncytial virus infection of lung epithelial cells in vitro. Am. J. Physiol. Cell. Mol. Physiol. 298, L558–L563.10.1152/ajplung.00225.2009Search in Google Scholar PubMed PubMed Central

Li, X., Cullere, X., Nishi, H., Saggu, G., Durand, E., Mansour, M.K., Tam, J.M., Song, X., Lin, X., Vyas, J.M., et al. (2016). PKC-δ activation in neutrophils promotes fungal clearance. J. Leukoc. Biol. 100, 581–588.10.1189/jlb.4A0915-405RSearch in Google Scholar PubMed PubMed Central

Liu, C.-I., Liu, G.Y., Song, Y., Yin, F., Hensler, M.E., Jeng, W.-Y., Nizet, V., Wang, A.H.-J., and Oldfield, E. (2008). A cholesterol biosynthesis inhibitor blocks Staphylococcus aureus virulence. Science 319, 1391–1394.10.1126/science.1153018Search in Google Scholar PubMed PubMed Central

Lin, F.-Y., Zhang, Y., Hensler, M., Liu, Y.-L., Chow, O.A., Zhu, W., Wang, K., Pang, R., Thienphrapa, W., Nizet, V., et al. (2012). Dual dehydrosqualene/squalene synthase inhibitors: leads for innate immune system-based therapeutics. ChemMedChem. 7, 561–564.10.1002/cmdc.201100589Search in Google Scholar PubMed PubMed Central

Makris, S., Bajorek, M., Culley, F.J., Goritzka, M., and Johansson, C. (2016). Alveolar macrophages can control respiratory syncytial virus infection in the absence of type I interferons. J. Innate Immun. 8, 452–463.10.1159/000446824Search in Google Scholar PubMed PubMed Central

Månsson Kvarnhammar, A., Tengroth, L., Adner, M., and Cardell, L.-O. (2013). Innate immune receptors in human airway smooth muscle cells: activation by TLR1/2, TLR3, TLR4, TLR7 and NOD1 agonists. PLoS One 8, e68701.10.1371/journal.pone.0068701Search in Google Scholar PubMed PubMed Central

Marakalala, M.J., Raju, R.M., Sharma, K., Zhang, Y.J., Eugenin, E.A., Prideaux, B., Daudelin, I.B., Chen, P.-Y., Booty, M.G., Kim, J.H., et al. (2016). Inflammatory signaling in human tuberculosis granulomas is spatially organized. Nat. Med. 22, 531–538.10.1038/nm.4073Search in Google Scholar PubMed PubMed Central

McAleer, J.P. and Kolls, J.K. (2014). Directing traffic: IL-17 and IL-22 coordinate pulmonary immune defense. Immunol. Rev. 260, 129–144.10.1111/imr.12183Search in Google Scholar PubMed PubMed Central

Mjösberg, J. and Spits, H. (2016). Human innate lymphoid cells. J. Allergy Clin. Immunol. 138, 1265–1276.10.1016/j.jaci.2016.09.009Search in Google Scholar PubMed

Monticelli, L.A., Sonnenberg, G.F., Abt, M.C., Alenghat, T., Ziegler, C.G.K., Doering, T.A., Angelosanto, J.M., Laidlaw, B.J., Yang, C.Y., Sathaliyawala, T., et al. (2011). Innate lymphoid cells promote lung-tissue homeostasis after infection with influenza virus. Nat. Immunol. 12, 1045–1054.10.1038/ni.2131Search in Google Scholar

Moore, T.A., Moore, B.B., Newstead, M.W., and Standiford, T.J. (2000). Gamma delta-T cells are critical for survival and early proinflammatory cytokine gene expression during murine Klebsiella pneumonia. J. Immunol. 165, 2643–2650.10.4049/jimmunol.165.5.2643Search in Google Scholar PubMed

Moyron-Quiroz, J.E., Rangel-Moreno, J., Kusser, K., Hartson, L., Sprague, F., Goodrich, S., Woodland, D.L., Lund, F.E., and Randall, T.D. (2004). Role of inducible bronchus associated lymphoid tissue (iBALT) in respiratory immunity. Nat. Med. 10, 927–934.10.1038/nm1091Search in Google Scholar PubMed

Moyron-Quiroz, J.E., Rangel-Moreno, J., Hartson, L., Kusser, K., Tighe, M.P., Klonowski, K.D., Lefrançois, L., Cauley, L.S., Harmsen, A.G., Lund, F.E., et al. (2006). Persistence and responsiveness of immunologic memory in the absence of secondary lymphoid organs. Immunity 25, 643–654.10.1016/j.immuni.2006.08.022Search in Google Scholar PubMed

Oldfield, E. and Feng, X. (2014). Resistance-resistant antibiotics. Trends Pharmacol. Sci. 35, 664–674.10.1016/j.tips.2014.10.007Search in Google Scholar PubMed PubMed Central

Oliphant, C.J., Hwang, Y.Y., Walker, J.A., Salimi, M., Wong, S.H., Brewer, J.M., Englezakis, A., Barlow, J.L., Hams, E., Scanlon, S.T., et al. (2014). MHCII-Mediated dialog between group 2 innate lymphoid cells and CD4+ T cells potentiates type 2 immunity and promotes parasitic helminth expulsion. Immunity 41, 283–295.10.1016/j.immuni.2014.06.016Search in Google Scholar PubMed PubMed Central

Paclik, D., Stehle, C., Lahmann, A., Hutloff, A., and Romagnani, C. (2015). ICOS regulates the pool of group 2 innate lymphoid cells under homeostatic and inflammatory conditions in mice. Eur. J. Immunol. 45, 2766–2772.10.1002/eji.201545635Search in Google Scholar PubMed

Pahari, S., Kaur, G., Negi, S., Aqdas, M., Das, D.K., Bashir, H., Singh, S., Nagare, M., Khan, J., Agrewala, J.N., et al. (2018). Reinforcing the functionality of mononuclear phagocyte system to control tuberculosis. Front. Immunol. 9, 193.10.3389/fimmu.2018.00193Search in Google Scholar PubMed PubMed Central

Petrov, V., Funderburg, N., Weinberg, A., and Sieg, S. (2013). Human β defensin-3 induces chemokines from monocytes and macrophages: diminished activity in cells from HIV-infected persons. Immunology 140, 413–420.10.1111/imm.12148Search in Google Scholar PubMed PubMed Central

Pociask, D.A., Scheller, E.V., Mandalapu, S., McHugh, K.J., Enelow, R.I., Fattman, C.L., Kolls, J.K., and Alcorn, J.F. (2013). IL-22 is essential for lung epithelial repair following influenza infection. Am. J. Pathol. 182, 1286–1296.10.1016/j.ajpath.2012.12.007Search in Google Scholar PubMed PubMed Central

Poerio, N., Bugli, F., Taus, F., Santucci, M.B., Rodolfo, C., Cecconi, F., Torelli, R., Varone, F., Inchingolo, R., Majo, F., et al. (2017). Liposomes loaded with bioactive lipids enhance antibacterial innate immunity irrespective of drug resistance. Sci. Rep. 7, 45120.10.1038/srep45120Search in Google Scholar PubMed PubMed Central

Proebstl, D., Voisin, M.-B., Woodfin, A., Whiteford, J., D’Acquisto, F., Jones, G.E., Rowe, D., and Nourshargh, S. (2012). Pericytes support neutrophil subendothelial cell crawling and breaching of venular walls in vivo. J. Exp. Med. 209, 1219–1234.10.1084/jem.20111622Search in Google Scholar PubMed PubMed Central

Rangel-Moreno, J., Moyron-Quiroz, J.E., Hartson, L., Kusser, K., and Randall, T.D. (2007). Pulmonary expression of CXC chemokine ligand 13, CC chemokine ligand 19, and CC chemokine ligand 21 is essential for local immunity to influenza. Proc. Natl. Acad. Sci. USA 104, 10577–10582.10.1073/pnas.0700591104Search in Google Scholar PubMed PubMed Central

Rangel-Moreno, J., Carragher, D.M., de la Luz Garcia-Hernandez, M., Hwang, J.Y., Kusser, K., Hartson, L., Kolls, J.K., Khader, S.A., and Randall, T.D. (2011). The development of inducible bronchus-associated lymphoid tissue depends on IL-17. Nat. Immunol. 12, 639–646.10.1038/ni.2053Search in Google Scholar PubMed PubMed Central

Rathinam, V.A.K., Vanaja, S.K., and Fitzgerald, K.A. (2012). Regulation of inflammasome signaling. Nat. Immunol. 13, 333–342.10.1038/ni.2237Search in Google Scholar PubMed PubMed Central

Ray, A., Chakraborty, K., and Ray, P. (2013). Immunosuppressive MDSCs induced by TLR signaling during infection and role in resolution of inflammation. Front. Cell. Infect. Microbiol. 3, 52.10.3389/fcimb.2013.00052Search in Google Scholar PubMed PubMed Central

Richards, C.D. (2017). Innate immune cytokines, fibroblast phenotypes, and regulation of extracellular matrix in lung. J. Interf. Cytokine Res. 37, 52–61.10.1089/jir.2016.0112Search in Google Scholar PubMed

Sandra Tjabringa, G., Rabe, K.F., and Hiemstra, P.S. (2005). The human cathelicidin LL-37: a multifunctional peptide involved in infection and inflammation in the lung. Pulm. Pharmacol. Ther. 18, 321–327.10.1016/j.pupt.2005.01.001Search in Google Scholar PubMed

Sawant, K.V., Xu, R., Cox, R., Hawkins, H., Sbrana, E., Kolli, D., Garofalo, R.P., and Rajarathnam, K. (2015). Chemokine CXCL1-mediated neutrophil trafficking in the lung: role of CXCR2 activation. J. Innate Immun. 7, 647–658.10.1159/000430914Search in Google Scholar PubMed PubMed Central

Scharf, S., Zahlten, J., Szymanski, K., Hippenstiel, S., Suttorp, N., and N’Guessan, P.D. (2012). Streptococcus pneumoniae induces human β-defensin-2 and -3 in human lung epithelium. Exp. Lung Res. 38, 100–110.10.3109/01902148.2011.652802Search in Google Scholar PubMed

Semple, F. and Dorin, J.R. (2012). β-Defensins: multifunctional modulators of infection, inflammation and more? J. Innate Immun. 4, 337–348.10.1159/000336619Search in Google Scholar PubMed PubMed Central

Sha, Q., Truong-Tran, A.Q., Plitt, J.R., Beck, L.A., and Schleimer, R.P. (2004). Activation of airway epithelial cells by Toll-like receptor agonists. Am. J. Respir. Cell Mol. Biol. 31, 358–364.10.1165/rcmb.2003-0388OCSearch in Google Scholar PubMed

Shaughnessy, J., Gulati, S., Agarwal, S., Unemo, M., Ohnishi, M., Su, X.-H., Monks, B.G., Visintin, A., Madico, G., Lewis, L.A., et al. (2016). A novel factor H-Fc chimeric immunotherapeutic molecule against Neisseria gonorrhoeae. J. Immunol. 196, 1732–1740.10.1016/j.imbio.2012.08.008Search in Google Scholar

Short, K.R., Kasper, J., van der Aa, S., Andeweg, A.C., Zaaraoui-Boutahar, F., Goeijenbier, M., Richard, M., Herold, S., Becker, C., Scott, D.P., et al. (2016). Influenza virus damages the alveolar barrier by disrupting epithelial cell tight junctions. Eur. Respir. J. 47, 954–966.10.1183/13993003.01282-2015Search in Google Scholar PubMed

Silver, J.S., Kearley, J., Copenhaver, A.M., Sanden, C., Mori, M., Yu, L., Pritchard, G.H., Berlin, A.A., Hunter, C.A., Bowler, R., et al. (2016). Inflammatory triggers associated with exacerbations of COPD orchestrate plasticity of group 2 innate lymphoid cells in the lungs. Nat. Immunol. 17, 626–635.10.1038/ni.3443Search in Google Scholar PubMed PubMed Central

Soloff, A.C., Bissel, S.J., Junecko, B.F., Giles, B.M., Reinhart, T.A., Ross, T.M., and Barratt-Boyes, S.M. (2014). Massive mobilization of dendritic cells during influenza A virus subtype H5N1 infection of nonhuman primates. J. Infect. Dis. 209, 2012–2016.10.1093/infdis/jiu009Search in Google Scholar PubMed PubMed Central

Spits, H., Artis, D., Colonna, M., Diefenbach, A., Di Santo, J.P., Eberl, G., Koyasu, S., Locksley, R.M., McKenzie, A.N., Mebius, R.E., et al. (2013). Innate lymphoid cells – a proposal for uniform nomenclature. Nat. Rev. Immunol. 13, 145–149.10.1038/nri3365Search in Google Scholar PubMed

Steele, C., Zheng, M., Young, E., Marrero, L., Shellito, J.E., and Kolls, J.K. (2002). Increased host resistance against Pneumocystis carinii pneumonia in gammadelta T-cell-deficient mice: protective role of gamma interferon and CD8+ T cells. Infect. Immun. 70, 5208–5215.10.1128/IAI.70.9.5208-5215.2002Search in Google Scholar PubMed PubMed Central

Steinmüller, C., Franke-Ullmann, G., Lohmann-Matthes, M.-L., and Emmendörffer, A. (2000). Local activation of nonspecific defense against a respiratory model infection by application of interferon-γ. Am. J. Respir. Cell Mol. Biol. 22, 481–490.10.1165/ajrcmb.22.4.3336Search in Google Scholar PubMed

Sugimoto, C., Hasegawa, A., Saito, Y., Fukuyo, Y., Chiu, K.B., Cai, Y., Breed, M.W., Mori, K., Roy, C.J., Lackner, A.A., et al. (2015). Differentiation kinetics of blood monocytes and dendritic cells in macaques: insights to understanding human myeloid cell development. J. Immunol. 195, 1774–1781.10.4049/jimmunol.1500522Search in Google Scholar PubMed PubMed Central

Sun, S., Zhao, G., Liu, C., Wu, X., Guo, Y., Yu, H., Song, H., Du, L., Jiang, S., Guo, R., et al. (2013). Inhibition of complement activation alleviates acute lung injury induced by highly pathogenic avian influenza H5N1 virus infection. Am. J. Respir. Cell Mol. Biol. 49, 221–230.10.1165/rcmb.2012-0428OCSearch in Google Scholar PubMed

Tait Wojno, E.D. and Artis, D. (2016). Emerging concepts and future challenges in innate lymphoid cell biology. J. Exp. Med. 213, 2229–2248.10.1084/jem.20160525Search in Google Scholar PubMed PubMed Central

Taube, C., Tertilt, C., Gyülveszi, G., Dehzad, N., Kreymborg, K., Schneeweiss, K., Michel, E., Reuter, S., Renauld, J.C., Arnold-Schild, D., et al. (2011). IL-22 is produced by innate lymphoid cells and limits inflammation in allergic airway disease. PLoS One 6, e21799.10.1371/journal.pone.0021799Search in Google Scholar PubMed PubMed Central

Thornton, D.J., Rousseau, K., and McGuckin, M.A. (2008). Structure and function of the polymeric mucins in airways mucus. Annu. Rev. Physiol. 70, 459–486.10.1146/annurev.physiol.70.113006.100702Search in Google Scholar PubMed

Trevejo-Nunez, G., Elsegeiny, W., Conboy, P., Chen, K., and Kolls, J.K. (2016). Critical role of IL-22/IL22-RA1 signaling in pneumococcal pneumonia. J. Immunol. 197, 1877–1883.10.4049/jimmunol.1600528Search in Google Scholar PubMed PubMed Central

Tukhvatulin, A.I., Dzharullaeva, A.S., Tukhvatulina, N.M., Shcheblyakov, D.V., Shmarov, M.M., Dolzhikova, I.V., Stanhope-Baker, P., Naroditsky, B.S., Gudkov, A.V., Logunov, D.Y., et al. (2016). Powerful complex immunoadjuvant based on synergistic effect of combined TLR4 and NOD2 activation significantly enhances magnitude of humoral and cellular adaptive immune responses. PLoS One 11, e0155650.10.1371/journal.pone.0155650Search in Google Scholar PubMed PubMed Central

Vazquez, D.E., Niño, D.F., De Maio, A., and Cauvi, D.M. (2015). Sustained expression of lipocalin-2 during polymicrobial sepsis. Innate Immun. 21, 477–489.10.1177/1753425914548491Search in Google Scholar PubMed

Wagage, S., John, B., Krock, B.L., Hall, A.O., Randall, L.M., Karp, C.L., Simon, M.C., and Hunter, C.A. (2014). The aryl hydrocarbon receptor promotes IL-10 production by NK cells. J. Immunol. 192, 1661–1670.10.4049/jimmunol.1300497Search in Google Scholar PubMed PubMed Central

Wang, J., Nikrad, M.P., Travanty, E.A., Zhou, B., Phang, T., Gao, B., Alford, T., Ito, Y., Nahreini, P., Hartshorn, K., et al. (2012). Innate immune response of human alveolar macrophages during influenza A infection. PLoS One 7, e29879.10.1371/journal.pone.0029879Search in Google Scholar PubMed PubMed Central

Warszawska, J.M., Gawish, R., Sharif, O., Sigel, S., Doninger, B., Lakovits, K., Mesteri, I., Nairz, M., Boon, L., Spiel, A., et al. (2013). Lipocalin 2 deactivates macrophages and worsens pneumococcal pneumonia outcomes. J. Clin. Invest. 123, 3363–3372.10.1172/JCI67911Search in Google Scholar PubMed PubMed Central

Welsh, K.J., Lewis, C.T., Boyd, S., Braun, M.C., and Actor, J.K. (2012). Complement factor C7 contributes to lung immunopathology caused by Mycobacterium tuberculosis. Clin. Dev. Immunol. 2012, 429675.10.1155/2012/429675Search in Google Scholar PubMed PubMed Central

Wilson, S.S., Wiens, M.E., and Smith, J.G. (2013). Antiviral mechanisms of human defensins. J. Mol. Biol. 425, 4965–4980.10.1016/j.jmb.2013.09.038Search in Google Scholar PubMed PubMed Central

Wittekindt, O.H. (2017). Tight junctions in pulmonary epithelia during lung inflammation. Pflüger’s Arch. 469, 135–147.10.1007/s00424-016-1917-3Search in Google Scholar PubMed PubMed Central

Wong, S.M., Shaughnessy, J., Ram, S., and Akerley, B.J. (2016). Defining the binding region in Factor H to develop a therapeutic Factor H-Fc fusion protein against Non-typeable Haemophilus influenzae. Front. Cell. Infect. Microbiol. 6, 40.10.3389/fcimb.2016.00040Search in Google Scholar PubMed PubMed Central

Workalemahu, G., Wang, H., Puan, K.-J., Nada, M.H., Kuzuyama, T., Jones, B.D., Jin, C., and Morita, C.T. (2014). Metabolic engineering of Salmonella vaccine bacteria to boost human Vγ2Vδ2 T cell immunity. J. Immunol. 193, 708–721.10.4049/jimmunol.1302746Search in Google Scholar PubMed PubMed Central

Wright, J.R. (2005). Immunoregulatory functions of surfactant proteins. Nat. Rev. Immunol. 5, 58–68.10.1038/nri1528Search in Google Scholar PubMed

Xia, M., Hesser, D.C., De, P., Sakala, I.G., Spencer, C.T., Kirkwood, J.S., Abate, G., Chatterjee, D., Dobos, K.M., and Hoft, D.F. (2016). A subset of protective γ 9 δ 2 T cells is activated by novel mycobacterial glycolipid components. Infect. Immun. 84, 2449–2462.10.1128/IAI.01322-15Search in Google Scholar PubMed PubMed Central

Yona, S., Kim, K.-W., Wolf, Y., Mildner, A., Varol, D., Breker, M., Strauss-Ayali, D., Viukov, S., Guilliams, M., Misharin, A., et al. (2013). Fate mapping reveals origins and dynamics of monocytes and tissue macrophages under homeostasis. Immunity 38, 79–91.10.1016/j.immuni.2012.12.001Search in Google Scholar PubMed PubMed Central

Zasloff, M. (2002). Antimicrobial peptides of multicellular organisms. Nature 415, 389–395.10.1038/415389aSearch in Google Scholar PubMed

Received: 2018-06-28
Accepted: 2018-09-19
Published Online: 2018-09-29
Published in Print: 2019-04-24

©2019 Walter de Gruyter GmbH, Berlin/Boston

Downloaded on 20.4.2024 from https://www.degruyter.com/document/doi/10.1515/hsz-2018-0304/html
Scroll to top button