Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 95
Filter
1.
Front Immunol ; 12: 681639, 2021.
Article in English | MEDLINE | ID: mdl-34484180

ABSTRACT

The human innate host defense molecules, SP-A1 and SP-A2 variants, differentially affect survival after infection in mice and in lung transplant patients. SP-A interacts with the sentinel innate immune cell in the alveolus, the alveolar macrophage (AM), and modulates its function and regulation. SP-A also plays a role in pulmonary surfactant-related aspects, including surfactant structure and reorganization. For most (if not all) pulmonary diseases there is a dysregulation of host defense and inflammatory processes and/or surfactant dysfunction or deficiency. Because SP-A plays a role in both of these general processes where one or both may become aberrant in pulmonary disease, SP-A stands to be an important molecule in health and disease. In humans (unlike in rodents) SP-A is encoded by two genes (SFTPA1 and SFTPA2) and each has been identified with extensive genetic and epigenetic complexity. In this review, we focus on functional, structural, and regulatory differences between the two SP-A gene-specific products, SP-A1 and SP-A2, and among their corresponding variants. We discuss the differential impact of these variants on the surfactant structure, the alveolar microenvironment, the regulation of epithelial type II miRNome, the regulation and function of the AM, the overall survival of the organism after infection, and others. Although there have been a number of reviews on SP-A, this is the first review that provides such a comprehensive account of the differences between human SP-A1 and SP-A2.


Subject(s)
Cellular Microenvironment/immunology , Genetic Variation , Macrophages, Alveolar/immunology , Macrophages, Alveolar/metabolism , Pulmonary Surfactant-Associated Protein A/genetics , Amino Acid Sequence , Amino Acid Substitution , Animals , Biomarkers , Disease Susceptibility , Humans , Mice , Models, Biological , Proteome , Proteomics/methods , Pulmonary Surfactant-Associated Protein A/chemistry , Pulmonary Surfactant-Associated Protein A/metabolism , Pulmonary Surfactants/metabolism , Structure-Activity Relationship
2.
Immunohorizons ; 5(8): 659-674, 2021 08 24.
Article in English | MEDLINE | ID: mdl-34429343

ABSTRACT

The TLR4-interacting SPA4 peptide suppresses inflammation. We assessed the structural and physicochemical properties and binding of SPA4 peptide to TLR4-MD2. We also studied the changes at the whole transcriptome level, cell morphology, viability, secreted cytokines and chemokines, and cell influx in cell systems and mouse models challenged with LPS and treated with SPA4 peptide. Our results demonstrated that the SPA4 peptide did not alter the cell viability and size and only moderately affected the transcriptome of the cells. Computational docking and rendering suggested that the SPA4 peptide intercalates with LPS-induced TLR4-MD2 complex. Results with alanine mutations of D-2 amino acid and NYTXXXRG-12-19 motif of SPA4 peptide suggested their role in binding to TLR4 and in reducing the cytokine response against LPS stimulus. Furthermore, therapeutically administered SPA4 peptide significantly suppressed the secreted levels of cytokines and chemokines in cells and bronchoalveolar lavage fluids of LPS-challenged mice. The results suggest that the SPA4 peptide intercalates with LPS-induced TLR4 complex and signaling for the suppression of inflammation.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Inflammation/prevention & control , Peptide Fragments/pharmacology , Pulmonary Surfactant-Associated Protein A/metabolism , Toll-Like Receptor 4/metabolism , Amino Acid Sequence , Animals , Anti-Inflammatory Agents/metabolism , Cell Line , Cell Survival/drug effects , Cell Survival/genetics , Cytokines/metabolism , Dendritic Cells/drug effects , Dendritic Cells/metabolism , Female , Gene Expression Regulation/drug effects , HEK293 Cells , Humans , Inflammation/genetics , Inflammation/metabolism , Lipopolysaccharides/pharmacology , Mice, Inbred C57BL , Mutation , Peptide Fragments/genetics , Peptide Fragments/metabolism , Protein Binding , Pulmonary Surfactant-Associated Protein A/chemistry , Pulmonary Surfactant-Associated Protein A/genetics
3.
Histochem Cell Biol ; 155(2): 261-269, 2021 Feb.
Article in English | MEDLINE | ID: mdl-32880000

ABSTRACT

Plate bodies are facultative organelles occasionally described in the adult lungs of various species, including sheep and goat. They consist of multiple layers of plate-like cisterns with an electron dense middle bar. The present study was performed to elucidate the three-dimensional (3D) characteristics of this organelle and its presumed function in surfactant protein A (SP-A) biology. Archived material of four adult goat lungs and PFA-fixed lung samples of two adult sheep lungs were used for the morphological and immunocytochemical parts of this study, respectively. 3D imaging was performed by electron tomography and focused ion beam scanning electron microscopy (FIB-SEM). Immuno gold labeling was used to analyze whether plate bodies are positive for SP-A. Transmission electron microscopy revealed the presence of plate bodies in three of four goat lungs and in both sheep lungs. Electron tomography and FIB-SEM characterized the plate bodies as layers of two up to over ten layers of membranous cisterns with the characteristic electron dense middle bar. The membranes of the plates were in connection with the rough endoplasmic reticulum and showed vesicular inclusions in the middle of the plates and a vesicular network at the sides of the organelle. Immuno gold labeling revealed the presence of SP-A in the vesicular network of plate bodies but not in the characteristic plates themselves. In conclusion, the present study clearly proves the connection of plate bodies with the rough endoplasmic reticulum and the presence of a vesicular network as part of the organelle involved in SP-A trafficking.


Subject(s)
Alveolar Epithelial Cells/chemistry , Imaging, Three-Dimensional , Organelles/metabolism , Organelles/ultrastructure , Pulmonary Surfactant-Associated Protein A/metabolism , Animals , Electron Microscope Tomography , Goats , Microscopy, Electron, Scanning , Organelles/chemistry , Pulmonary Surfactant-Associated Protein A/chemistry
4.
DNA Cell Biol ; 40(1): 126-131, 2021 Jan.
Article in English | MEDLINE | ID: mdl-33181027

ABSTRACT

Interstitial lung disease (ILD) is a large group of disorders, most of which lead to progressive scarring of lung tissue. The scarring associated with ILD eventually affects your ability to breathe and get enough oxygen into your bloodstream. The typical symptoms of ILD are shortness of breath at rest or aggravated by exertion and dry cough. In this study, we enrolled a family with ILDs from central south region of China. Three patients suffered from repeated cough and shortness of breath. The high resolution computed tomography (HRCT) testing further confirmed the diagnosis of interstitial lung lesions. Whole exome sequencing (WES) and Sanger sequencing were applied to detect the genetic lesion of the family. By employing WES, a novel heterozygous mutation (NM_001098668: c.554C>T/p.A185V) of surfactant protein A2 (SFTPA2) was identified in the affected individuals and absent in the healthy members. Bioinformatics analysis predicted that this mutation is disease-causing mutation and located in an evolutionarily conserved site of SFTPA2 protein. The novel mutation may disrupt the stability of SFTPA2 protein and induce endoplasmic reticulum stress, finally leading to ILD under the influence of microorganisms. Our study not only expands the spectrum of SFTPA2 mutations but also helps the family members to mitigate ILD risk factors. The study also supplements and improves genetic testing strategies and ILD risk estimation methodologies for China.


Subject(s)
Lung Diseases, Interstitial/genetics , Mutation, Missense , Pulmonary Surfactant-Associated Protein A/genetics , Aged , Female , Humans , Lung Diseases, Interstitial/pathology , Male , Middle Aged , Pedigree , Protein Domains , Protein Stability , Pulmonary Surfactant-Associated Protein A/chemistry
5.
Protein Expr Purif ; 176: 105727, 2020 12.
Article in English | MEDLINE | ID: mdl-32835791

ABSTRACT

Pulmonary surfactant protein-A (SP-A) is expressed by lung alveolar and bronchiolar epithelial cells and plays a critical role in innate immunity of the lung. Exposure of the lung to various environmental insults alters SP-A homeostasis. To investigate the cellular mechanisms involved in these alterations, we added the FLAG octapeptide (DYKDDDDK) to the carboxy-terminus (SP-A/C-FLAG) or near the amino-terminus (SP-A/N-FLAG) of mouse SP-A (WT-SP-A) to tag specific pools of protein. We hypothesized that addition of FLAG would have negligible effects on SP-A expression, oligomerization and secretion. Analysis of Chinese hamster ovary cells expressing these proteins indicated that tagged SP-A mRNA could be distinguished from WT-SP-A by northern analysis and RT-PCR using sequence-specific oligonucleotides. Tagged SP-A protein could be differentiated from WT-SP-A by western analysis using antibodies specific for the FLAG epitope. Subcellular fractionation and immunocytochemistry indicated the majority of each protein was present in punctuate (presumably endocytic) vesicles, and all forms of SP-A protein were secreted. These results suggest that a FLAG epitope added to the carboxy-terminus or inserted into the amino-terminus of the mature SP-A protein has little effect on its expression and cellular processing. However, disruptions of the amino-terminal end of SP-A prevents proper oligomerization, suggesting that this region of mature SP-A is critical in proper oligomeric assembly and is not useful for studies intended to define mechanisms underlying SP-A homeostasis.


Subject(s)
Gene Expression , Oligopeptides/chemistry , Protein Multimerization , Pulmonary Surfactant-Associated Protein A/chemistry , Recombinant Fusion Proteins/chemistry , Animals , Mice , Oligopeptides/biosynthesis , Oligopeptides/genetics , Oligopeptides/isolation & purification , Pulmonary Surfactant-Associated Protein A/biosynthesis , Pulmonary Surfactant-Associated Protein A/genetics , Pulmonary Surfactant-Associated Protein A/isolation & purification , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/isolation & purification
6.
Infect Immun ; 88(9)2020 08 19.
Article in English | MEDLINE | ID: mdl-32513852

ABSTRACT

Human surfactant protein-A2 (hSP-A2) is a component of pulmonary surfactant that plays an important role in the lung's immune system by interacting with viruses, bacteria, and fungi to facilitate pathogen clearance and by downregulating inflammatory responses after an allergic challenge. Genetic variation in SP-A2 at position Gln223Lys is present in up to ∼30% of the population and has been associated with several lung diseases, such as asthma, pulmonary fibrosis, and lung cancer (M. M. Pettigrew, J. F. Gent, Y. Zhu, E. W. Triche, et al., BMC Med Genet 8:15, 2007, https://bmcmedgenet.biomedcentral.com/articles/10.1186/1471-2350-8-15; Y. Wang, P. J. Kuan, C. Zing, J. T. Cronkhite, et al., Am J Hum Genet 84:52-59, 2009, https://www.cell.com/ajhg/fulltext/S0002-9297(08)00595-8). Previous work performed by our group showed differences in levels of SP-A binding to non-live mycoplasma membrane fractions that were dependent on the presence of a lysine (K) or a glutamine (Q) at amino acid position 223 in the carbohydrate region of SP-A2. On the basis of these differences, we have derived 20-amino-acid peptides flanking this region of interest in order to test the ability of each to regulate various immune responses to live Mycoplasma pneumoniae in SP-A knockout mice and RAW 264.7 cells. In both models, the 20-mer containing 223Q significantly decreased both tumor necrosis factor alpha (TNF-α) mRNA levels and protein levels in comparison to the 20-mer containing 223K during M. pneumoniae infection. While neither of the 20-mer peptides (223Q and 223K) had an effect on p38 phosphorylation during M. pneumoniae infection, the 223Q-20mer peptide significantly reduced NF-κB p65 phosphorylation in both models. Taken together, our data suggest that small peptides derived from the lectin domain of SP-A2 that contain the major allelic variant (223Q) maintain activity in reducing TNF-α induction during M. pneumoniae infection.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Host Microbial Interactions/immunology , Mycoplasma pneumoniae/immunology , Peptides/pharmacology , Pneumonia, Mycoplasma/drug therapy , Pulmonary Surfactant-Associated Protein A/immunology , Tumor Necrosis Factor-alpha/immunology , Adhesins, Bacterial/genetics , Adhesins, Bacterial/immunology , Animals , Anti-Inflammatory Agents/chemical synthesis , Disease Models, Animal , Gene Expression Regulation , Host Microbial Interactions/genetics , Humans , Lung/drug effects , Lung/immunology , Lung/microbiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mycoplasma pneumoniae/drug effects , Mycoplasma pneumoniae/pathogenicity , Neutrophils/drug effects , Neutrophils/immunology , Neutrophils/microbiology , Peptides/chemical synthesis , Pneumonia, Mycoplasma/genetics , Pneumonia, Mycoplasma/immunology , Pneumonia, Mycoplasma/microbiology , Protein Domains , Pulmonary Surfactant-Associated Protein A/chemistry , Pulmonary Surfactant-Associated Protein A/deficiency , Pulmonary Surfactant-Associated Protein A/genetics , RAW 264.7 Cells , RNA, Messenger/genetics , RNA, Messenger/immunology , Signal Transduction , Transcription Factor RelA/genetics , Transcription Factor RelA/immunology , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/genetics , p38 Mitogen-Activated Protein Kinases/genetics , p38 Mitogen-Activated Protein Kinases/immunology
7.
Front Immunol ; 10: 2613, 2019.
Article in English | MEDLINE | ID: mdl-31781112

ABSTRACT

Surfactant protein A (SP-A) provides surfactant stability, first line host defense, and lung homeostasis by binding surfactant phospholipids, pathogens, alveolar macrophages (AMs), and epithelial cells. Non-primates express one SP-A protein whereas humans express two: SP-A1 and SP-A2 with core intra- and inter-species differences in the collagen-like domain. Here, we used macrophages and solid phase binding assays to discern structural correlates of rat (r) and human (h) SP-A function. Binding assays using recombinant rSP-A expressed in insect cells showed that lack of proline hydroxylation, truncations of amino-terminal oligomerization domains, and site-directed serine (S) or alanine (A) mutagenesis of cysteine 6 (C6S), glutamate 195 (E195A), and glutamate 171 (E171A) in the carbohydrate recognition domain (CRD) all impaired SP-A binding. Replacement of arginine 197 with alanine found in hSP-A (R197A), however, restored the binding of hydroxyproline-deficient rSP-A to the SP-A receptor SP-R210 similar to native rat and human SP-A. In silico calculation of Ca++ coordination bond length and solvent accessibility surface area revealed that the "humanized" R197A substitution alters topology and solvent accessibility of the Ca++ coordination residues of the CRD domain. Binding assays in mouse AMs that were exposed to either endogenous SP-A or hSP-A1 (6A2) and hSP-A2 (1A0) isoforms in vivo revealed that mouse SP-A is a functional hybrid of hSP-A1 and hSP-A2 in regulating SP-A receptor occupancy and binding affinity. Binding assays using neonatal and adult human AMs indicates that the interaction of SP-A1 and SP-A2 with AMs is developmentally regulated. Furthermore, our data indicate that the auxiliary ion coordination loop encompassing the conserved E171 residue may comprise a conserved site of interaction with macrophages, and SP-R210 specifically, that merits further investigation to discern conserved and divergent SP-A functions between species. In summary, our findings support the notion that complex structural adaptation of SP-A regulate conserved and species specific AM functions in vertebrates.


Subject(s)
Pulmonary Surfactant-Associated Protein A/chemistry , Animals , Humans , Macrophages, Alveolar/metabolism , Mice , Mice, Inbred C57BL , Molecular Dynamics Simulation , Protein Domains , Protein Multimerization , Pulmonary Surfactant-Associated Protein A/physiology , Rats , Rats, Sprague-Dawley , Species Specificity
8.
Int J Biochem Cell Biol ; 116: 105612, 2019 11.
Article in English | MEDLINE | ID: mdl-31546020

ABSTRACT

BACKGROUND/AIMS: Epigallocatechin-3-gallate (EGCG), a major catechin found in green tea, plays an important anti-tumor role and is involved in various other biological processes, such as, neuroprotection by prevention of aggregation of misfolded proteins generated because of genetic defects. Surfactant protein A2 mutations (G231V and F198S) have been identified to be associated with pulmonary fibrosis and lung cancer, and these mutations cause protein aggregation, instability as well as secretion deficiency. The present study focused on investigating the inhibitory effects of EGCG on aggregation of mutant SP-A2 and elucidating the potential mechanisms underlying this action. METHODS: Wild-type and mutant SP-A2 were transiently expressed in CHO-K1 cells. The aggregated and soluble proteins were separated into NP-40-insoluble and NP-40-soluble fractions. Protein stability was validated by chymotrypsin limited proteolysis assay. Western blot and RT-PCR were used to determine the protein and mRNA expression level, respectively. RESULTS: Mutant SP-A2 alone or wild-type SP-A2 co-expressed with G231V formed NP-40-insoluble aggregates in CHO-K1 cells. EGCG significantly suppressed this aggregation and alleviated mutant SP-A2 accumulation in the ER. When combined with 4-PBA, EGCG treatment completely blocked mutant SP-A2 aggregate formation. Though secretion of mutant protein was not affected, EGCG facilitated protein instability in both wild-type and mutant protein. Importantly, MG132, a proteasome inhibitor, reversed EGCG-induced aggregate reduction. CONCLUSIONS: EGCG inhibits aggregation of misfolded SP-A2 via induction of protein instability and activation of proteasomal pathway for aggregate degradation.


Subject(s)
Catechin/analogs & derivatives , Proteasome Endopeptidase Complex/drug effects , Protein Aggregates/drug effects , Proteolysis/drug effects , Pulmonary Surfactant-Associated Protein A/chemistry , Animals , Butylamines/pharmacology , CHO Cells , Catechin/pharmacology , Cricetulus , Cysteine Proteinase Inhibitors/pharmacology , Detergents/pharmacology , Gene Expression , Leupeptins/pharmacology , Mutation , Octoxynol/pharmacology , Protein Stability , Pulmonary Fibrosis/metabolism , Pulmonary Surfactant-Associated Protein A/genetics , Pulmonary Surfactant-Associated Protein A/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Solubility
9.
J Innate Immun ; 11(1): 13-28, 2019.
Article in English | MEDLINE | ID: mdl-30293076

ABSTRACT

Innate recognition of viruses is an essential part of the immune response to viral pathogens. This is integral to the maintenance of healthy lungs, which are free from infection and efficient at gaseous exchange. An important component of innate immunity for identifying viruses is the family of C-type collagen-containing lectins, also known as collectins. These secreted, soluble proteins are pattern recognition receptors (PRRs) which recognise pathogen-associated molecular patterns (PAMPs), including viral glycoproteins. These innate immune proteins are composed of trimerized units which oligomerise into higher-order structures and facilitate the clearance of viral pathogens through multiple mechanisms. Similarly, many viral surface proteins form trimeric configurations, despite not showing primary protein sequence similarities across the virus classes and families to which they belong. In this review, we discuss the role of the lung collectins, i.e., surfactant proteins A and D (SP-A and SP-D) in viral recognition. We focus particularly on the structural similarity and complementarity of these trimeric collectins with the trimeric viral fusion proteins with which, we hypothesise, they have elegantly co-evolved. Recombinant versions of these innate immune proteins may have therapeutic potential in a range of infectious and inflammatory lung diseases including anti-viral therapeutics.


Subject(s)
Collectins , Immunity, Innate , Pulmonary Surfactant-Associated Protein A , Pulmonary Surfactant-Associated Protein D , Receptors, Pattern Recognition , Viral Fusion Proteins/immunology , Animals , Collectins/chemistry , Collectins/immunology , Humans , Lung/immunology , Lung Diseases/immunology , Lung Diseases/therapy , Lung Diseases/virology , Protein Multimerization , Pulmonary Surfactant-Associated Protein A/chemistry , Pulmonary Surfactant-Associated Protein A/immunology , Pulmonary Surfactant-Associated Protein D/chemistry , Pulmonary Surfactant-Associated Protein D/immunology , Receptors, Pattern Recognition/chemistry , Receptors, Pattern Recognition/immunology , Viral Fusion Proteins/chemistry , Viruses/immunology
10.
Drug Deliv ; 24(1): 1770-1781, 2017 Nov.
Article in English | MEDLINE | ID: mdl-29160134

ABSTRACT

The advent of nanomedicine requires novel delivery vehicles to actively target their site of action. Here, we demonstrate the development of lung-targeting drug-loaded liposomes and their efficacy, specificity and safety. Our study focuses on glucocorticoids methylprednisolone (MPS), a commonly used drug to treat lung injuries. The steroidal molecule was loaded into functionalized nano-sterically stabilized unilamellar liposomes (NSSLs). Targeting functionality was performed through conjugation of surfactant protein A (SPANb) nanobodies to form MPS-NSSLs-SPANb. MPS-NSSLs-SPANb exhibited good size distribution, morphology, and encapsulation efficiency. Animal experiments demonstrated the high specificity of MPS-NSSLs-SPANb to the lung. Treatment with MPS-NSSLs-SPANb reduced the levels of TNF-α, IL-8, and TGF-ß1 in rat bronchoalveolar lavage fluid and the expression of NK-κB in the lung tissues, thereby alleviating lung injuries and increasing rat survival. The nanobody functionalized nanoparticles demonstrate superior performance to treat lung injury when compared to that of antibody functionalized systems.


Subject(s)
Acute Lung Injury/drug therapy , Liposomes/chemistry , Methylprednisolone/chemistry , Methylprednisolone/pharmacology , Nanoparticles/chemistry , Pulmonary Surfactant-Associated Protein A/chemistry , Animals , Bronchoalveolar Lavage Fluid/chemistry , Drug Delivery Systems/methods , Glucocorticoids/chemistry , Glucocorticoids/pharmacology , Interleukin-8/metabolism , Lung/drug effects , Male , Pulmonary Surfactants/chemistry , Pulmonary Surfactants/pharmacology , Rats , Rats, Sprague-Dawley , Transforming Growth Factor beta1/metabolism , Tumor Necrosis Factor-alpha/metabolism
11.
Sci Rep ; 7(1): 8304, 2017 08 16.
Article in English | MEDLINE | ID: mdl-28814727

ABSTRACT

The extent to which defective innate immune responses contribute to chronic obstructive pulmonary disease (COPD) is not fully understood. Pulmonary surfactant protein A (SP-A) plays an important role in regulating innate immunity in the lungs. In this study, we hypothesised that cigarette smoke (CS) and its component acrolein might influence pulmonary innate immunity by affecting the function of SP-A. Indeed, acrolein-modified SP-A was detected in the lungs of mice exposed to CS for 1 week. To further confirm this finding, recombinant human SP-A (hSP-A) was incubated with CS extract (CSE) or acrolein and then analysed by western blotting and nanoscale liquid chromatography-matrix-assisted laser desorption/ionisation time-of-flight tandem mass spectrometry. These analyses revealed that CSE and acrolein induced hSP-A oligomerisation and that acrolein induced the modification of six residues in hSP-A: His39, His116, Cys155, Lys180, Lys221, and Cys224. These modifications had significant effects on the innate immune functions of hSP-A. CSE- or acrolein-induced modification of hSP-A significantly decreased hSP-A's ability to inhibit bacterial growth and to enhance macrophage phagocytosis. These findings suggest that CS-induced structural and functional defects in SP-A contribute to the dysfunctional innate immune responses observed in the lung during cigarette smoking.


Subject(s)
Acrolein/chemistry , Nicotiana/chemistry , Pulmonary Surfactant-Associated Protein A/chemistry , Pulmonary Surfactant-Associated Protein A/metabolism , Aldehydes/chemistry , Animals , CHO Cells , Cigarette Smoking/adverse effects , Cricetulus , Female , Macrophages/immunology , Macrophages/metabolism , Mice , Models, Biological , Molecular Structure , Phagocytosis , Protein Conformation , RAW 264.7 Cells , Sulfhydryl Compounds/chemistry
12.
Biochemistry ; 56(31): 4095-4105, 2017 08 08.
Article in English | MEDLINE | ID: mdl-28719181

ABSTRACT

Lung surfactant proteins (SPs) play critical roles in surfactant function and innate immunity. SP-A and SP-D, members of the collectin family of C-type lectins, exhibit distinct ligand specificities, effects on surfactant structure, and host defense functions despite extensive structural homology. SP-A binds to dipalmitoylphosphatidylcholine (DPPC), the major surfactant lipid component, but not phosphatidylinositol (PI), whereas SP-D shows the opposite preference. Additionally, SP-A and SP-D recognize widely divergent pathogen-associated molecular patterns. Previous studies suggested that a ligand-induced surface loop conformational change unique to SP-A contributes to lipid binding affinity. To test this hypothesis and define the structural features of SP-A and SP-D that determine their ligand binding specificities, a structure-guided approach was used to introduce key features of SP-D into SP-A. A quadruple mutant (E171D/P175E/R197N/K203D) that introduced an SP-D-like loop-stabilizing calcium binding site into the carbohydrate recognition domain was found to interconvert SP-A ligand binding preferences to an SP-D phenotype, exchanging DPPC for PI specificity, and resulting in the loss of lipid A binding and the acquisition of more avid mannan binding properties. Mutants with constituent single or triple mutations showed alterations in their lipid and sugar binding properties that were intermediate between those of SP-A and SP-D. Structures of mutant complexes with inositol or methyl-mannose revealed an attenuation of the ligand-induced conformational change relative to wild-type SP-A. These studies suggest that flexibility in a key surface loop supports the distinctive lipid binding functions of SP-A, thus contributing to its multiple functions in surfactant structure and regulation, and host defense.


Subject(s)
Models, Molecular , Pulmonary Surfactant-Associated Protein A/metabolism , Pulmonary Surfactant-Associated Protein D/metabolism , 1,2-Dipalmitoylphosphatidylcholine/chemistry , 1,2-Dipalmitoylphosphatidylcholine/metabolism , Amino Acid Substitution , Animals , Binding Sites , Crystallography, X-Ray , Kinetics , Lectins, C-Type/chemistry , Lectins, C-Type/metabolism , Ligands , Lipid A/chemistry , Lipid A/metabolism , Liposomes , Mutagenesis, Site-Directed , Mutation , Phosphatidylinositols/chemistry , Phosphatidylinositols/metabolism , Protein Conformation , Protein Interaction Domains and Motifs , Protein Refolding , Protein Stability , Pulmonary Surfactant-Associated Protein A/chemistry , Pulmonary Surfactant-Associated Protein A/genetics , Pulmonary Surfactant-Associated Protein D/chemistry , Pulmonary Surfactant-Associated Protein D/genetics , Rats , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism
13.
Ann Anat ; 211: 184-201, 2017 May.
Article in English | MEDLINE | ID: mdl-28351530

ABSTRACT

The collectins family encompasses several collagenous Ca2+-dependent defense lectins that are described as pathogen recognition molecules. They play an important role in both adaptive and innate immunity. Surfactant proteins A and D are two of these proteins which were initially discovered in association with surfactant in the pulmonary system. The structure, immune and inflammatory functions, and genetic variations have been well described in relation to their roles, function and pathophysiology in the pulmonary system. Subsequently, these proteins have been discovered in a wide range of other organs and organ systems. The role of these proteins outside the pulmonary system is currently an active area of research. This review intends to provide a current overview of the genetics, structure and extra-pulmonary functions of the surfactant collectin proteins.


Subject(s)
Immunity, Innate/genetics , Immunity, Innate/immunology , Pulmonary Surfactant-Associated Protein A/genetics , Pulmonary Surfactant-Associated Protein A/immunology , Pulmonary Surfactant-Associated Protein D/genetics , Pulmonary Surfactant-Associated Protein D/immunology , Animals , Humans , Polymorphism, Single Nucleotide/genetics , Pulmonary Surfactant-Associated Protein A/chemistry , Pulmonary Surfactant-Associated Protein D/chemistry , Structure-Activity Relationship
14.
Biochem Biophys Res Commun ; 485(1): 107-112, 2017 03 25.
Article in English | MEDLINE | ID: mdl-28188794

ABSTRACT

Human ß-defensin 3 (hBD3) is known to be involved in mast cell activation. However, molecular mechanisms underlying the regulation of hBD3-induced mast cell activation have been poorly understood. We previously reported that SP-A and SP-A-derived peptide 01 (SAP01) regulate the function of hBD3. In this study, we focused on the effects of SP-A and SAP01 on the activation of mast cells induced by hBD3. SAP01 directly bound to hBD3. Mast cell-mediated vascular permeability and edema in hBD3 administered rat ears were decreased when injected with SP-A or SAP01. Compatible with the results in rat ear model, both SP-A and SAP01 inhibited hBD3-induced chemotaxis of mast cells in vitro. Direct interaction between SP-A or SAP01 and hBD3 seemed to be responsible for the inhibitory effects on chemotaxis. Furthermore, SAP01 attenuated hBD3-induced accumulation of mast cells and eosinophils in tracheas of the OVA-sensitized inflammatory model. SP-A might contribute to the regulation of inflammatory responses mediated by mast cells during infection.


Subject(s)
Chemotaxis/drug effects , Inflammation/immunology , Mast Cells/immunology , Pulmonary Surfactant-Associated Protein A/immunology , beta-Defensins/immunology , Animals , Capillary Permeability/drug effects , Edema/drug therapy , Edema/immunology , Humans , Inflammation/drug therapy , Male , Mast Cells/cytology , Mast Cells/drug effects , Peptides/chemistry , Peptides/pharmacology , Pulmonary Surfactant-Associated Protein A/chemistry , Pulmonary Surfactant-Associated Protein A/pharmacology , Rats, Sprague-Dawley
15.
Immunobiology ; 222(2): 111-118, 2017 02.
Article in English | MEDLINE | ID: mdl-27793398

ABSTRACT

Respiratory syncytial virus (RSV) is the leading cause of bronchiolitis and hospitalisation of infants in developed countries. Surfactant protein A (SP-A) is an important innate immune molecule, localized in pulmonary surfactant. SP-A binds to carbohydrates on the surface of pathogens in a calcium-dependent manner to enable neutralisation, agglutination and clearance of pathogens including RSV. SP-A forms trimeric units and further oligomerises through interactions between its N-terminal domains. Whilst a recombinant trimeric fragment of the closely related molecule (surfactant protein D) has been shown to retain many of the native protein's functions, the importance of the SP-A oligomeric structure in its interaction with RSV has not been determined. The aim of this study was to produce a functional trimeric recombinant fragment of human (rfh)SP-A, which lacks the N-terminal domain (and the capacity to oligomerise) and test its ability to neutralise RSV in an in vitro model of human bronchial epithelial infection. We used a novel expression tag derived from spider silk proteins ('NT') to produce rfhSP-A in Escherichia coli, which we found to be trimeric and to bind to mannan in a calcium-dependent manner. Trimeric rfhSP-A reduced infection levels of human bronchial epithelial (AALEB) cells by RSV by up to a mean (±SD) of 96.4 (±1.9) % at 5µg/ml, which was significantly more effective than dimeric rfhSP-A (34.3 (±20.5) %) (p<0.0001). Comparatively, native human SP-A reduced RSV infection by up to 38.5 (±28.4) %. For the first time we report the development of a functional trimeric rfhSP-A molecule which is highly efficacious in neutralising RSV, despite lacking the N-terminal domain and capacity to oligomerise.


Subject(s)
Gene Expression , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Protein Multimerization , Pulmonary Surfactant-Associated Protein A/genetics , Respiratory Syncytial Virus Infections/genetics , Respiratory Syncytial Virus Infections/immunology , Respiratory Syncytial Virus, Human/immunology , Carbohydrate Metabolism , Carbohydrates/chemistry , Cells, Cultured , Humans , Protein Binding , Protein Multimerization/genetics , Pulmonary Surfactant-Associated Protein A/chemistry , Pulmonary Surfactant-Associated Protein A/isolation & purification , Pulmonary Surfactant-Associated Protein A/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/isolation & purification , Recombinant Fusion Proteins/metabolism , Respiratory Mucosa/immunology , Respiratory Mucosa/metabolism , Respiratory Mucosa/virology , Respiratory Syncytial Virus Infections/metabolism
16.
Alcohol Clin Exp Res ; 40(12): 2563-2572, 2016 12.
Article in English | MEDLINE | ID: mdl-27783409

ABSTRACT

BACKGROUND: Reactive aldehydes such as acetaldehyde and malondialdehyde generated as a result of alcohol metabolism and cigarette smoke exposure lead to the formation of malondialdehyde-acetaldehyde-adducted proteins (MAA adducts). These aldehydes can adduct to different proteins such as bovine serum albumin and surfactant protein A or surfactant protein D (SPD). Macrophages play an important role in innate immunity, but the effect of MAA adducts on macrophage function has not yet been examined. Because macrophage scavenger receptor A (SRA; CD204) mediates the uptake of modified proteins, we hypothesized that the effects of MAA-modified proteins on macrophage function are primarily mediated through SRA. METHODS: We tested this hypothesis by exposing SPD-MAA to macrophages and measuring functions. SPD-MAA treatment significantly stimulated pro-inflammatory cytokine tumor necrosis factor-alpha (TNF-α) release in the macrophage cell line, RAW 264.7. RESULTS: A significant reduction in phagocytosis of zymosan particles was also observed. SPD-MAA stimulated a significant dose-dependent increase in TNF-α and interleukin (IL)-6 release from peritoneal macrophages (PMs) of wild-type (WT) mice. But significantly less TNF-α and IL-6 were released from PMs of SRA-/- mice. We observed a significant reduction in phagocytosis of zymosan particles in PMs from WT mice treated with SPD-MAA. No further SPD-MAA-induced reduction was seen in PMs from SRA-/- mice. SPD-MAA treatment significantly increased SRA mRNA expression, but had no effect on surface receptor protein expression. Protein kinase C alpha inhibitor and NF-κB inhibitor significantly reduced pro-inflammatory cytokine release in response to SPD-MAA. CONCLUSIONS: In conclusion, our data demonstrate that SRA is important for MAA-adducted protein-mediated effect on macrophage functions.


Subject(s)
Acetaldehyde/chemistry , Macrophages/drug effects , Macrophages/metabolism , Malondialdehyde/chemistry , Pulmonary Surfactant-Associated Protein A/pharmacology , Pulmonary Surfactant-Associated Protein D/pharmacology , Scavenger Receptors, Class A/genetics , Animals , Cells, Cultured , Dose-Response Relationship, Drug , Humans , Interleukin-6/metabolism , Mice , Mice, Knockout , Phagocytosis/drug effects , Pulmonary Surfactant-Associated Protein A/chemistry , Pulmonary Surfactant-Associated Protein D/chemistry , Rats , Scavenger Receptors, Class A/biosynthesis , Tumor Necrosis Factor-alpha/metabolism
17.
Biophys J ; 111(3): 524-536, 2016 Aug 09.
Article in English | MEDLINE | ID: mdl-27508436

ABSTRACT

Pulmonary surfactant is a lipoprotein complex that reduces surface tension to prevent alveolar collapse and contributes to the protection of the respiratory surface from the entry of pathogens. Surfactant protein A (SP-A) is a hydrophilic glycoprotein of the collectin family, and its main function is related to host defense. However, previous studies have shown that SP-A also aids in the formation and biophysical properties of pulmonary surfactant films at the air-water interface. Humans, unlike rodents, have two genes, SFTPA1 and SFTPA2. The encoded proteins, SP-A1 and SP-A2, differ quantitatively or qualitatively in function. It has been shown that both gene products are necessary for tubular myelin formation, an extracellular structural form of lung surfactant. The goal of this study was to investigate potential differences in the biophysical properties of surfactants containing human SP-A1, SP-A2, or both. For this purpose, we have studied for the first time, to our knowledge, the biophysical properties of pulmonary surfactant from individual humanized transgenic mice expressing human SP-A1, SP-A2, or both SP-A1 and SP-A2, in the captive bubble surfactometer. We observed that pulmonary surfactant containing SP-A1 reaches lower surface tension after postexpansion interfacial adsorption than surfactants containing no SP-A or only SP-A2. Under interfacial compression-expansion cycling conditions, surfactant films containing SP-A1 also performed better, particularly with respect to the reorganization of the films that takes place during compression. On the other hand, addition of recombinant SP-A1 to a surfactant preparation reconstituted from the hydrophobic fraction of a porcine surfactant made it more resistant to inhibition by serum than the addition of equivalent amounts of SP-A2. We conclude that the presence of SP-A1 allows pulmonary surfactant to adopt a particularly favorable structure with optimal biophysical properties.


Subject(s)
Lung/metabolism , Pulmonary Surfactant-Associated Protein A/metabolism , Animals , Gene Knockout Techniques , Humans , Hydrophobic and Hydrophilic Interactions , Mice , Mice, Transgenic , Pulmonary Surfactant-Associated Protein A/chemistry , Pulmonary Surfactant-Associated Protein A/genetics , Surface Tension , Swine
18.
Biochemistry ; 55(26): 3692-701, 2016 07 05.
Article in English | MEDLINE | ID: mdl-27324153

ABSTRACT

Surfactant protein A (SP-A) is a collagenous C-type lectin (collectin) that is critical for pulmonary defense against inhaled microorganisms. Bifunctional avidity of SP-A for pathogen-associated molecular patterns (PAMPs) such as lipid A and for dipalmitoylphosphatidylcholine (DPPC), the major component of surfactant membranes lining the air-liquid interface of the lung, ensures that the protein is poised for first-line interactions with inhaled pathogens. To improve our understanding of the motifs that are required for interactions with microbes and surfactant structures, we explored the role of the tyrosine-rich binding surface on the carbohydrate recognition domain of SP-A in the interaction with DPPC and lipid A using crystallography, site-directed mutagenesis, and molecular dynamics simulations. Critical binding features for DPPC binding include a three-walled tyrosine cage that binds the choline headgroup through cation-π interactions and a positively charged cluster that binds the phosphoryl group. This basic cluster is also critical for binding of lipid A, a bacterial PAMP and target for SP-A. Molecular dynamics simulations further predict that SP-A binds lipid A more tightly than DPPC. These results suggest that the differential binding properties of SP-A favor transfer of the protein from surfactant DPPC to pathogen membranes containing appropriate lipid PAMPs to effect key host defense functions.


Subject(s)
Crystallography, X-Ray/methods , Proteolipids/metabolism , Pulmonary Surfactant-Associated Protein A/chemistry , Pulmonary Surfactant-Associated Protein A/metabolism , Pulmonary Surfactants/chemistry , Pulmonary Surfactants/metabolism , 1,2-Dipalmitoylphosphatidylcholine/metabolism , Animals , Binding Sites , Molecular Dynamics Simulation , Mutagenesis, Site-Directed , Mutation/genetics , Protein Conformation , Pulmonary Surfactant-Associated Protein A/genetics , Rats
19.
Biochim Biophys Acta ; 1864(4): 419-25, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26723227

ABSTRACT

Peroxiredoxin 6 (Prdx6) is a bifunctional enzyme with peroxidase and phospholipase A2 (PLA2) activities. This protein participates in the degradation and remodeling of internalized dipalmitoylphosphatidylcholine (DPPC), the major phospholipid component of lung surfactant. We have shown previously that the PLA2 activity of Prdx6 is inhibited by the lung surfactant-associated protein called surfactant protein A (SP-A) through direct protein-protein interaction. Docking of SPA and Prdx6 was modeled using the ZDOCK (zlab.bu.edu) program in order to predict molecular sites for binding of the two proteins. The predicted peptide sequences were evaluated for binding to the opposite protein using isothermal titration calorimetry and circular dichroism measurement followed by determination of the effect of the SP-A peptide on the PLA2 activity of Prdx6. The sequences 195EEEAKKLFPK204.in the Prdx6 helix and 83DEELQTELYEIKHQIL99 in SP-A were identified as the sites for hydrophobic interaction and H(+)-bonding between the 2 proteins. Treatment of mouse endothelial cells with the SP-A peptide inhibited their recovery from lipid peroxidation associated with oxidative stress indicating inhibition of Prdx6 activity by the peptide in the intact cell.


Subject(s)
Peroxiredoxin VI/chemistry , Pulmonary Surfactant-Associated Protein A/chemistry , Amino Acid Sequence , Binding Sites , Circular Dichroism , Molecular Sequence Data , Phospholipases A2/metabolism
20.
Pediatr Res ; 79(1-1): 13-21, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26372515

ABSTRACT

BACKGROUND: Male preterm infants are more likely to experience respiratory distress syndrome than females. Our objectives were to determine if sex-related differences in physiological adaptation after preterm birth increase with time after birth and if the use of continuous positive airway pressure (CPAP) reduces these differences. METHODS: Unanesthetized lambs (9F, 8M) were delivered at 0.90 of term. Blood gases, metabolites, and cardiovascular and respiratory parameters were monitored in spontaneously breathing lambs for 8 h. Supplemental oxygen was administered via a face mask at 4 cmH2O CPAP. At 8 h, lung compliance was determined, and bronchoalveolar lavage fluid (BALF) was analyzed for total protein and surfactant phospholipids. Surfactant protein (SP) gene expression and protein expression of SP-A and pro-SP-C were determined in lung tissue. RESULTS: For 8 h after delivery, males had significantly lower arterial pH and higher Paco2, and a greater percentage of males were dependent on supplemental oxygen than females. Inspiratory effort was greater and lung compliance was lower in male lambs. Total protein concentration in BALF, SP gene expression, and SP-A protein levels were not different between sexes; pro-SP-C was 24% lower in males. CONCLUSION: The use of CPAP did not eliminate the male disadvantage, which continues for up to 8 h after preterm birth.


Subject(s)
Premature Birth/physiopathology , Pulmonary Surfactant-Associated Protein A/chemistry , Pulmonary Surfactant-Associated Protein B/chemistry , Respiratory Distress Syndrome, Newborn/physiopathology , Respiratory System/physiopathology , Sex Characteristics , Adaptation, Physiological , Animals , Animals, Newborn , Body Weight , Bronchoalveolar Lavage Fluid/chemistry , Carbon Dioxide/blood , Continuous Positive Airway Pressure , Female , Fetal Organ Maturity , Gene Expression Regulation, Developmental , Hydrogen-Ion Concentration , Lung/embryology , Lung/metabolism , Lung Compliance , Male , Organ Size , Oxygen/administration & dosage , Oxygen/blood , Phospholipids/analysis , Premature Birth/metabolism , Pulmonary Surfactant-Associated Protein A/biosynthesis , Pulmonary Surfactant-Associated Protein A/genetics , Pulmonary Surfactant-Associated Protein B/biosynthesis , Pulmonary Surfactant-Associated Protein B/genetics , Respiratory Distress Syndrome, Newborn/blood , Sheep , Viscera/anatomy & histology
SELECTION OF CITATIONS
SEARCH DETAIL
...