Back to Journals » Journal of Inflammation Research » Volume 18
The Immune Modulatory Role of Surfactants in Mycoplasma pneumoniae Infection
Authors Li X , Zeng Q , Liu C , Yi X , Luo H, Tong Q, Chen H, You X
Received 20 November 2024
Accepted for publication 8 February 2025
Published 26 February 2025 Volume 2025:18 Pages 2909—2922
DOI https://doi.org/10.2147/JIR.S507526
Checked for plagiarism Yes
Review by Single anonymous peer review
Peer reviewer comments 2
Editor who approved publication: Dr Subhasis Chattopadhyay
Xinru Li,1,* Qianrui Zeng,1,* Chang Liu,1 Xinchao Yi,2 Haodang Luo,2 Qin Tong,3 Hongliang Chen,4 Xiaoxing You1
1Institute of Pathogenic Biology, Hengyang Medical College, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, University of South China, Hengyang, People’s Republic of China; 2Department of Clinical Laboratory, The Affiliated Nanhua Hospital, Hengyang Medical College, University of South China, Hengyang, People’s Republic of China; 3Department of Oncology, The First Affiliated Hospital, Hengyang Medical College, University of South China, Hengyang, 421001, People’s Republic of China; 4Chenzhou No. 1 People’s Hospital (The Affiliated Chenzhou Hospital), Hengyang Medical College, University of South China, Chenzhou, People’s Republic of China
*These authors contributed equally to this work
Correspondence: Hongliang Chen, Chenzhou No. 1 People’s Hospital (The Affiliated Chenzhou Hospital), Hengyang Medical College, University of South China, Chenzhou, People’s Republic of China, Tel/Fax +86-735-2343595, Email [email protected] Xiaoxing You, Institute of Pathogenic Biology, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Hengyang Medical College, University of South China, Hengyang, People’s Republic of China, Tel/Fax +86-734-8283366, Email [email protected]
Abstract: Mycoplasma pneumoniae is a prevalent respiratory microbe that causes acute inflammation in the respiratory system. Surfactant proteins (SP), particularly SP-A and SP-D, are essential for the immunological protection against M. pneumoniae infection. Variant SP-A2 may lead to immune reactions, which could account for the variability in clinical manifestations among individuals. Mechanistically, these surfactant proteins may act as candidate receptors, facilitating both the adhesion of M. pneumoniae and internalization of community-acquired respiratory distress syndrome toxin. They also exhibit a high affinity for lipid ligands on the surface of M. pneumoniae membranes via their carbohydrate recognition domains, which aid in the direct clearing of the bacteria. In addition, SP-A and SP-D demonstrated synergistic effects in augmenting the intake and elimination of M. pneumoniae by alveolar macrophages. Furthermore, these surfactant proteins negatively regulate pulmonary inflammation by influencing lymphocyte and dendritic cell activities, reducing airway eosinophilic infiltration, and managing asthma-related inflammatory responses. A thorough understanding of the immunomodulatory roles of surfactant proteins in M. pneumoniae infection will shed light on how homeostasis is preserved during mycoplasma pneumonia and may guide the development of novel therapeutic strategies against this organism.
Keywords: Mycoplasma pneumoniae, surfactant protein-A, surfactant protein-D, immune modulation
Introduction
Mycoplasma pneumoniae is the principal causative agent of pneumonia and many respiratory disorders, including tracheobronchitis, bronchiolitis, laryngitis, and pharyngitis, and often worsens many respiratory diseases, such as asthma, emphysema, and chronic obstructive pulmonary disease.1–4 M. pneumoniae also acts as an undesirable contaminant in advanced therapy medicinal products (ATMPs), posing significant challenges for detection, particularly in immunosuppressed patients.5,6 In the normal population, approximately 30% of pneumonia cases are associated with M. pneumoniae. However, in vivo experiments have revealed significant differences in bacterial load following M. pneumoniae infection across various mouse strains,7 highlighting the importance of host genetic background over the innate immune status. The pronounced difference in susceptibility between C57BL/6NCr and C3H/HeNCr mice represents different immune responses to M. pneumoniae infection.8 Early resistance to M. pneumoniae in C57BL mice may stem from innate defense mechanisms associated with alveolar macrophages (AMs) enriched with pulmonary surfactants, whereas impairment of these mechanisms is observed in C3H/HeNCr mice.9 Although M. pneumoniae develops multiple strategies to escape the host immune system and even cause persistent infection, it can still be cleared by the host immune immunity,10 in which surfactants play a pivotal role prior to the activation of antibody-mediated adaptive immunity.11
Pulmonary surfactants are vital components of the bronchoalveolar microenvironment and are composed of proteins, neutral lipids, and phospholipids.12 Surfactants are predominantly produced by alveolar type II cells and disseminate across the molecular layer of the alveolar fluid.13 Their role is essential in alleviating tension at the interface between gas and liquid in the alveoli, preventing atelectasis upon exhalation, and facilitating effective gas exchange.14 In addition to their biomechanical roles, the protein constituents of pulmonary surfactants serve as distinctive elements of the natural immune system.15 Surfactant proteins can be classified into four categories: surfactant protein A (SP-A), SP-B, SP-C, and SP-D.16,17 Among them, SP-A and SP-D belong to the C-type lectin family and are particularly important for their interactions with bacteria, fungi, and viruses, thereby playing crucial roles in immune defense against inhalation pathogens.18 For instance, SP-A can facilitate pathogen recognition and modulate the activity of immune cells, including AMs, neutrophils, and dendritic cells (DCs), thereby promoting or suppressing the overall inflammatory response, depending on the infection environment.19 Similarly, SP-D is crucial for adaptive and innate immunity, as it aggregates pathogens and promotes their uptake by AMs, while also regulating T and B cell activity to enhance the efficiency of the immune response.20
While pulmonary surfactants are not selective for the bacteria with which they interact, they exhibit a notable capacity to facilitate the internalization of community-acquired respiratory distress syndrome toxin (CARDS TX), which is a major virulence factor of M. pneumoniae.21 Given that pulmonary surfactants serve as frontline defenders against infections, exploring their role in the immunological response to mycoplasma pneumonia is important. Since the 1990s, extensive literature has shown that surfactants contribute to various aspects of M. pneumoniae infections, including receptor recognition, bactericidal activity, and immune modulation (Figure 1). This study aimed to elucidate the functions of SP-A and SP-D in M. pneumoniae infections and immune responses. Understanding the relationship between surfactant proteins and M. pneumoniae is crucial for the development of targeted therapies aimed at restoring normal lung function and improving patient outcomes.
![]() |
Figure 1 Overview of the immune modulation by pulmonary surfactant proteins upon Mycoplasma pneumoniae infection. Pulmonary surfactants contribute to various aspects of M. pneumoniae infection. SP-A and SP-D exhibit high affinity for live M. pneumoniae and MMF, enabling them to directly kill M. pneumoniae. In contrast, SP-A acts as a co-receptor for CARDS TX, resulting in ADP ribosylation and vacuolation in host cells after internalization (A). SP-A and SP-D attach to alveolar macrophages (AMs) with high specificity, release ROS and nitric oxide metabolites, increase chemotactic activity, and augment the phagocytic capacity of AMs to eliminate M. pneumoniae (B). The prominent function of surfactant proteins is the negative regulation of the immune system, including interaction with epidermal growth factor receptor to suppress epithelial cell secretion of mucin (C), alleviation of airway eosinophil infiltration and inflammation by inhibiting the release of EPO, or inhibition of eosinophil chemotaxis (D). In addition, SP-A is suggested to inhibit mast cells recruitment and the subsequent release of TNF-α to regulate the exacerbation of airway hyperresponsiveness and the associated influx of inflamed cells in reaction to M. pneumoniae infection (E). Other candidate functions include inhibiting dendritic cells maturation and suppressing the expression of HMGB-1 (F) or attenuating cytokine production by T lymphocytes, thereby directly or indirectly influencing T cell proliferation and increasing the number of activated B cells during M. pneumoniae infection (G). These factors enable the host immune system to fine-tune homeostasis during M. pneumoniae infection. Created in BioRender. You, X. (2025) https://BioRender.com/g33c697. |
Chemical Composition and Classification of Surfactant Proteins
The isolated surfactant contains approximately 80% lipids, 10% neutral lipids (predominantly cholesterol), and 10% proteins. The lipids of surfactants are mainly phospholipids (95%), of which phosphatidylcholine (PC) is the most common, accounting for 70–80%.22 The main component of PC is saturated dipalmitoylphosphatidylcholine (DPPC), which can accumulate at very high densities at the air-liquid interface, allowing for the lung exchange of gases by reducing alveolar surface tension.23 In addition, about half of the phospholipid components are desaturated, with approximately 15% acidic phospholipids such as phosphatidylglycerol (PG), phosphatidylinositol (PI), and bis(monoacylglycerol) phosphate, alongside minor quantities of phosphatidylethanolamine (PE), sphingomyelin (SM), and lysophosphatidylcholine (LPC).14 These lipids are believed to be involved in selected interactions with cationic hydrophobic proteins.24
In contrast, the protein content of surfactants is significantly lower than that of lipids and surfactants are synthesized and secreted mainly by bronchiolar Clara cells and alveolar type II cells.25 Currently, four common surfactant proteins are directly involved in reducing alveolar surface tension and regulating the host immune defense. These proteins can be categorized into macromolecular hydrophilic proteins, SP-A and SP-D, and smaller hydrophobic proteins, SP-B and SP-C, based on their hydrophilic properties and molecular weight.17 SP-B and SP-C are tiny molecules that react with surfactant lipids to lower the surface tension between the gas and liquid and enhance lung compliance, thereby maintaining normal alveolar expansion.26 Conversely, SP-A (26–36 kDa) and SP-D (39 kDa) tend to be large soluble hydrophilic proteins present on the majority of mucosal surfaces, with critical overlap and multiple functions in the natural defense and immune homeostasis of the lung.27 Intriguingly, despite the fact that SP-A and SP-D are equally crucial in immune responses, the level of SP-D in the surfactant of the alveolar epithelium is significantly lower than that of SP-A (approximately 10 times).28 SP-A and SP-D, along with mannose-binding lectins (MBL) and serum proteins conglutinin and collectin-43 (CL-43), belong to the C-type lectin family,29 an archaic class of carbohydrate-binding proteins that perform diverse biological functions, including immune defense, blood coagulation, and cellular interaction mediation.30
Structure of SP-A and SP-D
SP-A and SP-D are classified as collectins, because their lectin domains are associated with collagen-like regions. Like other types of lectins, both possess four distinct domains: the cysteine-rich N-terminal domain, the original triple helix collagen domain characterized by repeating Gly-X-Y triplets, the neck region featuring short hydrophobic amino acid segments as well amphiphilic helices, and the C-terminal carbohydrate recognition domain (CRD).28 The triple-helical collagen-like regions of SP-A and SP-D consist of 23 and 59 repeated Gly-X-Y motifs, respectively, where X and Y may represent any amino acid, usually proline or hydroxyproline.17,28 Similar to other Ca2+-dependent lectins, the CRD of both SP-A and SP-D contain four conserved cysteine residues that serve as Ca2+-dependent specific carbohydrate-binding sites.31 Both SP-A and SP-D spatially construct effective trimers that are maintained by disulfide bonds in the N-terminal region, triple-helix collagen-like segments, and hydrophobic interactions that generate coiled helical regions by strong engagement of the three-helix neck segments.27 These functional trimers can subsequently oligomerize into an octadecameric-like structure for SP-A (similar to a bunch of tulips) and a dodecameric cruciform-like structure, which may further reassemble into star multimers for SP-D.19,27 The affinity and specificity of SP-A and SP-D for bacterial surfaces are derived from their distinctive carbohydrate-binding motifs and the spatial arrangement of their multiple CRDs, positioned at the C-terminus of every trimer unit at certain angles and intervals, ultimately augmenting the overall affinity for binding to carbohydrate targets on microorganisms and facilitating bacterial phagocytosis.32 Therefore, SP-A and SP-D, also known as “carbohydrate pattern recognition molecules”, predominantly interact with glycoconjugates and lipids on microbial surfaces through their CRDs.27
Functions of Surfactant Proteins in Immune System
The host lung defense system comprises AMs, antimicrobial peptides, surfactant lipids, and protein constituents.14 Pulmonary proteins, specifically SP-A and SP-D, engage with diverse cell surface ligands on leukocytes, thereby modulating cellular functions related to phagocytosis and immune responses. Specifically, SP-A demonstrates anti-inflammatory effects in vitro by lowering the generation of cytokines, notably TNF-α and IL-1β, from AMs induced by LPS,33 as well as inhibiting mitogen-induced T cell proliferation.34 Furthermore, SP-A modulates the creation of major mediators, such as TNF and IL-2, in lung lymphocytes, which subsequently affects AM function within the innate pulmonary immune system.35 Besides their functions as opsonins, SP-A and SP-D moonlight towards driving factors for apoptotic cell clearance and bacterial killing11 (Table 1).
![]() |
Table 1 Potential Functions of SP-A and SP-D in M. pneumoniae Infection |
Augment Phagocytic Activity of Phagocytes
Surfactant proteins facilitate the absorption of germs, viruses, and allergens by AMs, monocytes, neutrophils, and DCs; delayed microbial clearance has been observed in surfactant-deficient mice.45 Nevertheless, under specific circumstances, pulmonary surfactants can exert opposing effects, such as inhibiting immune cell phagocytosis of Pneumocystis yersoni.11
Under normal conditions, SP-A and SP-D are believed to impede phagocytosis in macrophages by engaging their CRDs interacting with the transmembrane receptor signal inhibition regulator-α (SIRP-α) and result in the activation of their downstream effectors like SHP-1 and RhoA.46 After infection, the functions of SP-A and SP-D undergo a notable inversion, markedly enhancing the phagocytic abilities of macrophages. The mechanisms driving this shift include, but are not limited to ①Function as opsonins by interacting with membrane receptors such as SP-R210, Toll-like receptor 2 (TLR2), TLR4, CD14, SIRP-α, and CD91-calreticulin,19 thereby augmenting the phagocytic capacity of macrophages against invading pathogens, including Aspergillus fumigatus (conidia),47 Cryptococcus neoformans,48,49 Candida albicans,50 and Pneumocystis jirovecii,51,52 or directly interact with the phagocytosis receptor, scavenger receptor A, and mannose-receptor on macrophages to increase phagocytosis of microbes and apoptotic cells.53 ②Acting as an activation ligand to sense pathogens through their CRDs region to stimulate the phagocytic function of macrophages and monocytes.45 ③Behave as C1q to drive phagocytosis absorption of apoptotic cells through its interaction with CD91-calreticulin (C1qR),54 a receptor complex common to the collagen lectin family55 present on the surface of AMs owing to their homology with C1q,56 although SP-D has a stronger mediating phagocytosis compared to SP-A.55 ④Enhance Fc receptor- (FcR) and C3b/C4b receptor (CR1)-facilitated phagocytosis in monocytes and macrophages in vitro.57,58 ⑤Binding to viscous DNA released by dead microbes or necrotic cells to facilitate the absorption of DNA or apoptotic cells by macrophages. SP-D-deficient mice exhibit impaired clearance of free DNA from the lungs.59 Recently, a newly discovered secretory surfactant candidate, surfactant-associated protein 3 (SFTA3), was shown to enhance phagocytic efficiency in the lungs and may contribute to the clearance of particles and pathogenic microbes.60
Negatively Regulate the Inflammatory Response
Accumulating evidence shows that phosphatidylglycerol and phosphatidylinositol, the main components of pulmonary surfactants, exert anti-inflammatory action by inhibiting the activation of a variety of TLRs (such as TLR2/1, TLR3, TLR4, and TLR2/6), resulting in the secretion of anti-inflammatory mediators that weaken and destroy various respiratory RNA viruses.61 Additionally, SP-A and SP-D are reported to be associated with CD1462 or TLR263 and to block inflammatory cellular responses induced by stimuli such as smooth LPS, peptidoglycan, or yeast glycan. Borron et al reported that intratracheal administration of LPS leads to the production of TNF-α and nitric oxide (NO) in bronchoalveolar lavage fluid (BALF) in SP-A-deficient mice, which were significantly higher compared to those of wild mice.64 Moreover, SP-A knockout models exhibit heightened vulnerability to various bacterial and viral infections, such as Pseudomonas aeruginosa,65 Group B streptococcus,66 Influenza A virus,67 Haemophilus influenzae indissoluble,36 and Respiratory syncytial virus (RSV),68 and an enhanced inflammatory response to pathogen attack. From a mechanistic perspective, SP-A and SP-D may communicate with membrane receptors, including TLRs, SIRPa, and CD91-calreticulin, on leukocytes, thereby inhibiting microbial recognition and influencing the generation of cytokines and inflammatory agents in a microbial ligand-specific form.45,69 This process then alters the activity of innate immune cells, such as AMs, that produce IFN-γ and inducible nitric oxide synthase (iNOS)70 to negatively regulate the inflammatory balance during infection.
Modulation of Lymphocytes and DCs
SP-A may activate host immunological responses during the early stages of infection. When the anti-infective response is sufficient, SP-A subsequently inhibits the production of inflammatory mediators through AMs and ultimately influences T-lymphocyte activity, thereby safeguarding fragile lung tissue from T cell-mediated damage in the alveolar space.70 Both SP-A and SP-D can directly regulate lymphocyte functions at various stages, such as inducing a CD4+CD25+Foxp3+ suppressor regulatory T cell population through a TGF-β-dependent mechanism71 and inhibiting the generation of cytokines by T lymphocytes, such as IL-2 and IFN-γ, thereby directly or indirectly influencing T cell proliferation, lymphokine-activated killer cell activation, and adhesion molecule expression,72–74 ultimately reducing tissue damage caused by overactive immune responses. Furthermore, SP-A and SP-D inhibit DC maturation. For instance, SP-A attaches to DCs and inhibits their maturation in culture, consequently diminishing their capacity to stimulate allogeneic T cells.75 However, surfactant proteins seem to demonstrate different effects in augmenting particle antigen presentation, as SP-A inhibits DCs maturation, whereas SP-D promotes bone marrow-derived DCs maturation and enhances its antigen-presenting capacity by binding to specific receptors, such as the SP-D receptor and CD91.76 The diversity of SP-A and SP-D ensures the accurate regulation of immunity under various infection conditions.
Functions of Surfactants During M. pneumoniae Infection
Functions as Receptor for M. pneumoniae
Common virulence factors of M. pneumoniae include membrane lipoproteins, metabolites, and secretory toxins.77 The newly discovered CARDS TX is a 68 kDa membrane-binding protein, referred to as MPN372, which is thought to have a subunit similar to the S1 component of the pertussis toxin.78,79 The expression level of CARDS TX can be markedly elevated once M. pneumoniae infects host cells and demonstrates a strong affinity for SP-A in AMs, alveolar epithelial cells, and various other tissue cells.80 Recombinant CARDS TX is a Ca2+-dependent protein that interacts with SP-A in a concentration-dependent manner. The CARDS TX antiserum has been shown to block the adhesion of M. pneumoniae to SP-A,21 suggesting that SP-A may serve as a candidate receptor that facilitates both M. pneumoniae adhesion and CARDS TX internalization. Furthermore, CARDS TX binds to SP-A in airway epithelial cells through the AnxA2 receptor-mediated pathway, leading to its internalization and transport, which results in ADP ribosylation and vacuolation activity in mammalian cells.81 Furthermore, the inhibition of AnxA2 and SP-A diminished the binding of CARDS TX and the ensuing vacuolation of cells.21 Krishnan et al revealed that SP-A-deficient cells did not appear to influence CARDS TX internalization,82 suggesting the existence of other receptors that could substitute for the absence of SP-A. Conversely, the suppression of AnxA2 significantly diminishes CARDS TX adherence, internalization, and vacuolation toxicity.83 This indicates that SP-A may be merely one of the multiple receptors for CARDS TX, and its precise physiological function remains ambiguous, perhaps reflecting a larger dimension of the host’s antimicrobial immunological response.
Direct Antibacterial Action by High Affinity Bind M. pneumoniae
In vitro investigations have indicated that SP-A and SP-D exert direct antimicrobial properties on various pathogenic organisms, including Escherichia coli, Klebsiella pneumoniae, and Histoplasma capsulatum by increasing their membrane permeability.11,30 SP-A and SP-D exhibit opsonic activity within their immunological activities and bind to and aggregate with pathogens through their CRDs.20 This process enhances the uptake of microorganisms by immune cells including macrophages and neutrophils, thus facilitating efficient pathogen clearance.53 The membrane components of M. pneumoniae could promote eicosanoid synthesis in macrophages via TLR2 receptors. This pathogen-induced reaction can be eliminated by the anionic surfactant palmitoyl-oleoyl-phosphatidylglycerol,84 which has generated great interest owing to the significance of surfactant proteins in M. pneumoniae infection. Specifically, the CRDs of SP-A and SP-D exhibit a strong affinity for lipid ligands on the membrane of M. pneumoniae, particularly a subset of unsaturated phosphatidylglycerols.37,38 This is a crucial determinant in the relationship between SP (SP-A and SP-D) and ligands present on the membrane of complete M. pneumoniae85 and is strictly dependent on Ca.2+86 This interaction can be completely inhibited by the divalent cation-chelating agents EGTA and dipalmitoyl phosphatidylglycerol87 and is unaffected by other portion of the surfactant proteins (ie, lipid or hydrophobic surfactant proteins).14 This interaction not only impedes the growth of M. pneumoniae, as evidenced by decreased colony formation, metabolism, and DNA replication, but also significantly contributes to the establishment of antibody-independent immunity against the bacteria,86 thus enabling SP-A and SP-D to facilitate the direct elimination of M. pneumoniae. Meanwhile, Kannan et al found that mannose did not impede the binding of M. pneumoniae to hSP-A,21 revealing that CRDs and mannose-related components may not be directly involved in this interaction, but rather serve to enhance the clearance of pathogens by macrophages. Interestingly, the lipid profile recognized by SP-D differs from that of SP-A, although both share a certain degree of overlap.38 Moreover, bacterial recognition by SP-D involves single nucleotide polymorphisms (SNPs), since tandem mutants (E321Q/N323D) with altered SP-D carbohydrate content were unable to bind the lipid ligands of M. pneumoniae.38
Enhancing the Eradication of M. pneumoniae by AMs
AMs, situated within a phospholipid-rich milieu alongside SP-A and SP-D, act as key effectors that facilitate early clearance of M. pneumoniae both in vivo and in vitro, significantly contributing to the ensuing acquired immune response.19 SP-A and SP-D have been documented to attach to AMs with high specificity,38,44 affect the release of reactive oxygen species (ROS)88 and NO metabolites,69 boost chemotactic activity,89 and augment the phagocytic capacity of AMs to eliminate M. pneumoniae. The mechanism by which SP-A and SP-D eliminate M. pneumoniae may be linked to a temperature-sensitive, NO-dependent pathway,90 as pharmacological inhibition of iNOS markedly diminished SP-A-induced eradication of M. pneumoniae.40 Hickman-Davis et al further revealed the SP-A-mediated death of M. pneumoniae with AMs via the generation of peroxynitrite.40 In vivo tests also demonstrated that, following M. pneumoniae infection, the concentrations of NO in the BALF within C57BL/6 mice were significantly higher than those in SP-A-deficient mice,91 and the M. pneumoniae load and inflammation severity in iNOS-deficient mice were considerably greater than those in wild-type mice.92 However, the absence of iNOS expression cannot be offset by alternative NO sources.40 Furthermore, as the synthesis of NO is facilitated by TNF-α, SP-A may also impede NO production by inhibiting TNF-α secretion in Ams.93 Interestingly, the function of SP-A in the eradication of M. pneumoniae appears to be restricted to lower bacterial doses and the initial phases of infection. SP-A regulates NO production in a stimulus-specific manner; it inhibits NO production in the uninfected state but promotes NO formation during M. pneumoniae infection.91 Furthermore, SP-A and SP-D modulated ROS generation in AMs. ROS, including hydrogen peroxide, hydroxyl radicals, and superoxide, enhanced the antibacterial efficacy of SP-A and SP-D against M. pneumoniae.94 Simultaneously, superoxide anions and hydrogen peroxide may kill M. pneumoniae directly or indirectly by generating extremely reactive oxygen-nitrogen intermediates, including hydroxyl radicals and peroxynitrite.40
Negative Modulation of M. pneumoniae-Induced Inflammation
Polymorphisms of SP-A and M. pneumoniae Pneumonia
Allelic variations in SP-A significantly influence its immunomodulatory functions. Unlike rodents, human SP-A is encoded by two functional genes, SFTPA1 (SP-A1) and SFTPA2 (SP-A2), which are transcribed in opposite directions.95 Each gene exhibits considerable genetic and epigenetic complexity, which variably influences alveolar cell activity and the composition of surfactants.96 The distinction between the gene products of SP-A1 and SP-A2, along with their corresponding coding variations, was determined at four specific amino acid positions: 66, 73, 81, and 85.96 Notably, a particular gene allelic variant in SP-A2 (Gln 223 Lys) is prevalent in the population, where the allelic variation corresponds to the substitution of Gln (Q) with Lys (K) at site 223 in the lectin domain.97 Notwithstanding the structural similarities between SP-A1 and SP-A2, their functional mechanisms diverge.98 Ledford et al established that the recombinant human isoform of SP-A2 with a lysine substitution at position 223 (rhSP-A2 Lys223) binds to M. pneumoniae membrane components (MMF) with high affinity, whereas the rhSP-A2 variant with glutamine at position 223 (rhSP-A2 Gln223) shows markedly reduced affinity for MMF.43 Furthermore, mice expressing SP-A2 Gln223 exhibited enhanced neutrophil chemotaxis in response to MMF challenge compared to those expressing SP-A2 Lys223.43 Additionally, SP-A2 displays superior biological activity to SP-A1 in several key areas; it binds more effectively to neutrophils, promotes bacterial engulfment by AMs, enhances the production of proinflammatory factors by macrophage-like cell lines, and improves survival rates in lung transplantation patients.99–102 In summary, SP-A2 polymorphisms may lead to differential immune responses following M. pneumoniae infection, potentially contributing to the variability in clinical manifestations among individuals.
Restricts Mucin Production Induced by M. pneumoniae Components
MMF are predominantly identified via TLR1, TLR2, and TLR6.103 MMF can bind to these receptors and activate downstream signaling pathways including NF-κB,104 mitogen-activated protein kinases (MAPKs),105 and the epidermal growth factor receptor (EGFR).106 EGFR has long been recognized as a prominent receptor that regulates mucin production through endogenous and exogenous ligand signaling cascades, including the Ras/ERK or PI3K/Akt pathways.43 Previous reports indicated that SP-A exhibits a high affinity for live M. pneumoniae and MMF.107 In the absence or downregulation of SP-A, MMF induces increased activation of the EGFR signaling pathway, resulting in enhanced mucin production.43 In vitro experiments have indicated that SP-A can inhibit EGF-induced phosphorylation of EGFR, ERK, and Akt in a dose-dependent way, thereby inhibiting cell proliferation and motility.43,108 Additionally, SP-A-deficient mice show heightened sensitivity to MMF exposure, and the pharmacological inhibition of EGFR before MMF stimulation significantly decreases mucin production and neutrophil infiltration in SP-A-deficient mice.109 Mechanistically, SP-A may interact with EGFR via the neck region of the CRD, effectively blocking the binding of EGF to EGFR and suppressing mucin production.107 These findings indicate that SP-A may contribute to the negative regulation of pulmonary inflammation to a certain degree.
Alleviates Airway Eosinophilic Infiltration and Inflammation
The differences between non-allergic and allergic inflammation largely revolve around eosinophils, which are frequently observed in the airways of patients with asthma and interact with invading M. pneumoniae.110 In vivo investigations have demonstrated that local allergen exposure in patients with asthma leads to a substantial influx of eosinophils, accompanied by elevated SP-D and reduced SP-A levels in BALF.111 This shift in surfactant protein dynamics underscores their potential role in regulating the immune reactions during allergic inflammation.
The dysregulation of SP-A and SP-D metabolism appears to be a critical underlying illness characterized by eosinophil dominance.112 Studies have shown that both SP-A and SP-D modulate eosinophil chemotaxis in inflammatory airway diseases, thereby mitigating the inflammatory response and pathological injury associated with M. pneumoniae infection.113 Consequently, SP-A appeared to disrupt the host’s natural mechanism of clearing M. pneumoniae. One proposed mechanism involves SP-A interacting to eosinophils, limiting the release of eosinophil peroxidase (EPO) upon encountering M. pneumoniae.41 SP-A polymorphisms may influence eosinophil regulation. The variant SP-A Lys223, stemming from the substitution of glutamine (Q) with lysine (K), significantly enhances eosinophilic granuloma formation and offers a nuanced rationale for the capacity of SP-A to limit EPO release,114 highlighting the genotypic-phenotypic correlation in airway diseases.115 Additionally, the CRD of SP-A binds to eosinophils to modulate their degranulation and apoptosis, promoting phagocytosis by macrophages, which inhibits extracellular trap formation and reduces airway inflammation.116 In contrast, SP-D plays a relatively minor role, although it can bind directly to the eosinophil surface and inhibit chemotaxis.116,117 Certain dysfunctions in SP-A and SP-D are hypothesized to worsen M. pneumoniae infection, as elevated reactive nitrogen species in eosinophilic diseases caused by M. pneumoniae disrupt the normal oligomerization of these proteins.112 In summary, SP-A and SP-D promote homeostasis during M. pneumoniae infection while mitigating excessive responses that may lead to harm.
Regulation of Asthmatic Inflammation
Asthma is a persistent respiratory condition that can be exacerbated by M. pneumoniae infection.118 In patients with asthma, a significant link exists between the number of airway mast cells (MCs) in the airway smooth muscle and airway hyperresponsiveness (AHR).119 MCs serve a crucial role as reservoirs of TNF-α,120 amplifying local inflammation and worsening conditions such as asthma following M. pneumoniae infection. SP-A is suggested to significantly influence immunological responses by inhibiting the recruitment of MCs and the subsequent generation of TNF-α following M. pneumoniae infection. Meanwhile, in SP-A-deficient mice infected with M. pneumoniae, MCs further exacerbate AHR and the recruitment of inflammatory cells, which is accompanied by a substantial increase in TNF-α production.39 These findings highlight the protective role of SP-A against asthma. In patients with asthma with SP-A dysfunction or deficiency, M. pneumoniae infection can lead to overproduction of TNF-α by epithelial cells, AMs, and MCs, contributing to increased inflammation and tissue damage.121 In contrast, SNPs in the SP-A and SP-D genes, particularly the SP-A2 SNP rs1965708122 and SP-D SNP rs721917,123 correlate with a heightened risk of asthma. Individuals with asthma harboring these SNPs exhibited reduced lung function and poor asthma control.124 This suggests that these genetic variants influence the expression or function of surfactant proteins, thereby altering the immune response and potentially contributing to the development or severity of asthma.116
Inhibition of DCs Maturation and Lymphocytes Activation
Although SP-A has various immunomodulatory functions that influence the phenotype and activity of adaptive immune cells,125 its role in DCs maturation and lymphocyte activation following M. pneumoniae infection remains unclear. The only available reports, based on research by Ledford, indicate no significant variation in the overall amount of myeloid cells discovered in lung digests of M. pneumoniae infected SP-A-deficient mice in relation to WT C57BL/6 mice. However, the composition of cells, such as antigen-presenting cells, inflammatory monocytes, and neutrophils in BALF, along with the number of DCs, was markedly elevated in SP-A-deficient mice. Concurrently, concentrations of chemotactic factors for immature DCs, eg MCP-1, MIP-1α, and GM-CSF, were elevated in the BAL of SP-A-deficient mice following M. pneumoniae infection. Further studies confirmed that SP-A-deficient mice exhibited more mature DCs and an elevation in CD3+CD4+ and CD3+CD8+ T cells during M. pneumoniae infection. Nonetheless, the elevated burden of M. pneumoniae was not consistent with an increase in T lymphocytes. Additionally, the quantity of activated B cells (B220+IgM+CD69+) extracted from the lungs increased significantly, along with increased levels of high mobility group box 1 (HMGB-1) in SP-A-deficient lungs and mediastinal lymph nodes. HMGB-1, typically linked to necrotic cells, which are indicative of tissue damage and inflammatory signaling responses, has been recognized as a potent pro-inflammatory cytokine that actively regulates DCs maturation.125,126 Since the exogenous addition of SP-A could suppress HMGB-1 release from THP-1 cells activated by M. pneumoniae, it is speculated that SP-A contact with M. pneumoniae may similarly limit HMGB-1 secretion from DCs, which is consistent with the finding that the experimental group recovered with SP-A failed to induce HMGB-1 production from DCs.42 Therefore, SP-A reduces lung inflammation following M. pneumoniae infection by inhibiting DCs and activating T and B cells through the regulation of HMGB-1 expression.
Perspective and Conclusion
Surfactant proteins, particularly SP-A and SP-D, are crucial for maintaining pulmonary homeostasis and resisting pathogenic infections. A wide range of their functions not only influence the immune response during pathogen encounters but also significantly impact the inflammatory response. Due to this complexity, investigators often find it challenging to identify the specific roles of surfactant proteins under various infectious conditions. For instance, while SP-A may exhibit protective effects in models of M. pneumoniae infection, it can also demonstrate pro-inflammatory effects when confronted with other pathogens such as Influenza A virus, S. pneumoniae, RSV, P. aeruginosa, and HIV.19 This functional diversity complicates the interpretation of experimental results and the investigation of the underlying mechanisms.
Studies investigating the mechanisms of action of surfactant proteins often rely on cellular or animal models. However, cellular models struggle to fully replicate the complex host immune environment, and existing animal models such as mice exhibit large gaps in the genetic background of the human immune system. Notably, variations in the sequence and structure of mouse surfactant proteins compared to those of humans introduce uncertainties in research on M. pneumoniae, complicating the interpretation of the study results. With the advent of organoids and humanized cell models, it has become feasible to recreate the lung microenvironment and immune response in vitro,127 address the limitations of animal models, and provide a reliable experimental platform for investigating the pathogenesis of M. pneumoniae.
SP-A exhibits high polymorphism and is unevenly distributed among different individuals and ethnic groups, making it challenging to draw universal conclusions regarding the investigation of surfactant proteins. This polymorphism results in varying reactivity across patients with M. pneumoniae infection, further increasing the complexity of studies. Future research should investigate how this genetic variation influences individual susceptibility to M. pneumoniae and the outcomes of infection. By conducting studies on individuals with different genotypes, we may uncover the relationship between genetic polymorphisms and susceptibility to M. pneumoniae. This knowledge could facilitate the creation of personalized treatment strategies for individual genetic profiles, thereby enhancing the precision of anti-infection therapies.
Future research should focus on the processes that allow SP-A and SP-D to recognize M. pneumoniae. Advanced structural biology techniques, such as cryo-electron microscopy, allow direct observation of the binding process between surfactant proteins and M. pneumoniae at the atomic level, offering new insights into the diverse processes of surfactant proteins activity. Additionally, by comparing surfactant proteins in the context of M. pneumoniae and other pathogens, we can identify specific immune responses further elucidated their roles in the infectious environment. Investigating the regulatory mechanisms and immunomodulatory effects of surfactant proteins will help to clarify their specific functions in M. pneumoniae infection. Understanding these mechanisms will be instrumental in developing customized anti-infection therapies.
In the future, surfactant proteins should be integrated with other pulmonary immune factors, such as chemokines and cytokines, to investigate their synergistic effects on the regulation of M. pneumoniae infection. By systematically studying the interactions among these factors, we aimed to construct a more comprehensive lung immune regulatory network to provide new insights into the treatment of M. pneumoniae pneumonia.
Funding
This work was supported by the Natural Science Foundation of Hunan Province (2022JJ30488 and 2022JJ40406), Scientific Research Fund of Education Department of Hunan Province (21B0430), Science and Technology Innovation Project of Hengyang (202330046180) and Clinical Medical Technology Innovation Guidance Project of Hunan Province (2021SK51906 and 2021SK51808).
Disclosure
The authors report no conflicts of interest in this work.
References
1. Guo ZQ, Gu SY, Tian ZH, Du BY. A comprehensive review of Mycoplasma pneumoniae infection in chronic lung diseases: recent advances in understanding asthma, COPD, and bronchiectasis. Front Med Lausanne. 2024;11:1437731. doi:10.3389/fmed.2024.1437731
2. Chen M, He S, Miles P, et al. Nasal Bacterial Microbiome Differs Between Healthy Controls and Those With Asthma and Allergic Rhinitis. Front Cell Infect Microbiol. 2022;12:841995. doi:10.3389/fcimb.2022.841995
3. Chan ED, Kalayanamit T, Lynch DA, et al. Mycoplasma pneumoniae-associated bronchiolitis causing severe restrictive lung disease in adults: report of three cases and literature review. Chest. 1999;115(4):1188–1194. doi:10.1378/chest.115.4.1188
4. Esposito S, Cavagna R, Bosis S, Droghetti R, Faelli N, Principi N. Emerging role of Mycoplasma pneumoniae in children with acute pharyngitis. Eur J Clin Microbiol Infect Dis. 2002;21(8):607–610. doi:10.1007/s10096-002-0780-7
5. D’Apolito D, D’Aiello L, Pasqua S, et al. Strategy and validation of a consistent and reproducible nucleic acid technique for mycoplasma detection in advanced therapy medicinal products. Biologicals. 2020;64:49–57. doi:10.1016/j.biologicals.2020.01.001
6. Dabrazhynetskaya A, Furtak V, Volokhov D, Beck B, Chizhikov V. Preparation of reference stocks suitable for evaluation of alternative NAT-based mycoplasma detection methods. J Appl Microbiol. 2014;116(1):100–108. doi:10.1111/jam.12352
7. Cartner SC, Simecka JW, Lindsey JR, Cassell GH, Davis JK. Chronic respiratory mycoplasmosis in C3H/HeN and C57BL/6N mice: lesion severity and antibody response. Infect Immun. 1995;63(10):4138–4142. doi:10.1128/iai.63.10.4138-4142.1995
8. Parker RF, Davis JK, Blalock DK, Thorp RB, Simecka JW, Cassell GH. Pulmonary clearance of Mycoplasma pulmonis in C57BL/6N and C3H/HeN mice. Infect Immun. 1987;55(11):2631–2635. doi:10.1128/iai.55.11.2631-2635.1987
9. Hickman-Davis JM, Michalek SM, Gibbs-Erwin J, Lindsey JR. Depletion of alveolar macrophages exacerbates respiratory mycoplasmosis in mycoplasma-resistant C57BL mice but not mycoplasma-susceptible C3H mice. Infect Immun. 1997;65(6):2278–2282. doi:10.1128/iai.65.6.2278-2282.1997
10. Hu J, Ye Y, Chen X, Xiong L, Xie W, Liu P. Insight into the Pathogenic Mechanism of Mycoplasma pneumoniae. Curr Microbiol. 2022;80(1):14. doi:10.1007/s00284-022-03103-0
11. Wright JR. Immunoregulatory functions of surfactant proteins. Nat Rev Immunol. 2005;5(1):58–68. doi:10.1038/nri1528
12. Goerke J. Pulmonary surfactant: functions and molecular composition. Biochim Biophys Acta. 1998;1408(2–3):79–89. doi:10.1016/S0925-4439(98)00060-X
13. Olmeda B, Martinez-Calle M, Perez-Gil J. Pulmonary surfactant metabolism in the alveolar airspace: biogenesis, extracellular conversions, recycling. Ann Anat. 2017;209:78–92. doi:10.1016/j.aanat.2016.09.008
14. Possmayer F, Zuo YY, Veldhuizen RAW, Petersen NO. Pulmonary Surfactant: a Mighty Thin Film. Chem Rev. 2023;123(23):13209–13290. doi:10.1021/acs.chemrev.3c00146
15. Canadas O, Olmeda B, Alonso A, Perez-Gil J. Lipid-Protein and Protein-Protein Interactions in the Pulmonary Surfactant System and Their Role in Lung Homeostasis. Int J mol Sci. 2020;21(10):3708. doi:10.3390/ijms21103708
16. Whitsett JA, Weaver TE. Hydrophobic surfactant proteins in lung function and disease. N Engl J Med. 2002;347(26):2141–2148. doi:10.1056/NEJMra022387
17. Johansson J, Curstedt T. Molecular structures and interactions of pulmonary surfactant components. Eur J Biochem. 1997;244(3):675–693. doi:10.1111/j.1432-1033.1997.00675.x
18. Haagsman HP, Hogenkamp A, van Eijk M, Veldhuizen EJ. Surfactant collectins and innate immunity. Neonatology. 2008;93(4):288–294. doi:10.1159/000121454
19. Dong S, Pang H, Li F, Hua M, Liang M, Song C. Immunoregulatory function of SP-A. Mol Immunol. 2024;166:58–64. doi:10.1016/j.molimm.2024.01.005
20. Kingma PS, Whitsett JA. In defense of the lung: surfactant protein A and surfactant protein D. Curr Opin Pharmacol. 2006;6(3):277–283. doi:10.1016/j.coph.2006.02.003
21. Kannan TR, Provenzano D, Wright JR, Baseman JB. Identification and characterization of human surfactant protein A binding protein of Mycoplasma pneumoniae. Infect Immun. 2005;73(5):2828–2834. doi:10.1128/IAI.73.5.2828-2834.2005
22. Veldhuizen R, Nag K, Orgeig S, Possmayer F. The role of lipids in pulmonary surfactant. Biochim Biophys Acta. 1998;1408(2–3):90–108. doi:10.1016/S0925-4439(98)00061-1
23. Lopez-Rodriguez E, Perez-Gil J. Structure-function relationships in pulmonary surfactant membranes: from biophysics to therapy. Biochim Biophys Acta. 2014;1838(6):1568–1585. doi:10.1016/j.bbamem.2014.01.028
24. Casals C, Canadas O. Role of lipid ordered/disordered phase coexistence in pulmonary surfactant function. Biochim Biophys Acta. 2012;1818(11):2550–2562. doi:10.1016/j.bbamem.2012.05.024
25. Voorhout WF, Veenendaal T, Kuroki Y, Ogasawara Y, van Golde LM, Geuze HJ. Immunocytochemical localization of surfactant protein D (SP-D) in type II cells, Clara cells, and alveolar macrophages of rat lung. J Histochem Cytochem. 1992;40(10):1589–1597. doi:10.1177/40.10.1527377
26. Liekkinen J, Olzynska A, Cwiklik L, Bernardino de la Serna J, Vattulainen I, Javanainen M. Surfactant Proteins SP-B and SP-C in Pulmonary Surfactant Monolayers: physical Properties Controlled by Specific Protein-Lipid Interactions. Langmuir. 2023;39(12):4338–4350. doi:10.1021/acs.langmuir.2c03349
27. Watson A, Madsen J, Clark HW. SP-A and SP-D: dual Functioning Immune Molecules With Antiviral and Immunomodulatory Properties. Front Immunol. 2020;11:622598. doi:10.3389/fimmu.2020.622598
28. Kishore U, Greenhough TJ, Waters P, et al. Surfactant proteins SP-A and SP-D: structure, function and receptors. Mol Immunol. 2006;43(9):1293–1315. doi:10.1016/j.molimm.2005.08.004
29. Holmskov U, Jensenius JC. Structure and function of collectins: humoral C-type lectins with collagenous regions. Behring Inst Mitt. 1993;1993(93):224–235.
30. Wu H, Kuzmenko A, Wan S, et al. Surfactant proteins A and D inhibit the growth of Gram-negative bacteria by increasing membrane permeability. J Clin Invest. 2003;111(10):1589–1602. doi:10.1172/JCI16889
31. Haagsman HP. Interactions of surfactant protein A with pathogens. Biochim Biophys Acta. 1998;1408(2–3):264–277. doi:10.1016/S0925-4439(98)00072-6
32. Awasthi S. Surfactant protein (SP)-A and SP-D as antimicrobial and immunotherapeutic agents. Recent Pat Antiinfect Drug Discov. 2010;5(2):115–123. doi:10.2174/157489110791233559
33. Chroneos ZC, Sever-Chroneos Z, Shepherd VL. Pulmonary surfactant: an immunological perspective. Cell Physiol Biochem. 2010;25(1):13–26. doi:10.1159/000272047
34. Borron P, McCormack FX, Elhalwagi BM, et al. Surfactant protein A inhibits T cell proliferation via its collagen-like tail and a 210-kDa receptor. Am J Physiol. 1998;275(4):L679–686. doi:10.1152/ajplung.1998.275.4.L679
35. Yang S, Milla C, Panoskaltsis-Mortari A, Ingbar DH, Blazar BR, Haddad IY. Human surfactant protein a suppresses T cell-dependent inflammation and attenuates the manifestations of idiopathic pneumonia syndrome in mice. Am J Respir Cell mol Biol. 2001;24(5):527–536. doi:10.1165/ajrcmb.24.5.4400
36. LeVine AM, Whitsett JA. Pulmonary collectins and innate host defense of the lung. Microbes Infect. 2001;3(2):161–166. doi:10.1016/S1286-4579(00)01363-0
37. Chiba H, Pattanajitvilai S, Mitsuzawa H, Kuroki Y, Evans A, Voelker DR. Pulmonary surfactant proteins A and D recognize lipid ligands on Mycoplasma pneumoniae and markedly augment the innate immune response to the organism. Chest. 2003;123(3 Suppl):426S.
38. Chiba H, Pattanajitvilai S, Evans AJ, Harbeck RJ, Voelker DR. Human surfactant protein D (SP-D) binds Mycoplasma pneumoniae by high affinity interactions with lipids. J Biol Chem. 2002;277(23):20379–20385.
39. Ledford JG, Goto H, Potts EN, et al. SP-A preserves airway homeostasis during Mycoplasma pneumoniae infection in mice. J Immunol. 2009;182(12):7818–7827. doi:10.4049/jimmunol.0900452
40. Hickman-Davis J, Gibbs-Erwin J, Lindsey JR, Matalon S. Surfactant protein A mediates mycoplasmacidal activity of alveolar macrophages by production of peroxynitrite. Proc Natl Acad Sci U S A. 1999;96(9):4953–4958.
41. Ledford JG, Mukherjee S, Kislan MM, Nugent JL, Hollingsworth JW, Wright JR. Surfactant protein-A suppresses eosinophil-mediated killing of Mycoplasma pneumoniae in allergic lungs. PLoS One. 2012;7(2):e32436. doi:10.1371/journal.pone.0032436
42. Ledford JG, Lo B, Kislan MM, et al. Surfactant protein-A inhibits mycoplasma-induced dendritic cell maturation through regulation of HMGB-1 cytokine activity. J Immunol. 2010;185(7):3884–3894. doi:10.4049/jimmunol.1000387
43. Ledford JG, Voelker DR, Addison KJ, et al. Genetic variation in SP-A2 leads to differential binding to Mycoplasma pneumoniae membranes and regulation of host responses. J Immunol. 2015;194(12):6123–6132.
44. Tino MJ, Wright JR. Surfactant protein A stimulates phagocytosis of specific pulmonary pathogens by alveolar macrophages. Am J Physiol. 1996;270(4):L677–688.
45. Kuroki Y, Takahashi M, Nishitani C. Pulmonary collectins in innate immunity of the lung. Cell Microbiol. 2007;9(8):1871–1879. doi:10.1111/j.1462-5822.2007.00953.x
46. Janssen WJ, McPhillips KA, Dickinson MG, et al. Surfactant proteins A and D suppress alveolar macrophage phagocytosis via interaction with SIRP alpha. Am J Respir Crit Care Med. 2008;178(2):158–167. doi:10.1164/rccm.200711-1661OC
47. Madan T, Eggleton P, Kishore U, et al. Binding of pulmonary surfactant proteins A and D to Aspergillus fumigatus conidia enhances phagocytosis and killing by human neutrophils and alveolar macrophages. Infect Immun. 1997;65(8):3171–3179. doi:10.1128/iai.65.8.3171-3179.1997
48. Walenkamp AM, Verheul AF, Scharringa J, Hoepelman IM. Pulmonary surfactant protein A binds to Cryptococcus neoformans without promoting phagocytosis. Eur J Clin Invest. 1999;29(1):83–92. doi:10.1046/j.1365-2362.1999.00429.x
49. van de Wetering JK, Coenjaerts FE, Vaandrager AB, van Golde LM, Batenburg JJ. Aggregation of Cryptococcus neoformans by surfactant protein D is inhibited by its capsular component glucuronoxylomannan. Infect Immun. 2004;72(1):145–153. doi:10.1128/IAI.72.1.145-153.2004
50. van Rozendaal B, van Spriel AB, van De Winkel JG, Haagsman HP. Role of Pulmonary Surfactant Protein D in Innate Defense against Candida albicans. J Infect Dis. 2000;182(3):917–922. doi:10.1086/315799
51. Vuk-Pavlovic Z, Standing JE, Crouch EC, Limper AH. Carbohydrate recognition domain of surfactant protein D mediates interactions with Pneumocystis carinii glycoprotein A. Am J Respir Cell mol Biol. 2001;24(4):475–484. doi:10.1165/ajrcmb.24.4.3504
52. Zimmerman PE, Voelker DR, McCormack FX, Paulsrud JR, Martin WJ. 120-kD surface glycoprotein of Pneumocystis carinii is a ligand for surfactant protein A. J Clin Invest. 1992;89(1):143–149. doi:10.1172/JCI115554
53. Sano H, Kuroki Y. The lung collectins, SP-A and SP-D, modulate pulmonary innate immunity. Mol Immunol. 2005;42(3):279–287. doi:10.1016/j.molimm.2004.07.014
54. Ogden CA, deCathelineau A, Hoffmann PR, et al. C1q and mannose binding lectin engagement of cell surface calreticulin and CD91 initiates macropinocytosis and uptake of apoptotic cells. J Exp Med. 2001;194(6):781–795. doi:10.1084/jem.194.6.781
55. Vandivier RW, Ogden CA, Fadok VA, et al. Role of surfactant proteins A, D, and C1q in the clearance of apoptotic cells in vivo and in vitro: calreticulin and CD91 as a common collectin receptor complex. J Immunol. 2002;169(7):3978–3986. doi:10.4049/jimmunol.169.7.3978
56. Nepomuceno RR, Henschen-Edman AH, Burgess WH, Tenner AJ. cDNA cloning and primary structure analysis of C1qR(P), the human C1q/MBL/SPA receptor that mediates enhanced phagocytosis in vitro. Immunity. 1997;6(2):119–129. doi:10.1016/S1074-7613(00)80419-7
57. Tenner AJ, Robinson SL, Borchelt J, Wright JR. Human pulmonary surfactant protein (SP-A), a protein structurally homologous to C1q, can enhance FcR- and CR1-mediated phagocytosis. J Biol Chem. 1989;264(23):13923–13928. doi:10.1016/S0021-9258(18)80088-6
58. Bobak DA, Frank MM, Tenner AJ. C1q acts synergistically with phorbol dibutyrate to activate CR1-mediated phagocytosis by human mononuclear phagocytes. Eur J Immunol. 1988;18(12):2001–2007. doi:10.1002/eji.1830181220
59. Palaniyar N, Clark H, Nadesalingam J, Shih MJ, Hawgood S, Reid KB. Innate immune collectin surfactant protein D enhances the clearance of DNA by macrophages and minimizes anti-DNA antibody generation. J Immunol. 2005;174(11):7352–7358. doi:10.4049/jimmunol.174.11.7352
60. Tschernig T, Veith NT, Diler E, Bischoff M, Meier C, Schicht M. The importance of surfactant proteins-New aspects on macrophage phagocytosis. Ann Anat. 2016;208:142–145. doi:10.1016/j.aanat.2016.07.005
61. Numata M, Kandasamy P, Voelker DR. The anti-inflammatory and antiviral properties of anionic pulmonary surfactant phospholipids. Immunol Rev. 2023;317(1):166–186. doi:10.1111/imr.13207
62. Garcia-Verdugo I, Sanchez-Barbero F, Soldau K, Tobias PS, Casals C. Interaction of SP-A (surfactant protein A) with bacterial rough lipopolysaccharide (Re-LPS), and effects of SP-A on the binding of Re-LPS to CD14 and LPS-binding protein. Biochem J. 2005;391(Pt 1):115–124. doi:10.1042/BJ20050529
63. Agrawal V, Smart K, Jilling T, Hirsch E. Surfactant protein (SP)-A suppresses preterm delivery and inflammation via TLR2. PLoS One. 2013;8(5):e63990. doi:10.1371/journal.pone.0063990
64. Borron P, McIntosh JC, Korfhagen TR, Whitsett JA, Taylor J, Wright JR. Surfactant-associated protein A inhibits LPS-induced cytokine and nitric oxide production in vivo. Am J Physiol Lung Cell mol Physiol. 2000;278(4):L840–847. doi:10.1152/ajplung.2000.278.4.L840
65. Chen X, Guo J, Mahmoud S, et al. Regulatory roles of SP-A and exosomes in pneumonia-induced acute lung and kidney injuries. Front Immunol. 2023;14:1188023. doi:10.3389/fimmu.2023.1188023
66. LeVine AM, Whitsett JA, Gwozdz JA, et al. Distinct effects of surfactant protein A or D deficiency during bacterial infection on the lung. J Immunol. 2000;165(7):3934–3940. doi:10.4049/jimmunol.165.7.3934
67. LeVine AM, Hartshorn K, Elliott J, Whitsett J, Korfhagen T. Absence of SP-A modulates innate and adaptive defense responses to pulmonary influenza infection. Am J Physiol Lung Cell mol Physiol. 2002;282(3):L563–572. doi:10.1152/ajplung.00280.2001
68. LeVine AM, Gwozdz J, Stark J, Bruno M, Whitsett J, Korfhagen T. Surfactant protein-A enhances respiratory syncytial virus clearance in vivo. J Clin Invest. 1999;103(7):1015–1021. doi:10.1172/JCI5849
69. Crouch E, Wright JR. Surfactant proteins a and d and pulmonary host defense. Annu Rev Physiol. 2001;63(1):521–554. doi:10.1146/annurev.physiol.63.1.521
70. Wright JR, Borron P, Brinker KG, Folz RJ. Surfactant Protein A: regulation of innate and adaptive immune responses in lung inflammation. Am J Respir Cell mol Biol. 2001;24(5):513–517. doi:10.1165/ajrcmb.24.5.f208
71. Mukherjee S, Giamberardino C, Thomas JM, Gowdy K, Pastva AM, Wright JR. Surfactant protein A modulates induction of regulatory T cells via TGF-beta. J Immunol. 2012;188(9):4376–4384. doi:10.4049/jimmunol.1101775
72. Faulkner CB, Simecka JW, Davidson MK, et al. Gene expression and production of tumor necrosis factor alpha, interleukin 1, interleukin 6, and gamma interferon in C3H/HeN and C57BL/6N mice in acute Mycoplasma pulmonis disease. Infect Immun. 1995;63(10):4084–4090. doi:10.1128/iai.63.10.4084-4090.1995
73. Borron P, Veldhuizen RA, Lewis JF, et al. Surfactant associated protein-A inhibits human lymphocyte proliferation and IL-2 production. Am J Respir Cell mol Biol. 1996;15(1):115–121. doi:10.1165/ajrcmb.15.1.8679215
74. Borron PJ, Crouch EC, Lewis JF, Wright JR, Possmayer F, Fraher LJ. Recombinant rat surfactant-associated protein D inhibits human T lymphocyte proliferation and IL-2 production. J Immunol. 1998;161(9):4599–4603. doi:10.4049/jimmunol.161.9.4599
75. Brinker KG, Garner H, Wright JR. Surfactant protein A modulates the differentiation of murine bone marrow-derived dendritic cells. Am J Physiol Lung Cell mol Physiol. 2003;284(1):L232–241. doi:10.1152/ajplung.00187.2002
76. Brinker KG, Martin E, Borron P, et al. Surfactant protein D enhances bacterial antigen presentation by bone marrow-derived dendritic cells. Am J Physiol Lung Cell mol Physiol. 2001;281(6):L1453–1463. doi:10.1152/ajplung.2001.281.6.L1453
77. Yiwen C, Yueyue W, Lianmei Q, Cuiming Z, Xiaoxing Y. Infection strategies of mycoplasmas: unraveling the panoply of virulence factors. Virulence. 2021;12(1):788–817. doi:10.1080/21505594.2021.1889813
78. Kannan TR, Baseman JB. ADP-ribosylating and vacuolating cytotoxin of Mycoplasma pneumoniae represents unique virulence determinant among bacterial pathogens. Proc Natl Acad Sci U S A. 2006;103(17):6724–6729. doi:10.1073/pnas.0510644103
79. Dandekar T, Huynen M, Regula JT, et al. Re-annotating the Mycoplasma pneumoniae genome sequence: adding value, function and reading frames. Nucleic Acids Res. 2000;28(17):3278–3288. doi:10.1093/nar/28.17.3278
80. Kannan TR, Musatovova O, Balasubramanian S, et al. Mycoplasma pneumoniae Community Acquired Respiratory Distress Syndrome toxin expression reveals growth phase and infection-dependent regulation. Mol Microbiol. 2010;76(5):1127–1141. doi:10.1111/j.1365-2958.2010.07092.x
81. Becker A, Kannan TR, Taylor AB, et al. Structure of CARDS toxin, a unique ADP-ribosylating and vacuolating cytotoxin from Mycoplasma pneumoniae. Proc Natl Acad Sci U S A. 2015;112(16):5165–5170.
82. Krishnan M, Kannan TR, Baseman JB. Mycoplasma pneumoniae CARDS toxin is internalized via clathrin-mediated endocytosis. PLoS One. 2013;8(5):e62706. doi:10.1371/journal.pone.0062706
83. Somarajan SR, Al-Asadi F, Ramasamy K, Pandranki L, Baseman JB, Kannan TR. Annexin A2 mediates Mycoplasma pneumoniae community-acquired respiratory distress syndrome toxin binding to eukaryotic cells. mBio. 2014;5(4). doi:10.1128/mBio.01497-14
84. Kandasamy P, Zarini S, Chan ED, Leslie CC, Murphy RC, Voelker DR. Pulmonary surfactant phosphatidylglycerol inhibits Mycoplasma pneumoniae-stimulated eicosanoid production from human and mouse macrophages. J Biol Chem. 2011;286(10):7841–7853. doi:10.1074/jbc.M110.170241
85. Chmura K, Bai X, Nakamura M, et al. Induction of IL-8 by Mycoplasma pneumoniae membrane in BEAS-2B cells. Am J Physiol Lung Cell mol Physiol. 2008;295(1):L220–230.
86. Piboonpocanun S, Chiba H, Mitsuzawa H, et al. Surfactant protein A binds Mycoplasma pneumoniae with high affinity and attenuates its growth by recognition of disaturated phosphatidylglycerols. J Biol Chem. 2005;280(1):9–17.
87. Persson A, Chang D, Crouch E. Surfactant protein D is a divalent cation-dependent carbohydrate-binding protein. J Biol Chem. 1990;265(10):5755–5760.
88. Weissbach S, Neuendank A, Pettersson M, Schaberg T, Pison U. Surfactant protein A modulates release of reactive oxygen species from alveolar macrophages. Am J Physiol. 1994;267(6 Pt 1):L660–666. doi:10.1152/ajplung.1994.267.6.L660
89. Wright JR, Youmans DC. Pulmonary surfactant protein A stimulates chemotaxis of alveolar macrophage. Am J Physiol. 1993;264(4 Pt 1):L338–344. doi:10.1152/ajplung.1993.264.4.L338
90. Zhao B, Collins MT, Czuprynski CJ. Effects of gamma interferon and nitric oxide on the interaction of Mycobacterium avium subsp. paratuberculosis with bovine monocytes. Infect Immun. 1997;65(5):1761–1766. doi:10.1128/iai.65.5.1761-1766.1997
91. Hickman-Davis JM, Gibbs-Erwin J, Lindsey JR, Matalon S. Role of surfactant protein-A in nitric oxide production and mycoplasma killing in congenic C57BL/6 mice. Am J Respir Cell mol Biol. 2004;30(3):319–325. doi:10.1165/rcmb.2003-0246OC
92. Hickman-Davis JM, Wang Z, Fierro-Perez GA, et al. Surfactant dysfunction in SP-A-/- and iNOS-/- mice with mycoplasma infection. Am J Respir Cell mol Biol. 2007;36(1):103–113. doi:10.1165/rcmb.2006-0049OC
93. Hussain S, Wright JR, Martin WJ. Surfactant protein A decreases nitric oxide production by macrophages in a tumor necrosis factor-alpha-dependent mechanism. Am J Respir Cell mol Biol. 2003;28(4):520–527. doi:10.1165/rcmb.2002-0072OC
94. Hickman-Davis JM, Lindsey JR, Zhu S, Matalon S. Surfactant protein A mediates mycoplasmacidal activity of alveolar macrophages. Am J Physiol. 1998;274(2):L270–277.
95. Silveyra P, Floros J. Genetic complexity of the human surfactant-associated proteins SP-A1 and SP-A2. Gene. 2013;531(2):126–132. doi:10.1016/j.gene.2012.09.111
96. Floros J, Thorenoor N, Tsotakos N, Phelps DS. Human Surfactant Protein SP-A1 and SP-A2 Variants Differentially Affect the Alveolar Microenvironment, Surfactant Structure, Regulation and Function of the Alveolar Macrophage, and Animal and Human Survival Under Various Conditions. Front Immunol. 2021;12:681639.
97. Pederson WP, Cyphert-Daly JM, Tighe RM, Que LG, Ledford JG. Genetic variation in surfactant protein-A2 alters responses to ozone. PLoS One. 2021;16(2):e0247504. doi:10.1371/journal.pone.0247504
98. Sanchez-Barbero F, Rivas G, Steinhilber W, Casals C. Structural and functional differences among human surfactant proteins SP-A1, SP-A2 and co-expressed SP-A1/SP-A2: role of supratrimeric oligomerization. Biochem J. 2007;406(3):479–489. doi:10.1042/BJ20070275
99. Thorenoor N, Umstead TM, Zhang X, Phelps DS, Floros J. Survival of Surfactant Protein-A1 and SP-A2 Transgenic Mice After Klebsiella pneumoniae Infection, Exhibits Sex-, Gene-, and Variant Specific Differences; Treatment With Surfactant Protein Improves Survival. Front Immunol. 2018;9:2404. doi:10.3389/fimmu.2018.02404
100. Mikerov AN, Wang G, Umstead TM, et al. Surfactant protein A2 (SP-A2) variants expressed in CHO cells stimulate phagocytosis of Pseudomonas aeruginosa more than do SP-A1 variants. Infect Immun. 2007;75(3):1403–1412. doi:10.1128/IAI.01341-06
101. Wang G, Phelps DS, Umstead TM, Floros J. Human SP-A protein variants derived from one or both genes stimulate TNF-alpha production in the THP-1 cell line. Am J Physiol Lung Cell mol Physiol. 2000;278(5):L946–954.
102. D’Ovidio F, Floros J, Aramini B, et al. Donor surfactant protein A2 polymorphism and lung transplant survival. Eur Respir J. 2020;55(3):1.
103. Zuo LL, Wu YM, You XX. Mycoplasma lipoproteins and Toll-like receptors. J Zhejiang Univ Sci B. 2009;10(1):67–76. doi:10.1631/jzus.B0820256
104. Shimizu T, Kida Y, Kuwano K. A dipalmitoylated lipoprotein from Mycoplasma pneumoniae activates NF-kappa B through TLR1, TLR2, and TLR6. J Immunol. 2005;175(7):4641–4646. doi:10.4049/jimmunol.175.7.4641
105. Ma C, Hao X, Gao L, et al. Extracellular Vesicles Released from Macrophages Infected with Mycoplasma pneumoniae Stimulate Proinflammatory Response via the TLR2-NF-kappaB/JNK Signaling Pathway. Int J mol Sci. 2023;24(10):1.
106. Hao Y, Kuang Z, Jing J, et al. Mycoplasma pneumoniae modulates STAT3-STAT6/EGFR-FOXA2 signaling to induce overexpression of airway mucins. Infect Immun. 2014;82(12):5246–5255. doi:10.1128/IAI.01989-14
107. Younis US, Chu HW, Kraft M, Ledford JG. A 20-Mer Peptide Derived from the Lectin Domain of SP-A2 Decreases Tumor Necrosis Factor Alpha Production during Mycoplasma pneumoniae Infection. Infect Immun. 2020;88(9). doi:10.1128/IAI.00099-20
108. Hasegawa Y, Takahashi M, Ariki S, et al. Surfactant protein A down-regulates epidermal growth factor receptor by mechanisms different from those of surfactant protein D. J Biol Chem. 2017;292(45):18565–18576. doi:10.1074/jbc.M117.800771
109. Inoue A, Xin H, Suzuki T, et al. Suppression of surfactant protein A by an epidermal growth factor receptor tyrosine kinase inhibitor exacerbates lung inflammation. Cancer Sci. 2008;99(8):1679–1684.
110. Simon HU. Allergic inflammation: focus on eosinophils. Allergy. 2013;68(7):823–824. doi:10.1111/all.12231
111. Erpenbeck VJ, Schmidt R, Gunther A, Krug N, Hohlfeld JM. Surfactant protein levels in bronchoalveolar lavage after segmental allergen challenge in patients with asthma. Allergy. 2006;61(5):598–604. doi:10.1111/j.1398-9995.2006.01062.x
112. Ledford JG, Addison KJ, Foster MW, Que LG. Eosinophil-associated lung diseases. A cry for surfactant proteins A and D help? Am J Respir Cell mol Biol. 2014;51(5):604–614. doi:10.1165/rcmb.2014-0095TR
113. Ledford JG, Pastva AM, Wright JR. Review: collectins link innate and adaptive immunity in allergic airway disease. Innate Immun. 2010;16(3):183–190. doi:10.1177/1753425910368446
114. ABC D, Tanyaratsrisakul S, Voelker DR, Ledford JG. The Emerging Roles of Surfactant Protein-A in Asthma. J Clin Cell Immunol. 2018;9(4). doi:10.4172/2155-9899.1000553
115. Chang EH. The role of surfactant protein-A in sinusitis. Curr Opin Allergy Clin Immunol. 2019;19(1):26–29. doi:10.1097/ACI.0000000000000500
116. Choi Y, Jang J, Park HS. Pulmonary Surfactants: a New Therapeutic Target in Asthma. Curr Allergy Asthma Rep. 2020;20(11):70. doi:10.1007/s11882-020-00968-8
117. Hohlfeld JM, Schmiedl A, Erpenbeck VJ, Venge P, Krug N. Eosinophil cationic protein alters pulmonary surfactant structure and function in asthma. J Allergy Clin Immunol. 2004;113(3):496–502. doi:10.1016/j.jaci.2003.12.008
118. Haddad IY, Pataki G, Hu P, Galliani C, Beckman JS, Matalon S. Quantitation of nitrotyrosine levels in lung sections of patients and animals with acute lung injury. J Clin Invest. 1994;94(6):2407–2413. doi:10.1172/JCI117607
119. Brightling CE, Bradding P, Symon FA, Holgate ST, Wardlaw AJ, Pavord ID. Mast-cell infiltration of airway smooth muscle in asthma. N Engl J Med. 2002;346(22):1699–1705. doi:10.1056/NEJMoa012705
120. Elieh Ali Komi D, Wohrl S, Bielory L. Mast Cell Biology at Molecular Level: a Comprehensive Review. Clin Rev Allergy Immunol. 2020;58(3):342–365. doi:10.1007/s12016-019-08769-2
121. Hsia BJ, Ledford JG, Potts-Kant EN, et al. Mast cell TNF receptors regulate responses to Mycoplasma pneumoniae in surfactant protein A (SP-A)-/- mice. J Allergy Clin Immunol. 2012;130(1):205–214e202. doi:10.1016/j.jaci.2012.03.002
122. Kraft M, Ledford J. Surfactant Protein A: an Innate Immune Modulator and Therapeutic in Asthma. Trans Am Clin Climatol Assoc. 2024;134:94–112. doi:10.1513/AnnalsATS.201703-259OC.
123. Fakih D, Akiki Z, Junker K, et al. Surfactant protein D multimerization and gene polymorphism in COPD and asthma. Respirology. 2018;23(3):298–305. doi:10.1111/resp.13193
124. Hohlfeld JM. The role of surfactant in asthma. Respir Res. 2002;3(1):4. doi:10.1186/rr176
125. Telusma G, Datta S, Mihajlov I, et al. Dendritic cell activating peptides induce distinct cytokine profiles. Int Immunol. 2006;18(11):1563–1573. doi:10.1093/intimm/dxl089
126. Dumitriu IE, Baruah P, Bianchi ME, Manfredi AA, Rovere-Querini P. Requirement of HMGB1 and RAGE for the maturation of human plasmacytoid dendritic cells. Eur J Immunol. 2005;35(7):2184–2190. doi:10.1002/eji.200526066
127. Mahieu L, Van Moll L, De Vooght L, Delputte P, Cos P. In vitro modelling of bacterial pneumonia: a comparative analysis of widely applied complex cell culture models. FEMS Microbiol Rev. 2024;48(2). doi:10.1093/femsre/fuae007
© 2025 The Author(s). This work is published and licensed by Dove Medical Press Limited. The
full terms of this license are available at https://www.dovepress.com/terms.php
and incorporate the Creative Commons Attribution
- Non Commercial (unported, 3.0) License.
By accessing the work you hereby accept the Terms. Non-commercial uses of the work are permitted
without any further permission from Dove Medical Press Limited, provided the work is properly
attributed. For permission for commercial use of this work, please see paragraphs 4.2 and 5 of our Terms.